51
|
Manshori M, Kazemnejad S, Naderi N, Darzi M, Aboutaleb N, Golshahi H. Systemic delivery of menstrual blood stem cells is more effective in preventing remote organ injuries following myocardial infarction in comparison with bone marrow stem cells in rat. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2023; 26:645-652. [PMID: 37275762 PMCID: PMC10237164 DOI: 10.22038/ijbms.2023.67574.14809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 03/15/2023] [Indexed: 06/07/2023]
Abstract
Objectives Remote organ injury is a phenomenon that could happen following myocardial infarction (MI). We evaluated the potency of menstrual blood stromal (stem) cells (MenSCs) and bone marrow stem cells (BMSCs) to alleviate remote organ injuries following MI in rats. Materials and Methods 2 × 106 MenSCs or BMSCs were administrated seven days after MI induction via the tail vein. Four weeks after cell therapy, activities of aspartate aminotransferase (AST), urea, creatinine, and Blood Urea Nitrogen (BUN) were evaluated. The level of tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, and IL-6 were determined by ELISA assay. The expression of Nuclear Factor-κB (NF-κB) was evaluated by immunohistochemical staining. Apoptosis activity and tissue damage were also determined by TUNEL and H&E staining, respectively. Results MenSCs and BMSCs administration caused a significant reduction in AST, urea, and BUN levels compared with the MI group. In addition, systemic injection of MenSCs significantly decreased the IL-1β level compared with BMSCs and MI groups (P<0.05 and P<0.01 respectively). Apoptosis in injured kidneys was noticeably diminished in MenSCs-treated rats compared with BMSCs administrated and MI groups (P<0.05 and P<0.05, respectively). In hepatic tissue, limited numbers of TUNEL-positive cells were detected in all groups. Interestingly, MenSCs therapy evoked inhibition of NF-κB in the kidney strikingly. Although, no significant NF-κB expression was observed in hepatic tissue in any group (P>0.05). Conclusion MenSCs are probably more protective than BMSCs on remote organ injuries following MI via decreasing cell death and immunoregulatory properties.
Collapse
Affiliation(s)
- Mahmood Manshori
- Nanobiotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Somaieh Kazemnejad
- Nanobiotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Nasim Naderi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Darzi
- Nanobiotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Nahid Aboutaleb
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Physiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hannaneh Golshahi
- Nanobiotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| |
Collapse
|
52
|
Spartalis M. Genome Editing and Atrial Fibrillation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1396:129-137. [DOI: 10.1007/978-981-19-5642-3_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
|
53
|
El-Ashmawy NE, Khedr NF, Shaban MN, Al-Ashmawy GM. Diallyl trisulfide modulated autophagy in isoproterenol induced acute myocardial infarction. CLINICAL PHYTOSCIENCE 2022. [DOI: 10.1186/s40816-022-00351-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2022] Open
Abstract
Abstract
Background
Acute myocardial infarction (AMI) is the most serious manifestation of coronary artery disease. The initial ischemia in AMI causes biochemical and metabolic alterations in cardiomyocytes.
Objectives
The present study aimed to investigate the biomolecular mechanisms underlying cardioprotective effects of diallyl trisulfide (DATS) as well as captopril (CAP) in isoproterenol (ISO) induced AMI focusing on autophagy & PI3K/Akt signaling.
Methods
Seventy male Albino rats were divided into seven groups as follows: Normal control, ISO, ISO + LY294002 (PI3K inhibitor), DATS+ISO, CAP+ISO, DATS+LY294002 + ISO, and CAP+LY294002 + ISO. All treatments (40 mg/kg DATS, 50 mg/kg CAP & 0.3 mg/kg LY294002) were given daily for two weeks before ISO injection (85 mg/kg for 2 days). At the end of the experiment, serum and cardiac tissues were collected. Serum cardiac troponin I (cTnI), and creatine kinase MB (CK-MB) were measured. Cardiac glutathione peroxidase (GSH-px), malondialdehyde (MDA), hypoxia-inducible factor 1 alpha (HIF-1α), autophagy proteins (P62 & LC3IIB) and gene expression of PI3K, Akt, FOXO-1, and eNOS were assessed. Histopathological examination of heart tissue was performed.
Results
DATS and CAP significantly (p < 0.01) decreased serum CK-MB and cTnI, cardiac levels of MDA, HIF-1α, p62 and LC3IIB along with an increase in GSH-px activity compared with ISO group. Moreover, DATS and CAP significantly up-regulated PI3K, Akt, and eNOS gene expression but down-regulated FOXO-1 expression compared to ISO group. However, LY294002 reversed DATS and CAP cardioprotective effects.
Conclusion
DATS and CAP prior treatment proved cardioprotective effects via modulation of autophagy, PI3K/Akt signaling, eNOS and FOXO-1 downregulation in ISO induced AMI rat model.
Collapse
|
54
|
Mir R, Elfaki I, Javid J, Barnawi J, Altayar MA, Albalawi SO, Jalal MM, Tayeb FJ, Yousif A, Ullah MF, AbuDuhier FM. Genetic Determinants of Cardiovascular Disease: The Endothelial Nitric Oxide Synthase 3 (eNOS3), Krüppel-Like Factor-14 (KLF-14), Methylenetetrahydrofolate Reductase (MTHFR), MiRNAs27a and Their Association with the Predisposition and Susceptibility to Coronary Artery Disease. Life (Basel) 2022; 12:life12111905. [PMID: 36431040 PMCID: PMC9697170 DOI: 10.3390/life12111905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/11/2022] [Accepted: 11/11/2022] [Indexed: 11/18/2022] Open
Abstract
Coronary artery disease (CAD) is an important cause of death worldwide. CAD is caused by genetic and other factors including hypertension, hyperlipidemia, obesity, stress, unhealthy diet, physical inactively, smoking and Type 2 diabetes (T2D). The genome wide association studies (GWASs) have revealed the association of many loci with risk to diseases such as cancers, T2D and CAD. Nitric oxide (NO) is a potent vasodilator and is required for normal vascular health. It is produced in the endothelial cells in a reaction catalyzed by the endothelial NO synthase (eNOS). Methylenetetrahydrofolate reductase (MTHFR) is a very important enzyme involved in metabolism of folate and homocysteine, and its reduced function leads to cardiovascular disease. The Krüppel-like factor-14 (KLF-14) is an important transcriptional regulator that has been implicated in metabolic syndrome. MicroRNA (MiRNAs) are short non-coding RNAs that regulate the gene expression of proteins involved in important physiological processes including cell cycle and metabolism. In the present study, we have investigated the potential impact of germline pathogenic variants of endothelial eNOS, KLF-14, MTHFR, MiRNA-27a and their association with risk to CAD in the Saudi population. Methods: Amplification Refractory Mutation System (ARMS) PCR was used to detect MTHFR, KLF-14, miRNA-27a and eNOS3 genotyping in CAD patients and healthy controls. About 125 CAD cases and 125 controls were enrolled in this study and statistical associations were calculated including p-value, risk ratio (RR), and odds ratio (OD). Results: There were statistically significant differences (p < 0.05) in genotype distributions of MTHFR 677 C>T, KLF-14 rs972283 G>A, miRNAs27a rs895819 A>G and eNOS3 rs1799983 G>T between CAD patients and controls. In addition, our results indicated that the MTHFR-TT genotype was associated with increased CAD susceptibility with an OR 2.75 (95%) and p < 0.049, and the KLF14-AA genotype was also associated with increased CAD susceptibility with an OR of 2.24 (95%) and p < 0.024. Moreover, the miRNAs27a-GG genotype protects from CAD risk with an OR = 0.31 (0.016), p = 0.016. Our results also indicated that eNOS3 -GT genotype is associated with CAD susceptibility with an OR = 2.65, and p < 0.0003. Conclusion: The MTHFR 677C>T, KLF14 rs972283 G>A, miRNAs27a A>G, and eNOS3 rs1799983 G>T genotypes were associated with CAD susceptibility (p < 0.05). These findings require verification in future large-scale population based studies before these loci are used for the prediction and identification of individuals at risk to CAD. Weight control, physical activity, and smoking cessation are very influential recommendations given by clinicians to the at risk individuals to reduce or delay the development of CAD.
Collapse
Affiliation(s)
- Rashid Mir
- Prince Fahd Bin Sultan Research Chair, Department of Medical Lab Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia
- Correspondence: (R.M.); (I.E.)
| | - Imadeldin Elfaki
- Department of Biochemistry, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia
- Correspondence: (R.M.); (I.E.)
| | - Jamsheed Javid
- Prince Fahd Bin Sultan Research Chair, Department of Medical Lab Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Jameel Barnawi
- Prince Fahd Bin Sultan Research Chair, Department of Medical Lab Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Malik A. Altayar
- Prince Fahd Bin Sultan Research Chair, Department of Medical Lab Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Salem Owaid Albalawi
- Department of Cardiology, King Fahd Specialist Hospital, Tabuk 71491, Saudi Arabia
| | - Mohammed M. Jalal
- Prince Fahd Bin Sultan Research Chair, Department of Medical Lab Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Faris J. Tayeb
- Prince Fahd Bin Sultan Research Chair, Department of Medical Lab Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Aadil Yousif
- Prince Fahd Bin Sultan Research Chair, Department of Medical Lab Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Mohammad Fahad Ullah
- Prince Fahd Bin Sultan Research Chair, Department of Medical Lab Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Faisel M. AbuDuhier
- Prince Fahd Bin Sultan Research Chair, Department of Medical Lab Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia
| |
Collapse
|
55
|
Miao R, Wang L, Chen Z, Ge S, Li L, Zhang K, Chen Y, Guo W, Duan X, Zhu M, Zhao G, Lin F. Advances in the study of nicotinamide adenine dinucleotide phosphate oxidase in myocardial remodeling. Front Cardiovasc Med 2022; 9:1000578. [PMID: 36407440 PMCID: PMC9669076 DOI: 10.3389/fcvm.2022.1000578] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 10/18/2022] [Indexed: 07/30/2023] Open
Abstract
Myocardial remodeling is a key pathophysiological basis of heart failure, which seriously threatens human health and causes a severe economic burden worldwide. During chronic stress, the heart undergoes myocardial remodeling, mainly manifested by cardiomyocyte hypertrophy, apoptosis, interstitial fibrosis, chamber enlargement, and cardiac dysfunction. The NADPH oxidase family (NOXs) are multisubunit transmembrane enzyme complexes involved in the generation of redox signals. Studies have shown that NOXs are highly expressed in the heart and are involved in the pathological development process of myocardial remodeling, which influences the development of heart failure. This review summarizes the progress of research on the pathophysiological processes related to the regulation of myocardial remodeling by NOXs, suggesting that NOXs-dependent regulatory mechanisms of myocardial remodeling are promising new therapeutic targets for the treatment of heart failure.
Collapse
Affiliation(s)
- Runran Miao
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Heart Center of Xinxiang Medical University, Xinxiang, China
| | - Libo Wang
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Heart Center of Xinxiang Medical University, Xinxiang, China
- College of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, China
| | - Zhigang Chen
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Heart Center of Xinxiang Medical University, Xinxiang, China
- Cardiovascular Repair Engineering Technology Research Center, The First Affifiliated Hospital of Xinxiang Medical University, Xinxiang, China
- International Joint Laboratory of Cardiovascular Injury and Repair, The First Affifiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Shiqi Ge
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Heart Center of Xinxiang Medical University, Xinxiang, China
| | - Li Li
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Heart Center of Xinxiang Medical University, Xinxiang, China
| | - Kai Zhang
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Heart Center of Xinxiang Medical University, Xinxiang, China
| | - Yingen Chen
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Heart Center of Xinxiang Medical University, Xinxiang, China
| | - Wenjing Guo
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Heart Center of Xinxiang Medical University, Xinxiang, China
| | - Xulei Duan
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Heart Center of Xinxiang Medical University, Xinxiang, China
| | - Mingyang Zhu
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Heart Center of Xinxiang Medical University, Xinxiang, China
| | - Guoan Zhao
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Heart Center of Xinxiang Medical University, Xinxiang, China
- Cardiovascular Repair Engineering Technology Research Center, The First Affifiliated Hospital of Xinxiang Medical University, Xinxiang, China
- International Joint Laboratory of Cardiovascular Injury and Repair, The First Affifiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Fei Lin
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Heart Center of Xinxiang Medical University, Xinxiang, China
- Cardiovascular Repair Engineering Technology Research Center, The First Affifiliated Hospital of Xinxiang Medical University, Xinxiang, China
- International Joint Laboratory of Cardiovascular Injury and Repair, The First Affifiliated Hospital of Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
56
|
Zhang X, Wang A, Wang X, Zhao Q, Xing H. Evaluation of L-Selenomethionine on Ameliorating Cardiac Injury Induced by Environmental Ammonia. Biol Trace Elem Res 2022; 200:4712-4725. [PMID: 35094233 DOI: 10.1007/s12011-021-03071-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 12/15/2021] [Indexed: 12/27/2022]
Abstract
L-Selenomethionine is one of the important organic selenium sources. The supplementation of L-selenomethionine in diets is significant to improve the health of pigs. Ammonia is a major pollutant in the atmosphere and piggery, posing a threat to human and animal health. Although ammonia exposure can damage the heart, the mechanism of cardiac toxicity by ammonia is still unknown. In this study, we investigated the mechanism of cardiac injury induced by ammonia exposure in pigs and the protective effect of L-selenomethionine on its cardiotoxicity. The results showed that the blood ammonia content of pig increased significantly in ammonia group, the expressions of energy metabolism-related genes (LDHA, PDK4, HK2, and CPTIB) and the oxidative stress indexes were significantly changed (P < 0.05), the AMPK/PPAR-γ/NF-κB signaling pathways were activated, the chromatin edge aggregation and nuclear pyknosis were observed in ultrastructure, the apoptotic cells were significantly increased (P < 0.05), and the mRNA and protein expressions of apoptosis-related genes (Bcl-2, Bax, Cyt-c, caspase-3, and caspase-9) were significantly affected (P < 0.05). The above changes were significantly alleviated in ammonia + L-selenomethionine group, but there were still significant differences compared with the C group (P < 0.05). Our results indicated that ammonia exposure could cause energy metabolism disorder and oxidative stress and induce apoptosis of cardiomyocytes through AMPK/PPAR-γ/NF-κB pathways, which could lead to cardiac injury and affect cardiac function. L-Selenomethionine could effectively alleviate the cardiac damage caused by ammonia and antagonize the cardiotoxicity of ammonia.
Collapse
Affiliation(s)
- Xinxin Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Anqi Wang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Xinqiao Wang
- College of Chemical Engineering and Environment, China University of Petroleum, Beijing, 102249, People's Republic of China
| | - Qian Zhao
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Houjuan Xing
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, People's Republic of China.
| |
Collapse
|
57
|
An S, Moon S, Park SK. Association of metabolic comorbidity with myocardial infarction in individuals with a family history of cardiovascular disease: a prospective cohort study. BMC Public Health 2022; 22:1992. [PMID: 36316766 PMCID: PMC9624008 DOI: 10.1186/s12889-022-14330-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 10/04/2022] [Indexed: 12/07/2022] Open
Abstract
Background The association between metabolic comorbidity and myocardial infarction (MI) among individuals with a family history of cardiovascular disease (CVD) is yet to be elucidated. We aimed to examine the combined effects of metabolic comorbidities, including diabetes mellitus, hypertension, and dyslipidemia, with a family history of CVD in first-degree on the risk of incident MI. Methods This cohort study consisted of 81,803 participants aged 40–89 years without a previous history of MI at baseline from the Korean Genome and Epidemiology Study. We performed Cox proportional hazard regression analysis to estimate the hazard ratios (HRs) and 95% confidence intervals (CIs) for MI and early-onset MI risk associated with metabolic comorbidity in individuals with a family history of CVD. Results During a median follow-up of 5 years, 1,075 and 479 cases of total and early-onset MI were reported, respectively. According to the disease score, among individuals who had a positive family history of CVD, the HRs for MI were 1.92 (95% CI: 1.47–2.51) in individuals with one disease, 2.75 (95% CI: 2.09–3.61) in those with two diseases, and 3.74 (95% CI: 2.45–5.71) in those with three diseases at baseline compared to individuals without a family history of CVD and metabolic diseases. Similarly, an increase of the disease score among individuals with a positive family history of CVD was associated with an increase in early-onset MI risk. Conclusion Metabolic comorbidity was significantly associated with an increased risk of MI among individuals with a family history of CVD. Supplementary Information The online version contains supplementary material available at 10.1186/s12889-022-14330-2.
Collapse
Affiliation(s)
- Seokyung An
- grid.31501.360000 0004 0470 5905Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea ,grid.31501.360000 0004 0470 5905Department of Preventive Medicine, Seoul National University College of Medicine, Seoul, Korea ,grid.31501.360000 0004 0470 5905Cancer Research Institute, Seoul National University, Seoul, Korea
| | - Sungji Moon
- grid.31501.360000 0004 0470 5905Department of Preventive Medicine, Seoul National University College of Medicine, Seoul, Korea ,grid.31501.360000 0004 0470 5905Cancer Research Institute, Seoul National University, Seoul, Korea ,grid.31501.360000 0004 0470 5905Interdisciplinary Program in Cancer Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Sue K. Park
- grid.31501.360000 0004 0470 5905Department of Preventive Medicine, Seoul National University College of Medicine, Seoul, Korea ,grid.31501.360000 0004 0470 5905Cancer Research Institute, Seoul National University, Seoul, Korea ,grid.31501.360000 0004 0470 5905Integrated Major in Innovative Medical Science, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
58
|
Zeng C, Chen Z, Yang M, Lv J, Li H, Gao J, Yuan Z. A Hydroxytricyanopyrrole-Based Fluorescent Probe for Sensitive and Selective Detection of Hypochlorous Acid. Molecules 2022; 27:7237. [PMID: 36364062 PMCID: PMC9656901 DOI: 10.3390/molecules27217237] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 10/20/2022] [Accepted: 10/22/2022] [Indexed: 07/29/2023] Open
Abstract
Hypochlorous acid (HOCl) is a reactive substance that reacts with most biomolecules and is essential in physiological and pathological processes. Abnormally elevated HOCl levels may cause inflammation and other disease responses. To further understand its key role in inflammation, HOCl must be detected in situ. Here, we designed a hydroxytricyanopyrrole-based small-molecule fluorescent probe (HTCP-NTC) to monitor and identify trace amounts of HOCl in biological systems. In the presence of HOCl, HTCP-NTC released hydroxyl groups that emit strong fluorescence covering a wide wavelength range from the visible to near-infrared region owing to the resumption of the intramolecular charge transfer process. Additionally, HTCP-NTC demonstrated a 202-fold fluorescence enhancement accompanied by a large Stokes shift and a low detection limit (21.7 nM). Furthermore, HTCP-NTC provided a rapid response to HOCl within 18 s, allowing real-time monitoring of intracellular HOCl. HTCP-NTC exhibited rapid kinetics and biocompatibility, allowing effective monitoring of the exogenous and endogenous HOCl fluctuations in living cells. Finally, based on fluorescence imaging, HTCP-NTC is a potential method for understanding the relationship between inflammation and HOCl.
Collapse
Affiliation(s)
- Chunhua Zeng
- Key Laboratory of Basic Pharmacology of the Ministry of Education and Joint International Research Laboratory of Ethnomedicine of the Ministry of Education, Zunyi Medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi 563000, China
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, School of Pharmacy, Zunyi Medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi 563000, China
- Guizhou International Scientific and Technological Cooperation Base for Medical Photo-Theranostics Technology and Innovative Drug Development, Zunyi Medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi 563000, China
| | - Zhengjun Chen
- Key Laboratory of Basic Pharmacology of the Ministry of Education and Joint International Research Laboratory of Ethnomedicine of the Ministry of Education, Zunyi Medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi 563000, China
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, School of Pharmacy, Zunyi Medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi 563000, China
- Guizhou International Scientific and Technological Cooperation Base for Medical Photo-Theranostics Technology and Innovative Drug Development, Zunyi Medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi 563000, China
| | - Mingyan Yang
- Key Laboratory of Basic Pharmacology of the Ministry of Education and Joint International Research Laboratory of Ethnomedicine of the Ministry of Education, Zunyi Medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi 563000, China
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, School of Pharmacy, Zunyi Medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi 563000, China
- Guizhou International Scientific and Technological Cooperation Base for Medical Photo-Theranostics Technology and Innovative Drug Development, Zunyi Medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi 563000, China
| | - Jiajia Lv
- Key Laboratory of Basic Pharmacology of the Ministry of Education and Joint International Research Laboratory of Ethnomedicine of the Ministry of Education, Zunyi Medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi 563000, China
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, School of Pharmacy, Zunyi Medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi 563000, China
- Guizhou International Scientific and Technological Cooperation Base for Medical Photo-Theranostics Technology and Innovative Drug Development, Zunyi Medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi 563000, China
| | - Hongyu Li
- Key Laboratory of Basic Pharmacology of the Ministry of Education and Joint International Research Laboratory of Ethnomedicine of the Ministry of Education, Zunyi Medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi 563000, China
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, School of Pharmacy, Zunyi Medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi 563000, China
- Guizhou International Scientific and Technological Cooperation Base for Medical Photo-Theranostics Technology and Innovative Drug Development, Zunyi Medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi 563000, China
| | - Jie Gao
- Key Laboratory of Basic Pharmacology of the Ministry of Education and Joint International Research Laboratory of Ethnomedicine of the Ministry of Education, Zunyi Medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi 563000, China
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, School of Pharmacy, Zunyi Medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi 563000, China
- Guizhou International Scientific and Technological Cooperation Base for Medical Photo-Theranostics Technology and Innovative Drug Development, Zunyi Medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi 563000, China
| | - Zeli Yuan
- Key Laboratory of Basic Pharmacology of the Ministry of Education and Joint International Research Laboratory of Ethnomedicine of the Ministry of Education, Zunyi Medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi 563000, China
- Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, School of Pharmacy, Zunyi Medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi 563000, China
- Guizhou International Scientific and Technological Cooperation Base for Medical Photo-Theranostics Technology and Innovative Drug Development, Zunyi Medical University, No. 6 West Xuefu Road, Xinpu District, Zunyi 563000, China
| |
Collapse
|
59
|
Viloria MAD, Li Q, Lu W, Nhu NT, Liu Y, Cui ZY, Cheng YJ, Lee SD. Effect of exercise training on cardiac mitochondrial respiration, biogenesis, dynamics, and mitophagy in ischemic heart disease. Front Cardiovasc Med 2022; 9:949744. [PMID: 36304547 PMCID: PMC9592995 DOI: 10.3389/fcvm.2022.949744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 08/12/2022] [Indexed: 12/07/2022] Open
Abstract
Objective Cardiac mitochondrial dysfunction was found in ischemic heart disease (IHD). Hence, this study determined the effects of exercise training (ET) on cardiac mitochondrial respiration and cardiac mitochondrial quality control in IHD. Methods A narrative synthesis was conducted after searching animal studies written in English in three databases (PubMed, Web of Science, and EMBASE) until December 2020. Studies that used aerobic exercise as an intervention for at least 3 weeks and had at least normal, negative (sedentary IHD), and positive (exercise-trained IHD) groups were included. The CAMARADES checklist was used to check the quality of the included studies. Results The 10 included studies (CAMARADES score: 6–7/10) used swimming or treadmill exercise for 3–8 weeks. Seven studies showed that ET ameliorated cardiac mitochondrial respiratory function as manifested by decreased reactive oxygen species (ROS) production and increased complexes I-V activity, superoxide dismutase 2 (SOD2), respiratory control ratio (RCR), NADH dehydrogenase subunits 1 and 6 (ND1/6), Cytochrome B (CytB), and adenosine triphosphate (ATP) production. Ten studies showed that ET improved cardiac mitochondrial quality control in IHD as manifested by enhanced and/or controlled mitochondrial biogenesis, dynamics, and mitophagy. Four other studies showed that ET resulted in better cardiac mitochondrial physiological characteristics. Conclusion Exercise training could improve cardiac mitochondrial functions, including respiration, biogenesis, dynamics, and mitophagy in IHD. Systematic review registration https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=226817, identifier: CRD42021226817.
Collapse
Affiliation(s)
- Mary Audrey D. Viloria
- Department of Physical Therapy, Graduate Institute of Rehabilitation Science, China Medical University, Taichung, Taiwan,Department of Physical Therapy, College of Health Sciences, Mariano Marcos State University, Batac, Philippines
| | - Qing Li
- Department of Rehabilitation, Shanghai Xuhui Central Hospital, Shanghai, China
| | - Wang Lu
- Department of Traditional Treatment, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Nguyen Thanh Nhu
- Faculty of Medicine, Can Tho University of Medicine and Pharmacy, Can Tho, Vietnam
| | - Yijie Liu
- School of Rehabilitation Medicine, Shanghai University of Traditional Medicine, Shanghai, China,Institute of Rehabilitation Medicine, Shanghai University of Traditional Medicine, Shanghai, China
| | - Zhen-Yang Cui
- School of Rehabilitation Medicine, Weifang Medical University, Weifang, China
| | - Yu-Jung Cheng
- Department of Physical Therapy, Graduate Institute of Rehabilitation Science, China Medical University, Taichung, Taiwan,Yu-Jung Cheng
| | - Shin-Da Lee
- Department of Physical Therapy, Graduate Institute of Rehabilitation Science, China Medical University, Taichung, Taiwan,School of Rehabilitation Medicine, Weifang Medical University, Weifang, China,Department of Physical Therapy, Asia University, Taichung, Taiwan,*Correspondence: Shin-Da Lee
| |
Collapse
|
60
|
Amerizadeh A, Vaseghi G, Esmaeilian N, Asgary S. Cardiovascular Effects of Cydonia oblonga Miller (Quince). EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:3185442. [PMID: 36262168 PMCID: PMC9576383 DOI: 10.1155/2022/3185442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 07/28/2022] [Accepted: 08/21/2022] [Indexed: 11/07/2022]
Abstract
Cydonia oblonga Miller (quince) is a monotypic genus in the Rosaceae family which used to treat or prevent many medical conditions such as diabetes, cancer, infections, and ulcer. This review will summarize the current state of knowledge available on botany, phytochemistry, and pharmacology of this plant with a focus on its effect on some cardiovascular risk factors such as diabetes, blood pressure, lipid profile, and body weight. Databases of Google Scholar, Web of Science, PubMed, Scopus, and SID were searched systematically for English published articles with no date limitation. There were no human studies found, and all of the studies were conducted on animals or in vitro models. Reviewing of all 12 included articles showed that different types of quince extract have positive effects on cardiovascular-related factors such as blood pressure, diabetes, glucose metabolism, lipid profile, ROS, body weight, liver dysfunction, and thrombosis. An antihypertensive effect of quince showed to be a magnitude that is comparable to captopril and the lipid-lowering effect of quince showed to be a magnitude that is comparable to simvastatin. However, two studies evaluated the effect of quince fruit extract on insulin levels; one of them reported no positive effect, and the other one reported a significant positive effect. It can be concluded that different parts of quince including leaf, seed, and fruit could be used for improving cardiovascular-related factors including blood pressure, glucose metabolism and diabetes, obesity, and lipid-adjusting purposes. Quince was also found to have strong anti-inflammatory and antioxidant capacity. This study paves the way for further studies on the cardiovascular effect of quince consumption as a beneficial nutraceutical in humans.
Collapse
Affiliation(s)
- Atefeh Amerizadeh
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Golnaz Vaseghi
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nazgol Esmaeilian
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sedigheh Asgary
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
61
|
Verma H, Bhattacharjee A, Shivavedi N, Nayak PK. Evaluation of rosmarinic acid against myocardial infarction in maternally separated rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2022; 395:1189-1207. [PMID: 35876905 DOI: 10.1007/s00210-022-02273-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 07/10/2022] [Indexed: 12/07/2022]
Abstract
Depression and coronary heart diseases are the common comorbid disorder affecting humans globally. The present study evaluated the effectiveness of rosmarinic acid (RA) against myocardial infarction (MI) in comorbid depression induced by maternal separation in rats. Maternal stress is one of the childhood crises that may be a potential risk factor for coronary heart disease in later part of life. As per protocol, 70-80% of pups were separated daily for 3 h between postnatal day 1 (PND1) and postnatal day 21 (PND21). Forced-swim test, sucrose preference test, and electrocardiography were performed during the experiment. Body weight was measured on PND0, PND35, and PND55. Orally rosmarinic acid (25 mg/kg and 50 mg/kg) and fluoxetine (10 mg/kg) was done from PND35 to PND55. On PND53 and PND54, isoproterenol (100 mg/kg, subcutaneously) was administered to induce myocardial infarction. On PND55, blood was collected and animals sacrificed, and plasma corticosterone, brain-derived neurotrophic factor, cardiac biomarkers, interleukine-10, and anti-oxidant parameters were measured. Rosmarinic acid and fluoxetine ameliorated the maternal separation-induced increase in immobility period, anhedonia, body weight, ST elevation, corticosterone, creatine kinase-MB (CK-MB), and lactate dehydrogenase (LDH). At the same time, both drugs elevated the tissue levels of BDNF, IL-10, glutathione, and superoxide dismutase activity. This study provides the first experimental evidence that maternal stress is an independent risk factor of cardiac abnormalities in rats. Moreover, maternal stress synergistically increases the severity of cardiac abnormalities induced by isoproterenol. Interestingly, fluoxetine and rosmarinic acid effectively ameliorated behavioral anomalies and myocardial infarction in maternally separated rats. Schematic representation of possible molecular mechanism of action of rosmarinic acid against MS-induced myocardial infarction. RA, rosmarinic acid; MS, maternal separation; PND, postnatal days; ISO, isoproterenol; BDNF, brain-derived neurotrophic factor; GSH, glutathione; SOD, superoxide dismutase; IL-10, interleukin-10; MI, myocardial infarction.
Collapse
Affiliation(s)
- Himanshu Verma
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University (BHU), Uttar Pradesh, Varanasi, 221005, India
| | - Anindita Bhattacharjee
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Uttar Pradesh, Varanasi, 221005, India
| | - Naveen Shivavedi
- Shri Ram Group of Institutions, Faculty of Pharmacy, Jabalpur, Madhya Pradesh, India
| | - Prasanta Kumar Nayak
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University (BHU), Uttar Pradesh, Varanasi, 221005, India.
| |
Collapse
|
62
|
Lim P, Le Maistre M, Campanini LB, De Roux Q, Mongardon N, Landon V, Bouguerra H, Aouate D, Woerther PL, Vincent F, Galy A, Tacher V, Galien S, Ennezat PV, Fiore A, Folliguet T, Huguet R, Mekontso-Dessap A, Iung B, Lepeule R. Vasoplegic Syndrome after Cardiac Surgery for Infective Endocarditis. J Clin Med 2022; 11:5523. [PMID: 36233404 PMCID: PMC9573652 DOI: 10.3390/jcm11195523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/16/2022] [Accepted: 09/16/2022] [Indexed: 12/07/2022] Open
Abstract
Purpose: Post-operative vasoplegic syndrome is a dreaded complication in infective endocarditis (IE). Methods and Results: This retrospective study included 166 consecutive patients referred to cardiac surgery for non-shocked IE. Post-operative vasoplegic syndrome was defined as a persistent hypotension (mean blood pressure < 65 mmHg) refractory to fluid loading and cardiac output restoration. Cardiac surgery was performed 7 (5−12) days after the beginning of antibiotic treatment, 4 (1−9) days after negative blood culture and in 72.3% patients with adapted anti-biotherapy. Timing of cardiac surgery was based on ESC guidelines and operating room availability. Most patients required valve replacement (80%) and cardiopulmonary bypass (CPB) duration was 106 (95−184) min. Multivalvular surgery was performed in 43 patients, 32 had tricuspid valve surgery. Post-operative vasoplegic syndrome was reported in 53/166 patients (31.9%, 95% confidence interval of 24.8−39.0%) of the whole population; only 15.1% (n = 8) of vasoplegic patients had a post-operative documented infection (6 positive blood cultures) and no difference was reported between vasoplegic and non-vasoplegic patients for valve culture and the timing of cardiac surgery. Of the 23 (13.8%) in hospital-deaths, 87.0% (n = 20) occurred in the vasoplegic group and the main causes of death were multiorgan failure (n = 17) and neurological complications (n = 3). Variables independently associated with vasoplegic syndrome were CPB duration (1.82 (1.16−2.88) per tertile) and NTproBNP level (2.11 (1.35−3.30) per tertile). Conclusions: Post-operative vasoplegic syndrome is frequent and is the main cause of death after IE cardiac surgery. Our data suggested that the mechanism of vasoplegic syndrome was more related to inflammatory cardiovascular injury rather than the consequence of ongoing bacteremia.
Collapse
Affiliation(s)
- Pascal Lim
- Service de Cardiologie, DMU Médecine, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires Henri Mondor, Faculté de Santé, Univ. Paris Est Créteil, F-94010 Créteil, France
| | - Margaux Le Maistre
- Service de Cardiologie, DMU Médecine, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires Henri Mondor, Faculté de Santé, Univ. Paris Est Créteil, F-94010 Créteil, France
| | - Lucas Benoudiba Campanini
- Service de Cardiologie, DMU Médecine, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires Henri Mondor, Faculté de Santé, Univ. Paris Est Créteil, F-94010 Créteil, France
| | - Quentin De Roux
- Service d’anesthésie-Réanimation Chirurgicale, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires Henri Mondor, Faculté de Santé, Univ. Paris Est Créteil, F-94010 Créteil, France
| | - Nicolas Mongardon
- Service d’anesthésie-Réanimation Chirurgicale, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires Henri Mondor, Faculté de Santé, Univ. Paris Est Créteil, F-94010 Créteil, France
| | - Valentin Landon
- Service de Cardiologie, DMU Médecine, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires Henri Mondor, Faculté de Santé, Univ. Paris Est Créteil, F-94010 Créteil, France
| | - Hassina Bouguerra
- Service de Cardiologie, DMU Médecine, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires Henri Mondor, Faculté de Santé, Univ. Paris Est Créteil, F-94010 Créteil, France
| | - David Aouate
- Service de Cardiologie, DMU Médecine, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires Henri Mondor, Faculté de Santé, Univ. Paris Est Créteil, F-94010 Créteil, France
| | - Paul-Louis Woerther
- Laboratoire de Bactériologie et Virologie, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires Henri Mondor, Faculté de Santé, Univ. Paris Est Créteil, F-94010 Créteil, France
| | - Fihman Vincent
- Laboratoire de Bactériologie et Virologie, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires Henri Mondor, Faculté de Santé, Univ. Paris Est Créteil, F-94010 Créteil, France
| | - Adrien Galy
- Unité Transversale de Traitement des Infections, DMU PDTI, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires Henri Mondor, Faculté de Santé, Univ. Paris Est Créteil, F-94010 Créteil, France
| | - Vania Tacher
- Service de Radiologie, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires Henri Mondor, Faculté de Santé, Univ. Paris Est Créteil, F-94010 Créteil, France
| | - Sébastien Galien
- Service de Maladies Infectieuses et Immunologie, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires Henri Mondor, Faculté de Santé, Univ. Paris Est Créteil, F-94010 Créteil, France
| | - Pierre-Vladimir Ennezat
- Service de Cardiologie, DMU Médecine, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires Henri Mondor, Faculté de Santé, Univ. Paris Est Créteil, F-94010 Créteil, France
| | - Antonio Fiore
- Service de Chirurgie Cardiaque, DMU CARE, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires Henri Mondor, Faculté de Santé, Univ. Paris Est Créteil, F-94010 Créteil, France
| | - Thierry Folliguet
- Service de Chirurgie Cardiaque, DMU CARE, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires Henri Mondor, Faculté de Santé, Univ. Paris Est Créteil, F-94010 Créteil, France
| | - Raphaelle Huguet
- Service de Cardiologie, DMU Médecine, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires Henri Mondor, Faculté de Santé, Univ. Paris Est Créteil, F-94010 Créteil, France
| | - Armand Mekontso-Dessap
- Service de Médecine Intensive Réanimation, DMU Médecine, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires Henri Mondor, Faculté de Santé, Univ. Paris Est Créteil, F-94010 Créteil, France
| | - Bernard Iung
- Service de Cardiologie, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires Bichat et Université Paris Cité, Assistance Publique-Hôpitaux de Paris (AP-HP), F-75018 Paris, France
| | - Raphael Lepeule
- Unité Transversale de Traitement des Infections, DMU PDTI, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires Henri Mondor, Faculté de Santé, Univ. Paris Est Créteil, F-94010 Créteil, France
| |
Collapse
|
63
|
Zhang Z, Dalan R, Hu Z, Wang JW, Chew NW, Poh KK, Tan RS, Soong TW, Dai Y, Ye L, Chen X. Reactive Oxygen Species Scavenging Nanomedicine for the Treatment of Ischemic Heart Disease. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2202169. [PMID: 35470476 DOI: 10.1002/adma.202202169] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/08/2022] [Indexed: 06/14/2023]
Abstract
Ischemic heart disease (IHD) is the leading cause of disability and mortality worldwide. Reactive oxygen species (ROS) have been shown to play key roles in the progression of diabetes, hypertension, and hypercholesterolemia, which are independent risk factors that lead to atherosclerosis and the development of IHD. Engineered biomaterial-based nanomedicines are under extensive investigation and exploration, serving as smart and multifunctional nanocarriers for synergistic therapeutic effect. Capitalizing on cell/molecule-targeting drug delivery, nanomedicines present enhanced specificity and safety with favorable pharmacokinetics and pharmacodynamics. Herein, the roles of ROS in both IHD and its risk factors are discussed, highlighting cardiovascular medications that have antioxidant properties, and summarizing the advantages, properties, and recent achievements of nanomedicines that have ROS scavenging capacity for the treatment of diabetes, hypertension, hypercholesterolemia, atherosclerosis, ischemia/reperfusion, and myocardial infarction. Finally, the current challenges of nanomedicines for ROS-scavenging treatment of IHD and possible future directions are discussed from a clinical perspective.
Collapse
Affiliation(s)
- Zhan Zhang
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, 999078, China
| | - Rinkoo Dalan
- Department of Endocrinology, Tan Tock Seng Hospital, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 408433, Singapore
| | - Zhenyu Hu
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Jiong-Wei Wang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Department of Diagnostic Radiology and Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Nicholas Ws Chew
- Department of Cardiology, National University Heart Centre, National University Hospital, Singapore, 119074, Singapore
| | - Kian-Keong Poh
- Department of Cardiology, National University Heart Centre, National University Hospital, Singapore, 119074, Singapore
| | - Ru-San Tan
- Department of Cardiology, National Heart Centre Singapore, Singapore, 119609, Singapore
| | - Tuck Wah Soong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Yunlu Dai
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, 999078, China
- MoE Frontiers Science Center for Precision Oncology, University of Macao, Taipa, Macau SAR, 999078, China
| | - Lei Ye
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Xiaoyuan Chen
- Department of Diagnostic Radiology and Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Department of Chemical and Biomolecular Engineering and Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| |
Collapse
|
64
|
Arozal W, Monayo ER, Barinda AJ, Perkasa DP, Soetikno V, Nafrialdi N, Louisa M. Protective effects of silver nanoparticles in isoproterenol-induced myocardial infarction in rats. Front Med (Lausanne) 2022; 9:867497. [PMID: 36091690 PMCID: PMC9454814 DOI: 10.3389/fmed.2022.867497] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 08/04/2022] [Indexed: 12/07/2022] Open
Abstract
Background Silver nanoparticles (AgNPs) are widely used in the medical field, including cardiovascular. However, limited research has investigated the effect of AgNPs on the protection of myocardial infarction (MI). Objectives Isoproterenol (Iso)-induced MI and the cardiac protection offered by AgNPs were investigated in the present study. Additionally, we characterized the profile of Ag in the form of nanoparticles. Methods Twenty-four male Wistar rats were randomly divided into four groups as follows: normal, Iso, Iso + AgNO3, and Iso + AgNP groups. AgNPs and silver ion (AgNO3) were administered intraperitoneally at 2.5 mg/kg BW for 14 days. Iso induction was performed using two doses of 85 mg/kg BW given subcutaneously on days 13 and 14. Blood and cardiac tissue samples were taken 24 h after the last dose of Iso and checked for Creatine Kinase-MB (CK-MB), lactate dehydrogenase in plasma along with oxidative stress parameters, mitochondria biogenesis markers, and inflammation representative genes in cardiac tissue. Additionally, we analyzed the histopathological features in cardiac tissue. Results The silver was confirmed in the form of nanoparticles by its size at intervals of 8.72-37.84 nm. Both AgNO3 and AgNPs showed similar cardioprotective effects, as shown by the decrease in biochemical markers of cardiac toxicity, namely, CK-MB. Additionally, AgNPs group have better efficacy compared with AgNO3 group in ameliorating Iso-mediated oxidative stress production, as evidenced by the significant decrease in malondialdehyde level and increased superoxide dismutase activity (P < 0.0001 and P < 0.01, respectively) in cardiac tissue compared with the Iso group. Mechanistically, AgNPs, but not AgNO3, enhanced the expression levels of mitochondrial transcription factor A and peroxisome proliferator-activated receptor-gamma coactivator 1-alpha in post-MI heart and reduced the protein expression of nuclear factor-kappa B (NF-κB) assessed by western blot analysis. Furthermore, these results were confirmed with the histopathological evaluation of cardiac tissue. Nevertheless, pretreatment with either AgNO3 or AgNPs improved the aspartate aminotransferase level. Conclusion These results suggested that AgNPs have more superior cardioprotective effect compared with AgNO3 against Iso-induced MI, at least in part through amelioration of NF-κB expression level induced by oxidative stress overproduction.
Collapse
Affiliation(s)
- Wawaimuli Arozal
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Edwina Rogayah Monayo
- Doctoral Program in Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Faculty of Medicine Universitas Negeri Gorontalo, Gorontalo, Indonesia
| | - Agian Jeffilano Barinda
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Metabolic, Cardiovascular and Aging Cluster, The Indonesian Medical Education and Research Institute, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
| | - Dian Pribadi Perkasa
- Doctoral Program in Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Research Center for Radiation Process Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency, Jakarta, Indonesia
| | - Vivian Soetikno
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Nafrialdi Nafrialdi
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Melva Louisa
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| |
Collapse
|
65
|
Detection and Quantification of Nanoparticle-Induced Intracellular ROS in Live Cells by Laser Scanning Confocal Microscopy. Methods 2022; 207:11-19. [PMID: 36028162 DOI: 10.1016/j.ymeth.2022.08.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 07/29/2022] [Accepted: 08/12/2022] [Indexed: 12/07/2022] Open
Abstract
All living organisms utilise reactive oxygen species (ROS) for essential cellular functions, the majority of which involve signal transduction pathways, such as enzyme regulation, cell growth and differentiation signalling, and inflammation mediation. Increased ROS in cancer cells can be caused by abnormalities in the tumor environment. However, using fluorescence microscopy to detect and quantify ROS changes in biological systems is difficult for several reasons: (1) lack of specificity of ROS-sensitive probes, (2) high turnover of ROS species, (3) rapid decrease in ROS fluorescence with time, and (4) detection and quantification techniques with insufficient sensitivity. Existing approaches to ROS measurement using confocal microscopy imaging focus solely on ROS detection rather than quantification. A novel fluorescence-based ROS detection and quantification technique has been developed for the purpose of resolving the limitations of existing methods. In general, ROS is detected by fluorescence using instrumentation such as flow cytometry and laser scanning confocal microscopy. However, these approaches confirm only the presence or absence of ROS; they are not quantitative, which is essential for therapeutic applications. In the newly developed technique, cerium-based ROS-generating nanoparticles have been used to elevate the ROS in HT-1080 fibrosarcoma cells. The elevated ROS levels are detected using an H2DCFDA fluorescence probe, which is used widely for this application, and captured as digital images using a 488 nm fluorescence channel. Quantification of the ROS is achieved using script in MATLAB software to convert the fluorescence intensities to numerical values. Thus, this technique nearly simultaneously integrates both detection and quantification of the ROS, which provides the statistical justification necessary to support therapeutic translation.
Collapse
|
66
|
Dutta A, Das M, Ghosh A, Rana S. Molecular and cellular pathophysiology of circulating cardiomyocyte-specific cell free DNA (cfDNA): Biomarkers of heart failure and potential therapeutic targets. Genes Dis 2022. [DOI: 10.1016/j.gendis.2022.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022] Open
|
67
|
Rotariu D, Babes EE, Tit DM, Moisi M, Bustea C, Stoicescu M, Radu AF, Vesa CM, Behl T, Bungau AF, Bungau SG. Oxidative stress - Complex pathological issues concerning the hallmark of cardiovascular and metabolic disorders. Biomed Pharmacother 2022; 152:113238. [PMID: 35687909 DOI: 10.1016/j.biopha.2022.113238] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/30/2022] [Accepted: 06/02/2022] [Indexed: 12/07/2022] Open
Abstract
Oxidative stress is a complex biological process characterized by the excessive production of reactive oxygen species (ROS) that act as destroyers of the REDOX balance in the body and, implicitly, inducing oxidative damage. All the metabolisms are impaired in oxidative stress and even nucleic acid balance is influenced. ROS will promote structural changes of the tissues and organs due to interaction with proteins and phospholipids. The constellation of the cardiovascular risk factors (CVRFs) will usually develop in subjects with predisposition to cardiac disorders. Oxidative stress is usually related with hypertension (HTN), diabetes mellitus (DM), obesity and cardiovascular diseases (CVDs) like coronary artery disease (CAD), cardiomyopathy or heart failure (HF), that can develop in subjects with the above-mentioned diseases. Elements describing the complex relationship between CVD and oxidative stress should be properly explored and described because prevention may be the optimal approach. Our paper aims to expose in detail the complex physiopathology of oxidative stress in CVD occurrence and novelties regarding the phenomenon. Biomarkers assessing oxidative stress or therapy targeting specific pathways represent a major progress that actually change the outcome of subjects with CVD. New antioxidants therapy specific for each CVD represents a captivating and interesting future perspective with tremendous benefits on subject's outcome.
Collapse
Affiliation(s)
- Dragos Rotariu
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania.
| | - Emilia Elena Babes
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy of Oradea, University of Oradea, 410073 Oradea, Romania.
| | - Delia Mirela Tit
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania; Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania.
| | - Madalina Moisi
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy of Oradea, University of Oradea, 410073 Oradea, Romania.
| | - Cristiana Bustea
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy of Oradea, University of Oradea, 410073 Oradea, Romania.
| | - Manuela Stoicescu
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy of Oradea, University of Oradea, 410073 Oradea, Romania.
| | - Andrei-Flavius Radu
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania; Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy of Oradea, University of Oradea, 410073 Oradea, Romania.
| | - Cosmin Mihai Vesa
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy of Oradea, University of Oradea, 410073 Oradea, Romania.
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India.
| | | | - Simona Gabriela Bungau
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania; Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania.
| |
Collapse
|
68
|
Khan A, Iqubal A, Wasim M, Syed MA, Haque SE. D-pinitol attenuates isoproterenol induced myocardial infarction by alleviating cardiac inflammation, oxidative stress and ultrastructural changes in Swiss Albino mice. Clin Exp Pharmacol Physiol 2022; 49:1232-1245. [PMID: 35866379 DOI: 10.1111/1440-1681.13703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 06/25/2022] [Accepted: 07/18/2022] [Indexed: 12/07/2022]
Abstract
Cardiovascular diseases are the most disturbing problems throughout the world. The side effects of existing drugs are continuously compelling the scientist to look for better options in terms of safety, efficacy and cost-effectiveness. Our study is also a move in this direction. We have chosen D-pinitol to see its cardioprotective role in isoproterenol-induced myocardial infarction in Swiss Albino mice. Grouping was made by dividing mice into eight groups (n = 6). Group I - control; Group II - Isoproterenol (ISO) (150 mg/kg, i.p.); Group III - D-pinitol (PIN) (25 mg), Group IV - PIN (50 mg), Group V - PIN (100 mg) per kg per oral, respectively with ISO; Group VI - PIN per se (100 mg D-pinitol only); Group VII - Propranolol (PRO) (20 mg/kg/oral) with ISO; and Group VIII - PRO per se (20 mg/kg, p.o.). After 24 hrs of the last dose, the blood sample was collected for biochemical parameters, then mice were, euthanised through cervical dislocation under anesthesia and cardiac tissue was collected for biochemical, histopathological and ultrastructural evaluation. Administration of ISO in mice altered the level of antioxidant markers, cardiac injury markers and inflammatory markers, which were significantly restored towards normal by D-pinitol at the dose of 50 and 100 mg. 25 mg of D-pinitol dosage, did not produce significant cardio protection. The histopathological and ultrastructural analysis further confirmed these findings. Our study showed that D-pinitol significantly protected myocardial damage which was induced by ISO and reverted oxidative stress and inflammation considerably.
Collapse
Affiliation(s)
- Aamir Khan
- Department of Pharmacology, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi, India
| | - Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi, India
| | - Mohd Wasim
- Department of Pharmacology, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi, India
| | - Mansoor Ali Syed
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| | - Syed Ehtaishamul Haque
- Department of Pharmacology, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi, India
| |
Collapse
|
69
|
Spartalis M, Pantelidis P, Kontogiannis C, Paschou SA, Spartalis E, Iliopoulos DC, Siasos G. The complex relationship between diabetes and cardiac arrhythmias: Pathophysiology and mechanisms. Curr Pharm Des 2022; 28:2129-2130. [PMID: 35864792 DOI: 10.2174/1381612828666220720095433] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 06/10/2022] [Indexed: 12/07/2022]
Abstract
Coronary artery disease, autonomic neuropathy, and diabetic cardiomyopathy are the most common cardiovascular complications of diabetes. However, emerging evidence demonstrates that diabetes also affects the heart's electrical conduction system, culminating in lethal arrhythmias and sudden cardiac death. Diabetes and rhythm disturbances have a complex relationship, and arrhythmias cannot be attributed to ischemia and autonomic neuropathy only. Hypoglycemia, hyperglycemia, and glucose fluctuations can potentially induce arrhythmias by activating various pathways. Structural remodeling can accelerate and exacerbate disease development. Mitochondrial dysfunction can also alter the structure and metabolism of cardiomyocytes and contribute to disease progression through oxidative stress and inflammation.
Collapse
Affiliation(s)
- Michael Spartalis
- 3rd Department of Cardiology, Sotiria Thoracic Diseases General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Panteleimon Pantelidis
- 3rd Department of Cardiology, Sotiria Thoracic Diseases General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Christos Kontogiannis
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Stavroula A Paschou
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Eleftherios Spartalis
- Laboratory of Experimental Surgery and Surgical Research 'N. S. Christeas', National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Dimitrios C Iliopoulos
- Laboratory of Experimental Surgery and Surgical Research 'N. S. Christeas', National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Gerasimos Siasos
- 3rd Department of Cardiology, Sotiria Thoracic Diseases General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
70
|
Hepatoprotective Role of Carvedilol against Ischemic Hepatitis Associated with Acute Heart Failure via Targeting miRNA-17 and Mitochondrial Dynamics-Related Proteins: An In Vivo and In Silico Study. Pharmaceuticals (Basel) 2022; 15:ph15070832. [PMID: 35890131 PMCID: PMC9319470 DOI: 10.3390/ph15070832] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/26/2022] [Accepted: 07/01/2022] [Indexed: 02/01/2023] Open
Abstract
Acute heart failure (AHF) is one of the most common diseases in old age that can lead to mortality. Systemic hypoperfusion is associated with hepatic ischemia–reperfusion injury, which may be irreversible. Ischemic hepatitis due to AHF has been linked to the pathogenesis of liver damage. In the present study, we extensively investigated the role of mitochondrial dynamics-related proteins and their epigenetic regulation in ischemic liver injury following AHF and explored the possible hepatoprotective role of carvedilol. The biochemical analysis revealed that the ischemic liver injury following AHF significantly elevated the activity of alanine aminotransferase (ALT), aspartate aminotransferase (AST), and alkaline phosphatase (ALP) enzymes, the level of total and direct bilirubin, and the expression of hepatic mitogen-activated protein kinase (MAPK), dynamin-1-like protein (DNM1L), and hepatic miRNA-17. At the same time, it significantly reduced the serum albumin level, the activity of hepatic superoxide dismutase (SOD), and the expression of mitochondrial peroxisome proliferator-activated receptor-1α (PGC-1α), and mitofusin 2 (Mtf2). The histological examination of the liver tissue revealed degenerated hepatocytes. Interestingly, administration of carvedilol either prior to or after isoprenaline-induced AHF significantly improved the liver function and reversed the deterioration effect of AHF-induced ischemic hepatitis, as demonstrated by biochemical, immunohistochemical, and histological analysis. Our results indicated that the hepatoprotective effect of carvedilol in ameliorating hepatic ischemic damage could be attributed to its ability to target the mitochondrial dynamics-related proteins (Mtf2, DNM1L and PGC-1α), but also their epigenetic regulator miRNA-17. To further explore the mode of action of carvedilol, we have investigated, in silico, the ability of carvedilol to target dynamin-1-like protein and mitochondrial dynamics protein (MID51). Our results revealed that carvedilol has a high binding affinity (−14.83 kcal/mol) toward the binding pocket of DNM1L protein. In conclusion, our study highlights the hepatoprotective pharmacological application of carvedilol to attenuate ischemic hepatitis associated with AHF.
Collapse
|
71
|
Gropler MRF, Lipshultz SE, Wilkinson JD, Towbin JA, Colan SD, Canter CE, Lavine KJ, Simpson KE. Pediatric and adult dilated cardiomyopathy are distinguished by distinct biomarker profiles. Pediatr Res 2022; 92:206-215. [PMID: 34404929 DOI: 10.1038/s41390-021-01698-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 07/29/2021] [Accepted: 08/04/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Emerging evidence suggests that pediatric and adult dilated cardiomyopathy (DCM) represent distinct diseases. Few diagnostic tools exist for pediatric cardiologists to assess clinical status and prognosis. We hypothesized that pediatric DCM would have a unique biomarker profile compared to adult DCM and controls. METHODS We utilized a DNA aptamer array (SOMAScan) to compare biomarker profiles between pediatric and adult DCM. We simultaneously measured 1310 plasma proteins and peptides from 39 healthy children (mean age 3 years, interquartile range (IQR) 1-14), 39 ambulatory subjects with pediatric DCM (mean age 2.7 years, IQR 1-13), and 40 ambulatory adults with DCM (mean age 53 years, IQR 46-63). RESULTS Pediatric and adult DCM patients displayed distinct biomarker profiles, despite similar clinical characteristics. We identified 20 plasma peptides and proteins that were increased in pediatric DCM compared to age- and sex-matched controls. Unbiased multidimensionality reduction analysis suggested previously unrecognized heterogeneity among pediatric DCM subjects. Biomarker profile analysis identified four subgroups of pediatric DCM with distinguishing clinical characteristics. CONCLUSIONS These findings support the emerging concept that pediatric and adult DCM are distinct disease entities, signify the need to develop pediatric-specific biomarkers for disease prognostication, and challenge the paradigm that pediatric DCM should be viewed as a single disease. IMPACT Pediatric and adult DCM patients displayed distinct biomarker profiles, despite similar clinical characteristics and outcomes. Our findings suggest that pediatric DCM may be a heterogeneous disease with various sub-phenotypes, including differing biomarker profiles and clinical findings. These data provide prerequisite information for future prospective studies that validate the identified pediatric DCM biomarkers, address their diagnostic accuracy and prognostic significance, and explore the full extent of heterogeneity amongst pediatric DCM patients.
Collapse
Affiliation(s)
- Melanie R F Gropler
- Division of Pediatric Cardiology, Department of Pediatrics, University of Colorado Anschutz Medical center, Aurora, CO, USA
| | - Steven E Lipshultz
- Department of Pediatrics, University of Buffalo Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY, USA
| | - James D Wilkinson
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Jeffrey A Towbin
- Division of Pediatric Cardiology, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Steven D Colan
- Department of Pediatric Cardiology, Boston Children's Hospital, Boston, MA, USA
| | - Charles E Canter
- Division of Pediatric Cardiology, Department of Pediatrics, Washington University School of Medicine, Saint Louis, MO, USA
| | - Kory J Lavine
- Division of Cardiology, Department of Medicine, Center for Cardiovascular Research, Washington University School of Medicine, Saint Louis, MO, USA
| | - Kathleen E Simpson
- Division of Pediatric Cardiology, Department of Pediatrics, University of Colorado Anschutz Medical center, Aurora, CO, USA.
| |
Collapse
|
72
|
Naghdi A, Goodarzi MT, Karimi J, Hashemnia M, Khodadadi I. Effects of curcumin and metformin on oxidative stress and apoptosis in heart tissue of type 1 diabetic rats. J Cardiovasc Thorac Res 2022; 14:128-137. [PMID: 35935389 PMCID: PMC9339728 DOI: 10.34172/jcvtr.2022.23] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 06/01/2022] [Indexed: 12/07/2022] Open
Abstract
Introduction: Hyperglycemia enhances oxidative stress and apoptosis and induces damages in heart tissue. Based on antioxidant properties of curcumin and metformin, we hypothesized that these agents may exhibit cardioprotective effects by attenuating oxidative stress and modulating expression of the genes involved in apoptosis in type-1 diabetes.
Methods: Thirty-six male rats were randomly divided into six groups; (N): control; (D): streptozotocin-induced diabetic rats; (D+Cur50) and (D+Cur150): diabetic rats treated with 50 and 150 milligram of curcumin per kilogram of body weight (mg/kg.bw), respectively; (D+Met300) and (D+Met500): diabetic rats received 300 and 500 mg/kg.bw of metformin, respectively. Heart tissues were dissected and gene expression levels of Bax, Bcl-2, and caspase-3 were analyzed. Total anti-oxidant capacity (TAC), total oxidant status (TOS), and malondialdehyde (MDA) level, and activities of catalase (CAT), superoxide dismutase (SOD) and glutathione peroxidase (GPx) were measured.
Results: Enhancement in TOS, OSI, and MDA levels as well as increased in the activity of CAT and reduction in SOD and GPx activities were observed in diabetic group (D) compared with control rats. Treatment of diabetic animals with either curcumin or metformin normalized TOS, OSI, and MDA levels and restored CAT, SOD, and GPx activities. Diabetes caused extensive damages in heart tissue of rats (group D) and increased expression of caspase-3 and Bax genes and enhanced ratio of Bax/Bcl-2 expression compared with controls. Treatment with curcumin or metformin mitigated histopathological changes and dampened apoptosis by normalizing Bax and caspase-3 expression.
Conclusion: Curcumin and metformin modulated diabetes-induced cardiac damage probably by reducing oxidative stress.
Collapse
Affiliation(s)
- Atefeh Naghdi
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Taghi Goodarzi
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Biochemistry, Shahrood Branch, Islamic Azad University, Shahrood, Iran
| | - Jamshid Karimi
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Hashemnia
- Department of Pathobiology, Faculty of Veterinary Medicine, Razi University, Kermanshah, Iran
| | - Iraj Khodadadi
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
73
|
Eberhardt DR, Lee SH, Yin X, Balynas AM, Rekate EC, Kraiss JN, Lang MJ, Walsh MA, Streiff ME, Corbin AC, Li Y, Funai K, Sachse FB, Chaudhuri D. EFHD1 ablation inhibits cardiac mitoflash activation and protects cardiomyocytes from ischemia. J Mol Cell Cardiol 2022; 167:1-14. [PMID: 35304170 PMCID: PMC9107497 DOI: 10.1016/j.yjmcc.2022.03.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 02/23/2022] [Accepted: 03/11/2022] [Indexed: 12/07/2022]
Abstract
Altered levels of intracellular calcium (Ca2+) are a highly prevalent feature in different forms of cardiac injury, producing changes in contractility, arrhythmias, and mitochondrial dysfunction. In cardiac ischemia-reperfusion injury, mitochondrial Ca2+ overload leads to pathological production of reactive oxygen species (ROS), activates the permeability transition, and cardiomyocyte death. Here we investigated the cardiac phenotype caused by deletion of EF-hand domain-containing protein D1 (Efhd1-/-), a Ca2+-binding mitochondrial protein whose function is poorly understood. Efhd1-/- mice are viable and have no adverse cardiac phenotypes. They feature reductions in basal ROS levels and mitoflash events, both important precursors for mitochondrial injury, though cardiac mitochondria have normal susceptibility to Ca2+ overload. Notably, we also find that Efhd1-/- mice and their cardiomyocytes are resistant to hypoxic injury.
Collapse
Affiliation(s)
- David R Eberhardt
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah, United States of America
| | - Sandra H Lee
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah, United States of America
| | - Xue Yin
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah, United States of America
| | - Anthony M Balynas
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah, United States of America
| | - Emma C Rekate
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah, United States of America
| | - Jackie N Kraiss
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah, United States of America
| | - Marisa J Lang
- Diabetes & Metabolism Research Center, University of Utah, United States of America
| | - Maureen A Walsh
- Diabetes & Metabolism Research Center, University of Utah, United States of America
| | - Molly E Streiff
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah, United States of America; Department of Biomedical Engineering, University of Utah, United States of America
| | - Andrea C Corbin
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah, United States of America; Department of Biomedical Engineering, University of Utah, United States of America
| | - Ying Li
- Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, United States of America
| | - Katsuhiko Funai
- Diabetes & Metabolism Research Center, University of Utah, United States of America
| | - Frank B Sachse
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah, United States of America; Department of Biomedical Engineering, University of Utah, United States of America
| | - Dipayan Chaudhuri
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah, United States of America; Department of Biomedical Engineering, University of Utah, United States of America; Department of Internal Medicine, Division of Cardiovascular Medicine, University of Utah, Salt Lake City, UT, United States of America.
| |
Collapse
|
74
|
Zhao X, He Y, Zhang Y, Wan H, Wan H, Yang J. Inhibition of Oxidative Stress: An Important Molecular Mechanism of Chinese Herbal Medicine (Astragalus membranaceus, Carthamus tinctorius L., Radix Salvia Miltiorrhizae, etc.) in the Treatment of Ischemic Stroke by Regulating the Antioxidant System. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1425369. [PMID: 35651725 PMCID: PMC9151006 DOI: 10.1155/2022/1425369] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/11/2022] [Indexed: 01/07/2023]
Abstract
Ischemic stroke is a severe cerebrovascular disease with high mortality and morbidity. Traditional Chinese medicine (TCM) has been utilized for thousands of years in China and is becoming increasingly popular all over the world, especially for the treatments of ischemic stroke. More and more evidences have implicated that oxidative stress has been closely related with ischemic stroke. This review will concentrate on the evidence of the action mechanism of Chinese herbal medicine and its active ingredient in preventing ischemic stroke by modulating redox signaling and oxidative stress pathways and providing references for clinical treatment and scientific research applications.
Collapse
Affiliation(s)
- Xixi Zhao
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yu He
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yangyang Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Haofang Wan
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Haitong Wan
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jiehong Yang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| |
Collapse
|
75
|
The Regulatory Mechanism and Effect of RIPK3 on PE-induced Cardiomyocyte Hypertrophy. J Cardiovasc Pharmacol 2022; 80:236-250. [PMID: 35561290 DOI: 10.1097/fjc.0000000000001293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 04/26/2022] [Indexed: 12/07/2022]
Abstract
ABSTRACT As a critical regulatory molecule, receptor-interacting protein kinase 3 (RIPK3) can mediate the signaling pathway of programmed necrosis. Calcium/calmodulin-dependent protein kinase II (CaMKII) has been proved as a new substrate for RIPK3-induced necroptosis. In the present study, we aimed to investigate the regulatory mechanism of RIPK3 on phenylephrine (PE)-induced cardiomyocyte hypertrophy. Cardiomyocyte hypertrophy was induced by exposure to PE (100 μM) for 48 h. Primary cardiomyocytes were pretreated with RIPK3 inhibitor GSK'872 (10 μM), and RIPK3 siRNA was used to deplete the intracellular expression of RIPK3. The indexes related to myocardial hypertrophy, cell injury, necroptosis, CaMKII activation, gene expression, oxidative stress, and mitochondrial membrane potential were measured. We found that after cardiomyocytes were stimulated by PE, the expressions of hypertrophy markers, atrial and brain natriuretic peptides (ANP and BNP), were increased, the release of lactate dehydrogenase (LDH) was increased, the level of adenosine triphosphate (ATP)was decreased, the oxidation and phosphorylation levels of CaMKII were increased, and CaMKIIδ alternative splicing was disturbed. However, both GSK'872 and depletion of RIPK3 could reduce myocardial dysfunction, inhibit CaMKII activation and necroptosis, and finally alleviate myocardial hypertrophy. In addition, the pretreatment of RIPK3 could also lessen the accumulation of reactive oxygen species (ROS) induced by PE and stabilize the membrane potential of mitochondria. These results indicated that targeted inhibition of RIPK3 could suppress the activation of CaMKII and reduce necroptosis and oxidative stress, leading to alleviated myocardial hypertrophy. Collectively, our findings provided valuable insights into the clinical treatment of hypertrophic cardiomyopathy.
Collapse
|
76
|
Shi X, Dorsey A, Qiu H. New Progress in the Molecular Regulations and Therapeutic Applications in Cardiac Oxidative Damage Caused by Pressure Overload. Antioxidants (Basel) 2022; 11:antiox11050877. [PMID: 35624741 PMCID: PMC9137593 DOI: 10.3390/antiox11050877] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/25/2022] [Accepted: 04/26/2022] [Indexed: 12/11/2022] Open
Abstract
Chronic pressure overload is a key risk factor for mortality due to its subsequent development of heart failure, in which the underlying molecular mechanisms remain vastly undetermined. In this review, we updated the latest advancements for investigating the role and relevant mechanisms of oxidative stress involved in the pathogenesis of pressure-overload-induced cardiomyopathy and cardiac dysfunction, focusing on significant biological sources of reactive oxygen species (free radical) production, antioxidant defenses, and their association with the cardiac metabolic remodeling in the stressed heart. We also summarize the newly developed preclinical therapeutic approaches in animal models for pressure-overload-induced myocardial damage. This review aims to enhance the current understanding of the mechanisms of chronic hypertensive heart failure and potentially improve the development of better therapeutic strategies for the associated diseases.
Collapse
Affiliation(s)
| | | | - Hongyu Qiu
- Correspondence: ; Tel.: +1-404-413-3371; Fax: +1-404-413-9566
| |
Collapse
|
77
|
Xing Y, Sui Z, Liu Y, Wang MM, Wei X, Lu Q, Wang X, Liu N, Lu C, Chen R, Wu M, Wang Y, Zhao YH, Guo F, Cao JL, Qi J, Wang W. Blunting TRPML1 channels protects myocardial ischemia/reperfusion injury by restoring impaired cardiomyocyte autophagy. Basic Res Cardiol 2022; 117:20. [PMID: 35389129 DOI: 10.1007/s00395-022-00930-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 03/15/2022] [Accepted: 03/26/2022] [Indexed: 01/31/2023]
Abstract
Accumulating evidence suggests that autophagy dysfunction plays a critical role in myocardial ischemia/reperfusion (I/R) injury. However, the underling mechanism of malfunctional autophagy in the cardiomyocytes subjected to I/R has not been well defined. As a result, there is no effective therapeutic option by targeting autophagy to prevent myocardial I/R injury. Here, we used both an in vitro and an in vivo I/R model to monitor autophagic flux in the cardiomyocytes, by exposing neonatal rat ventricular myocytes to hypoxia/reoxygenation and by subjecting mice to I/R, respectively. We observed that the autophagic flux in the cardiomyocytes subjected to I/R was blocked in both in vitro and in vivo models. Down-regulating a lysosomal cationic channel, TRPML1, markedly restored the blocked myocardial autophagic flux induced by I/R, demonstrating that TRPML1 directly contributes to the blocked autophagic flux in the cardiomyocytes subjected to I/R. Mechanistically, TRPML1 is activated secondary to ROS elevation following ischemia/reperfusion, which in turn induces the release of lysosomal zinc into the cytosol and ultimately blocks the autophagic flux in cardiomyocytes, presumably by disrupting the fusion between autophagosomes and lysosomes. As a result, the inhibited myocardial autophagic flux induced by TRPML1 disrupted mitochondria turnover and resulted in mass accumulation of damaged mitochondria and further ROS release, which directly led to cardiomyocyte death. More importantly, pharmacological and genetic inhibition of TRPML1 channels greatly reduced infarct size and rescued heart function in mice subjected to I/R in vivo by restoring impaired myocardial autophagy. In summary, our study demonstrates that secondary to ROS elevation, activation of TRPML1 results in autophagy inhibition in the cardiomyocytes subjected to I/R, which directly leads to cardiomyocyte death by disrupting mitochondria turnover. Therefore, targeting TRPML1 represents a novel therapeutic strategy to protect against myocardial I/R injury.
Collapse
Affiliation(s)
- Yanhong Xing
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China
| | - Zhongheng Sui
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China
| | - Yucheng Liu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China
| | - Meng-Meng Wang
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital, China Medical University, Shenyang, 110122, Liaoning, China
| | - Xiangqing Wei
- Department of Anesthesiology, the Second Affiliated Hospital of Nantong University, Nantong, 226006, Jiangsu, China
| | - Qixia Lu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China
| | - Xinyan Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China
| | - Nan Liu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China
| | - Chen Lu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China
| | - Rong Chen
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China
| | - Mengmei Wu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China
| | - Yuqing Wang
- Department of Medicine and Biosystemic Science, Faculty of Medicine, Kyushu University, Fukuoka, Kyushu, 8128582, Japan
| | - Yu-Hong Zhao
- Department of Clinical Epidemiology, Clinical Research Center, Shengjing Hospital of China Medical University, No. 36 San Hao Street, Shenyang, 110004, Liaoning, China
| | - Feng Guo
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, Liaoning, China.
| | - Jun-Li Cao
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China.
| | - Jiansong Qi
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China. .,Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, NS, B3H 4R2, Canada.
| | - Wuyang Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
78
|
Reily-Bell M, Bahn A, Katare R. Reactive Oxygen Species-Mediated Diabetic Heart Disease: Mechanisms and Therapies. Antioxid Redox Signal 2022; 36:608-630. [PMID: 34011169 DOI: 10.1089/ars.2021.0098] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Significance: Diabetic heart disease (DHD) is the primary cause of mortality in people with diabetes. A significant contributor to the development of DHD is the disruption of redox balance due to reactive oxygen species (ROS) overproduction resulting from sustained high glucose levels. Therapies specifically focusing on the suppression of ROS will hugely benefit patients with DHD. Recent Advances: In addition to the gold standard pharmacological therapies, the recent development of gene therapy provides an exciting avenue for developing new therapeutics to treat ROS-mediated DHD. In particular, microRNAs (miRNAs) are gaining interest due to their crucial role in several physiological and pathological processes, including DHD. Critical Issues: miRNAs have many targets and differential function depending on the environment. Therefore, a proper understanding of the function of miRNAs in specific cell types and cell states is required for the successful application of this technology. In the present review, we first provide an overview of the role of ROS in contributing to DHD and the currently available treatments. We then discuss the newer gene therapies with a specific focus on the role of miRNAs as the causative factors and therapeutic targets to combat ROS-mediated DHD. Future Directions: The future of miRNA therapeutics in tackling ROS-mediated DHD is dependent on a complete understanding of how miRNAs behave in different cells and environments. Future research should also aim to develop conditional miRNA therapeutic platforms capable of switching on and off in response to disruptions in the redox state. Antioxid. Redox Signal. 36, 608-630.
Collapse
Affiliation(s)
- Matthew Reily-Bell
- Department of Physiology-HeartOtago, University of Otago, Dunedin, New Zealand
| | - Andrew Bahn
- Department of Physiology-HeartOtago, University of Otago, Dunedin, New Zealand
| | - Rajesh Katare
- Department of Physiology-HeartOtago, University of Otago, Dunedin, New Zealand
| |
Collapse
|
79
|
Temiz Artmann A, Kurulgan Demirci E, Fırat IS, Oflaz H, Artmann GM. Recombinant Activated Protein C (rhAPC) Affects Lipopolysaccharide-Induced Mechanical Compliance Changes and Beat Frequency of mESC-Derived Cardiomyocyte Monolayers. Shock 2022; 57:544-552. [PMID: 34416756 PMCID: PMC8906254 DOI: 10.1097/shk.0000000000001845] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/17/2021] [Accepted: 08/03/2021] [Indexed: 12/07/2022]
Abstract
BACKGROUND Septic cardiomyopathy increases mortality by 70% to 90% and results in mechanical dysfunction of cells. METHODS Here, we created a LPS-induced in-vitro sepsis model with mouse embryonic stem cell-derived cardiomyocytes (mESC-CM) using the CellDrum technology which simultaneously measures mechanical compliance and beat frequency of mESCs. Visualization of reactive oxygen species (ROS), actin stress fibers, and mRNA quantification of endothelial protein C receptor (EPCR) and protease-activated receptor 1 (PAR1) before/after LPS incubation were used for method validation. Since activated protein C (APC) has cardioprotective effects, samples were treated with human recombinant APC (rhAPC) with/-out LPS predamage to demonstrate the application in therapeutic studies. RESULTS Twelve hours LPS treatment (5 μg/mL) increased ROS and decreased actin stress fiber density and significantly downregulated EPCR and PAR1 compared to control samples (0.26, 0.39-fold respectively). rhAPC application (5 μg/mL, 12 h) decreased ROS and recovered actin density, EPCR, and PAR1 levels were significantly upregulated compared to LPS predamaged samples (4.79, 3.49-fold respectively). The beat frequencies were significantly decreased after 6- (86%) and 12 h (73%) of LPS application. Mechanical compliance of monolayers significantly increased in a time-dependent manner, up to eight times upon 12-h LPS incubation compared to controls. rhAPC incubation increased the beat frequency by 127% (6h-LPS) and 123% (12h-LPS) and decreased mechanical compliance by 68% (12h-LPS) compared to LPS predamaged samples. CONCLUSION LPS-induced contraction dysfunction and the reversal effects of rhAPC were successfully assessed by the mechanical properties of mESC-CMs. The CellDrum technology proved a decent tool to simulate sepsis in-vitro.
Collapse
Affiliation(s)
- Aysegül Temiz Artmann
- Institute for Bioengineering, University of Applied Sciences Aachen/Campus Juelich, Juelich, Germany
| | - Eylem Kurulgan Demirci
- Institute for Bioengineering, University of Applied Sciences Aachen/Campus Juelich, Juelich, Germany
- Department of Chemistry, Faculty of Science, Izmir Institute of Technology, Campus Gulbahce, URLA, Izmir, Turkey
| | - Ipek Seda Fırat
- Institute for Bioengineering, University of Applied Sciences Aachen/Campus Juelich, Juelich, Germany
| | - Hakan Oflaz
- Bioengineering Department, Faculty of Engineering, Gebze Technical University, Kocaeli, Turkey
| | - Gerhard M. Artmann
- Institute for Bioengineering, University of Applied Sciences Aachen/Campus Juelich, Juelich, Germany
| |
Collapse
|
80
|
Diab A, Valenzuela Ripoll C, Guo Z, Javaheri A. HDL Composition, Heart Failure, and Its Comorbidities. Front Cardiovasc Med 2022; 9:846990. [PMID: 35350538 PMCID: PMC8958020 DOI: 10.3389/fcvm.2022.846990] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/09/2022] [Indexed: 12/24/2022] Open
Abstract
Although research on high-density lipoprotein (HDL) has historically focused on atherosclerotic coronary disease, there exists untapped potential of HDL biology for the treatment of heart failure. Anti-oxidant, anti-inflammatory, and endothelial protective properties of HDL could impact heart failure pathogenesis. HDL-associated proteins such as apolipoprotein A-I and M may have significant therapeutic effects on the myocardium, in part by modulating signal transduction pathways and sphingosine-1-phosphate biology. Furthermore, because heart failure is a complex syndrome characterized by multiple comorbidities, there are complex interactions between heart failure, its comorbidities, and lipoprotein homeostatic mechanisms. In this review, we will discuss the effects of heart failure and associated comorbidities on HDL, explore potential cardioprotective properties of HDL, and review novel HDL therapeutic targets in heart failure.
Collapse
|
81
|
Septembre-Malaterre A, Boumendjel A, Seteyen ALS, Boina C, Gasque P, Guiraud P, Sélambarom J. Focus on the high therapeutic potentials of quercetin and its derivatives. PHYTOMEDICINE PLUS : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 2:100220. [PMID: 35403087 PMCID: PMC8759805 DOI: 10.1016/j.phyplu.2022.100220] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/10/2022] [Accepted: 01/13/2022] [Indexed: 04/15/2023]
Abstract
BACKGROUND Polyphenols and particularly flavonoids are of constant interest to the scientific community. Flavonoids are investigated for their biological and pharmacological purposes, notably as antioxidant, anticancer, antiviral and for their anti-inflammatory activities. Certainly, one of the best-known flavonols recognized for its therapeutic and preventive properties, is quercetin. Despite its biological interest, quercetin suffer from some drawbacks, mainly related to its bioavailability. Hence, its synthetic or biosynthetic derivatives have been the subject of intensive research. The health-promoting biological activities of flavonols and derivatives mainly arise from their capacity to disrupt the host-pathogen interactions and/or to regulate host cellular functions including oxidative processes and immunological responses. In the age of coronavirus pandemic, the anti-inflammatory and antiviral potential of flavonols should be put forward to explore these substances for decreasing the viral load and inflammatory storm caused by the infection. PURPOSE OF STUDY The present review will decipher and discuss the antioxidant, anti-inflammatory and antiviral capacities of major flavonol with a focus on the molecular basis and structure-activity relationships. STUDY DESIGN Current study used a combination of quercetin derivatives, pathway, antioxidant, anti-inflammatory, antiviral activities as keywords to retrieve the literature. This study critically reviewed the current literature and presented the ability of natural analogs of quercetin having superior antioxidant, anti-inflammatory and antiviral effects than the original molecule. RESULTS This review allowed the identification of relevant key structure-activity relationship elements and highlight approaches on the mechanisms governing the antioxidant, antiviral and anti-inflammatory activities. CONCLUSION Through a critical analysis of the literature, flavonols and more precisely quercetin derivatives reviewed and found to act simultaneously on inflammation, virus and oxidative stress, three key factors that may lead to life threatening diseases.
Collapse
Affiliation(s)
- Axelle Septembre-Malaterre
- Université de La Réunion, Unité de recherche Etudes Pharmaco-Immunologie (EPI), CHU La Réunion site Félix Guyon, Allée des Topazes, CS11021, 97400 Saint Denis de La Réunion, France
- Laboratoire d'immunologie clinique et expérimentale de la zone de l'océan indien (LICE-OI) CHU La Réunion site Félix Guyon, Allée des Topazes, CS11021, 97400 Saint Denis de La Réunion, France
| | | | - Anne-Laure Sandenon Seteyen
- Université de La Réunion, Unité de recherche Etudes Pharmaco-Immunologie (EPI), CHU La Réunion site Félix Guyon, Allée des Topazes, CS11021, 97400 Saint Denis de La Réunion, France
| | - Chailas Boina
- Université de La Réunion, Unité de recherche Etudes Pharmaco-Immunologie (EPI), CHU La Réunion site Félix Guyon, Allée des Topazes, CS11021, 97400 Saint Denis de La Réunion, France
- Laboratoire d'immunologie clinique et expérimentale de la zone de l'océan indien (LICE-OI) CHU La Réunion site Félix Guyon, Allée des Topazes, CS11021, 97400 Saint Denis de La Réunion, France
| | - Philippe Gasque
- Université de La Réunion, Unité de recherche Etudes Pharmaco-Immunologie (EPI), CHU La Réunion site Félix Guyon, Allée des Topazes, CS11021, 97400 Saint Denis de La Réunion, France
- Laboratoire d'immunologie clinique et expérimentale de la zone de l'océan indien (LICE-OI) CHU La Réunion site Félix Guyon, Allée des Topazes, CS11021, 97400 Saint Denis de La Réunion, France
| | - Pascale Guiraud
- Université de La Réunion, Unité de recherche Etudes Pharmaco-Immunologie (EPI), CHU La Réunion site Félix Guyon, Allée des Topazes, CS11021, 97400 Saint Denis de La Réunion, France
| | - Jimmy Sélambarom
- Université de La Réunion, Unité de recherche Etudes Pharmaco-Immunologie (EPI), CHU La Réunion site Félix Guyon, Allée des Topazes, CS11021, 97400 Saint Denis de La Réunion, France
| |
Collapse
|
82
|
|
83
|
Zeng X, Yu J, Zeng T, Liu Y, Li B. 3'-daidzein sulfonate protects myocardial cells from hypoxic-ischemic injury via the NRF2/HO-1 signaling pathway. J Thorac Dis 2022; 13:6897-6910. [PMID: 35070374 PMCID: PMC8743394 DOI: 10.21037/jtd-21-1909] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 12/17/2021] [Indexed: 12/11/2022]
Abstract
Background Myocardial infarction (MI) has a high mortality and disability rate and greatly affects human health. This study sought to explore the therapeutic effect and molecular mechanism of 3'-daidzein sulfonate (DSS) on MI. Methods A rat MI model was established and low and high doses of DSS were administered to the rats. An in vitro oxygen glucose deprivation model was used to verify the treatment role and mechanism of DSS. The establishment of the rat MI model was confirmed by electrocardiogram. The tissue changes were detected by HE, Masson’s trichrome, TUNEL and TTC staining. Cell viability was detected by CCK-8. The viable and dead cells were detected by Calcein-AM/PI. Apoptotic cells, ROS and JC-1 were detected by flow cytometry apoptosis. The level of proteins was detected by western blotting. MDA, SOD and GSH were detected by ELISA. Results The results of Hematoxylin and eosin, TUNEL, and Masson staining showed that the myocardial tissue of the MI group was repaired by DSS. The serum levels of cardiac troponin I (CTnI), lactate dehydrogenase (LDH), creatine kinase-MB (CK-MB), and malondialdehyde (MDA) were decreased by DSS, while the serum levels of superoxide dismutase and glutathione were promoted by DSS. The treatment of DSS activated the Nuclear Factor Erythroid 2-Related Factor 2 (NRF-2)/Heme Oxygenase 1 (HO-1) pathway and inhibited the caspase-3 apoptosis pathway. The in vitro experiment showed that DSS greatly restored cell viability and reduced cell apoptosis. DSS also greatly inhibited mitochondrial membrane potential depolarization, reactive oxygen species production, and oxidative stress. The application of the NRF-2 inhibitor, C29H25N3O4S (ML385), greatly inhibited the treatment role of DSS and the NRF-2/HO-1 pathway, and activated the caspase-3 apoptosis pathway. Conclusions In conclusion, this study first identified the beneficial role of DSS in MI. DSS protected myocardial cells by activating the NRF-2/HO-1 pathway and inhibiting cell apoptosis. DSS could be used as a novel drug in the treatment of MI.
Collapse
Affiliation(s)
- Xueliang Zeng
- Department of pharmacy, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Junjian Yu
- Cardiovascular and Thoracic Surgery Department 2, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Taohui Zeng
- Department of pharmacy, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Yuan Liu
- Department of pharmacy, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Bei Li
- Department of pharmacy, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| |
Collapse
|
84
|
Potential Implications of Rimonabant on Age-Related Oxidative Stress and Inflammation. Antioxidants (Basel) 2022; 11:antiox11010162. [PMID: 35052666 PMCID: PMC8773212 DOI: 10.3390/antiox11010162] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 02/01/2023] Open
Abstract
Over the last decades, growing interest has turned to preventive and therapeutic approaches for achieving successful aging. Oxidative stress and inflammation are fundamental features of cardiovascular diseases; therefore, potential targets of them can improve cardiac outcomes. Our study aimed to examine the involvement of the endocannabinoid system, especially the CB1 receptor blockade, on inflammatory and oxidant/antioxidant processes. Twenty-month-old female and male Wistar rats were divided into rimonabant-treated and aging control (untreated) groups. Rimonabant, a selective CB1 receptor antagonist, was administered at the dose of 1 mg/kg/day intraperitoneally for 2 weeks. Cardiac amounts of ROS, the antioxidant glutathione and superoxide dismutase (SOD), and the activity and concentration of the heme oxygenase (HO) enzyme were detected. Among inflammatory parameters, nuclear factor-kappa B (NF-κB), tumor necrosis factor-alpha (TNF-α), and myeloperoxidase (MPO) enzyme activity were measured. Two weeks of low dose rimonabant treatment significantly reduced the cardiac ROS via boosting of the antioxidant defense mechanisms as regards the HO system, and the SOD and glutathione content. Consistently, the age-related inflammatory response was alleviated. Rimonabant-treated animals showed significantly decreased NF-κB, TNF-α, and MPO levels. Our findings prove the beneficial involvement of CB1 receptor blocker rimonabant on inflammatory and oxidative damages to the aging heart.
Collapse
|
85
|
Effect of Combined Endurance Training and MitoQ on Cardiac Function and Serum Level of Antioxidants, NO, miR-126, and miR-27a in Hypertensive Individuals. BIOMED RESEARCH INTERNATIONAL 2022; 2022:8720661. [PMID: 35071600 PMCID: PMC8776465 DOI: 10.1155/2022/8720661] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 12/17/2021] [Indexed: 12/27/2022]
Abstract
Objectives Hypertension (HTN) is one of the most important risk factors for cardiovascular diseases. Despite advances in treatment and control of HTN, the prevalence of HTN is still increasing. MitoQ is a supplement that acts on mitochondria and attenuates reactive oxygen species (ROS), which plays an important role in cardiovascular health. miRNAs play an important role in the pathophysiology of HTN. We evaluated the effects of MitoQ supplementation and endurance training (ET), alone and in combination, on functional indices of the heart and serum levels of miR-126, miR-27a, antioxidants, and NO, in patients with HTN. Methods In a double-blind randomized clinical trial, 52 male participants (age 40-55 years) were randomly divided into four groups (n = 13) of placebo, MitoQ (20 mg/day, oral), ET (cycle ergometer, moderate intensity, 40-60% VO2 peak, heart rate 120-140 b/min, 45 min a day, three days/week for six weeks), and MitoQ+ET. Cardiac function indices were assessed by echocardiography before and after interventions. Results Systolic blood pressure (SBP) significantly decreased in all intervention groups (P < 0.001) while DBP (P < 0.01) and LV hypertrophy (P < 0.05) were significantly decreased only in the MitoQ+ET group. Serum levels of SOD, GPx, and NO and the level of miR-126 significantly increased in all treatment groups, while miR-27a reduced in the ET (P < 0.05) and MitoQ+ET (P < 0.01) groups. Conclusions Compared to MitoQ and ET alone, their combination has more prominent improving effects on cardiac health and amelioration of BP in the patients with HTN. These effects are through miR-126 and miR-27a modulation and ameliorating mitochondrial ROS production.
Collapse
|
86
|
Taha A, Ahmed S, Ahmed Mbbs R, Mohammed Md A. Type 2 Myocardial Infarction Related to Very Low Carbohydrate Ketogenic Diet. J Investig Med High Impact Case Rep 2022; 10:23247096221074879. [PMID: 35272529 PMCID: PMC8921742 DOI: 10.1177/23247096221074879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022] Open
Abstract
A 38-year-old male with no history of cardiac disease presented with chest pain typical of acute coronary syndrome. He assumed very-low-carbohydrate ketogenic diet (VLC-KD) 4 weeks prior to admission but no other lifestyle change. Workup showed dynamic ST-T changes on electrocardiogram, significantly elevated troponins, ketonuria, and mild rhabdomyolysis. Transthoracic echocardiogram revealed mild inferior wall hypokinesia and cardiac catheterization showed normal coronaries; hence, the diagnosis of type II myocardial infarction (MI) was established. Although the pathogenesis remains unclear, this temporal association between VLC-KD and type 2 MI raised some concerns about VLC-KD’s cardiovascular safety profile.
Collapse
Affiliation(s)
- Ahmed Taha
- School of Medicine, Indiana University, Indianapolis, IN, USA.,Deaconess Hospital, Evansville, IN, USA
| | - Selma Ahmed
- Philadelphia College of Osteopathic Medicine, Philadelphia, PA, USA
| | - Roaa Ahmed Mbbs
- School of Medicine, Ahfad University for Women, Omdurman, Sudan
| | | |
Collapse
|
87
|
Kim YE, Kim J. ROS-Scavenging Therapeutic Hydrogels for Modulation of the Inflammatory Response. ACS APPLIED MATERIALS & INTERFACES 2021; 14:23002-23021. [PMID: 34962774 DOI: 10.1021/acsami.1c18261] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Although reactive oxygen species (ROS) are essential for cellular processes, excessive ROS could be a major cause of various inflammatory diseases because of the oxidation of proteins, DNA, and membrane lipids. It has recently been suggested that the amount of ROS could thus be regulated to treat such physiological disorders. A ROS-scavenging hydrogel is a promising candidate for therapeutic applications because of its high biocompatibility, 3D matrix, and ability to be modified. Approaches to conferring antioxidant properties to normal hydrogels include embedding ROS-scavenging catalytic nanoparticles, modifying hydrogel polymer chains with ROS-adsorbing organic moieties, and incorporating ROS-labile linkers in polymer backbones. Such therapeutic hydrogels can be used for wound healing, cardiovascular diseases, bone repair, ocular diseases, and neurodegenerative disorders. ROS-scavenging hydrogels could eliminate oxidative stress, accelerate the regeneration process, and show synergetic effects with other drugs or therapeutic molecules. In this review, the mechanisms by which ROS are generated and scavenged in the body are outlined, and the effects of high levels of ROS and the resulting oxidative stress on inflammatory diseases are described. Next, the mechanism of ROS scavenging by hydrogels is explained depending on the ROS-scavenging agents embedded within the hydrogel. Lastly, the recent achievements in the development of ROS-scavenging hydrogels to treat various inflammation-associated diseases are presented.
Collapse
Affiliation(s)
- Ye Eun Kim
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Jaeyun Kim
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Institute of Quantum Biophysics (IQB), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| |
Collapse
|
88
|
Maiti BK, Maia LB, Moura JJG. Sulfide and transition metals - A partnership for life. J Inorg Biochem 2021; 227:111687. [PMID: 34953313 DOI: 10.1016/j.jinorgbio.2021.111687] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 11/24/2021] [Accepted: 11/28/2021] [Indexed: 12/13/2022]
Abstract
Sulfide and transition metals often came together in Biology. The variety of possible structural combinations enabled living organisms to evolve an array of highly versatile metal-sulfide centers to fulfill different physiological roles. The ubiquitous iron‑sulfur centers, with their structural, redox, and functional diversity, are certainly the best-known partners, but other metal-sulfide centers, involving copper, nickel, molybdenum or tungsten, are equally crucial for Life. This review provides a concise overview of the exclusive sulfide properties as a metal ligand, with emphasis on the structural aspects and biosynthesis. Sulfide as catalyst and as a substrate is discussed. Different enzymes are considered, including xanthine oxidase, formate dehydrogenases, nitrogenases and carbon monoxide dehydrogenases. The sulfide effect on the activity and function of iron‑sulfur, heme and zinc proteins is also addressed.
Collapse
Affiliation(s)
- Biplab K Maiti
- National Institute of Technology Sikkim, Department of Chemistry, Ravangla Campus, Barfung Block, Ravangla Sub Division, South Sikkim 737139, India.
| | - Luisa B Maia
- LAQV, REQUIMTE, Department of Chemistry, NOVA School of Science and Technology (FCT NOVA), Universidade NOVA de Lisboa, Campus de Caparica, Portugal.
| | - José J G Moura
- LAQV, REQUIMTE, Department of Chemistry, NOVA School of Science and Technology (FCT NOVA), Universidade NOVA de Lisboa, Campus de Caparica, Portugal.
| |
Collapse
|
89
|
Perveen S, Rossin D, Vitale E, Rosso R, Vanni R, Cristallini C, Rastaldo R, Giachino C. Therapeutic Acellular Scaffolds for Limiting Left Ventricular Remodelling-Current Status and Future Directions. Int J Mol Sci 2021; 22:ijms222313054. [PMID: 34884856 PMCID: PMC8658014 DOI: 10.3390/ijms222313054] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/26/2021] [Accepted: 11/29/2021] [Indexed: 12/14/2022] Open
Abstract
Myocardial infarction (MI) is one of the leading causes of heart-related deaths worldwide. Following MI, the hypoxic microenvironment triggers apoptosis, disrupts the extracellular matrix and forms a non-functional scar that leads towards adverse left ventricular (LV) remodelling. If left untreated this eventually leads to heart failure. Besides extensive advancement in medical therapy, complete functional recovery is never accomplished, as the heart possesses limited regenerative ability. In recent decades, the focus has shifted towards tissue engineering and regenerative strategies that provide an attractive option to improve cardiac regeneration, limit adverse LV remodelling and restore function in an infarcted heart. Acellular scaffolds possess attractive features that have made them a promising therapeutic candidate. Their application in infarcted areas has been shown to improve LV remodelling and enhance functional recovery in post-MI hearts. This review will summarise the updates on acellular scaffolds developed and tested in pre-clinical and clinical scenarios in the past five years with a focus on their ability to overcome damage caused by MI. It will also describe how acellular scaffolds alone or in combination with biomolecules have been employed for MI treatment. A better understanding of acellular scaffolds potentialities may guide the development of customised and optimised therapeutic strategies for MI treatment.
Collapse
Affiliation(s)
- Sadia Perveen
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (S.P.); (D.R.); (E.V.); (R.R.); (R.V.); (C.G.)
| | - Daniela Rossin
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (S.P.); (D.R.); (E.V.); (R.R.); (R.V.); (C.G.)
| | - Emanuela Vitale
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (S.P.); (D.R.); (E.V.); (R.R.); (R.V.); (C.G.)
| | - Rachele Rosso
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (S.P.); (D.R.); (E.V.); (R.R.); (R.V.); (C.G.)
| | - Roberto Vanni
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (S.P.); (D.R.); (E.V.); (R.R.); (R.V.); (C.G.)
| | | | - Raffaella Rastaldo
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (S.P.); (D.R.); (E.V.); (R.R.); (R.V.); (C.G.)
- Correspondence:
| | - Claudia Giachino
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (S.P.); (D.R.); (E.V.); (R.R.); (R.V.); (C.G.)
| |
Collapse
|
90
|
Syed Abd Halim SA, Abd Ghafar N, Das S, Zainalabidin S, Jubri Z. Cardioprotective effects of Gynura procumbens extract on oxidative status and myocardial injury in rats with isoproterenol-induced myocardial infarction. INTERNATIONAL FOOD RESEARCH JOURNAL 2021; 28:1223-1232. [DOI: 10.47836/ifrj.28.6.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Gynura procumbens (GP) grows abundantly in Southeast Asia. The present work was conducted to investigate the cardioprotective potential of ethanol extract of GP on cardiac markers, antioxidant levels, and histopathology of isoproterenol-induced myocardial infarction (MI). A total of 36 adult Sprague Dawley rats were randomly divided into six groups. Treatments were given via oral gavage for 28 days: two groups were given normal saline 0.9%; two groups were given GP250 mg/kg/day; and two groups were given GP500 mg/kg/day. On day 27 and 28, MI was induced with a subcutaneous injection of 85 mg/kg isoproterenol. The rats were sacrificed 48 h after the 1st injection. Cardiac markers, lipid peroxidation, oxidative status, and histopathological analyses were evaluated. Isoproterenol significantly increased the levels of troponin T, creatine kinase MB isoenzyme (CKMB), lactate dehydrogenase (LDH), and malondialdehyde (MDA), whereas the level of superoxide dismutase (SOD), catalase, and glutathione peroxidase were significantly decreased. In addition, the histopathological findings showed a necrosis of the myocardium as evidenced by neutrophil infiltration, and interstitial oedema with acceleration of apoptosis in MI. Interestingly, treatment with GP restored the levels of troponin T, LDH, MDA, SOD, and catalase significantly. Moreover, GP preserved the myocardial architecture while decreasing both necrosis and apoptosis. GP has the potential to limit myocardial injury after MI, and this is most likely achieved through its modulation of antioxidant enzyme activities.
Collapse
|
91
|
Alfhili MA, Aljuraiban GS. Lauric Acid, a Dietary Saturated Medium-Chain Fatty Acid, Elicits Calcium-Dependent Eryptosis. Cells 2021; 10:cells10123388. [PMID: 34943896 PMCID: PMC8699421 DOI: 10.3390/cells10123388] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/20/2021] [Accepted: 11/28/2021] [Indexed: 12/18/2022] Open
Abstract
Cardiovascular diseases (CVD) are a leading cause of mortality worldwide, and dietary habits represent a major risk factor for dyslipidemia; a hallmark of CVD. Saturated fatty acids contribute to CVD by aggravating dyslipidemia, and, in particular, lauric acid (LA) raises circulating cholesterol levels. The role of red blood cells (RBCs) in CVD is increasingly being appreciated, and eryptosis has recently been identified as a novel mechanism in CVD. However, the effect of LA on RBC physiology has not been thoroughly investigated. RBCs were isolated from heparin-anticoagulated whole blood (WB) and exposed to 50-250 μM of LA for 24 h at 37 °C. Hemoglobin was photometrically examined as an indicator of hemolysis, whereas eryptosis was assessed by Annexin V-FITC for phosphatidylserine (PS) exposure, Fluo4/AM for Ca2+, light scatter for cellular morphology, H2DCFDA for oxidative stress, and BODIPY 581/591 C11 for lipid peroxidation. WB was also examined for RBC, leukocyte, and platelet viability and indices. LA caused dose-responsive hemolysis, and Ca2+-dependent PS exposure, elevated erythrocyte sedimentation rate (ESR), cytosolic Ca2+ overload, cell shrinkage and granularity, oxidative stress, accumulation of lipid peroxides, and stimulation of casein kinase 1α (CK1α). In WB, LA disrupted leukocyte distribution with elevated neutrophil-lymphocyte ratio (NLR) due to selective toxicity to lymphocytes. In conclusion, this report provides the first evidence of the pro-eryptotic potential of LA and associated mechanisms, which informs dietary interventions aimed at CVD prevention and management.
Collapse
Affiliation(s)
- Mohammad A. Alfhili
- Chair of Medical and Molecular Genetics Research, Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 12372, Saudi Arabia
- Correspondence: ; Tel.: +966-504-262-597
| | - Ghadeer S. Aljuraiban
- Department of Community Health Sciences, King Saud University, Riyadh 12372, Saudi Arabia;
| |
Collapse
|
92
|
Alotaibi BS, Ijaz M, Buabeid M, Kharaba ZJ, Yaseen HS, Murtaza G. Therapeutic Effects and Safe Uses of Plant-Derived Polyphenolic Compounds in Cardiovascular Diseases: A Review. Drug Des Devel Ther 2021; 15:4713-4732. [PMID: 34848944 PMCID: PMC8619826 DOI: 10.2147/dddt.s327238] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/12/2021] [Indexed: 12/29/2022] Open
Abstract
Polyphenols have long been recognized as health-promoting entities, including beneficial effects on cardiovascular disease, but their reputation has been boosted recently following a number of encouraging clinical studies in multiple chronic pathologies, that seem to validate efficacy. Health benefits of polyphenols have been linked to their well-established powerful antioxidant activity. This review aims to provide comprehensive and up-to-date knowledge on the current therapeutic status of polyphenols having sufficient heed towards the treatment of cardiovascular diseases. Furthermore, data about the safety profile of highly efficacious polyphenols has also been investigated to further enhance their role in cardiac abnormalities. Evidence is presented to support the action of phenolic derivatives against cardiovascular pathologies by following receptors and signaling pathways which ultimately cause changes in endogenous antioxidant, antiplatelet, vasodilatory, and anti-inflammatory activities. In addition, in vitro antioxidant and pre-clinical and clinical experiments on anti-inflammatory as well as immunomodulatory attributes of polyphenols have revealed their role as cardioprotective agents. However, an obvious shortage of in vivo studies related to dose selection and toxicity of polyphenols makes these compounds a suitable target for clinical investigations. Further studies are needed for the development of safe and potent herbal products against cardiovascular diseases. The novelty of this review is to provide comprehensive knowledge on polyphenols safety and their health claims. It will help researchers to identify those moieties which likely exert protective and therapeutic effects towards cardiovascular diseases.
Collapse
Affiliation(s)
- Badriyah Shadid Alotaibi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Munazza Ijaz
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan
| | - Manal Buabeid
- Medical and Bio-Allied Health Sciences Research Centre, Ajman University, Ajman, United Arab Emirates
- Department of Clinical Sciences, Ajman University, Ajman, 346, United Arab Emirates
| | - Zelal Jaber Kharaba
- Department of Clinical Sciences, College of Pharmacy, Al-Ain University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Hafiza Sidra Yaseen
- Department of Pharmacy, COMSATS University Islamabad, Lahore Campus, Lahore, 54000, Pakistan
| | - Ghulam Murtaza
- Department of Pharmacy, COMSATS University Islamabad, Lahore Campus, Lahore, 54000, Pakistan
| |
Collapse
|
93
|
Shafiei-Irannejad V, Abbaszadeh S, Janssen PML, Soraya H. Memantine and its benefits for cancer, cardiovascular and neurological disorders. Eur J Pharmacol 2021; 910:174455. [PMID: 34461125 DOI: 10.1016/j.ejphar.2021.174455] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 08/19/2021] [Accepted: 08/26/2021] [Indexed: 01/10/2023]
Abstract
Memantine is a non-competitive N-methyl-D-aspartate (NMDA) receptor antagonist that was initially indicated for the treatment of moderate to severe Alzheimer's disease. It is now also considered for a variety of other pathologies in which activation of NMDA receptors apparently contributes to the pathogenesis and progression of disease. In addition to the central nervous system (CNS), NMDA receptors can be found in non-neuronal cells and tissues that recently have become an interesting research focus. Some studies have shown that glutamate signaling plays a role in cell transformation and cancer progression. In addition, these receptors may play a role in cardiovascular disorders. In this review, we focus on the most recent findings for memantine with respect to its pharmacological effects in a range of diseases, including inflammatory disorders, cardiovascular diseases, cancer, neuropathy, as well as retinopathy.
Collapse
Affiliation(s)
- Vahid Shafiei-Irannejad
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Samin Abbaszadeh
- Department of Pharmacology, Faculty of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Paul M L Janssen
- Department of Physiology and Cell Biology, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Hamid Soraya
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran; Department of Pharmacology, Faculty of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
94
|
Wagner KH, Khoei NS, Hana CA, Doberer D, Marculescu R, Bulmer AC, Hörmann-Wallner M, Mölzer C. Oxidative Stress and Related Biomarkers in Gilbert's Syndrome: A Secondary Analysis of Two Case-Control Studies. Antioxidants (Basel) 2021; 10:antiox10091474. [PMID: 34573106 PMCID: PMC8472792 DOI: 10.3390/antiox10091474] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/09/2021] [Accepted: 09/12/2021] [Indexed: 12/12/2022] Open
Abstract
Bilirubin is an important antioxidant and a modulator of biological functions. However, most of the protection against oxidative stress was shown in vitro or ex vivo. The aim of this case-control study was to investigate whether subjects with Gilbert’s syndrome (GS) experience different levels of lipid and protein oxidation (as well as differences in oxidative stress related markers) compared to healthy controls. GS subjects (n = 119) demonstrated higher serum levels of unconjugated bilirubin (p < 0.001), a lower BMI (p < 0.001), 37% higher antioxidant potential assessed as ferric reducing ability potential (p < 0.001), higher advanced oxidation protein products (p < 0.01) andlower apolipoprotein B (p < 0.05), hs-C-reactive protein (p < 0.05), interleukin 6 (p < 0.001) and interleukin 1 beta (p < 0.05) values compared to healthy controls (n = 119). Furthermore, the resting heart rate was significantly lower in the GS group (p < 0.05). Stronger protective effects for GS subjects were demonstrated in the older subgroup (n = 104, average age 50 years) compared to those of the younger group (n = 134, average age 27 years). Although not all markers related to oxidative stress were different between the groups (e.g., malondialdehyde, homocysteine, oxLDL, and myeloperoxidase; p > 0.05), the observed differences contribute to the explanation of why GS serves as an important protector in the pathogenesis of metabolic, oxidative stress related diseases.
Collapse
Affiliation(s)
- Karl-Heinz Wagner
- Department of Nutritional Sciences, Faculty of Life Sciences, University of Vienna, 1090 Vienna, Austria;
- Research Platform Active Ageing, University of Vienna, 1090 Vienna, Austria;
- Correspondence: ; Tel.: +43-1-4277 (ext. 54930)
| | | | - Claudia Anna Hana
- Department of Nutritional Sciences, Faculty of Life Sciences, University of Vienna, 1090 Vienna, Austria;
| | - Daniel Doberer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna General Hospital, 1090 Vienna, Austria;
| | - Rodrig Marculescu
- Clinical Institute of Laboratory Medicine, Medical University of Vienna, Vienna General Hospital, 1090 Vienna, Austria;
| | - Andrew Cameron Bulmer
- Menzies Health Institute Queensland, School of Medical Science, Griffith University, Brisbane, QLD 4222, Australia;
| | - Marlies Hörmann-Wallner
- Institute for Dietetics and Nutrition, University of Applied Sciences FH JOANNEUM, 8020 Graz, Austria;
| | - Christine Mölzer
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK;
| |
Collapse
|
95
|
Ajmal M, Ajmal A, Huang L, Zeng L. The Potential Therapeutic Role of Celastrol in Patients With Heart Failure With Preserved Ejection Fraction. Front Cardiovasc Med 2021; 8:725602. [PMID: 34490381 PMCID: PMC8418197 DOI: 10.3389/fcvm.2021.725602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 07/27/2021] [Indexed: 12/07/2022] Open
Abstract
Analysis of left ventricular systolic dysfunction remained at the centre of heart failure research for many years (also known as heart failure with reduced ejection fraction, HFrEF). Although more than 50% of all heart failure patients experience a form of heart failure characterised by preserved ejection fraction (HFpEF), the pathophysiological mechanisms leading to this form of heart failure remain not well-understood. Several evidence-based treatments for HFrEF are in routine use, but there are limited evidence-based therapies for HFpEF. The effects of these remain controversial, with current treatment options being limited to managing the associated symptoms and conditions. Accumulating evidence demonstrates that pro-inflammatory and oxidative stress pathways play key roles in the development and progression of HFpEF, such as the Unfolded Protein Response (UPR) and inducible nitric oxide synthase. Celastrol, derived from medicinal plants, is a bioactive compound with strong anti-inflammatory properties, which could deem it as fruitful in overcoming the effects of such dysregulated UPR. This literature review therefore focuses on Celastrol's anti-inflammatory and antioxidant activities, alongside its other potential therapeutic activities, and its ability to impede the pathways that are thought to be involved in the development of HFpEF, such as the JAK2/STAT pathway, to elucidate the potential therapeutic role of this bioactive compound, in the treatment of HFpEF.
Collapse
Affiliation(s)
- Maryam Ajmal
- GKT School of Medical Education, Faculty of Life Science and Medicine, King's College London, London, United Kingdom
| | - Aisha Ajmal
- St. George's Hospital Medical School, University of London, London, United Kingdom
| | - Lei Huang
- Department of Heart Center, Tianjin Third Central Hospital, Tianjin, China.,Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin Third Central Hospital, Tianjin, China
| | - Lingfang Zeng
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Excellence, Faculty of Life Science and Medicine, King's College London, London, United Kingdom
| |
Collapse
|
96
|
Raj P, Sayfee K, Parikh M, Yu L, Wigle J, Netticadan T, Zieroth S. Comparative and Combinatorial Effects of Resveratrol and Sacubitril/Valsartan alongside Valsartan on Cardiac Remodeling and Dysfunction in MI-Induced Rats. Molecules 2021; 26:5006. [PMID: 34443591 PMCID: PMC8401506 DOI: 10.3390/molecules26165006] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/05/2021] [Accepted: 08/09/2021] [Indexed: 12/11/2022] Open
Abstract
The development and progression of heart failure (HF) due to myocardial infarction (MI) is a major concern even with current optimal therapy. Resveratrol is a plant polyphenol with cardioprotective properties. Sacubitril/valsartan is known to be beneficial in chronic HF patients. In this study, we investigated the comparative and combinatorial benefits of resveratrol with sacubitril/valsartan alongside an active comparator valsartan in MI-induced male Sprague Dawley rats. MI-induced and sham-operated animals received vehicle, resveratrol, sacubitril/valsartan, valsartan alone or sacubitril/valsartan + resveratrol for 8 weeks. Echocardiography was performed at the endpoint to assess cardiac structure and function. Cardiac oxidative stress, inflammation, fibrosis, brain natriuretic peptide (BNP), creatinine and neutrophil gelatinase associated lipocalin were measured. Treatment with resveratrol, sacubitril/valsartan, valsartan and sacubitril/valsartan + resveratrol significantly prevented left ventricular (LV) dilatation and improved LV ejection fraction in MI-induced rats. All treatments also significantly reduced myocardial tissue oxidative stress, inflammation and fibrosis, as well as BNP. Treatment with the combination of sacubitril/valsartan and resveratrol did not show additive effects. In conclusion, resveratrol, sacubitril/valsartan, and valsartan significantly prevented cardiac remodeling and dysfunction in MI-induced rats. The reduction in cardiac remodeling and dysfunction in MI-induced rats was mediated by a reduction in cardiac oxidative stress, inflammation and fibrosis.
Collapse
Affiliation(s)
- Pema Raj
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada; (P.R.); (M.P.)
- Canadian Centre for Agri-Food Research in Health and Medicine, Winnipeg, MB R2H 2A6, Canada; (K.S.); (L.Y.)
- Agriculture and Agri-Food Canada, Winnipeg, MB R3T 2M9, Canada
| | - Karen Sayfee
- Canadian Centre for Agri-Food Research in Health and Medicine, Winnipeg, MB R2H 2A6, Canada; (K.S.); (L.Y.)
| | - Mihir Parikh
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada; (P.R.); (M.P.)
| | - Liping Yu
- Canadian Centre for Agri-Food Research in Health and Medicine, Winnipeg, MB R2H 2A6, Canada; (K.S.); (L.Y.)
| | - Jeffrey Wigle
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB R3E 0J9, Canada;
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
| | - Thomas Netticadan
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada; (P.R.); (M.P.)
- Canadian Centre for Agri-Food Research in Health and Medicine, Winnipeg, MB R2H 2A6, Canada; (K.S.); (L.Y.)
- Agriculture and Agri-Food Canada, Winnipeg, MB R3T 2M9, Canada
| | - Shelley Zieroth
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada; (P.R.); (M.P.)
- Section of Cardiology, Department of Medicine, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
| |
Collapse
|
97
|
Wu Z, Jankowski V, Jankowski J. Irreversible post-translational modifications - Emerging cardiovascular risk factors. Mol Aspects Med 2021; 86:101010. [PMID: 34404548 DOI: 10.1016/j.mam.2021.101010] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/19/2021] [Accepted: 08/12/2021] [Indexed: 12/23/2022]
Abstract
Despite the introduction of lipid-lowering drugs, antihypertensives, antiplatelet and anticoagulation therapies for primary prevention of cardiovascular and heart diseases (CVD), it remains the number one cause of death globally, raising the question for novel/further essential factors besides traditional risk factors such as cholesterol, blood pressure and coagulation. With continuous identification and characterization of non-enzymatic post-translationally modified isoforms of proteins and lipoproteins, it is becoming increasingly clear that irreversible non-enzymatic post-translational modifications (nPTMs) alter the biological functions of native proteins and lipoproteins thereby transforming innate serum components into CVD mediators. In particular renal insufficiency and metabolic imbalance are major contributors to the systemically increased concentration of reactive metabolites and thus increased frequency of nPTMs, promoting multi-morbid disease development centering around cardiovascular disease. nPTMs are significantly involved in the onset and progression of cardiovascular disease and represent a significant and novel risk factor. These insights represent potentially new avenues for risk assessment, prevention and therapy. This review chapter summarizes all forms of nPTMs found in CKD and under metabolic imbalance and discusses the biochemical connections between molecular alterations and the pathological impact on increased cardiovascular risk, novel nPTM-associated non-traditional cardiovascular risk factors, and clinical implication of nPTM in cardiovascular disease.
Collapse
Affiliation(s)
- Zhuojun Wu
- Institute for Molecular Cardiovascular Research, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Vera Jankowski
- Institute for Molecular Cardiovascular Research, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany; Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Universiteitssingel 50, Maastricht, the Netherlands.
| |
Collapse
|
98
|
Kwon IH, Kim IY, Heo MB, Park JW, Lee SW, Lee TG. Real-time heart rate monitoring system for cardiotoxicity assessment of Daphnia magna using high-speed digital holographic microscopy. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 780:146405. [PMID: 33774290 DOI: 10.1016/j.scitotenv.2021.146405] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/22/2021] [Accepted: 03/06/2021] [Indexed: 06/12/2023]
Abstract
Machine vision techniques for monitoring heart rates in aquatic bioassays have been applied to cardiotoxicity assessment. However, the requisite large data sizes and long calculation times make long-term observations of heart rates difficult. In this study, we developed a real-time heart rate monitoring system for individual Daphnia magna in a water chamber mounter that immobilizes their movement in 100 mL media. Heart rates are calculated from real-time, time-resolved relative phase information from digital holograms acquired with a 200 fps camera and parallel computation using a graphics processing unit. With this system, we monitored the real-time changes in the heart rates of individual D. magna specimens exposed to H2O2 as a positive control for reactive oxygen species (ROS) levels in an aquatic environment for 10 h, a period long enough to observe dynamic heart rate responses to the mounting process and exposure and to establish heart rate trends. An additional group analysis was conducted to compare to conventional cardiotoxicity assessment, with results of both assessments showing that the heart rates reduced according to ROS level by H2O2 exposure concentration. Notably, the results of our real-time dynamic heart rate monitoring in aquatic conditions indicated that establishing a relaxation heart rate before measurements could improve the accuracy of toxicity assessment. It is believed that the system developed in this study, achieving the simultaneous measurement, analysis, and display of reconstructed results, will find wide application in other aquatic bioassays.
Collapse
Affiliation(s)
- Ik Hwan Kwon
- Safety Measurement Institute, Korea Research Institute of Standards and Science, Daejeon 34113, Republic of Korea
| | - In Young Kim
- Safety Measurement Institute, Korea Research Institute of Standards and Science, Daejeon 34113, Republic of Korea
| | - Min Beom Heo
- Safety Measurement Institute, Korea Research Institute of Standards and Science, Daejeon 34113, Republic of Korea
| | - June-Woo Park
- Environmental Biology Research Group, Korea Institute of Toxicology, 17 Jegok-gil, Jinju 52834, Republic of Korea; Human and Environmental Toxicology Program, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Sang-Won Lee
- Safety Measurement Institute, Korea Research Institute of Standards and Science, Daejeon 34113, Republic of Korea; Department of Medical Physics, University of Science and Technology, Daejeon 34113, Republic of Korea.
| | - Tae Geol Lee
- Safety Measurement Institute, Korea Research Institute of Standards and Science, Daejeon 34113, Republic of Korea; Department of Nano Science, University of Science and Technology, Daejeon 34113, Republic of Korea.
| |
Collapse
|
99
|
Matuz-Mares D, Riveros-Rosas H, Vilchis-Landeros MM, Vázquez-Meza H. Glutathione Participation in the Prevention of Cardiovascular Diseases. Antioxidants (Basel) 2021; 10:1220. [PMID: 34439468 PMCID: PMC8389000 DOI: 10.3390/antiox10081220] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/18/2021] [Accepted: 07/23/2021] [Indexed: 01/31/2023] Open
Abstract
Cardiovascular diseases (CVD) (such as occlusion of the coronary arteries, hypertensive heart diseases and strokes) are diseases that generate thousands of patients with a high mortality rate worldwide. Many of these cardiovascular pathologies, during their development, generate a state of oxidative stress that leads to a deterioration in the patient's conditions associated with the generation of reactive oxygen species (ROS) and reactive nitrogen species (RNS). Within these reactive species we find superoxide anion (O2•-), hydroxyl radical (•OH), nitric oxide (NO•), as well as other species of non-free radicals such as hydrogen peroxide (H2O2), hypochlorous acid (HClO) and peroxynitrite (ONOO-). A molecule that actively participates in counteracting the oxidizing effect of reactive species is reduced glutathione (GSH), a tripeptide that is present in all tissues and that its synthesis and/or regeneration is very important to be able to respond to the increase in oxidizing agents. In this review, we will address the role of glutathione, its synthesis in both the heart and the liver, and its importance in preventing or reducing deleterious ROS effects in cardiovascular diseases.
Collapse
Affiliation(s)
| | | | - María Magdalena Vilchis-Landeros
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Avenida Universidad 3000, Cd. Universitaria, Coyoacán, Ciudad de México 04510, Mexico; (D.M.-M.); (H.R.-R.)
| | - Héctor Vázquez-Meza
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Avenida Universidad 3000, Cd. Universitaria, Coyoacán, Ciudad de México 04510, Mexico; (D.M.-M.); (H.R.-R.)
| |
Collapse
|
100
|
Hsu PY, Mammadova A, Benkirane-Jessel N, Désaubry L, Nebigil CG. Updates on Anticancer Therapy-Mediated Vascular Toxicity and New Horizons in Therapeutic Strategies. Front Cardiovasc Med 2021; 8:694711. [PMID: 34386529 PMCID: PMC8353082 DOI: 10.3389/fcvm.2021.694711] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 06/18/2021] [Indexed: 12/11/2022] Open
Abstract
Vascular toxicity is a frequent adverse effect of current anticancer chemotherapies and often results from endothelial dysfunction. Vascular endothelial growth factor inhibitors (VEGFi), anthracyclines, plant alkaloids, alkylating agents, antimetabolites, and radiation therapy evoke vascular toxicity. These anticancer treatments not only affect tumor vascularization in a beneficial manner, they also damage ECs in the heart. Cardiac ECs have a vital role in cardiovascular functions including hemostasis, inflammatory and coagulation responses, vasculogenesis, and angiogenesis. EC damage can be resulted from capturing angiogenic factors, inhibiting EC proliferation, survival and signal transduction, or altering vascular tone. EC dysfunction accounts for the pathogenesis of myocardial infarction, atherothrombosis, microangiopathies, and hypertension. In this review, we provide a comprehensive overview of the effects of chemotherapeutic agents on vascular toxicity leading to hypertension, microvascular rarefaction thrombosis and atherosclerosis, and affecting drug delivery. We also describe the potential therapeutic approaches such as vascular endothelial growth factor (VEGF)-B and prokineticin receptor-1 agonists to maintain endothelial function during or following treatments with chemotherapeutic agents, without affecting anti-tumor effectiveness.
Collapse
Affiliation(s)
| | | | | | | | - Canan G. Nebigil
- INSERM UMR 1260, Regenerative Nanomedicine, University of Strasbourg, FMTS (Fédération de Médecine Translationnelle de l'Université de Strasbourg), Strasbourg, France
| |
Collapse
|