1
|
Hedayati N, Yaghoobi A, Salami M, Gholinezhad Y, Aghadavood F, Eshraghi R, Aarabi MH, Homayoonfal M, Asemi Z, Mirzaei H, Hajijafari M, Mafi A, Rezaee M. Impact of polyphenols on heart failure and cardiac hypertrophy: clinical effects and molecular mechanisms. Front Cardiovasc Med 2023; 10:1174816. [PMID: 37293283 PMCID: PMC10244790 DOI: 10.3389/fcvm.2023.1174816] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 05/02/2023] [Indexed: 06/10/2023] Open
Abstract
Polyphenols are abundant in regular diets and possess antioxidant, anti-inflammatory, anti-cancer, neuroprotective, and cardioprotective effects. Regarding the inadequacy of the current treatments in preventing cardiac remodeling following cardiovascular diseases, attention has been focused on improving cardiac function with potential alternatives such as polyphenols. The following online databases were searched for relevant orginial published from 2000 to 2023: EMBASE, MEDLINE, and Web of Science databases. The search strategy aimed to assess the effects of polyphenols on heart failure and keywords were "heart failure" and "polyphenols" and "cardiac hypertrophy" and "molecular mechanisms". Our results indicated polyphenols are repeatedly indicated to regulate various heart failure-related vital molecules and signaling pathways, such as inactivating fibrotic and hypertrophic factors, preventing mitochondrial dysfunction and free radical production, the underlying causes of apoptosis, and also improving lipid profile and cellular metabolism. In the current study, we aimed to review the most recent literature and investigations on the underlying mechanism of actions of different polyphenols subclasses in cardiac hypertrophy and heart failure to provide deep insight into novel mechanistic treatments and direct future studies in this context. Moreover, due to polyphenols' low bioavailability from conventional oral and intravenous administration routes, in this study, we have also investigated the currently accessible nano-drug delivery methods to optimize the treatment outcomes by providing sufficient drug delivery, targeted therapy, and less off-target effects, as desired by precision medicine standards.
Collapse
Affiliation(s)
- Neda Hedayati
- School of Medicine, Iran University of Medical Science, Tehran, Iran
| | - Alireza Yaghoobi
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Marziyeh Salami
- Department of Clinical Biochemistry, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Yasaman Gholinezhad
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farnaz Aghadavood
- Student Research Committee, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Reza Eshraghi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad-Hossein Aarabi
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mina Homayoonfal
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Hajijafari
- Department of Anesthesiology, School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Alireza Mafi
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
- Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Malihe Rezaee
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Harwansh RK, Yadav P, Deshmukh R. Current Insight into Novel Delivery Approaches of Resveratrol for Improving Therapeutic Efficacy and Bioavailability with its Clinical Updates. Curr Pharm Des 2023; 29:2921-2939. [PMID: 38053352 DOI: 10.2174/0113816128282713231129094715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 10/22/2023] [Accepted: 11/02/2023] [Indexed: 12/07/2023]
Abstract
Resveratrol (RSV) is a polyphenolic phytoalexin, and belongs to the stilbene family. RSV has several therapeutic activities such as cardioprotective, anticancer, and antioxidant. Apart from its therapeutic benefits, its pharmacological uses are limited due to low solubility, poor bioavailability, and short biological halflife. A researcher continuously focuses on overcoming the limitations of RSV through nanotechnology platforms to get the optimum health benefits. In this context, nanocarriers are pioneering to overcome these drawbacks. Nanocarriers possess high drug loading capacity, thermal stability, low production cost, longer shelflife, etc. Fortunately, scientists were proficient in delivering resveratrol-based nanocarriers in the present scenario. Nanocarriers can deliver drugs to the target sites without compromising the bioavailability. Thus, this review highlights how the latest nanocarrier systems overcome the shortcomings of RSV, which will be good for improving therapeutic efficacy and bioavailability. Moreover, recent updates on resveratrol-based novel formulations and their clinical trials have been addressed to manage several health-related problems.
Collapse
Affiliation(s)
- Ranjit K Harwansh
- Institute of Pharmaceutical Research, GLA University, Mathura 281406, India
| | - Paras Yadav
- Institute of Pharmaceutical Research, GLA University, Mathura 281406, India
| | - Rohitas Deshmukh
- Institute of Pharmaceutical Research, GLA University, Mathura 281406, India
| |
Collapse
|
3
|
Fan D, Liu C, Zhang Z, Huang K, Wang T, Chen S, Li Z. Progress in the Preclinical and Clinical Study of Resveratrol for Vascular Metabolic Disease. Molecules 2022; 27:7524. [PMID: 36364370 PMCID: PMC9658204 DOI: 10.3390/molecules27217524] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 10/21/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022] Open
Abstract
Vascular metabolic dysfunction presents in various diseases, such as atherosclerosis, hypertension, and diabetes mellitus. Due to the high prevalence of these diseases, it is important to explore treatment strategies to protect vascular function. Resveratrol (RSV), a natural polyphenolic phytochemical, is regarded as an agent to regulate metabolic pathways. Many studies have proven that RSV has beneficial effects on improving metabolism in endothelial cells (ECs) and vascular smooth muscle cells (VSMCs), which provide new directions to treat vascular metabolic diseases. Herein, we overviewed that RSV could regulate cell metabolism activity by inhibiting glucose uptake, suppressing glycolysis, preventing cells from fatty acid-related damages, reducing lipogenesis, increasing fatty acid oxidation, enhancing lipolysis, elevating uptake and synthesis of glutamine, and increasing NO release. Furthermore, in clinical trials, although the results from different studies remain controversial, we proposed that RSV had better therapeutic effects at high concentrations and for patients with metabolic disorders.
Collapse
Affiliation(s)
- Dongxiao Fan
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Chenshu Liu
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Zhongyu Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Kan Huang
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Tengyao Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Sifan Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Zilun Li
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| |
Collapse
|
4
|
Liu H, Fu D, Luo Y, Peng D. Independent association of Lp(a) with platelet reactivity in subjects without statins or antiplatelet agents. Sci Rep 2022; 12:16609. [PMID: 36198899 PMCID: PMC9534895 DOI: 10.1038/s41598-022-21121-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 09/22/2022] [Indexed: 11/09/2022] Open
Abstract
The physiological effect of Lp(a) on platelet activity is unclear. Previous studies explored the relationship between Lp(a) and platelet aggregation in patients taking statins and antiplatelet agents, but few was conducted in individuals without the bias of those drugs that either influence Lp(a) or platelet activity. The aim of this study was to assess the relationship between Lp(a) levels and platelet aggregation in subjects not taking statins or antiplatelet drugs. A hospital-based cross-sectional study was conducted to investigate the independent contribution of Lp(a) to platelet activity by controlling the effects of potential confounding factors including lipoprotein-associated phospholipase A2 [Lp-PLA2]. Blood samples were collected from 92 subjects without statins or antiplatelet agents from the Second Xiangya Hospital. The univariate correlation analysis showed a significant correlation between AA-induced average aggregation rate [AAR] and ApoB (r = 0.324, P = 0.002), ApoA1 (r = 0.252, P = 0.015), Lp(a) (r = 0.370, P < 0.001), Lp-PLA2 (r = 0.233, P = 0.025) and platelet counts [PLT] (r = 0.389, P < 0.001). Multivariate regression analysis suggested that Lp(a) contributed independently to AA-induced average aggregation rate (β = 0.023, P = 0.027) after controlling for the effects of ApoB, Lp-PLA2 and platelet counts. Lp(a) is positively associated with platelet aggregation independent of Lp-PLA2, which may partly account for the atherothrombotic effect of Lp(a).
Collapse
Affiliation(s)
- Huixing Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Di Fu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Yonghong Luo
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Daoquan Peng
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Changsha, 410011, Hunan, China.
| |
Collapse
|
5
|
KARA Ö, KİLİTCİ A, DAĞLIOĞLU G. Resveratrolün sıçan böbreğinde cisplatine bağlı hasar üzerindeki koruyucu etkisi. CUKUROVA MEDICAL JOURNAL 2022. [DOI: 10.17826/cumj.1086261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Purpose: The aim of this study was to evaluate the protective effect of resveratrol on cisplatin induced damage in rat kidney.
Materials and Methods: 30 female Wistar-Albino rats were allocated to form three groups: In group 1 (control group), 1 mL of 0.9% NaCl (saline) was administered intraperitoneally for 3 days. In group 2 (cisplatin group), 7.5 mg / kg intraperitoneal cisplatin was given for 3 days. In group 3 (cisplatin + resveratrol group) 7.5 mg / kg cisplatin and 10 mg / kg resveratrol were given via intraperitoneal route. Right kidneys were surgically extirpated in all groups. Malondialdehyde (MDA) levels and activities of catalase (CAT) and superoxide dismutase (SOD) were measured in both blood and tissues. Also, toxicity markers such as vascular congestion, hemorrhage, tubule degeneration and glomerular damage were assessed by examining the slides prepared from kidney tissue with microscopy.
Results: Tissue damage was significantly higher in group 2 than other groups. The MDA levels were significantly higher and the activities of SOD, and CAT were lower in group 2 than other groups.
Conclusion: According to our short term findings, resveratrol might be an effective molecule to prevent the harmful effect of cisplatin in rat kidney.
Collapse
Affiliation(s)
- Özlem KARA
- Kirsehir Ahi Evran University School of Medicine, Department of Histology and Embryology
| | - Asuman KİLİTCİ
- DUZCE UNIVERSITY, SCHOOL OF MEDICINE, DEPARTMENT OF SURGICAL MEDICAL SCIENCES, DEPARTMENT OF PATHOLOGY
| | - Gülçin DAĞLIOĞLU
- 3 Cukurova University Training and Research Hospital, Clinic of Biochemistry
| |
Collapse
|
6
|
Waiz M, Alvi SS, Khan MS. Potential dual inhibitors of PCSK-9 and HMG-R from natural sources in cardiovascular risk management. EXCLI JOURNAL 2022; 21:47-76. [PMID: 35221836 PMCID: PMC8859648 DOI: 10.17179/excli2021-4453] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/08/2021] [Indexed: 12/11/2022]
Abstract
Atherosclerotic cardiovascular disease (ASCVD) stands amongst the leading causes of mortality worldwide and has attracted the attention of world's leading pharmaceutical companies in order to tackle such mortalities. The low-density lipoprotein-cholesterol (LDL-C) is considered the most prominent biomarker for the assessment of ASCVD risk. Distinct inhibitors of 3-hydroxy-3-methyl-glutaryl-CoA reductase (HMG-R), the chief hepatic cholesterogenic enzyme, are being used since last seven decades to manage hypercholesterolemia. On the other hand, discovery and the association of proprotein convertase subtilisin/kexin type-9 (PCSK-9) with increased ASCVD risk have established PCSK-9 as a novel therapeutic target in cardiovascular medicine. PCSK-9 is well reckoned to facilitate the LDL-receptor (LDL-R) degradation and compromised LDL-C clearance leading to the arterial atherosclerotic plaque formation. The currently available HMG-R inhibitors (statins) and PCSK-9 inhibitors (siRNA, anti-sense oligonucleotides, and monoclonal antibodies) have shown great promises in achieving LDL-C lowering goals, however, their life long prescriptions have raised significant concerns. These deficits associated with the synthetic HMG-R and PCSK-9 inhibitors called for the discovery of alternative therapeutic candidates with potential dual HMG-R and PCSK-9 inhibitory activities from natural origins. Therefore, this report firstly describes the mechanistic insights into the cholesterol homeostasis through HMG-R, PCSK-9, and LDL-R functionality and then compiles the pharmacological effects of natural secondary metabolites with special emphasis on their dual HMG-R and PCSK-9 inhibitory action. In conclusion, various natural products exhibit atheroprotective effects via targeting HMG-R and PCSK-9 activities and lipoprotein metabolism, however, further clinical assessments are still warranted prior their approval for ASCVD risk management in hypercholesterolemic patients.
Collapse
Affiliation(s)
- Mohd Waiz
- IIRC-5, Clinical Biochemistry and Natural Product Research Lab, Department of Biosciences, Integral University, Lucknow, U.P. 226026, India
| | - Sahir Sultan Alvi
- IIRC-5, Clinical Biochemistry and Natural Product Research Lab, Department of Biosciences, Integral University, Lucknow, U.P. 226026, India
| | - M Salman Khan
- IIRC-5, Clinical Biochemistry and Natural Product Research Lab, Department of Biosciences, Integral University, Lucknow, U.P. 226026, India
| |
Collapse
|
7
|
Ageing, Age-Related Cardiovascular Risk and the Beneficial Role of Natural Components Intake. Int J Mol Sci 2021; 23:ijms23010183. [PMID: 35008609 PMCID: PMC8745076 DOI: 10.3390/ijms23010183] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 12/14/2021] [Accepted: 12/16/2021] [Indexed: 12/18/2022] Open
Abstract
Ageing, in a natural way, leads to the gradual worsening of the functional capacity of all systems and, eventually, to death. This process is strongly associated with higher metabolic and oxidative stress, low-grade inflammation, accumulation of DNA mutations and increased levels of related damage. Detrimental changes that accumulate in body cells and tissues with time raise the vulnerability to environmental challenges and enhance the risk of major chronic diseases and mortality. There are several theses concerning the mechanisms of ageing: genetic, free radical telomerase, mitochondrial decline, metabolic damage, cellular senescence, neuroendocrine theory, Hay-flick limit and membrane theories, cellular death as well as the accumulation of toxic and non-toxic garbage. Moreover, ageing is associated with structural changes within the myocardium, cardiac conduction system, the endocardium as well as the vasculature. With time, the cardiac structures lose elasticity, and fibrotic changes occur in the heart valves. Ageing is also associated with a higher risk of atherosclerosis. The results of studies suggest that some natural compounds may slow down this process and protect against age-related diseases. Animal studies imply that some of them may prolong the lifespan; however, this trend is not so obvious in humans.
Collapse
|
8
|
Gonçalinho GHF, Roggerio A, Goes MFDS, Avakian SD, Leal DP, Strunz CMC, Mansur ADP. Comparison of Resveratrol Supplementation and Energy Restriction Effects on Sympathetic Nervous System Activity and Vascular Reactivity: A Randomized Clinical Trial. Molecules 2021; 26:molecules26113168. [PMID: 34073163 PMCID: PMC8199010 DOI: 10.3390/molecules26113168] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 01/22/2023] Open
Abstract
Background: Chronic sympathetic nervous system activation is associated with endothelial dysfunction and cardiometabolic disease, which may be modulated by resveratrol (RSV) and energy restriction (ER). This study aimed to examine the effects of RSV and ER on plasma noradrenaline (NA), flow-mediated vasodilation (ed-FMD), and endothelium-independent nitrate-mediated vasodilation (ei-NMD). Methods: The study included 48 healthy adults randomized to 30-days intervention of RSV or ER. Results: Waist circumference, total cholesterol, HDL-c, LDL-c, apoA-I, and plasma NA decreased in the ER group, whilst RSV increased apoB and total cholesterol, without changing plasma NA. No effects on vascular reactivity were observed in both groups. Plasma NA change was positively correlated with total cholesterol (r = 0.443; p = 0.002), triglycerides (r = 0.438; p = 0.002), apoA-I (r = 0.467; p = 0.001), apoB (r = 0.318; p = 0.032) changes, and ei-NMD (OR = 1.294; 95%CI: 1.021–1.640). Conclusions: RSV does not improve cardiometabolic risk factors, sympathetic activity, and endothelial function. ER decreases plasma NA and waist circumference as well as improves blood lipids, but does not modify endothelial function. Finally, plasma NA was associated with ei-NMD, which could be attributed to a higher response to nitrate in patients with greater resting sympathetic vasoconstriction.
Collapse
|
9
|
Predictors of Clopidogrel Hyper-Responsiveness in Neuro-Interventional Procedures. J Stroke Cerebrovasc Dis 2020; 30:105420. [PMID: 33161351 DOI: 10.1016/j.jstrokecerebrovasdis.2020.105420] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 10/09/2020] [Accepted: 10/18/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Hyper-responsiveness to clopidogrel abnormally inhibits platelet aggregation and increases hemorrhagic complications. The present study investigated clinical factors related to clopidogrel hyper-responsiveness in neuro-interventional procedures. METHODS Two hundred twenty-four patients receiving clopidogrel for coil embolization to treat unruptured cerebral aneurysm or carotid artery stenting to treat carotid artery stenosis at the internal carotid artery origin were retrospectively reviewed for their P2Y12 reactivity unit (PRU) values and clinical characteristics. Hyper-responsiveness to clopidogrel was defined as a PRU of <95. RESULTS The mean PRU was 218.2 ± 77.8. Hyper-responsiveness to clopidogrel was observed in 12 patients (5.4%). Hyper-responsiveness was observed in younger patients, patients with a lower concentration of hemoglobin A1c, and patients with a higher low-density lipoprotein cholesterol (LDL-C) concentration compared with non-hyper-responsive patients (P = 0.01, P < 0.01, P < 0.01, respectively). On analysis of concomitant drugs, the patients in the hyper-responsive group were less frequently administered calcium channel blockers (CCBs) compared with the non-hyper-responsive group (P = 0.01). No significant differences in the usage of proton pump inhibitors or statins were observed. A LDL-C concentration of >120 mg/dL and no usage of CCBs were significant independent predictors of hyper-responsiveness to clopidogrel with a multivariate analysis (OR; 6.16, 95% CI, 1.57-26.64, P = 0.01, OR; 0.09, 95% CI, 0.01-0.82, P = 0.03, respectively). CONCLUSION The present study shows that a higher LDL-C concentration and no usage of CCBs are independent predictors of clopidogrel hyper-responsiveness. These results are useful to predict perioperative hemorrhagic complications. Considering dose reduction of clopidogrel or alternative drugs in high risk cases is necessary to prevent perioperative hemorrhagic complications.
Collapse
|
10
|
Jiao M, Li J, Zhang Q, Xu X, Li R, Dong P, Meng C, Li Y, Wang L, Qi W, Kang K, Wang H, Wang T. Identification of Four Potential Biomarkers Associated With Coronary Artery Disease in Non-diabetic Patients by Gene Co-expression Network Analysis. Front Genet 2020; 11:542. [PMID: 32714363 PMCID: PMC7344232 DOI: 10.3389/fgene.2020.00542] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 05/05/2020] [Indexed: 12/17/2022] Open
Abstract
Background Coronary artery disease (CAD) is a type of cardiovascular disease that greatly hurts the health of human beings. Diabetic status is one of the largest clinical factors affecting CAD-associated gene expression changes. Most of the studies focus on diabetic patients, whereas few have been done for non-diabetic patients. Since the pathophysiological processes may vary among these patients, we cannot simply follow the standard based on the data from diabetic patients. Therefore, the prognostic and predictive diagnostic biomarkers for CAD in non-diabetic patient need to be fully recognized. Materials and Methods To screen out candidate genes associated with CAD in non-diabetic patients, weighted gene co-expression network analysis (WGCNA) was constructed to conduct an analysis of microarray expression profiling in patients with CAD. First, the microarray data GSE20680 and GSE20681 were downloaded from NCBI. We constructed co-expression modules via WGCNA after excluding the diabetic patients. As a result, 18 co-expression modules were screened out, including 1,225 differentially expressed genes (DEGs) that were obtained from 152 patients (luminal stenosis ≥50% in at least one major vessel) and 170 patients (stenosis of <50%). Subsequently, a Pearson's correlation analysis was conducted between the modules and clinical traits. Then, a functional enrichment analysis was conducted, and we used gene network analysis to reveal hub genes. Last, we validated the hub genes with peripheral blood samples in an independent patient cohort using RT-qPCR. Results The results showed that the midnight blue module and the yellow module played vital roles in the pathogenesis of CAD in non-diabetic patients. Additionally, CD40, F11R, TNRC18, and calcium/calmodulin-dependent protein kinase type II gamma (CAMK2G) were screened out and validated using enzyme-linked immunosorbent assay (ELISA) in an independent patient cohort and immunohistochemical (IHC) staining in an atherosclerosis mouse model. Conclusion Our findings demonstrate that hub genes, CD40, F11R, TNRC18, and CAMK2G, are surrogate diagnostic biomarkers and/or therapeutic targets for CAD in non-diabetic patients and require deeper validation.
Collapse
Affiliation(s)
- Min Jiao
- Department of Cardiology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Jingtian Li
- Department of Cardiology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Quan Zhang
- Department of Cardiology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Xiufeng Xu
- Department of Neurology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Ruidong Li
- Graduate Program in Genetics, Genomics, and Bioinformatics, University of California, Riverside, Riverside, CA, United States
| | - Peikang Dong
- Department of Cardiology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Chun Meng
- Department of Cardiology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Yi Li
- Department of Cardiology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Lijuan Wang
- Department of Cardiology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Wanpeng Qi
- Department of Cardiology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Kai Kang
- Department of Cardiology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Hongjie Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Wang
- Department of Cardiology, Affiliated Hospital of Weifang Medical University, Weifang, China
| |
Collapse
|
11
|
Jiménez-Moreno N, Volpe F, Moler JA, Esparza I, Ancín-Azpilicueta C. Impact of Extraction Conditions on the Phenolic Composition and Antioxidant Capacity of Grape Stem Extracts. Antioxidants (Basel) 2019; 8:antiox8120597. [PMID: 31795232 PMCID: PMC6943662 DOI: 10.3390/antiox8120597] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 11/22/2019] [Accepted: 11/26/2019] [Indexed: 12/19/2022] Open
Abstract
The use of grape stems for the extraction of bioactive compounds to be used in the pharmaceutical, food, and cosmetic industries is a promising objective. The aim of this work is to determine the influence of the different extraction conditions (temperature, ethanol concentration, and ratio of sample/solvent) on phenolic composition and antioxidant capacity of Mazuelo stem extracts. In general, the ethanol concentration of the extraction solvent was the factor that had the greatest influence on the extraction of different bioactive compounds. The greatest content of total phenolic compounds and the highest antioxidant activity of the extracts were obtained with 50% ethanol and at 40 °C. The most abundant compound found in the different extracts obtained from Mazuelo grape stem was (+)-catechin, but appreciable concentrations of gallic acid, a quercetin derivative, and stilbenes (trans-resveratrol and trans-ε-viniferin) were also extracted. Quercetin and malvidin-3-glucoside showed the highest correlation with the antioxidant capacity of the extracts, while stilbenes did not present such relation. The maximum concentration of gallic acid was extracted with water but the extraction of most of the compounds was maximum on using 50% ethanol. Consequently, the selection of the extraction method to be used will depend on the particular compound to be extracted in greatest quantity.
Collapse
Affiliation(s)
- Nerea Jiménez-Moreno
- Department of Sciences, Universidad Pública de Navarra, Campus Arrosadía s/n, 31006 Pamplona, Spain; (N.J.-M.); (F.V.)
| | - Francesca Volpe
- Department of Sciences, Universidad Pública de Navarra, Campus Arrosadía s/n, 31006 Pamplona, Spain; (N.J.-M.); (F.V.)
| | - Jose Antonio Moler
- Department of Statistics and Operational Research, Universidad Pública de Navarra, Campus Arrosadía s/n, 31006 Pamplona, Spain;
| | - Irene Esparza
- Department of Sciences, Universidad Pública de Navarra, Campus Arrosadía s/n, 31006 Pamplona, Spain; (N.J.-M.); (F.V.)
- Institute for Advanced Materials (InaMat), Universidad Pública de Navarra, 31006 Pamplona, Spain
- Correspondence: (I.E.); (C.A.-A.); Tel.: +34-948-169596 (C.A.-A.)
| | - Carmen Ancín-Azpilicueta
- Department of Sciences, Universidad Pública de Navarra, Campus Arrosadía s/n, 31006 Pamplona, Spain; (N.J.-M.); (F.V.)
- Institute for Advanced Materials (InaMat), Universidad Pública de Navarra, 31006 Pamplona, Spain
- Correspondence: (I.E.); (C.A.-A.); Tel.: +34-948-169596 (C.A.-A.)
| |
Collapse
|
12
|
Bagetta D, Maruca A, Lupia A, Mesiti F, Catalano R, Romeo I, Moraca F, Ambrosio FA, Costa G, Artese A, Ortuso F, Alcaro S, Rocca R. Mediterranean products as promising source of multi-target agents in the treatment of metabolic syndrome. Eur J Med Chem 2019; 186:111903. [PMID: 31787360 DOI: 10.1016/j.ejmech.2019.111903] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 10/02/2019] [Accepted: 11/18/2019] [Indexed: 01/19/2023]
Abstract
Alteration of nutritional habits play an essential role on the risk of developing Metabolic Syndrome (MetS). Several epidemiological studies have shown that assuming diets rich of foods included in the Mediterranean diet (MetDiet) pattern like, such as olive oil, nuts, fruit, fiber, vegetables, wine and grain cereals has protective effects on the different risk factors characterizing the MetS. The beneficial effects of the MetDiet in the MetS are mainly due to the antioxidant and anti-inflammatory properties of the most abundant phytochemical components of such foods as polyphenols like resveratrol and oleuropein, allyl sulfides, ellagic acid, mono- and poly-unsaturated fatty acids (MUFA and PUFA), tocopherols and flavonoids like quercetin, which have shown positive results in the prevention of cardiovascular diseases (CVDs), with related risk factors, like hypertension, hypercholesterolemia and obesity. In this review, we highlighted the multi-target activities of the bioactive components contained in some foods typical of the Mediterranean area like olive oil, onion, liquorice, rosemary, oregano, hazelnut, pistachio, "Melannurca" apple, red wine, hot pepper, Citrus sp. fruits, saffron and garlic, with particular focus on their impact on health outcomes in relation to MetS main key factors, such as insulin resistance (IR) and type 2 diabetes mellitus (T2DM), endothelial dysfunctions, inflammatory response, oxidative stress and dyslipidaemic and hypercholesterolemic effects.
Collapse
Affiliation(s)
- Donatella Bagetta
- Dipartimento di Scienze della Salute, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy; Net4Science srl, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy
| | - Annalisa Maruca
- Dipartimento di Scienze della Salute, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy; Net4Science srl, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy
| | - Antonio Lupia
- Dipartimento di Scienze della Salute, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy; Net4Science srl, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy
| | - Francesco Mesiti
- Dipartimento di Scienze della Salute, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy; Net4Science srl, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy
| | - Raffaella Catalano
- Dipartimento di Scienze della Salute, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy; Net4Science srl, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy
| | - Isabella Romeo
- Net4Science srl, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy; Department of Chemistry and Chemical Technologies, University of Calabria, via Pietro Bucci, 87036, Arcavacata di Rende, Italy
| | - Federica Moraca
- Net4Science srl, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy; Department of Pharmacy, University of Naples "Federico II", via D. Montesano 49, 80131, Naples, Italy.
| | - Francesca Alessandra Ambrosio
- Dipartimento di Scienze della Salute, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy.
| | - Giosuè Costa
- Dipartimento di Scienze della Salute, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy; Net4Science srl, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy
| | - Anna Artese
- Dipartimento di Scienze della Salute, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy; Net4Science srl, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy
| | - Francesco Ortuso
- Dipartimento di Scienze della Salute, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy; Net4Science srl, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy
| | - Stefano Alcaro
- Dipartimento di Scienze della Salute, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy; Net4Science srl, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy
| | - Roberta Rocca
- Net4Science srl, Università "Magna Græcia", Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy; Department of Experimental and Clinical Medicine, "Magna Græcia" University, Campus Salvatore Venuta, Viale Europa, 88100, Catanzaro, Italy
| |
Collapse
|
13
|
Gurung AB, Pamay P, Tripathy D, Biswas K, Chatterjee A, Joshi SR, Bhattacharjee A. Bioprospection of anti-inflammatory phytochemicals suggests rutaecarpine and quinine as promising 15-lipoxygenase inhibitors. J Cell Biochem 2019; 120:13598-13613. [PMID: 30937959 DOI: 10.1002/jcb.28634] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 01/29/2019] [Accepted: 02/04/2019] [Indexed: 01/31/2023]
Abstract
15-Lipoxygenase (15-LOX) belongs to the family of nonheme iron containing enzymes that catalyzes the peroxidation of polyunsaturated fatty acids (PUFAs) to generate eicosanoids that play an important role in signaling pathways. The role of 15-LOX has been demonstrated in atherosclerosis as well as other inflammatory diseases. In the present study, drug-like compounds were first screened from a set of anti-inflammatory phytochemicals based on Lipinski's rule of five (ROF) and in silico toxicity filters. Two lead compounds-quinine (QUIN) and rutaecarpine (RUT) were shortlisted by analyzing molecular interactions and binding energies of the filtered compounds with the target using molecular docking. Molecular dynamics simulation studies indicate stable trajectories of apo_15-LOX and docked complexes (15-LOX_QUIN and 15-LOX_RUT). In vitro 15-LOX inhibition studies shows that both QUIN and RUT have lower inhibitory concentration (IC50 ) value than the control (quercetin). Both QUIN and RUT exhibit moderate antioxidant activities. The cell viability study of these compounds suggests no significant toxicity in HEK-293 cell lines. Further, QUIN and RUT both did not show any inhibition against selected Gram-positive and Gram-negative bacterial species. Thus, based on our present findings, rutaecarpine and quinine may be suggested as promising 15-LOX inhibitor for the prevention of the atherosclerosis development.
Collapse
Affiliation(s)
- Arun Bahadur Gurung
- Computational Biology Laboratory, Department of Biotechnology and Bioinformatics, North-Eastern Hill University, Shillong, Meghalaya, India
| | - Pezaiwi Pamay
- Computational Biology Laboratory, Department of Biotechnology and Bioinformatics, North-Eastern Hill University, Shillong, Meghalaya, India
| | - Debabrata Tripathy
- Genetics and Molecular biology Laboratory, Department of Biotechnology and Bioinformatics, North-Eastern Hill University, Shillong, Meghalaya, India
| | - Koel Biswas
- Microbiology Laboratory, Department of Biotechnology and Bioinformatics, North-Eastern Hill University, Shillong, Meghalaya, India
| | - Anupam Chatterjee
- Genetics and Molecular biology Laboratory, Department of Biotechnology and Bioinformatics, North-Eastern Hill University, Shillong, Meghalaya, India
| | - S R Joshi
- Microbiology Laboratory, Department of Biotechnology and Bioinformatics, North-Eastern Hill University, Shillong, Meghalaya, India
| | - Atanu Bhattacharjee
- Computational Biology Laboratory, Department of Biotechnology and Bioinformatics, North-Eastern Hill University, Shillong, Meghalaya, India.,Bioinformatics Centre, North-Eastern Hill University, Shillong, Meghalaya, India
| |
Collapse
|
14
|
Liberale L, Bonaventura A, Montecucco F, Dallegri F, Carbone F. Impact of Red Wine Consumption on Cardiovascular Health. Curr Med Chem 2019; 26:3542-3566. [PMID: 28521683 DOI: 10.2174/0929867324666170518100606] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 03/05/2017] [Accepted: 03/05/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND The devastating effects of heavy alcohol drinking have been long time recognized. In the last decades, potential benefits of modest red wine drinking were suggested. In European countries in which red wide intake is not negligible (such as France), the association between cholesterol and cardiovascular (CV) risk was less evident, suggesting the action of some protective molecules in red wine or other foods and drinks. METHODS This narrative review is based on the material searched for and obtained via PubMed up to May 2016. The search terms we used were: "red wine, cardiovascular, alcohol" in combination with "polyphenols, heart failure, infarction". RESULTS Epidemiological and mechanistic evidence of a J-shaped relationship between red wine intake and CV risk further supported the "French paradox". Specific components of red wine both in vitro and in animal models were discovered. Polyphenols and especially resveratrol largely contribute to CV prevention mainly through antioxidant properties. They exert beneficial effects on endothelial dysfunction and hypertension, dyslipidemia, metabolic diseases, thus reducing the risk of adverse CV events such as myocardial infarction ischemic stroke and heart failure. Of interest, recent studies pointed out the role of ethanol itself as a potential cardioprotective agent, but a clear epidemiological evidence is still missing. The aim of this narrative review is to update current knowledge on the intracellular mechanism underlying the cardioprotective effects of polyphenols and ethanol. Furthermore, we summarized the results of epidemiological studies, emphasizing their methodological criticisms and the need for randomized clinical trials able to clarify the potential role of red wine consumption in reducing CV risk. CONCLUSION Caution in avowing underestimation of the global burden of alcohol-related diseases was particularly used.
Collapse
Affiliation(s)
- Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Aldo Bonaventura
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
- IRCCS AOU San Martino - IST, Genova, 10 Largo Benzi, 16132 Genoa, Italy
- Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 9 viale Benedetto XV, 16132 Genoa, Italy
| | - Franco Dallegri
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
- IRCCS AOU San Martino - IST, Genova, 10 Largo Benzi, 16132 Genoa, Italy
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| |
Collapse
|
15
|
Korakas E, Dimitriadis G, Raptis A, Lambadiari V. Dietary Composition and Cardiovascular Risk: A Mediator or a Bystander? Nutrients 2018; 10:E1912. [PMID: 30518065 PMCID: PMC6316552 DOI: 10.3390/nu10121912] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 11/28/2018] [Accepted: 11/29/2018] [Indexed: 02/07/2023] Open
Abstract
The role of nutrition in the pathogenesis of cardiovascular disease has long been debated. The established notion of the deleterious effects of fat is recently under question, with numerous studies demonstrating the benefits of low-carbohydrate, high-fat diets in terms of obesity, diabetes, dyslipidemia, and metabolic derangement. Monounsaturated and polyunsaturated fatty acids, especially n-3 PUFAs (polyunsaturated fatty acids), are the types of fat that favor metabolic markers and are key components of the Mediterranean Diet, which is considered an ideal dietary pattern with great cardioprotective effects. Except for macronutrients, however, micronutrients like polyphenols, carotenoids, and vitamins act on molecular pathways that affect oxidative stress, endothelial function, and lipid and glucose homeostasis. In relation to these metabolic markers, the human gut microbiome is constantly revealed, with its composition being altered by even small dietary changes and different microbial populations being associated with adverse cardiovascular outcomes, thus becoming the target for potential new treatment interventions. This review aims to present the most recent data concerning different dietary patterns at both the macro- and micronutrient level and their association with atherosclerosis, obesity, and other risk factors for cardiovascular disease.
Collapse
Affiliation(s)
- Emmanouil Korakas
- Second Department of Internal Medicine and Research Institute, University General Hospital Attikon, 124 62 Haidari, Greece.
| | - George Dimitriadis
- Second Department of Internal Medicine and Research Institute, University General Hospital Attikon, 124 62 Haidari, Greece.
| | - Athanasios Raptis
- Second Department of Internal Medicine and Research Institute, University General Hospital Attikon, 124 62 Haidari, Greece.
| | - Vaia Lambadiari
- Second Department of Internal Medicine and Research Institute, University General Hospital Attikon, 124 62 Haidari, Greece.
| |
Collapse
|
16
|
Koushki M, Amiri‐Dashatan N, Ahmadi N, Abbaszadeh H, Rezaei‐Tavirani M. Resveratrol: A miraculous natural compound for diseases treatment. Food Sci Nutr 2018; 6:2473-2490. [PMID: 30510749 PMCID: PMC6261232 DOI: 10.1002/fsn3.855] [Citation(s) in RCA: 171] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 09/24/2018] [Accepted: 09/26/2018] [Indexed: 12/11/2022] Open
Abstract
Resveratrol (3, 5, 4'-trihydroxystilbene) is a nonflavonoid polyphenol that naturally occurs as phytoalexin. It is produced by plant sources such as grapes, apples, blueberries, plums, and peanut. This compound has critical roles in human health and is well known for its diverse biological activities such as antioxidant and anti-inflammatory properties. Nowadays, due to rising incidence of different diseases such as cancer and diabetes, efforts to find novel and effective disease-protective agents have led to the identification of plant-derived compounds such as resveratrol. Furthermore, several in vitro and in vivo studies have revealed the effectiveness of resveratrol in various diseases such as diabetes mellitus, cardiovascular disease, metabolic syndrome, obesity, inflammatory, neurodegenerative, and age-related diseases. This review presents an overview of currently available studies on preventive properties and essential molecular mechanisms involved in various diseases.
Collapse
Affiliation(s)
- Mehdi Koushki
- Department of BiochemistryFaculty of MedicineTehran University of Medical SciencesTehranIran
| | - Nasrin Amiri‐Dashatan
- Student Research CommitteeProteomics Research CenterShahid Beheshti University of Medical SciencesTehranIran
| | - Nayebali Ahmadi
- Proteomics Research CenterFaculty of Paramedical SciencesShahid Beheshti University of Medical SciencesTehranIran
| | | | - Mostafa Rezaei‐Tavirani
- Proteomics Research CenterFaculty of Paramedical SciencesShahid Beheshti University of Medical SciencesTehranIran
| |
Collapse
|
17
|
Chen J, Li JH, Zhao SJ, Wang DY, Zhang WZ, Liang WJ. Clinical significance of costimulatory molecules CD40/CD40L and CD134/CD134L in coronary heart disease: A case-control study. Medicine (Baltimore) 2017; 96:e7634. [PMID: 28796044 PMCID: PMC5556210 DOI: 10.1097/md.0000000000007634] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The aim of the study was to evaluate the potential role of CD40/CD40 ligand (CD40L) and CD134/CD134 ligand (CD134L) in the development of coronary heart disease (CHD) via the performance of a case-control study.The research objects were 234 cases of CHD patients and 120 cases of well-matched normal controls. Following the separation of peripheral blood mononuclear cells (PBMCs), real-time quantitative PCR (qRT-PCR), Western blot, immunohistochemistry, and flow cytometry were applied for the detection of mRNA levels and expression levels of CD40/CD40L and CD134/CD134L; meanwhile, intercellular adhesion molecule-1 (ICAM-1) and Fas protein mRNA levels were detected using qRT-PCR.There was no statistical difference in the comparison of baseline characteristics between groups, indicating comparability between groups. qRT-PCR and Western blot analysis indicated that CD40/CD40L and CD134/CD134L mRNA and protein expression levels were all increased in the CHD group than those in the control group. Flow cytometry further confirmed the similar tendency. Meanwhile, ICAM-1 and Fas protein mRNA levels were elevated in the CHD group and positively correlated with the above parameters. Furthermore, CD40/CD40L expression rates were negatively correlated with gender and different types of CHD. Meanwhile, CD134/CD134L expressions were also higher in male patients, in patients with family history, previous history of hypertension, diabetes, and cerebrovascular diseases.CD40/CD40L and CD134/CD134L are increased and may have potential correlation with clinical pathological features of patients with CHD. Further in-depth exploration of costimulatory molecules for CHD guidance as well as intrinsic mechanisms are needed combined with in vivo and in vitro experiments.
Collapse
Affiliation(s)
- Jun Chen
- Department of Cardiovascular Medicine, Guangzhou Panyu Central Hospital,
- Panyu District Cardiovascular Disease Research Institute of Guangzhou, Guangzhou, P.R. China
| | - Jian-Hao Li
- Department of Cardiovascular Medicine, Guangzhou Panyu Central Hospital,
- Panyu District Cardiovascular Disease Research Institute of Guangzhou, Guangzhou, P.R. China
| | - Shan-Jun Zhao
- Department of Cardiovascular Medicine, Guangzhou Panyu Central Hospital,
- Panyu District Cardiovascular Disease Research Institute of Guangzhou, Guangzhou, P.R. China
| | - Da-Yu Wang
- Department of Cardiovascular Medicine, Guangzhou Panyu Central Hospital,
- Panyu District Cardiovascular Disease Research Institute of Guangzhou, Guangzhou, P.R. China
| | - Wen-Zhu Zhang
- Department of Cardiovascular Medicine, Guangzhou Panyu Central Hospital,
- Panyu District Cardiovascular Disease Research Institute of Guangzhou, Guangzhou, P.R. China
| | - Wei-Jie Liang
- Department of Cardiovascular Medicine, Guangzhou Panyu Central Hospital,
- Panyu District Cardiovascular Disease Research Institute of Guangzhou, Guangzhou, P.R. China
| |
Collapse
|
18
|
Alfaras I, Di Germanio C, Bernier M, Csiszar A, Ungvari Z, Lakatta EG, de Cabo R. Pharmacological Strategies to Retard Cardiovascular Aging. Circ Res 2017; 118:1626-42. [PMID: 27174954 DOI: 10.1161/circresaha.116.307475] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 04/08/2016] [Indexed: 01/10/2023]
Abstract
Aging is the major risk factor for cardiovascular diseases, which are the leading cause of death in the United States. Traditionally, the effort to prevent cardiovascular disease has been focused on addressing the conventional risk factors, including hypertension, hyperglycemia, hypercholesterolemia, and high circulating levels of triglycerides. However, recent preclinical studies have identified new approaches to combat cardiovascular disease. Calorie restriction has been reproducibly shown to prolong lifespan in various experimental model animals. This has led to the development of calorie restriction mimetics and other pharmacological interventions capable to delay age-related diseases. In this review, we will address the mechanistic effects of aging per se on the cardiovascular system and focus on the prolongevity benefits of various therapeutic strategies that support cardiovascular health.
Collapse
Affiliation(s)
- Irene Alfaras
- From the Experimental Gerontology Section, Translational Gerontology Branch (I.A., C.D.G., M.B., R.d.C.) and Laboratory of Cardiovascular Science (E.G.L.), National Institute on Aging, National Institutes of Health, Baltimore, MD; Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy (C.D.G.); and Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK (A.C., Z.U.)
| | - Clara Di Germanio
- From the Experimental Gerontology Section, Translational Gerontology Branch (I.A., C.D.G., M.B., R.d.C.) and Laboratory of Cardiovascular Science (E.G.L.), National Institute on Aging, National Institutes of Health, Baltimore, MD; Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy (C.D.G.); and Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK (A.C., Z.U.)
| | - Michel Bernier
- From the Experimental Gerontology Section, Translational Gerontology Branch (I.A., C.D.G., M.B., R.d.C.) and Laboratory of Cardiovascular Science (E.G.L.), National Institute on Aging, National Institutes of Health, Baltimore, MD; Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy (C.D.G.); and Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK (A.C., Z.U.)
| | - Anna Csiszar
- From the Experimental Gerontology Section, Translational Gerontology Branch (I.A., C.D.G., M.B., R.d.C.) and Laboratory of Cardiovascular Science (E.G.L.), National Institute on Aging, National Institutes of Health, Baltimore, MD; Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy (C.D.G.); and Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK (A.C., Z.U.)
| | - Zoltan Ungvari
- From the Experimental Gerontology Section, Translational Gerontology Branch (I.A., C.D.G., M.B., R.d.C.) and Laboratory of Cardiovascular Science (E.G.L.), National Institute on Aging, National Institutes of Health, Baltimore, MD; Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy (C.D.G.); and Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK (A.C., Z.U.)
| | - Edward G Lakatta
- From the Experimental Gerontology Section, Translational Gerontology Branch (I.A., C.D.G., M.B., R.d.C.) and Laboratory of Cardiovascular Science (E.G.L.), National Institute on Aging, National Institutes of Health, Baltimore, MD; Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy (C.D.G.); and Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK (A.C., Z.U.)
| | - Rafael de Cabo
- From the Experimental Gerontology Section, Translational Gerontology Branch (I.A., C.D.G., M.B., R.d.C.) and Laboratory of Cardiovascular Science (E.G.L.), National Institute on Aging, National Institutes of Health, Baltimore, MD; Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy (C.D.G.); and Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK (A.C., Z.U.).
| |
Collapse
|
19
|
High Triglycerides Are Associated with Low Thrombocyte Counts and High VEGF in Nephropathia Epidemica. J Immunol Res 2016; 2016:8528270. [PMID: 28053993 PMCID: PMC5178363 DOI: 10.1155/2016/8528270] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 10/24/2016] [Indexed: 01/05/2023] Open
Abstract
Nephropathia epidemica (NE) is a mild form of hemorrhagic fever with renal syndrome. Several reports have demonstrated a severe alteration in lipoprotein metabolism. However, little is known about changes in circulating lipids in NE. The objectives of this study were to evaluate changes in serum total cholesterol, high density cholesterol (HDCL), and triglycerides. In addition to evaluation of serum cytokine activation associations, changes in lipid profile and cytokine activation were determined for gender, thrombocyte counts, and VEGF. Elevated levels of triglycerides and decreased HDCL were observed in NE, while total cholesterol did not differ from controls. High triglycerides were associated with both the lowest thrombocyte counts and high serum VEGF, as well as a high severity score. Additionally, there were higher levels of triglycerides in male than female NE patients. Low triglycerides were associated with upregulation of IFN-γ and IL-12, suggesting activation of Th1 helper cells. Furthermore, levels of IFN-γ and IL-12 were increased in patients with lower severity scores, suggesting that a Th1 type immune response is playing protective role in NE. These combined data advance the understanding of NE pathogenesis and indicate a role for high triglycerides in disease severity.
Collapse
|
20
|
Negative regulation of NLRP3 inflammasome by SIRT1 in vascular endothelial cells. Immunobiology 2016; 222:552-561. [PMID: 27908642 DOI: 10.1016/j.imbio.2016.11.002] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 11/01/2016] [Accepted: 11/02/2016] [Indexed: 01/01/2023]
Abstract
NLRP3 inflammasome not only functions as a critical effector in innate immunity, but also triggers the production of proinflammatory cytokines involved in inflammation-associated diseases. Sirtuin 1 (SIRT1) plays an important role in the regulation of cellular inflammation. However, whether the activation of NLRP3 inflammasome is regulated by SIRT1 remains unknown. In this study, we investigated the regulatory effect of SIRT1 on NLRP3 inflammasome and the underlying mechanisms. We found that lipopolysaccharide (LPS) and adenosine triphosphate (ATP)-induced the activation of NLRP3 inflammasome in human umbilical vein endothelial cells (HUVECs). Activation of SIRT1 inhibited NLRP3 inflammasome activation and subsequent caspase-1 cleavage as well as interleukin (IL)-1β secretion, whereas SIRT1 knockdown obviously enhanced the activation of NLRP3 inflammasome in HUVECs. Importantly, gene silencing of SIRT1 abrogated the inhibitory effect of SIRT1 activator on NLRP3 inflammasome formation and IL-1β production in HUVECs stimulated with LPS plus ATP. Further study indicated that cluster of differentiation 40 (CD40) may be involved in the regulation of NLRP3 inflammasome by SIRT1. In vivo studies indicated that implantation of the periarterial carotid collar increased the arterial expression levels of CD40 and CD40 Ligand (CD40L), but inhibited arterial SIRT1 expression in the rabbits. Moreover, treatment with SIRT1 activator decreased CD40 and CD40L levels in collared arteries. Meanwhile, serum IL-1β level, the marker of inflammasome activation, was also inhibited by SIRT1 activation. Taken together, these findings revealed a novel regulatory mechanism of NLRP3 inflammasome by SIRT1, which may be related to suppression of CD40.
Collapse
|
21
|
Bonnefont-Rousselot D. Resveratrol and Cardiovascular Diseases. Nutrients 2016; 8:nu8050250. [PMID: 27144581 PMCID: PMC4882663 DOI: 10.3390/nu8050250] [Citation(s) in RCA: 290] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Revised: 04/10/2016] [Accepted: 04/20/2016] [Indexed: 02/06/2023] Open
Abstract
The increased incidence of cardiovascular diseases (CVDs) has stimulated research for substances that could improve cardiovascular health. Among them, resveratrol (RES), a polyphenolic compound notably present in grapes and red wine, has been involved in the “French paradox”. RES is known for its antioxidant and anti-inflammatory properties and for its ability to upregulate endothelial NO synthase (eNOS). RES was able to scavenge •OH/O2•− and peroxyl radicals, which can limit the lipid peroxidation processes. Moreover, in bovine aortic endothelial cells (BAEC) under glucose-induced oxidative stress, RES restored the activity of dimethylargininedimethylaminohydrolase (DDAH), an enzyme that degrades an endogenous inhibitor of eNOS named asymmetric dimethylarginine (ADMA). Thus, RES could improve •NO availability and decrease the endothelial dysfunction observed in diabetes. Preclinical studies have made it possible to identify molecular targets (SIRT-1, AMPK, Nrf2, NFκB…); however, there are limited human clinical trials, and difficulties in the interpretation of results arise from the use of high-dose RES supplements in research studies, whereas low RES concentrations are present in red wine. The discussions on potential beneficial effects of RES in CVDs (atherosclerosis, hypertension, stroke, myocardial infarction, heart failure) should compare the results of preclinical studies with those of clinical trials.
Collapse
Affiliation(s)
- Dominique Bonnefont-Rousselot
- Department of Biochemistry, Faculty of Pharmacy, Paris Descartes University, Sorbonne Paris Cité, Paris 75006, France.
- Department of Metabolic Biochemistry, Pitié-Salpêtrière-Charles Foix Hospital (AP-HP), Paris 75013, France.
- Inserm UMR_S 1166 ICAN, UPMC, La Pitié Hospital, Paris 75013, France.
| |
Collapse
|
22
|
Jayakumar T, Yang CH, Geraldine P, Yen TL, Sheu JR. The pharmacodynamics of antiplatelet compounds in thrombosis treatment. Expert Opin Drug Metab Toxicol 2016; 12:615-32. [PMID: 27055051 DOI: 10.1080/17425255.2016.1176141] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Thanasekaran Jayakumar
- Graduate Institute of Medical Sciences, Taipei Medical University, Taipei, Taiwan
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chih-Hao Yang
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Pitchairaj Geraldine
- Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, India
| | - Ting-Lin Yen
- Graduate Institute of Medical Sciences, Taipei Medical University, Taipei, Taiwan
| | - Joen-Rong Sheu
- Graduate Institute of Medical Sciences, Taipei Medical University, Taipei, Taiwan
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
23
|
Pan W, Yu H, Huang S, Zhu P. Resveratrol Protects against TNF-α-Induced Injury in Human Umbilical Endothelial Cells through Promoting Sirtuin-1-Induced Repression of NF-KB and p38 MAPK. PLoS One 2016; 11:e0147034. [PMID: 26799794 PMCID: PMC4723256 DOI: 10.1371/journal.pone.0147034] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 12/28/2015] [Indexed: 11/18/2022] Open
Abstract
Inflammation and reactive oxygen species (ROS) play important roles in the pathogenesis of atherosclerosis. Resveratrol has been shown to possess anti-inflammatory and antioxidative stress activities, but the underlying mechanisms are not fully understood. In the present study, we investigated the molecular basis associated with the protective effects of resveratrol on tumor necrosis factor-alpha (TNF-α)-induced injury in human umbilical endothelial cells (HUVECs) using a variety of approaches including a cell viability assay, reverse transcription and quantitative polymerase chain reaction, western blot, and immunofluorescence staining. We showed that TNF-α induced CD40 expression and ROS production in cultured HUVECs, which were attenuated by resveratrol treatment. Also, resveratrol increased the expression of sirtuin 1 (SIRT1); and repression of SIRT1 by small-interfering RNA (siRNA) and the SIRT1 inhibitor Ex527 reduced the inhibitory effects of resveratrol on CD40 expression and ROS generation. In addition, resveratrol downregulated the levels of p65 and phospho-p38 MAPK, but this inhibitory effect was attenuated by the suppression of SIRT1 activity. Moreover, the p38 MAPK inhibitor SD203580 and the nuclear factor (NF)-κB inhibitor pyrrolidine dithiocarbamate (PDTC) achieved similar repressive effects as resveratrol on TNF-α-induced ROS generation and CD40 expression. Thus, our study provides a mechanistic link between resveratrol and the activation of SIRT1, the latter of which is involved in resveratrol-mediated repression of the p38 MAPK/NF-κB pathway and ROS production in TNF-α-treated HUVECs.
Collapse
Affiliation(s)
- Wei Pan
- Provincial Clinical Medical College, Fujian Medical University, Fuzhou, China
| | - Huizhen Yu
- Provincial Clinical Medical College, Fujian Medical University, Fuzhou, China
- Department of Geriatrics, Fujian Provincial Hospital Key Laboratory of Geriatrics, Fujian Medical University, Fuzhou, China
- Fujian Institute of Clinical Geriatrics, Fuzhou, China
| | - Shujie Huang
- Fujian Institute of Clinical Geriatrics, Fuzhou, China
| | - Pengli Zhu
- Provincial Clinical Medical College, Fujian Medical University, Fuzhou, China
- Fujian Institute of Clinical Geriatrics, Fuzhou, China
- * E-mail:
| |
Collapse
|
24
|
Phytochemical Compounds and Protection from Cardiovascular Diseases: A State of the Art. BIOMED RESEARCH INTERNATIONAL 2015; 2015:918069. [PMID: 26504846 PMCID: PMC4609427 DOI: 10.1155/2015/918069] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 06/14/2015] [Indexed: 02/07/2023]
Abstract
Cardiovascular diseases represent a worldwide relevant socioeconomical problem. Cardiovascular disease prevention relies also on lifestyle changes, including dietary habits. The cardioprotective effects of several foods and dietary supplements in both animal models and in humans have been explored. It was found that beneficial effects are mainly dependent on antioxidant and anti-inflammatory properties, also involving modulation of mitochondrial function. Resveratrol is one of the most studied phytochemical compounds and it is provided with several benefits in cardiovascular diseases as well as in other pathological conditions (such as cancer). Other relevant compounds are Brassica oleracea, curcumin, and berberine, and they all exert beneficial effects in several diseases. In the attempt to provide a comprehensive reference tool for both researchers and clinicians, we summarized in the present paper the existing literature on both preclinical and clinical cardioprotective effects of each mentioned phytochemical. We structured the discussion of each compound by analyzing, first, its cellular molecular targets of action, subsequently focusing on results from applications in both ex vivo and in vivo models, finally discussing the relevance of the compound in the context of human diseases.
Collapse
|
25
|
Preclinical and clinical evidence for the role of resveratrol in the treatment of cardiovascular diseases. Biochim Biophys Acta Mol Basis Dis 2015; 1852:1155-77. [DOI: 10.1016/j.bbadis.2014.10.016] [Citation(s) in RCA: 211] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 10/24/2014] [Accepted: 10/27/2014] [Indexed: 12/12/2022]
|
26
|
Novelle MG, Wahl D, Diéguez C, Bernier M, de Cabo R. Resveratrol supplementation: Where are we now and where should we go? Ageing Res Rev 2015; 21:1-15. [PMID: 25625901 DOI: 10.1016/j.arr.2015.01.002] [Citation(s) in RCA: 177] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 01/09/2015] [Accepted: 01/15/2015] [Indexed: 01/15/2023]
Abstract
Pre-clinical findings have provided mounting evidence that resveratrol, a dietary polyphenol, may confer health benefits and protect against a variety of medical conditions and age-related complications. However, there is no consistent evidence of an increased protection against metabolic disorders and other ailments when comparing studies in laboratory animals and humans. A number of extraneous and potential confounding variables can affect the outcome of clinical research. To date, most of the studies that have investigated the effect of resveratrol administration on patient outcomes have been limited by their sample sizes. In this review, we will survey the latest advances regarding the timing, dosage, formulation, bioavailability, toxicity of resveratrol, and resveratrol-drug interactions in human studies. Moreover, the present report focuses on the actions of resveratrol treatment in combating diseases, such as cancer, diabetes, neurodegeneration, cardiovascular disease, and other age-related ailments.
Collapse
Affiliation(s)
- Marta G Novelle
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD 21224, USA; Research Center of Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain
| | - Devin Wahl
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Carlos Diéguez
- Research Center of Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain
| | - Michel Bernier
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD 21224, USA.
| |
Collapse
|
27
|
Chan LW, Luo XP, Ni HC, Shi HM, Liu L, Wen ZC, Gu XY, Qiao J, Li J. High levels of LDL-C combined with low levels of HDL-C further increase platelet activation in hypercholesterolemic patients. ACTA ACUST UNITED AC 2014; 48:167-73. [PMID: 25466164 PMCID: PMC4321223 DOI: 10.1590/1414-431x20144182] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 09/09/2014] [Indexed: 01/31/2023]
Abstract
High levels of low-density lipoprotein cholesterol (LDL-C) enhance platelet
activation, whereas high levels of high-density lipoprotein cholesterol (HDL-C) exert
a cardioprotective effect. However, the effects on platelet activation of high levels
of LDL-C combined with low levels of HDL-C (HLC) have not yet been reported. We aimed
to evaluate the platelet activation marker of HLC patients and investigate the
antiplatelet effect of atorvastatin on this population. Forty-eight patients with
high levels of LDL-C were enrolled. Among these, 23 had HLC and the other 25 had high
levels of LDL-C combined with normal levels of HDL-C (HNC). A total of 35
normocholesterolemic (NOMC) volunteers were included as controls. Whole blood flow
cytometry and platelet aggregation measurements were performed on all participants to
detect the following platelet activation markers: CD62p (P-selectin), PAC-1
(GPIIb/IIIa), and maximal platelet aggregation (MPAG). A daily dose of 20 mg
atorvastatin was administered to patients with high levels of LDL-C, and the above
assessments were obtained at baseline and after 1 and 2 months of treatment. The
expression of platelets CD62p and PAC-1 was increased in HNC patients compared to
NOMC volunteers (P<0.01 and P<0.05). Furthermore, the surface expression of
platelets CD62p and PAC-1 was greater among HLC patients than among HNC patients
(P<0.01 and P<0.05). Although the expression of CD62p and PAC-1 decreased
significantly after atorvastatin treatment, it remained higher in the HLC group than
in the HNC group (P<0.05 and P=0.116). The reduction of HDL-C further increased
platelet activation in patients with high levels of LDL-C. Platelet activation
remained higher among HLC patients regardless of atorvastatin treatment.
Collapse
Affiliation(s)
- L W Chan
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - X P Luo
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - H C Ni
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - H M Shi
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - L Liu
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Z C Wen
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - X Y Gu
- Department of Laboratory Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - J Qiao
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - J Li
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
28
|
Wang W, Bai L, Qiao H, Lu Y, Yang L, Zhang J, Lin R, Ren F, Zhang J, Ji M. The protective effect of fenofibrate against TNF-α-induced CD40 expression through SIRT1-mediated deacetylation of NF-κB in endothelial cells. Inflammation 2014; 37:177-85. [PMID: 24022598 DOI: 10.1007/s10753-013-9728-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Fenofibrate, as a lipid-lowering drug in clinic, participates in the regulation of inflammatory response. Recently, increasing studies have indicated that sirtuin1 (SIRT1), a NAD+-dependent deacetylase, has potential anti-inflammatory effect in endothelial cells. However, whether the regulatory effect of fenofibrate on inflammation response is mediated by SIRT1 remains unclear. The aim of this study was to investigate the effect of fenofibrate on the expressions of SIRT1 and pro-inflammatory cytokine CD40 in endothelial cells and explore the underlying mechanisms. The results showed that fenofibrate upregulated SIRT1 expression and inhibited CD40 expression in TNF-α-stimulated endothelial cells, but these effects were reversed by peroxisome proliferator-activated receptor-α (PPARα) antagonist GW6471. Furthermore, SIRT1 inhibitors sirtinol/nicotinamide (NAM) or SIRT1 knockdown could attenuate the effect of fenofibrate on CD40 expression in endothelial cells. Importantly, NF-κB inhibitor pyrrolidine dithiocarbamate (PDTC) augmented the effect of fenofibrate on CD40 expression. Further study found that fenofibrate decreased the expression of acetylated-NF-κB p65 (Ac-NF-κB p65) in TNF-α-stimulated endothelial cells, which was abolished by SIRT1 knockdown. These results indicate that fenofibrate has protective effect against TNF-α-induced CD40 expression through SIRT1-mediated deacetylation of the p65 subunit of NF-κB.
Collapse
|
29
|
Chromium picolinate inhibits cholesterol-induced stimulation of platelet aggregation in hypercholesterolemic rats. Ir J Med Sci 2014; 184:291-6. [DOI: 10.1007/s11845-014-1102-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Accepted: 02/26/2014] [Indexed: 01/18/2023]
|
30
|
Mohamed HE, El-Swefy SE, Hasan RA, Hasan AA. Neuroprotective effect of resveratrol in diabetic cerebral ischemic-reperfused rats through regulation of inflammatory and apoptotic events. Diabetol Metab Syndr 2014; 6:88. [PMID: 25191525 PMCID: PMC4153889 DOI: 10.1186/1758-5996-6-88] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 08/10/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Diabetes and cerebral ischemic-reperfusion are among the most common causes of neurological complications in Egypt. The prevalence of diabetes in Egypt is high and it can be considered as a major clinical and public health problem. METHODS Blood glucose, lipid profile, oxidative stress makers (cerebral MDA & GSH), cerebral interleukin-4 (IL-4) level and cerebral cyclooxygenase-2 (COX-2) gene expression were measured in male albino rats weighing 200 ± 20 g. The rats were divided into five groups, normal control group, diabetic group (diabetes was induced by single dose of streptozotocin [STZ]), diabetic cerebral ischemic-reperfused group, two treated groups (diabetic and diabetic ischemic-reperfused), both groups treated with resveratrol. Histological study was done using H&E, AgNOR and cresyl violet stains. Immunohistochemistry for Bax and COX-2 was done with morphometric study. RESULTS Diabetic and diabetic cerebral ischemic- reperfused rats showed significant increase in serum glucose level, serum TAG, serum LDL-C, atherogenic index, cerebral MDA and upregulation of COX-2 gene expression. These groups showed significant decrease in serum HDL, cerebral IL-4 and depletion of cerebral GSH when compared to normal control rats. Treating these groups with resveratrol resulted in significant decrease in serum glucose level, serum TAG, TC, serum LDL-C, atherogenic index, cerebral MDA and downregulation of COX-2 gene expression. The results of COX-2 gene expression were confirmed by COX-2 immunohistochemistry. Also, significant increase in serum HDL, cerebral IL-4 and cerebral GSH contents could be observed in these treated groups as compared to normal control group. Cerebral apoptotic index and optical density of Bax reaction revealed significant increase in diabetic and diabetic cerebral ischemic-reperfused rats while treatment of these groups with resveratrol resulted in significant decrease in cerebral apoptotic index and optical density of Bax reaction. These apoptotic results were confirmed with AgNOR and cresyl violet stains. CONCLUSION The results of this research suggest that upregulation of cerebral COX-2 gene along with the decrease in cerebral IL-4 and enhanced cerebral apoptosis is critically involved in cerebral damage associated with diabetes and cerebral ischemic-reperfusion. Resveratrol can ameliorate these effects and has promising neuroprotective effect in diabetic-induced cerebral complications.
Collapse
Affiliation(s)
- Hoda E Mohamed
- />Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Sahar E El-Swefy
- />Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Rehab A Hasan
- />Department of Histology, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Ahmed A Hasan
- />Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| |
Collapse
|
31
|
Wang W, Lin Q, Lin R, Zhang J, Ren F, Zhang J, Ji M, Li Y. PPARα agonist fenofibrate attenuates TNF-α-induced CD40 expression in 3T3-L1 adipocytes via the SIRT1-dependent signaling pathway. Exp Cell Res 2013; 319:1523-33. [DOI: 10.1016/j.yexcr.2013.04.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Revised: 03/14/2013] [Accepted: 04/09/2013] [Indexed: 11/30/2022]
|
32
|
Gocmen AY, Ocak GA, Ozbilim G, Delibas N, Gumuslu S. Effect of atorvastatin on atherosclerotic plaque formation and platelet activation in hypercholesterolemic rats. Can J Physiol Pharmacol 2013; 91:680-5. [PMID: 23984971 DOI: 10.1139/cjpp-2012-0325] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We aimed to investigate whether atorvastatin influenced the CD40-CD40L pathway in atherosclerosis formation in rats fed a high cholesterol diet. Thirty-six male Wistar rats were divided among 4 groups as follows: control (C), statin (S), 5% cholesterol fed (HC), and statin-administered hypercholesterolemic (HCS). Serum levels of lipids, soluble CD40L, platelet factor 4, and interleukin-6 were assayed with commercial kits. The number of platelets expressing surface P-selectin, CD40, and CD40L were determined by flow cytometry. Aortas were examined for fatty streaks. In the HC group, we observed a significant increase in serum lipid levels and platelet activation markers compared with the control group. Rats in the HCS group had a significant decrease in lipid levels and downregulation in the number of platelets expressing surface P-selectin, CD40, and CD40L compared with the HC group. We observed decreased fatty streak formations in aortas in HCS rats. A positive correlation was found for platelet activation markers and atherosclerotic fatty streak formations. Regression analysis revealed that the predictor of atherosclerosis was CD40L. Our study suggests that in a rat hypercholesterolemic model, statin treatment may influence the CD40-CD40L dyad, and that this effect is parallelled by a suppression of progression of atherosclerotic plaque formation.
Collapse
Affiliation(s)
- Ayse Yesim Gocmen
- Department of Biochemistry, Faculty of Medicine, Bozok University, 66200 Yozgat, Turkey
| | | | | | | | | |
Collapse
|
33
|
Dalaklioglu S, Genc GE, Aksoy NH, Akcit F, Gumuslu S. Resveratrol ameliorates methotrexate-induced hepatotoxicity in rats via inhibition of lipid peroxidation. Hum Exp Toxicol 2013; 32:662-71. [PMID: 23424212 DOI: 10.1177/0960327112468178] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Hepatotoxicity is one of the major complications of methotrexate (MTX) therapy. This study was carried out to evaluate the possible protective effect of resveratrol (trans-3,5,4'-trihydroxystilbene, RVT) against MTX-induced hepatotoxicity. Rats were randomly divided into four groups as control, MTX treated (7 mg/kg/day, intraperitoneally (i.p.), once daily for 3 consecutive days), MTX + RVT treated (20 mg/kg/day, i.p.), and RVT treated. First dose of RVT was administrated 3 days before the MTX injection and continued for 3 days. Histopathology of liver was evaluated by light microscopy. Aspartate aminotransferase (AST), alanine aminotransferase (ALT), and alkaline phosphatase (ALP) were used as biochemical markers of MTX-induced hepatic injury. The levels of thiobarbituric acid reactive substances (TBARS, a marker of lipid peroxidation) and activities of hepatic antioxidant enzymes such as catalase (CAT) and glutathione-S-transferase (GST) were used to analyze the oxidative stress-mediated lipid peroxidation in liver sections. Our results showed that MTX administration significantly increased ALT, ASP, and ALP levels. TBARS, CAT, and GST levels were also markedly increased in liver after MTX administration. RVT treatment significantly prevented MTX-induced hepatotoxicity, as indicated by AST, ALT, and ALP levels and liver histopathology. Moreover, administration of RVT significantly decreased the elevated levels of TBARS and activities of CAT and GST in the liver compared to MTX-treated group. These results revealed that RVT may have a protective effect against MTX-induced hepatotoxicity by inhibiting oxidative stress-mediated lipid peroxidation. Consequently, RVT treatment might be a promising strategy against MTX-induced hepatotoxicity.
Collapse
Affiliation(s)
- S Dalaklioglu
- Department of Pharmacology, Medical Faculty, Akdeniz University, Antalya, Turkey.
| | | | | | | | | |
Collapse
|
34
|
SIRT1 regulates TNF-α-induced expression of CD40 in 3T3-L1 adipocytes via NF-κB pathway. Cytokine 2012; 60:447-55. [DOI: 10.1016/j.cyto.2012.05.025] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Revised: 05/08/2012] [Accepted: 05/22/2012] [Indexed: 12/11/2022]
|
35
|
Esposito P, Rampino T, Dal Canton A. Soluble CD40 as a modulator of CD40 pathway. Immunol Lett 2012; 147:85-86. [PMID: 22819355 DOI: 10.1016/j.imlet.2012.06.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2012] [Revised: 05/30/2012] [Accepted: 06/20/2012] [Indexed: 02/07/2023]
|
36
|
Esposito P, Rampino T, Gregorini M, Gabanti E, Bianzina S, Dal Canton A. Mechanisms underlying sCD40 production in hemodialysis patients. Cell Immunol 2012; 278:10-15. [PMID: 23121970 DOI: 10.1016/j.cellimm.2012.06.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Revised: 05/08/2012] [Accepted: 06/19/2012] [Indexed: 02/07/2023]
Abstract
CD40 and its ligand (CD40L) regulate several cellular functions, including T and B-cell activation. The soluble form of CD40 (sCD40) antagonizes CD40/CD40L interaction. Patients undergoing hemodialysis (HD) present elevated sCD40 serum levels, which underlying molecular mechanisms are unknown. We studied sCD40 serum and urinary levels, CD40 membrane and gene expression and membrane shedding in HD, uremic not-HD patients (UR) and healthy subjects (N). We found that in HD sCD40 serum levels were higher than UR and N, being significantly elevated in anuric patients, and that sCD40 correlated to renal function in UR subjects, who presented also a reduced sCD40 urinary excretion rate. HD and UR presented reduced CD40 membrane and gene expression. The concentration of TNF-α converting enzyme (TACE), responsible for CD40 cleavage was not different between HD and N. Therefore the reduced renal clearance is the main cause of elevated sCD40 levels in HD. This finding could have relevant clinical implications.
Collapse
Affiliation(s)
- Pasquale Esposito
- Department of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, and University of Pavia, Italy.
| | | | | | | | | | | |
Collapse
|