1
|
Cheng Y, Zhao A, Li Y, Li C, Miao X, Yang W, Wang Y. Roles of SIRT3 in cardiovascular and neurodegenerative diseases. Ageing Res Rev 2025:102654. [PMID: 39755174 DOI: 10.1016/j.arr.2024.102654] [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: 11/20/2024] [Revised: 12/29/2024] [Accepted: 12/30/2024] [Indexed: 01/06/2025]
Abstract
Sirtuin-3 (SIRT3) in mitochondria has nicotinamide adenine dinucleotide (NAD+)-dependent protein deacetylase activity. As such, SIRT3 is crucial in cardiovascular and neurodegenerative diseases. Advanced proteomics and transcriptomics studies have revealed that SIRT3 expression becomes altered when the heart or brain is affected by external stimuli or disease, such as diabetic cardiomyopathy, atherosclerosis, myocardial infarction, Alzheimer's disease, Huntington's disease, and Parkinson's disease. More specifically, SIRT3 participates in the development of these disorders through its deacetylase activity and in combination with downstream signaling pathways. The paper reviews SIRT3's expression changes, roles, and mechanisms associated with the development of cardiovascular and neurodegenerative diseases. Additionally, strategies targeting SIRT3 to treat or regulate cardiovascular and neurodegenerative disease development are discussed.
Collapse
Affiliation(s)
- Yu Cheng
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, China; Department of Cardiovascular Center, TheFirst Hospital of Jilin University, Changchun,Jilin, China
| | - Anqi Zhao
- Laboratory of Basic Medicine, General Hospital of Northern Theatre Command, No. 83 Wenhua Road, Shenhe District, Shenyang 110016, Liaoning, China
| | - Ying Li
- Department of Medical clinic,Jilin women and children health hospital,Changchun,Jilin,China
| | - Cheng Li
- Department of Cardiovascular Center, TheFirst Hospital of Jilin University, Changchun,Jilin, China
| | - Xiao Miao
- The Second Hosptial of Jilin University,Changchun,Jilin,China.
| | - Wanshan Yang
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, China.
| | - Yonggang Wang
- Department of Cardiovascular Center, TheFirst Hospital of Jilin University, Changchun,Jilin, China
| |
Collapse
|
2
|
Li G, Wu G, Huang Q, Dong S, Zhou Y, Lu M, Liang J, Zhou X, Zhou Z. Carbon-based light addressable potential aptasensor based on the synergy of C-MXene@rGO and OPD@NGQDs for low-density lipoprotein detection. Mikrochim Acta 2024; 192:35. [PMID: 39729216 DOI: 10.1007/s00604-024-06909-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024]
Abstract
A novel carbon-based light-addressable potentiometric aptasensor (C-LAPS) was constructed for detection low-density lipoprotein (LDL) in serum. Carboxylated Ti3C2 MXene @reduced graphene oxide (C-MXene@rGO) was used as interface and o-phenylenediamine functionalized nitrogen-doped graphene quantum dots (OPD@NGQDs) as the photoelectric conversion element. The photosensitive layers composed of OPD@NGQDs/C-MXene@rGO exhibit superior photoelectric conversion efficiency and excellent biocompatibility, which contribute to an improved response signal. When LDL reacts with the LDL aptamer (LDLApt) immobilized on the photosensitive layers to form LDL-LDLApt complexes, the reaction process can induce the modification of the surface potential in the photosensitive layer, leading to potential shift observed through the I-V curves. The experimental conditions were successfully optimized with few planned tests by applying the Box-Behnken design and response surface methodology aspects of the Design-Expert software. Under the optimal condition, the potential shift had a linear relationship with concentrations of LDL from 0.02 to 0.30 μg/mL. The limit of detection (LOD) was 5.88 ng/mL (S/N = 3) and the sensitivity was 315.20 mV/μg·mL-1. In addition, the LDL C-LAPS demonstrated excellent specificity, reproducibility, and stability in detecting LDL. The sensor performed well in quantifying LDL in real samples. Therefore, the LDL C-LAPS has the potential for clinical applications.
Collapse
Affiliation(s)
- Guiyin Li
- College of Chemistry, Guangdong University of Petrochemical Technology, Guandu Road, Maoming, Guangdong, 525000, People's Republic of China
| | - Guangxiong Wu
- School of Life and Environmental Sciences, School of Intellectual Property, Guilin University of Electronic Technology, Guilin, Guangxi, 541004, People's Republic of China
| | - Qing Huang
- School of Life and Environmental Sciences, School of Intellectual Property, Guilin University of Electronic Technology, Guilin, Guangxi, 541004, People's Republic of China
| | - Shuaikang Dong
- School of Life and Environmental Sciences, School of Intellectual Property, Guilin University of Electronic Technology, Guilin, Guangxi, 541004, People's Republic of China
| | - Yu Zhou
- School of Life and Environmental Sciences, School of Intellectual Property, Guilin University of Electronic Technology, Guilin, Guangxi, 541004, People's Republic of China
| | - Mei Lu
- Department of Clinical Laboratory, The 924, Hospital of Chinese People's Liberation Army Joint Logistic Support Force, Guilin, Guangxi, 541002, People's Republic of China
| | - Jintao Liang
- School of Life and Environmental Sciences, School of Intellectual Property, Guilin University of Electronic Technology, Guilin, Guangxi, 541004, People's Republic of China.
| | - Xueqing Zhou
- Department of Clinical Laboratory, The 924, Hospital of Chinese People's Liberation Army Joint Logistic Support Force, Guilin, Guangxi, 541002, People's Republic of China.
| | - Zhide Zhou
- School of Life and Environmental Sciences, School of Intellectual Property, Guilin University of Electronic Technology, Guilin, Guangxi, 541004, People's Republic of China.
| |
Collapse
|
3
|
Milan KL, Anuradha M, Ramkumar KM. Role of miR-125b-5p in modulating placental SIRT7 expression and its implications for lipid metabolism in gestational diabetes. J Reprod Immunol 2024; 167:104422. [PMID: 39755065 DOI: 10.1016/j.jri.2024.104422] [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: 11/21/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 01/06/2025]
Abstract
Gestational diabetes is marked impaired glucose tolerance, poses various adverse outcomes including increased BMI and obesity. These outcomes results from excess lipid accumulation which is marked by elevated triglycerides. In GDM, placenta exhibits altered lipid metabolism, including reduced fatty acid oxidation and increased triglyceride accumulation. These elevated triglycerides can also contribute to oxidative stress in GDM. SIRT7 plays an important role in regulating lipid metabolism and triglycerides levels. This study aimed to investigate the potential of miRNA to regulate the placental SIRT7 in GDM. PCR analysis reveals that SIRT7 expression along with oxidative stress markers elevated in GDM placenta. These elevated SIRT7 levels were positively correlated with BMI and triglycerides levels in GDM subjects. miR-125b-5p was identified to regulate SIRT7 mRNA using in-silico approaches. Expression levels of miR-125b-5p were found to be downregulated in GDM placenta and found to be negatively correlated with SIRT7 mRNA expression. To confirm the hypothesis BeWo cells were transfected with anti-miR-125b and miR-125b-mimic. Anti-miR overexpressed the SIRT7 expression where mimic dysregulated it. Additionally, overexpressing miR-125b-5p controlled the elevated SIRT7 caused by the exposure of high glucose in BeWo cells. Collectively this study indicated that miR-125b-5p may regulate lipid metabolism via SIRT7 contributing to GDM. These findings highlights the warrant of further research to develop the therapeutic approaches that target miR-125b-5p to reduce lipid accumulation and obesity in GDM.
Collapse
Affiliation(s)
- K L Milan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603 203, India
| | - M Anuradha
- Department of Obstetrics & Gynaecology, SRM Medical College Hospital and Research Centre, Kattankulathur, Tamil Nadu 603203, India
| | - Kunka Mohanram Ramkumar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603 203, India.
| |
Collapse
|
4
|
Liu S, Huang J, Luo J, Gao X, Song S, Bian Q, Weng Y, Chen J. LOX-1-Based Assembly Layer on Devices Surface to Promote Endothelial Repair and Reduce Complications for In Situ Interventional Plaque. Adv Healthc Mater 2024:e2403060. [PMID: 39692170 DOI: 10.1002/adhm.202403060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/05/2024] [Indexed: 12/19/2024]
Abstract
Rapid endothelialization and functional recovery are considered as promising methods to extend the long-term effectiveness of cardiovascular implant materials. LOX-1 participates in the initiation and development of atherosclerosis and is highly expressed in a variety of cells involved in atherosclerosis, hence it is feasible to accelerate the recovery of endothelial function and inhibit the development of existing plaques by regulating LOX-1. Herein, the surface is modified with Poly I, a LOX-1 inhibitor, using rich amino dendritic macromolecules (PAMAM) as the linker coating, to against the pathological microenvironment. Poly I modified surface resisted endothelial damage caused by oxidative stress through the LOX-1-NADPH signaling pathway and inhibited endothelial inflammation via the LOX-1-NF-κB signaling pathway. It also promoted endothelial cell migration and inhibited platelet adhesion. Moreover, the Poly I modified surface can inhibit oxLDL-induced macrophage foam cell formation and alleviate inflammation by modulating macrophage phenotypes. Poly I modified surface significantly reduced plaque burden after treatment of atherosclerotic model rats, most importantly, it significantly inhibited post-implantation-induced restenosis and thrombosis. In vivo and in vitro evaluations confirmed its safety and therapeutic efficacy against atherosclerosis. Overall, the multifunctional Poly I with pathological microenvironment regulation exhibits potential application value in the surface engineering of cardiovascular devices.
Collapse
Affiliation(s)
- Sainan Liu
- Key Laboratory of Advanced Technology for Materials of Chinese Education Ministry, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Jinquan Huang
- Key Laboratory of Advanced Technology for Materials of Chinese Education Ministry, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Jiayan Luo
- Key Laboratory of Advanced Technology for Materials of Chinese Education Ministry, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Xiaowa Gao
- Key Laboratory of Advanced Technology for Materials of Chinese Education Ministry, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Siqi Song
- Key Laboratory of Advanced Technology for Materials of Chinese Education Ministry, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Qihao Bian
- Key Laboratory of Advanced Technology for Materials of Chinese Education Ministry, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Yajun Weng
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
| | - Junying Chen
- Key Laboratory of Advanced Technology for Materials of Chinese Education Ministry, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| |
Collapse
|
5
|
Wang A, Wang C, Xuan B, Sun Y, Li B, Zhao Q, Yu R, Wang X, Zhu M, Wei J. The role of splicing events in the inflammatory response of atherosclerosis: molecular mechanisms and modulation. Front Immunol 2024; 15:1507420. [PMID: 39742258 PMCID: PMC11685076 DOI: 10.3389/fimmu.2024.1507420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/02/2024] [Indexed: 01/03/2025] Open
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by persistent inflammatory responses throughout all stages of its progression. Modulating these inflammatory responses is a promising avenue for the development of cardiovascular disease therapies. Splicing events modulate gene expression and diversify protein functionality, exerting pivotal roles in the inflammatory mechanisms underlying atherosclerosis. These insights may provide novel opportunities for developing anti-inflammatory therapies for this disease. This article systematically discusses the diverse splice variants and how splicing events impact the inflammatory response in atherosclerosis via endothelial cells, macrophages, and vascular smooth muscle cells, highlighting their underlying molecular mechanisms and implications. Furthermore, this study summarizes clinical evidence supporting splicing-related molecules as diagnostic biomarkers and therapeutic targets in atherosclerosis. Lastly, we outline the current challenges and future research directions concerning splicing events and inflammatory responses in atherosclerosis. This offers a novel perspective and evidence for formulating new therapeutic strategies aimed at lowering the risk of atherosclerosis.
Collapse
Affiliation(s)
- Aolong Wang
- Heart Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- First Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Chengzhi Wang
- Second Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Bihan Xuan
- College of Integrated Traditional Chinese and Western Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yanqin Sun
- Heart Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Bin Li
- Heart Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- Henan Evidence-Based Medicine Center of Chinese Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Qifei Zhao
- Heart Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Rui Yu
- Heart Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Xinlu Wang
- Heart Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Mingjun Zhu
- Heart Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Jingjing Wei
- Heart Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
6
|
Tian B, Ji J, Jin C. BATF alleviates ox-LDL-induced HCAEC injury by regulating SIRT1 expression in coronary heart disease. PLoS One 2024; 19:e0306514. [PMID: 39680523 DOI: 10.1371/journal.pone.0306514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 06/19/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Coronary heart disease (CHD) represents a significant global health concern, arising from an intricate interplay between genetic predisposition and environmental influences, with a pivotal involvement of oxidized low-density lipoprotein (ox-LDL) in the pathophysiology of it. We aimed to elucidate the synergistic dynamics of B cell activating transcription factor (BATF) and Sirtuin 1 (SIRT1) in cell injury caused by ox-LDL, reveal potential therapeutic strategies for CHD. METHODS The GSE42148 dataset was used to analyze Differentially expressed genes (DEGs) to construct a gene co-expression network. Then bioinformatics analysis was performed on key modules to select the BATF gene. In vitro experiments were conducted to investigate the protective impact of BATF against human coronary artery endothelial cells (HCAEC) injury induced by ox-LDL. Further investigations probed the synergistic impact of BATF and SIRT1 modulation on cellular apoptosis and damage in the presence of ox-LDL. RESULTS BATF was significantly down-regulated in the CHD sample of the GSE42148 dataset. In vitro assays have proven that BATF alleviates ox-LDL-induced HCAEC injury. Notably, BATF emerged as a pivotal regulator of SIRT1 expression post ox-LDL exposure. Subsequent experiments underscored the interplay between BATF and SIRT1 in mitigating ox-LDL-induced apoptosis and Lactate Dehydrogenase (LDH) activity elevation, highlighting their collaborative role in cellular protection. CONCLUSION The research findings suggested a prospective protective function of BATF in HCAEC injury induced by ox-LDL, likely through the mediation of SIRT1 regulation. These results could offer fresh perspectives on the etiology of CHD and possible treatment avenues.
Collapse
Affiliation(s)
- Bei Tian
- Nursing Department, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Jingyu Ji
- Nursing Department, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Can Jin
- Department of Cardiovascular Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| |
Collapse
|
7
|
Citrin KM, Chaube B, Fernández-Hernando C, Suárez Y. Intracellular endothelial cell metabolism in vascular function and dysfunction. Trends Endocrinol Metab 2024:S1043-2760(24)00296-0. [PMID: 39672762 DOI: 10.1016/j.tem.2024.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 11/04/2024] [Accepted: 11/11/2024] [Indexed: 12/15/2024]
Abstract
Endothelial cells (ECs) form the inner lining of blood vessels that is crucial for vascular function and homeostasis. They regulate vascular tone, oxidative stress, and permeability. Dysfunction leads to increased permeability, leukocyte adhesion, and thrombosis. ECs undergo metabolic changes in conditions such as wound healing, cancer, atherosclerosis, and diabetes, and can influence disease progression. We discuss recent research that has revealed diverse intracellular metabolic pathways in ECs that are tailored to their functional needs, including lipid handling, glycolysis, and fatty acid oxidation (FAO). Understanding EC metabolic signatures in health and disease will be crucial not only for basic biology but can also be exploited when designing new therapies to target EC-related functions in different vascular diseases.
Collapse
Affiliation(s)
- Kathryn M Citrin
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA; Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA; Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, CT, USA; Department of Cellular and Molecular Physiology, Yale University, New Haven, CT, USA
| | - Balkrishna Chaube
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA; Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA; Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, CT, USA; Indian Institute of Technology Dharwad, Karnataka, India
| | - Carlos Fernández-Hernando
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA; Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA; Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, CT, USA; Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Yajaira Suárez
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA; Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA; Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, CT, USA; Department of Pathology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
8
|
Lee YN, Wu YJ, Su CH, Wang BJ, Yang SH, Lee HI, Chou YH, Tien TY, Lin CF, Chan WH, Chung CH, Wang SW, Yeh HI. Fluorescent gold nanoclusters possess multiple actions against atherosclerosis. Redox Biol 2024; 78:103427. [PMID: 39566163 PMCID: PMC11612375 DOI: 10.1016/j.redox.2024.103427] [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: 09/20/2024] [Revised: 11/06/2024] [Accepted: 11/11/2024] [Indexed: 11/22/2024] Open
Abstract
Atherosclerosis caused major morbidity and mortality worldwide. Molecules possessing lipid-lowering and/or anti-inflammatory properties are potential druggable targets against atherosclerosis. We examined the anti-atherosclerotic effects of fluorescent gold nanoclusters (FANC), which were dihydrolipoic acid (DHLA)-capped 2-nm gold nanoparticles. We evaluated the 8-week effects of FANC in Western-type diet-fed ApoE-deficient mice by either continuous intraperitoneal delivery (20 μM, 50 μl weekly) or via drinking water (300 nM). FANC reduced aortic atheroma burden, serum total cholesterol, and oxidative stress markers malondialdehyde and 4-hydroxynonenal levels. FANC attenuated hepatic lipid deposit, with changed expression of lipid homeostasis-related genes HMGCR, SREBP, PCSK9, and LDLR in a pattern similar to mice treated with ezetimibe. FANC also inhibited intestinal cholesterol absorption, resembling the action of ezetimibe. The lipid-lowering and anti-atherosclerotic effects of FANC reappeared in Western-type diet-fed LDLr-deficient mice. FANC bound insulin receptor β (IRβ) via DHLA, leading to AKT activation. However, unlike insulin, which also bound IRβ to activate AKT to induce HO-1, activation of AKT by FANC was independent of HO-1 expression in human aortic endothelial cells (HAECs). Alternatively, FANC up-regulated NRF2, interfered the binding of KEAP1 to NRF2, and promoted KEAP1 degradation to free NRF2 for nuclear entry to induce HO-1 that suppressed the expression of ICAM-1 and VCAM-1. Consistently, FANC suppressed ox-LDL-induced enhanced attachment of THP-derived macrophages onto HAECs. In macrophages, FANC up-regulated ABCA1, and reversed ox-LDL-induced suppression of cholesterol efflux. FANC effected in vitro at nano moles. In conclusion, our findings showed novel actions and multiple mechanisms of FANC worked coherently against atherosclerosis.
Collapse
Affiliation(s)
- Yi-Nan Lee
- Cardiovascular Center, Department of Internal Medicine, and Department of Medical Research, MacKay Memorial Hospital, Taipei, 10449, Taiwan
| | - Yih-Jer Wu
- Cardiovascular Center, Department of Internal Medicine, and Department of Medical Research, MacKay Memorial Hospital, Taipei, 10449, Taiwan; Department of Medicine, MacKay Medical College, New Taipei City, 25245, Taiwan
| | - Cheng-Huang Su
- Cardiovascular Center, Department of Internal Medicine, and Department of Medical Research, MacKay Memorial Hospital, Taipei, 10449, Taiwan
| | - Bo-Jeng Wang
- Cardiovascular Center, Department of Internal Medicine, and Department of Medical Research, MacKay Memorial Hospital, Taipei, 10449, Taiwan
| | - Sheng-Hsun Yang
- Cardiovascular Center, Department of Internal Medicine, and Department of Medical Research, MacKay Memorial Hospital, Taipei, 10449, Taiwan
| | - Hsin-I Lee
- Department of Medicine, MacKay Medical College, New Taipei City, 25245, Taiwan
| | - Yen-Hung Chou
- Department of Medicine, MacKay Medical College, New Taipei City, 25245, Taiwan
| | - Ting-Yi Tien
- Cardiovascular Center, Department of Internal Medicine, and Department of Medical Research, MacKay Memorial Hospital, Taipei, 10449, Taiwan
| | - Chao-Feng Lin
- Cardiovascular Center, Department of Internal Medicine, and Department of Medical Research, MacKay Memorial Hospital, Taipei, 10449, Taiwan; Department of Medicine, MacKay Medical College, New Taipei City, 25245, Taiwan
| | - Wen-Hsiung Chan
- Department of Bioscience Technology and Center for Nanotechnology, Chung Yuan Christian University, Zhongbei Road, Zhongli District, Taoyuan City, 32023, Taiwan
| | - Ching-Hu Chung
- Department of Medicine, MacKay Medical College, New Taipei City, 25245, Taiwan
| | - Shin-Wei Wang
- Institute of Biomedical Sciences, MacKay Medical College, New Taipei City, 25245, Taiwan
| | - Hung-I Yeh
- Cardiovascular Center, Department of Internal Medicine, and Department of Medical Research, MacKay Memorial Hospital, Taipei, 10449, Taiwan.
| |
Collapse
|
9
|
Shilenok I, Kobzeva K, Soldatov V, Deykin A, Bushueva O. C11orf58 (Hero20) Gene Polymorphism: Contribution to Ischemic Stroke Risk and Interactions with Other Heat-Resistant Obscure Chaperones. Biomedicines 2024; 12:2603. [PMID: 39595169 PMCID: PMC11592265 DOI: 10.3390/biomedicines12112603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Recently identified Hero proteins, which possess chaperone-like functions, are promising candidates for research into atherosclerosis-related diseases, including ischemic stroke (IS). Methods: 2204 Russian subjects (917 IS patients and 1287 controls) were genotyped for fifteen common SNPs in Hero20 gene C11orf58 using probe-based PCR and the MassArray-4 system. Results: Six C11orf58 SNPs were significantly associated with an increased risk of IS in the overall group (OG) and significantly modified by smoking (SMK) and low fruit/vegetable intake (LFVI): rs10766342 (effect allele (EA) A; P(OG = 0.02; SMK = 0.009; LFVI = 0.04)), rs11024032 (EA T; P(OG = 0.01; SMK = 0.01; LFVI = 0.036)), rs11826990 (EA G; P(OG = 0.007; SMK = 0.004; LFVI = 0.03)), rs3203295 (EA C; P(OG = 0.016; SMK = 0.01; LFVI = 0.04)), rs10832676 (EA G; P(OG = 0.006; SMK = 0.002; LFVI = 0.01)), rs4757429 (EA T; P(OG = 0.02; SMK = 0.04; LFVI = 0.04)). The top ten intergenic interactions of Hero genes (two-, three-, and four-locus models) involved exclusively polymorphic loci of C11orf58 and C19orf53 and were characterized by synergic and additive (independent) effects between SNPs. Conclusions: Thus, C11orf58 gene polymorphism represents a major risk factor for IS. Bioinformatic analysis showed the involvement of C11orf58 SNPs in molecular mechanisms of IS mediated by their role in the regulation of redox homeostasis, inflammation, vascular remodeling, apoptosis, vasculogenesis, neurogenesis, lipid metabolism, proteostasis, hypoxia, cell signaling, and stress response. In terms of intergenic interactions, C11orf58 interacts most closely with C19orf53.
Collapse
Affiliation(s)
- Irina Shilenok
- Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 305041 Kursk, Russia
- Division of Neurology, Kursk Emergency Hospital, 305035 Kursk, Russia
| | - Ksenia Kobzeva
- Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 305041 Kursk, Russia
| | - Vladislav Soldatov
- Laboratory of Genome Editing for Biomedicine and Animal Health, Belgorod State National Research University, 308015 Belgorod, Russia
- Department of Pharmacology and Clinical Pharmacology, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Alexey Deykin
- Laboratory of Genome Editing for Biomedicine and Animal Health, Belgorod State National Research University, 308015 Belgorod, Russia
- Department of Pharmacology and Clinical Pharmacology, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Olga Bushueva
- Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 305041 Kursk, Russia
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 305041 Kursk, Russia
| |
Collapse
|
10
|
Chen F, Jiang R, Yu X. Circular RNA circ_0002984 Facilitates the Proliferation and Migration of Ox-LDL-Induced Vascular Smooth Muscle Cells via the Let-7a-5p/KLF5 Pathway. Cardiovasc Toxicol 2024; 24:1253-1267. [PMID: 39181944 DOI: 10.1007/s12012-024-09911-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 08/06/2024] [Indexed: 08/27/2024]
Abstract
Circular RNAs (circRNAs) play an important role in the progression of atherosclerosis (AS). This study aimed to explore the exact role and mechanism of circ_0002984 in oxidized low-density lipoprotein (ox-LDL)-mediated human vascular smooth muscle cells (HVSMCs). The model of smooth muscle cell phenotype switching was constructed by treating HVSMCs with ox-LDL. The levels of circ_0002984, let-7a-5p, and kruppel-like factor 5 (KLF5) were measured by quantitative real-time PCR or western blot assay. Cell proliferation, migration, and apoptosis were detected by Cell Counting Kit-8 (CCK-8), EdU staining, wound healing assay, transwell assay, and flow cytometry. The expression of cleaved-caspase-3 and KLF5 was examined by western blot. The relationship between let-7a-5p and circ_0002984 or KLF5 was verified by dual-luciferase reporter assay or RIP assay. The results showed that circ_0002984 and KLF5 were up-regulated, while let-7a-5p was down-regulated in AS patients and ox-LDL-disposed HVSMCs. Silence of circ_0002984 suppressed proliferation and migration, and promoted apoptosis in ox-LDL-stimulated HVSMCs. Moreover, circ_0002984 sponged let-7a-5p to regulate the proliferation, migration, and apoptosis in ox-LDL-resulted HVSMCs. In addition, KLF5 was a target of let-7a-5p and its overexpression reversed the effect of let-7a-5p on the proliferation, migration, and apoptosis in ox-LDL-treated HVSMCs. Also, circ_0002984 positively regulated KLF5 expression by absorbing let-7a-5p. The promotion effect of circ_0002984 on the proliferation and migration of ox-LDL-treated HVSMCs was reversed by KLF5 silencing. Taken together, depletion of circ_0002984 inhibited the proliferation and migration of ox-LDL-stimulated HVSMCs, which might be achieved by modulating the let-7a-5p/KLF5 axis.
Collapse
MESH Headings
- Lipoproteins, LDL/toxicity
- Humans
- Cell Proliferation/drug effects
- RNA, Circular/metabolism
- RNA, Circular/genetics
- Kruppel-Like Transcription Factors/genetics
- Kruppel-Like Transcription Factors/metabolism
- Cell Movement/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/drug effects
- Signal Transduction
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Apoptosis/drug effects
- Cells, Cultured
- Atherosclerosis/pathology
- Atherosclerosis/metabolism
- Atherosclerosis/genetics
- Gene Expression Regulation
Collapse
Affiliation(s)
- Feng Chen
- Department of Cardiovascular Medicine, Lishui People's Hospital, Lishui, Zhejiang, China
| | - Ruilai Jiang
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xiufeng Yu
- Department of Emergency Medicine, Lishui People's Hospital, No. 1188 Liyang Street, Yanquan Avenue, Liandu District, Lishui, 323050, Zhejiang, China.
| |
Collapse
|
11
|
Martino F, Niglio T, Martino E, Barilla' F, Guardamagna O, Paravati V, Bassareo PP. Awareness of cholesterol levels in 46,309 Italian children and adolescents unveils the tip of the iceberg. Eur J Pediatr 2024; 183:4747-4754. [PMID: 39207459 DOI: 10.1007/s00431-024-05745-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 08/17/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND AND AIMS Cardiovascular diseases (CVD) risk factors include high cholesterol. Children with total cholesterol (TC) levels ≥ 170 mg/dL are usually considered hypercholesterolemic. This study aimed at investigating the awareness of TC levels in a large Italian paediatric population and at looking for a possible correlation between their TC and TC in their parents' blood. METHODS AND RESULTS A survey was carried out in 46,309 subjects (mean age 9.7 ± 2.3 years; age range 6-14 years) to check the awareness of their own TC levels by using a personal and family medical history questionnaire. In 95.67% of the sample TC value was unknown. In 2.69% TC was < 170 mg/dL, whereas 1.64% were hypercholesterolemic (TC ≥ 170 mg/dL). A statistically significant correlation was found between children with normal TC values and physiological TC values in both parents (p < 0.0001). Again, a significant association between children with high TC and their parents with high TC was detected when parents were analysed separately (i.e. children with TC ≥ 170 mg/dl vs maternal TC ≥ 200 mg/dL: OR 2.01 (95% CI 1.61-2.49, p < 0.001); children with TC ≥ 200 mg/dl vs maternal TC ≥ 240 mg/dL: OR 3.14 (95% CI 2.14-4.6, p < 0.001); children with TC ≥ 170 mg/dl vs paternal TC ≥ 200 mg/dL: OR 2.39 (95% CI 1.91-2.98, p < 0.001); children with TC ≥ 200 mg/dl vs paternal TC ≥ 240 mg/dL: OR 3.85 (95% CI 2.70-5,.50, p < 0.001). CONCLUSION Just a minority of the investigated young patients knew their TC. This is worrisome. Children with normal TC values are more likely to be born from healthy parents with physiological TC. In addition, high TC in the enrolled subjects is significantly associated with high TC in their parents. Overall, these findings seem to highlight the importance of health education and genetics in TC pathogenesis.
Collapse
Affiliation(s)
- Francesco Martino
- Department of Internal Medicine, Anaesthesiology, and Cardiovascular Sciences, La Sapienza University of Rome, Rome, Italy
| | | | - Eliana Martino
- Department of Internal Medicine, Anaesthesiology, and Cardiovascular Sciences, La Sapienza University of Rome, Rome, Italy
| | - Francesco Barilla'
- Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Ornella Guardamagna
- Department of Public Health and Paediatric Sciences, University of Turin, Turin, Italy
| | - Vincenzo Paravati
- Department of Internal Medicine, Anaesthesiology, and Cardiovascular Sciences, La Sapienza University of Rome, Rome, Italy
| | - Pier Paolo Bassareo
- School of Medicine, University College of Dublin, Mater Misericordiae University Hospital and Children's Health Ireland at Crumlin, Dublin, Ireland.
| |
Collapse
|
12
|
He L, Chen S, Zhu X, He F. Gamma-glutamyl transferase to high-density lipoprotein cholesterol ratio: A valuable predictor of coronary heart disease incidence. Nutr Metab Cardiovasc Dis 2024:103775. [PMID: 39638678 DOI: 10.1016/j.numecd.2024.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 09/25/2024] [Accepted: 10/23/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND AND AIM Existing studies have found that serological markers for predicting coronary heart disease (CHD) have relatively low predictive value for the severity of coronary arteries and the types of CHD. GGT to HDL-C ratio (GHR) has been shown to be associated with T2DM and non-alcoholic fatty liver disease. Therefore, we explore the relationship among GHR, CHD and its subgroups. METHODS AND RESULTS The study retrospectively analyzed 2703 participants from August 2022 to August 2023. The patients were divided into CHD group (N = 1911) and control group (N = 792) according to the diagnostic criteria of CHD. Adjustments for all covariates found that GHR was an independent risk factor for CHD (OR: 1.025, 95 % CI 1.016-1.033) and had the highest AUC of 0.767 (95 % CI 0.744-0.790) in identifying CHD. Additionally, GHR was significantly associated with multi-vessel CHD (OR: 1.018, 95 % CI 1.012-1.023) and showed excellent diagnostic capability for patients with multi-vessel CHD (AUC: 0.638). Moreover, compared with chronic coronary syndromes (CCS) and unstable angina (UA) groups, the level of GHR was significantly increased in acute myocardial infarction (AMI) (ST elevation myocardial infarction and Non-ST elevation myocardial infarction) group (P < 0.05). GHR had the higher AUC in STMETI [0.819 (95 % CI 0.796-0.854)] and NASTEMI [0.792 (95 % CI 0.766-0.816)] than the CCS and UA groups. CONCLUSIONS Our study analyses found that GHR is an independent risk factor for CHD and can predict the severity of coronary artery stenosis. Moreover, GHR has a high predictive value for AMI than CCS and UA in CHD patients.
Collapse
Affiliation(s)
- Li He
- Department of Emergency, Wuhan Fourth Hospital, No. 473 Hanzheng Street, Qiaokou District, Wuhan, Hubei, 430033, China.
| | - Sisi Chen
- Department of Emergency, Wuhan Fourth Hospital, No. 473 Hanzheng Street, Qiaokou District, Wuhan, Hubei, 430033, China
| | - Xuan Zhu
- Department of Emergency, Wuhan Fourth Hospital, No. 473 Hanzheng Street, Qiaokou District, Wuhan, Hubei, 430033, China
| | - Fang He
- Department of Emergency, Wuhan Fourth Hospital, No. 473 Hanzheng Street, Qiaokou District, Wuhan, Hubei, 430033, China
| |
Collapse
|
13
|
Mosalmanzadeh N, Pence BD. Oxidized Low-Density Lipoprotein and Its Role in Immunometabolism. Int J Mol Sci 2024; 25:11386. [PMID: 39518939 PMCID: PMC11545486 DOI: 10.3390/ijms252111386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/04/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Modified cholesterols such as oxidized low-density lipoprotein (OxLDL) contribute to atherosclerosis and other disorders through the promotion of foam cell formation and inflammation. In recent years, it has become evident that immune cell responses to inflammatory molecules such as OxLDLs depend on cellular metabolic functions. This review examines the known effects of OxLDL on immunometabolism and immune cell responses in atherosclerosis and several other diseases. We additionally provide context on the relationship between OxLDL and aging/senescence and identify gaps in the literature and our current understanding in these areas.
Collapse
Affiliation(s)
| | - Brandt D. Pence
- College of Health Sciences and Center for Nutraceutical and Dietary Supplement Research, University of Memphis, Memphis, TN 38111, USA
| |
Collapse
|
14
|
Godinez-Mora S, Campos-Perez W, Perez-Robles M, Robles-Jimarez C, Muñoz-Hernandez A, Torres-Vanegas J, Martinez-Lopez E. Positive correlation between n-6 : n-3 PUFA ratio intake with serum oxHDL/HDL-c ratio in patients with coronary artery disease. Coron Artery Dis 2024:00019501-990000000-00280. [PMID: 39729580 DOI: 10.1097/mca.0000000000001437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2024]
Abstract
BACKGROUND Coronary artery disease (CAD) is one of the most prevalent cardiovascular diseases where serum lipoprotein oxidation plays a significant role. Polyunsaturated fatty acids (PUFA) n-6 : n-3 unbalance ratio consumption, affects lipoprotein oxidation, and inflammation processes. This study aimed to analyze the relationship between n-6 : n-3 PUFA ratio intake with oxidized lipoproteins in individuals with CAD. METHODS A cross-sectional study was performed including 105 subjects (51 diagnosed with CAD and 54 non-CAD) from western Mexico. Dietary information was collected using a habitual day food record. Serum oxidized low-density lipoprotein (oxLDL) and oxidized high-density lipoprotein (oxHDL) concentrations were quantified by enzyme linked immunosorbent assay. RESULTS CAD subjects had higher oxHDL/HDL cholesterol (HDL-c) ratio [0.102 (0.092-0.112) vs. 0.080 (0.070-0.090), P = 0.004] and oxLDL/LDL cholesterol (LDL-c) ratio [129.2 (108-150.4) vs. 59.7 (39.3-80), P < 0.001] compared to non-CAD subjects. Risk factors associated with CAD were a high n-6 : n-3 PUFA ratio (odds ratio, OR = 2.3, P = 0.046), hypoalphalipoproteinemia in men (OR = 3.2, P = 0.014), moderate/high tobacco index (OR = 6.33, P = 0.003), elevated waist circumference in women (OR = 7, P = 0.004), hypertension (OR = 21.14, P < 0.001), and type 2 diabetes (OR: 25, P < 0.001). The oxHDL/HDL-c ratio was positively associated with the n-6 : n-3 PUFA ratio [r2 = 28.3, B = 0.002 (0.001-0.003), P < 0.001] in CAD patients. CONCLUSIONS This study showed that a higher n-6 : n-3 PUFA ratio intake correlates with higher serum oxHDL/HDL-c in CAD patients.
Collapse
Affiliation(s)
- Sissi Godinez-Mora
- Departamento de Biología Molecular y Genómica, Instituto de Nutrigenética y Nutrigenómica Traslacional
- Doctorado en Ciencias de la Nutrición Traslacional, Departamento de la Reproducción Humana, Crecimiento y Desarrollo Infantil, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, México
| | - Wendy Campos-Perez
- Departamento de Biología Molecular y Genómica, Instituto de Nutrigenética y Nutrigenómica Traslacional
| | - Mariana Perez-Robles
- Departamento de Biología Molecular y Genómica, Instituto de Nutrigenética y Nutrigenómica Traslacional
| | - Cesar Robles-Jimarez
- Departamento de Biología Molecular y Genómica, Instituto de Nutrigenética y Nutrigenómica Traslacional
| | - Alejandra Muñoz-Hernandez
- Departamento de Biología Molecular y Genómica, Instituto de Nutrigenética y Nutrigenómica Traslacional
| | - Joel Torres-Vanegas
- Departamento de Biología Molecular y Genómica, Instituto de Nutrigenética y Nutrigenómica Traslacional
- Doctorado en Ciencias de la Nutrición Traslacional, Departamento de la Reproducción Humana, Crecimiento y Desarrollo Infantil, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, México
| | - Erika Martinez-Lopez
- Departamento de Biología Molecular y Genómica, Instituto de Nutrigenética y Nutrigenómica Traslacional
| |
Collapse
|
15
|
Udayan SP, Hariharan S, Nevin KG. Multifaceted bioactivity of marine fungal derived secondary metabolite, xyloketal B -a review. Toxicol Res (Camb) 2024; 13:tfae156. [PMID: 39345795 PMCID: PMC11425363 DOI: 10.1093/toxres/tfae156] [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: 04/01/2024] [Revised: 09/03/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024] Open
Abstract
Background A growing number of findings have focused on the distinctive physiochemical characteristics that marine microorganisms have acquired as a result of their adaptation to the challenging conditions inherent in the marine environment. It has been established that the marine environment is a very rich source of bioactive substances with a variety of biological effects and structural diversity. A major discovery was the extraction of xyloketals from Xylaria sp. Numerous thorough studies have subsequently been carried out to determine the medicinal potential of these bioactive components. Xyloketals are thought to be a very promising and significant class of naturally occurring substances with a wide range of potent biological activities, such as radical scavenging, suppression of cell proliferation, reduction of neonatal hypoxic-ischemic brain injury, antioxidant activity, inhibition of acetylcholine esterase, inhibition of L-calcium channels, and others. Xyloketal B is one of the most potent molecules with significant therapeutic properties among the numerous variants discovered. Conclusion This review summarizes the structural characterization of all naturally occurring xyloketal compounds, especially the B derivative with an emphasis on their bioactivity and provides an outline of how xyloketals operate in diverse disease scenarios.
Collapse
Affiliation(s)
- Sreelekshmi Puthuvalnikarthil Udayan
- Department of Marine Biosciences, Faculty of Ocean Science and Technology, Kerala University of Fisheries and Ocean Studies, Panangad PO, Kochi, Ernakulam District, Kerala 682506, India
| | - Sini Hariharan
- Department of Biochemistry, Government College, Kariavattom PO, Thiruvananthapuram District, Kerala 695581, India
| | - Kottayath Govindan Nevin
- Department of Marine Biosciences, Faculty of Ocean Science and Technology, Kerala University of Fisheries and Ocean Studies, Panangad PO, Kochi, Ernakulam District, Kerala 682506, India
- Centre for Bioactive Substances from Marine Organisms, Kerala University of Fisheries and Ocean Studies, Kochi 682506, India
| |
Collapse
|
16
|
Al-Rashed F, AlSaeed H, Almansour N, Al-Mulla F, Hannun YA, Ahmad R. IL-6R (trans-signaling) is a key regulator of reverse cholesterol transport in lipid-laden macrophages. Clin Immunol 2024; 267:110351. [PMID: 39216780 PMCID: PMC11402558 DOI: 10.1016/j.clim.2024.110351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/14/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Atherosclerosis is a cardiovascular disease caused by cholesterol-laden arterial plaques. This study evaluated the correlation between interleukin-6 (IL-6), its receptors (IL6R/CD126), and glycoprotein 130 (gp130) alongside atherosclerosis biomarkers in a cohort of 142 subjects, equally divided between lean and obese individuals. Subsequent analyses used THP-1-derived macrophages to assess the biochemical impact of inhibiting IL-6 receptors. IL-6 secretion increased with atherosclerosis in obese subjects, while IL6R/CD126 and gp130 on monocytes decreased. Pharmacological gp130 inhibition altered lipid metabolism, increasing LDLR gene expression and cholesterol synthesis via SREBF2 and mevalonate kinase, along with HMG-CoA reductase at protein levels. gp130-deficient cells produced more cholesterol and had lower ABCA1 levels, suggesting hindered cholesterol efflux. Filipin III staining confirmed cholesterol retention in gp130-inhibited cells. Ex-vivo investigation on lean PBMCs further defined the impact of gp130 inhibition on the reduction of cholesterol efflux. Our results indicates gp130 is crucial for macrophage reverse cholesterol transport and may be a target for atherosclerosis treatments.
Collapse
Affiliation(s)
- Fatema Al-Rashed
- Immunology and Microbiology Department, Dasman Diabetes Institute, Al-Soor Street, Dasman, Kuwait, PO BOX 1180, Dasman 15462, Kuwait.
| | - Halemah AlSaeed
- Immunology and Microbiology Department, Dasman Diabetes Institute, Al-Soor Street, Dasman, Kuwait, PO BOX 1180, Dasman 15462, Kuwait
| | - Nourah Almansour
- Immunology and Microbiology Department, Dasman Diabetes Institute, Al-Soor Street, Dasman, Kuwait, PO BOX 1180, Dasman 15462, Kuwait
| | - Fahd Al-Mulla
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Kuwait, Kuwait
| | - Yusuf A Hannun
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11794, USA
| | - Rasheed Ahmad
- Immunology and Microbiology Department, Dasman Diabetes Institute, Al-Soor Street, Dasman, Kuwait, PO BOX 1180, Dasman 15462, Kuwait
| |
Collapse
|
17
|
Ajoolabady A, Pratico D, Ren J. Endothelial dysfunction: mechanisms and contribution to diseases. Acta Pharmacol Sin 2024; 45:2023-2031. [PMID: 38773228 PMCID: PMC11420364 DOI: 10.1038/s41401-024-01295-8] [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: 02/18/2024] [Accepted: 04/16/2024] [Indexed: 05/23/2024] Open
Abstract
The endothelium, lining the inner surface of blood vessels and spanning approximately 3 m2, serves as the largest organ in the body. Comprised of endothelial cells, the endothelium interacts with other bodily components including the bloodstream, circulating cells, and the lymphatic system. Functionally, the endothelium primarily synchronizes vascular tone (by balancing vasodilation and vasoconstriction) and prevents vascular inflammation and pathologies. Consequently, endothelial dysfunction disrupts vascular homeostasis, leading to vascular injuries and diseases such as cardiovascular, cerebral, and metabolic diseases. In this opinion/perspective piece, we explore the recently identified mechanisms of endothelial dysfunction across various disease subsets and critically evaluate the strengths and limitations of current therapeutic interventions at the pre-clinical level.
Collapse
Affiliation(s)
- Amir Ajoolabady
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Domenico Pratico
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Jun Ren
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
| |
Collapse
|
18
|
Vass Z, Shenker-Horváth K, Bányai B, Vető KN, Török V, Gém JB, Nádasy GL, Kovács KB, Horváth EM, Jakus Z, Hunyady L, Szekeres M, Dörnyei G. Investigating the Role of Cannabinoid Type 1 Receptors in Vascular Function and Remodeling in a Hypercholesterolemic Mouse Model with Low-Density Lipoprotein-Cannabinoid Type 1 Receptor Double Knockout Animals. Int J Mol Sci 2024; 25:9537. [PMID: 39273484 PMCID: PMC11395437 DOI: 10.3390/ijms25179537] [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: 07/16/2024] [Revised: 08/24/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
Hypercholesterolemia forms the background of several cardiovascular pathologies. LDL receptor-knockout (LDLR-KO) mice kept on a high-fat diet (HFD) develop high cholesterol levels and atherosclerosis (AS). Cannabinoid type 1 receptors (CB1Rs) induce vasodilation, although their role in cardiovascular pathologies is still controversial. We aimed to reveal the effects of CB1Rs on vascular function and remodeling in hypercholesterolemic AS-prone LDLR-KO mice. Experiments were performed on a newly established LDLR and CB1R double-knockout (KO) mouse model, in which KO and wild-type (WT) mice were kept on an HFD or a control diet (CD) for 5 months. The vascular functions of abdominal aorta rings were tested with wire myography. The vasorelaxation effects of acetylcholine (Ach, 1 nM-1 µM) were obtained after phenylephrine precontraction, which was repeated with inhibitors of nitric oxide synthase (NOS) and cyclooxygenase (COX), Nω-nitro-L-arginine (LNA), and indomethacin (INDO), respectively. Blood pressure was measured with the tail-cuff method. Immunostaining of endothelial NOS (eNOS) was carried out. An HFD significantly elevated the cholesterol levels in the LDLR-KO mice more than in the corresponding WT mice (mean values: 1039 ± 162 mg/dL vs. 91 ± 18 mg/dL), and they were not influenced by the presence of the CB1R gene. However, with the defect of the CB1R gene, damage to the Ach relaxation ability was moderated. The blood pressure was higher in the LDLR-KO mice compared to their WT counterparts (systolic/diastolic values: 110/84 ± 5.8/6.8 vs. 102/80 ± 3.3/2.5 mmHg), which was significantly elevated with an HFD (118/96 ± 1.9/2 vs. 100/77 ± 3.4/3.1 mmHg, p < 0.05) but attenuated in the CB1R-KO HFD mice. The expression of eNOS was depressed in the HFD WT mice compared to those on the CD, but it was augmented if CB1R was knocked out. This newly established double-knockout mouse model provides a tool for studying the involvement of CB1Rs in the development of hypercholesterolemia and atherosclerosis. Our results indicate that knocking out the CB1R gene significantly attenuates vascular damage in hypercholesterolemic mice.
Collapse
MESH Headings
- Animals
- Male
- Mice
- Acetylcholine/pharmacology
- Atherosclerosis/metabolism
- Atherosclerosis/genetics
- Atherosclerosis/pathology
- Atherosclerosis/etiology
- Diet, High-Fat/adverse effects
- Disease Models, Animal
- Hypercholesterolemia/metabolism
- Hypercholesterolemia/genetics
- Hypercholesterolemia/pathology
- Mice, Inbred C57BL
- Mice, Knockout
- Nitric Oxide Synthase Type III/metabolism
- Receptor, Cannabinoid, CB1/metabolism
- Receptor, Cannabinoid, CB1/genetics
- Receptors, LDL/genetics
- Receptors, LDL/metabolism
- Receptors, LDL/deficiency
- Vascular Remodeling/drug effects
- Vasodilation/drug effects
Collapse
Affiliation(s)
- Zsolt Vass
- Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, 17 Vas Street, 1088 Budapest, Hungary; (Z.V.); (K.S.-H.); (K.N.V.); (V.T.); (G.D.)
| | - Kinga Shenker-Horváth
- Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, 17 Vas Street, 1088 Budapest, Hungary; (Z.V.); (K.S.-H.); (K.N.V.); (V.T.); (G.D.)
- Center for Sports Nutrition Science, Hungarian University of Sports Science, 42-48 Alkotás Street, 1123 Budapest, Hungary
| | - Bálint Bányai
- Department of Physiology, Faculty of Medicine, Semmelweis University, 37-47 Tűzoltó Street, 1094 Budapest, Hungary; (B.B.); (J.B.G.); (G.L.N.); (K.B.K.); (E.M.H.); (Z.J.); (L.H.)
| | - Kinga Nóra Vető
- Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, 17 Vas Street, 1088 Budapest, Hungary; (Z.V.); (K.S.-H.); (K.N.V.); (V.T.); (G.D.)
| | - Viktória Török
- Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, 17 Vas Street, 1088 Budapest, Hungary; (Z.V.); (K.S.-H.); (K.N.V.); (V.T.); (G.D.)
| | - Janka Borbála Gém
- Department of Physiology, Faculty of Medicine, Semmelweis University, 37-47 Tűzoltó Street, 1094 Budapest, Hungary; (B.B.); (J.B.G.); (G.L.N.); (K.B.K.); (E.M.H.); (Z.J.); (L.H.)
| | - György L. Nádasy
- Department of Physiology, Faculty of Medicine, Semmelweis University, 37-47 Tűzoltó Street, 1094 Budapest, Hungary; (B.B.); (J.B.G.); (G.L.N.); (K.B.K.); (E.M.H.); (Z.J.); (L.H.)
| | - Kinga Bernadett Kovács
- Department of Physiology, Faculty of Medicine, Semmelweis University, 37-47 Tűzoltó Street, 1094 Budapest, Hungary; (B.B.); (J.B.G.); (G.L.N.); (K.B.K.); (E.M.H.); (Z.J.); (L.H.)
| | - Eszter Mária Horváth
- Department of Physiology, Faculty of Medicine, Semmelweis University, 37-47 Tűzoltó Street, 1094 Budapest, Hungary; (B.B.); (J.B.G.); (G.L.N.); (K.B.K.); (E.M.H.); (Z.J.); (L.H.)
| | - Zoltán Jakus
- Department of Physiology, Faculty of Medicine, Semmelweis University, 37-47 Tűzoltó Street, 1094 Budapest, Hungary; (B.B.); (J.B.G.); (G.L.N.); (K.B.K.); (E.M.H.); (Z.J.); (L.H.)
| | - László Hunyady
- Department of Physiology, Faculty of Medicine, Semmelweis University, 37-47 Tűzoltó Street, 1094 Budapest, Hungary; (B.B.); (J.B.G.); (G.L.N.); (K.B.K.); (E.M.H.); (Z.J.); (L.H.)
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, 2 Magyar Tudósok Körútja, 1117 Budapest, Hungary
| | - Mária Szekeres
- Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, 17 Vas Street, 1088 Budapest, Hungary; (Z.V.); (K.S.-H.); (K.N.V.); (V.T.); (G.D.)
- Department of Physiology, Faculty of Medicine, Semmelweis University, 37-47 Tűzoltó Street, 1094 Budapest, Hungary; (B.B.); (J.B.G.); (G.L.N.); (K.B.K.); (E.M.H.); (Z.J.); (L.H.)
| | - Gabriella Dörnyei
- Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, 17 Vas Street, 1088 Budapest, Hungary; (Z.V.); (K.S.-H.); (K.N.V.); (V.T.); (G.D.)
| |
Collapse
|
19
|
Zhang M, Li J, Hu W. The complex interplay between ferroptosis and atherosclerosis. Biomed Pharmacother 2024; 178:117183. [PMID: 39079265 DOI: 10.1016/j.biopha.2024.117183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 08/25/2024] Open
Abstract
Atherosclerosis, characterized by the accumulation of plaque within the arterial walls, is an intricate cardiovascular disease that often results in severe health issues. Recent studies have emphasized the importance of ferroptosis, a controlled type of cell death dependent on iron, as a critical factor in this disease state. Ferroptosis, distinguished by its reliance on iron and the accumulation of lipid hydroperoxides, offers a unique insight into the pathology of atherosclerotic lesions. This summary encapsulates the current knowledge of the intricate role ferroptosis plays in the onset and progression of atherosclerosis. It explores the molecular processes through which lipid peroxidation and iron metabolism contribute to the development of atheromatous plaques and evaluates the possibility of utilizing ferroptosis as a novel treatment approach for atherosclerosis. By illuminating the intricate relationship between ferroptosis-related processes and atherosclerosis, this review paves the way for future clinical applications and personalized medicine approaches aimed at alleviating the effects of atherosclerosis.
Collapse
Affiliation(s)
- Mao Zhang
- Department of Vascular Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Jiangping Li
- Department of Oncological Radiotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Hu
- Department of Vascular Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
20
|
Thej C, Kishore R. Epigenetic regulation of sex dimorphism in cardiovascular health. Can J Physiol Pharmacol 2024; 102:498-510. [PMID: 38427976 DOI: 10.1139/cjpp-2023-0406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2024]
Abstract
Cardiovascular diseases (CVDs) remain the leading cause of morbidity and mortality, affecting people of all races, ages, and sexes. Substantial sex dimorphism exists in the prevalence, manifestation, and outcomes of CVDs. Understanding the role of sex hormones as well as sex-hormone-independent epigenetic mechanisms could play a crucial role in developing effective and sex-specific cardiovascular therapeutics. Existing research highlights significant disparities in sex hormones, epigenetic regulators, and gene expression related to cardiac health, emphasizing the need for a nuanced understanding of these variations between men and women. Despite these differences, current treatment approaches for CVDs often lack sex-specific considerations. A pivotal shift toward personalized medicine, informed by comprehensive insights into sex-specific DNA methylation, histone modifications, and non-coding RNA dynamics, holds the potential to revolutionize CVD management. By understanding sex-specific epigenetic complexities, independent of sex hormone influence, future cardiovascular research can be tailored to achieve effective diagnostic and therapeutic interventions for both men and women. This review summarizes the current knowledge and gaps in epigenetic mechanisms and sex dimorphism implicated in CVDs.
Collapse
Affiliation(s)
- Charan Thej
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Raj Kishore
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
- Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
21
|
Zhao Y, Wang L, Huang Y, Evans PC, Little PJ, Tian X, Weng J, Xu S. Anthocyanins in Vascular Health and Disease: Mechanisms of Action and Therapeutic Potential. J Cardiovasc Pharmacol 2024; 84:289-302. [PMID: 39240726 DOI: 10.1097/fjc.0000000000001602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 05/31/2024] [Indexed: 09/08/2024]
Abstract
ABSTRACT Unhealthy lifestyles have placed a significant burden on individuals' cardiovascular health. Anthocyanins are water-soluble flavonoid pigments found in a wide array of common foods and fruits. Anthocyanins have the potential to contribute to the prevention and treatment of cardiovascular disease by improving lipid profiles and vascular function, reducing blood glucose levels and blood pressure, and inhibiting inflammation. These actions have been demonstrated in numerous clinical and preclinical studies. At the cellular and molecular level, anthocyanins and their metabolites could protect endothelial cells from senescence, apoptosis, and inflammation by activating the phosphoinositide 3-kinase/protein kinase B/endothelial nitric oxide synthases, silent information regulator 1 (SIRT1), or nuclear factor erythroid2-related factor 2 pathways and inhibiting the nuclear factor kappa B, Bax, or P38 mitogen-activated protein kinase pathways. Furthermore, anthocyanins prevent vascular smooth muscle cell from platelet-derived growth factor -induced or tumor necrosis factor-α-induced proliferation and migration by inhibiting the focal adhesion kinase and extracellular regulated protein kinases signaling pathways. Anthocyanins could also attenuate vascular inflammation by reducing the formation of oxidized lipids, preventing leukocyte adhesion and infiltration of the vessel wall, and macrophage phagocytosis of deposited lipids through reducing the expression of cluster of differentiation 36 and increasing the expression of ATP-binding cassette subfamily A member 1 and ATP-binding cassette subfamily G member 1. At the same time, anthocyanins could lower the risk of thrombosis by inhibiting platelet activation and aggregation through down-regulating P-selectin, transforming growth factor-1, and CD40L. Thus, the development of anthocyanin-based supplements or derivative drugs could provide new therapeutic approaches to the prevention and treatment of vascular diseases.
Collapse
Affiliation(s)
- Yaping Zhao
- Department of Endocrinology, Institute of Endocrine and Metabolic Disease, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, Anhui, China
- Anhui Provincial Key Laboratory of Metabolic Health and Panvascular Diseases, Hefei, Anhui, China
| | - Li Wang
- Anhui Provincial Key Laboratory of Metabolic Health and Panvascular Diseases, Hefei, Anhui, China
| | - Yu Huang
- Anhui Provincial Key Laboratory of Metabolic Health and Panvascular Diseases, Hefei, Anhui, China
| | - Paul C Evans
- Department of Biomedical Sciences, City University of Hong Kong, China
| | - Peter J Little
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Xiaoyu Tian
- School of Pharmacy, The University of Queensland, Queensland, Australia; and
| | - Jianping Weng
- Department of Endocrinology, Institute of Endocrine and Metabolic Disease, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, Anhui, China
- Anhui Provincial Key Laboratory of Metabolic Health and Panvascular Diseases, Hefei, Anhui, China
| | - Suowen Xu
- Department of Endocrinology, Institute of Endocrine and Metabolic Disease, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, Anhui, China
- Anhui Provincial Key Laboratory of Metabolic Health and Panvascular Diseases, Hefei, Anhui, China
| |
Collapse
|
22
|
Gouaref I, Otmane A, Makrelouf M, Abderrhmane SA, Haddam AEM, Koceir EA. Crucial Interactions between Altered Plasma Trace Elements and Fatty Acids Unbalance Ratio to Management of Systemic Arterial Hypertension in Diabetic Patients: Focus on Endothelial Dysfunction. Int J Mol Sci 2024; 25:9288. [PMID: 39273236 PMCID: PMC11395650 DOI: 10.3390/ijms25179288] [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: 07/18/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
The coexistence of SAH with T2DM is a common comorbidity. In this study, we investigated the link between altered plasma antioxidant trace elements (ATE: manganese, selenium, zinc, and copper) and fatty acids ratio (FAR: polyunsaturated/saturated) imbalance as transition biomarkers between vascular pathology (SAH) to metabolic pathology (T2DM). Our data revealed strong correlation between plasma ATE and FAR profile, which is modified during SAH-T2DM association compared to the healthy group. This relationship is mediated by lipotoxicity (simultaneously prominent visceral adipose tissue lipolysis, significant flow of non-esterified free fatty acids release, TG-Chol-dyslipidemia, high association of total SFA, palmitic acid, arachidonic acid, and PUFA ω6/PUFA ω3; drop in tandem of PUFA/SFA and EPA + DHA); oxidative stress (lipid peroxidation confirmed by TAS depletion and MDA rise, concurrent drop of Zn/Cu-SOD, GPx, GSH, Se, Zn, Se/Mn, Zn/Cu; concomitant enhancement of Cu, Mn, and Fe); endothelial dysfunction (endotheline-1 increase); athero-thrombogenesis risk (concomitant rise of ApoB100/ApoA1, Ox-LDL, tHcy, and Lp(a)), and inflammation (higher of Hs-CRP, fibrinogen and ferritin). Our study opens to new therapeutic targets and to better dietary management, such as to establishing dietary ATE and PUFA ω6/PUFA ω3 or PUFA/SFA reference values for atherosclerotic risk prevention in hypertensive/diabetic patients.
Collapse
Affiliation(s)
- Ines Gouaref
- Bioenergetics and Intermediary Metabolism Team, Laboratory of Biology and Organism Physiology, Biological Sciences Faculty, Nutrition and Pathologies Post Graduate School, Houari Boumediene University of Sciences and Technology (USTHB), Bab Ezzouar, Algiers 16123, Algeria
- Tamayouz Laboratory, Centre de Recherche en Biotechnologie (CRBT), Ali Mendjli Nouvelle Ville UV 03 BP E73, Constantine 25000, Algeria
| | - Amel Otmane
- Biochemistry and Genetics Laboratory, University Hospital Center, Mohamed Lamine Debaghine, Bab El Oued, Algiers 16000, Algeria
| | - Mohamed Makrelouf
- Biochemistry and Genetics Laboratory, University Hospital Center, Mohamed Lamine Debaghine, Bab El Oued, Algiers 16000, Algeria
| | - Samir Ait Abderrhmane
- Diabetology Unit, University Hospital Center, Mohamed Seghir Nekkache (ex. HCA de Aïn Naâdja), Algiers 16208, Algeria
| | - Ali El Mahdi Haddam
- Diabetology Unit, University Hospital Center, Mohamed Lamine Debaghine, Algiers I-University, Bab El Oued, Algiers 16000, Algeria
| | - Elhadj-Ahmed Koceir
- Bioenergetics and Intermediary Metabolism Team, Laboratory of Biology and Organism Physiology, Biological Sciences Faculty, Nutrition and Pathologies Post Graduate School, Houari Boumediene University of Sciences and Technology (USTHB), Bab Ezzouar, Algiers 16123, Algeria
- Tamayouz Laboratory, Centre de Recherche en Biotechnologie (CRBT), Ali Mendjli Nouvelle Ville UV 03 BP E73, Constantine 25000, Algeria
| |
Collapse
|
23
|
Nam U, Kim J, Yi HG, Jeon JS. Investigation of the Dysfunction Caused by High Glucose, Advanced Glycation End Products, and Interleukin-1 Beta and the Effects of Therapeutic Agents on the Microphysiological Artery Model. Adv Healthc Mater 2024; 13:e2302682. [PMID: 38575148 DOI: 10.1002/adhm.202302682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 03/31/2024] [Indexed: 04/06/2024]
Abstract
Diabetes mellitus (DM) has substantial global implications and contributes to vascular inflammation and the onset of atherosclerotic cardiovascular diseases. However, translating the findings from animal models to humans has inherent limitations, necessitating a novel platform. Therefore, herein, an arterial model is established using a microphysiological system. This model successfully replicates the stratified characteristics of human arteries by integrating collagen, endothelial cells (ECs), and vascular smooth muscle cells (VSMCs). Perfusion via a peristaltic pump shows dynamic characteristics distinct from those of static culture models. High glucose, advanced glycation end products (AGEs), and interleukin-1 beta are employed to stimulate diabetic conditions, resulting in notable cellular changes and different levels of cytokines and nitric oxide. Additionally, the interactions between the disease models and oxidized low-density lipoproteins (LDL) are examined. Finally, the potential therapeutic effects of metformin, atorvastatin, and diphenyleneiodonium are investigated. Metformin and diphenyleneiodonium mitigate high-glucose- and AGE-associated pathological changes, whereas atorvastatin affects only the morphology of ECs. Altogether, the arterial model represents a pivotal advancement, offering a robust and insightful platform for investigating cardiovascular diseases and their corresponding drug development.
Collapse
Affiliation(s)
- Ungsig Nam
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
- Center for Scientific Instrumentation, Korea Basic Science Institute (KBSI), Daejeon, 34133, Republic of Korea
| | - Jaesang Kim
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Hee-Gyeong Yi
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Jessie S Jeon
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| |
Collapse
|
24
|
Stringham NT, Green M, Roche W, Prado-Cabrero A, Mulcahy R, Nolan J. Lutein, zeaxanthin, and meso-zeaxanthin supplementation attenuates inflammatory cytokines and markers of oxidative cardiovascular processes in humans. Nutr Metab Cardiovasc Dis 2024; 34:1976-1983. [PMID: 38890092 DOI: 10.1016/j.numecd.2024.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 05/03/2024] [Accepted: 05/06/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND AND AIMS Systemic inflammation and oxidation are primary contributors to the development of atherosclerosis. Oxidation of low-density lipoprotein (LDL) particles within the vascular endothelium has been hypothesized to be an initial step in the formation of atherosclerotic plaques, with inflammatory cytokines serving as the signaling mechanism for concomitant macrophage activation. Supplementation with the antioxidative macular xanthophylls (lutein [L], zeaxanthin [Z], and meso-zeaxanthin [MZ]) has been shown to aid in the reduction of inflammatory physiologic responses; therefore, we hypothesized that in our study population, supplementation with these xanthophylls would facilitate a systemic reduction in markers of inflammation and cardiovascular lipid oxidation. METHODS AND RESULTS In this double-blind placebo-controlled supplementation study, participants were randomly allocated to receive the active intervention containing L (10 mg) + MZ (10 mg) + Z (2 mg) or placebo (containing sunflower oil). Serum concentrations of carotenoids (assessed by HPLC), inflammatory cytokines (IL-6, IL-1β, TNF-α) and oxidized LDL (OxLDL; by solid-phase sandwich ELISA) were measured at baseline and at 6-months. Results showed that over the supplementation period, compared to placebo, the active group demonstrated statistically significant increases in serum concentrations of L, Z, & MZ (p < 0.05), reductions in inflammatory cytokines IL-1β (p < 0.001) and TNF-α (p = 0.003), as well as a corresponding reduction in serum OxLDL (p = 0.009). CONCLUSIONS Our data show that L, Z, & MZ supplementation results in decreased serum IL-1β, TNF-α, and OxLDL. This suggests that these carotenoids are acting systemically to attenuate oxidative lipid products and inflammation, thus reducing their contribution to atherosclerotic plaque formation.
Collapse
Affiliation(s)
- Nicole T Stringham
- Nutrition Research Centre Ireland (NRCI), Southeast Technical University, Waterford, Ireland; Northern Arizona University, Flagstaff, AZ, USA.
| | - Marina Green
- Nutrition Research Centre Ireland (NRCI), Southeast Technical University, Waterford, Ireland
| | - Warren Roche
- Nutrition Research Centre Ireland (NRCI), Southeast Technical University, Waterford, Ireland
| | - Alfonso Prado-Cabrero
- Nutrition Research Centre Ireland (NRCI), Southeast Technical University, Waterford, Ireland
| | - Riona Mulcahy
- Nutrition Research Centre Ireland (NRCI), Southeast Technical University, Waterford, Ireland
| | - John Nolan
- Nutrition Research Centre Ireland (NRCI), Southeast Technical University, Waterford, Ireland
| |
Collapse
|
25
|
Ma Y, Lai J, Wan Q, Sun L, Wang Y, Li X, Zhang Q, Wu J. Exploring the common mechanisms and biomarker ST8SIA4 of atherosclerosis and ankylosing spondylitis through bioinformatics analysis and machine learning. Front Cardiovasc Med 2024; 11:1421071. [PMID: 39131703 PMCID: PMC11310936 DOI: 10.3389/fcvm.2024.1421071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 07/08/2024] [Indexed: 08/13/2024] Open
Abstract
Background Atherosclerosis (AS) is a major contributor to cerebrovascular and cardiovascular events. There is growing evidence that ankylosing spondylitis is closely linked to AS, often co-occurring with it; however, the shared pathogenic mechanisms between the two conditions are not well understood. This study employs bioinformatics approaches to identify common biomarkers and pathways between AS and ankylosing spondylitis. Methods Gene expression datasets for AS (GSE100927, GSE28829, GSE155512) and ankylosing spondylitis (GSE73754, GSE25101) were obtained from the Gene Expression Omnibus (GEO). Differential expression genes (DEGs) and module genes for AS and ankylosing spondylitis were identified using the Limma R package and weighted gene co-expression network analysis (WGCNA) techniques, respectively. The machine learning algorithm SVM-RFE was applied to pinpoint promising biomarkers, which were then validated in terms of their expression levels and diagnostic efficacy in AS and ankylosing spondylitis, using two separate GEO datasets. Furthermore, the interaction of the key biomarker with the immune microenvironment was investigated via the CIBERSORT algorithm, single-cell analysis was used to identify the locations of common diagnostic markers. Results The dataset GSE100927 contains 524 DEGs associated with AS, whereas dataset GSE73754 includes 1,384 genes categorized into modules specific to ankylosing spondylitis. Analysis of these datasets revealed an overlap of 71 genes between the DEGs of AS and the modular genes of ankylosing spondylitis. Utilizing the SVM-RFE algorithm, 15 and 24 central diagnostic genes were identified in datasets GSE100927 and GSE73754, respectively. Further validation of six key genes using external datasets confirmed ST8SIA4 as a common diagnostic marker for both conditions. Notably, ST8SIA4 is upregulated in samples from both diseases. Additionally, ROC analysis confirmed the robust diagnostic utility of ST8SIA4. Moreover, analysis through CIBERSORT suggested an association of the ST8SIA4 gene with the immune microenvironment in both disease contexts. Single-cell analysis revealed that ST8SIA4 is primarily expressed in Macrophages, Monocytes, T cells, and CMPs. Conclusion This study investigates the role of ST8SIA4 as a common diagnostic gene and the involvement of the lysosomal pathway in both AS and ankylosing spondylitis. The findings may yield potential diagnostic biomarkers and offer new insights into the shared pathogenic mechanisms underlying these conditions.
Collapse
Affiliation(s)
- Yirong Ma
- Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Junyu Lai
- Cardiology Department, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Qiang Wan
- Cardiology Department, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Liqiang Sun
- Cardiology Department, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Yang Wang
- Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Xingliang Li
- Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Qinhe Zhang
- Department of Acupuncture and Tuina, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Jianguang Wu
- Cardiology Department, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| |
Collapse
|
26
|
Shrivastav D, Kumbhakar SK, Srivastava S, Singh DD. Natural product-based treatment potential for type 2 diabetes mellitus and cardiovascular disease. World J Diabetes 2024; 15:1603-1614. [PMID: 39099809 PMCID: PMC11292323 DOI: 10.4239/wjd.v15.i7.1603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/22/2024] [Accepted: 05/16/2024] [Indexed: 07/08/2024] Open
Abstract
BACKGROUND Type 2 diabetes (T2D) is a metabolic disease of impaired glucose utilization and a major cause of cardiovascular disease (CVD). The pathogenesis of both diseases shares common risk factors and mechanisms, and both are significant contributors to global morbidity and mortality. Supplements of natural products for T2D mellitus (T2DM) and CVD can be seen as a potential preventive and effective therapeutic strategy. AIM To critically evaluate the therapeutic potential of natural products in T2D and coronary artery disease (CAD). METHODS By using specific keywords, we strategically searched the PubMed database. Randomized controlled trials (RCTs) were searched as the primary focus that examined the effect of natural products on glycemic control, oxidative stress, and antioxidant levels. We focused on outcomes such as low blood glucose levels, adjustment on markers of oxidative stress and antioxidants. After screening full-length papers, we included 9 RCTs in our review that met our inclusion criteria. RESULTS In the literature search on the database, we found that various natural products like plant secondary metabolites play a diverse role in the management of CAD. American ginseng, sesame oil and cocoa flavanols proved effective in lowering blood glucose levels and controlling blood pressure, which are key factors in managing T2DM and CVD. In diabetic patients Melissa officinalis effectively reduce inflammation and shows diabetes prevention. Both fish oil and flaxseed oil reduced insulin levels and inflammatory markers, suggesting benefits for both conditions. The lipid profile and endothelial function were enhanced by Nigella sativa oil and Terminalia chebula, which is significant for the management of cardiovascular risk factors in T2DM. Additionally Bilberry extract also showed promise for improving glycemic control in patients with T2DM. CONCLUSION The high level of antioxidant, anti-inflammatory, and anti-angiogenic properties found in natural products makes them promising therapeutic options for the management of CAD, with the potential benefit of lowering the risk of CAD.
Collapse
Affiliation(s)
- Dharmsheel Shrivastav
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur 303002, Rajasthan, India
| | - Satyam Kumar Kumbhakar
- Department of Biotechnology, Govt Veer Surendra Say P.G. College, Gariaband 493889, Chhattisgarh, India
| | - Shivangi Srivastava
- Department of Life Science, Chhatrapati Shahu ji Maharaj University, Kanpur 208024, Uttar Pradesh, India
| | - Desh Deepak Singh
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur 303002, Rajasthan, India
| |
Collapse
|
27
|
Singh A, Bhatt KS, Nguyen HC, Frisbee JC, Singh KK. Endothelial-to-Mesenchymal Transition in Cardiovascular Pathophysiology. Int J Mol Sci 2024; 25:6180. [PMID: 38892367 PMCID: PMC11173124 DOI: 10.3390/ijms25116180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
Under different pathophysiological conditions, endothelial cells lose endothelial phenotype and gain mesenchymal cell-like phenotype via a process known as endothelial-to-mesenchymal transition (EndMT). At the molecular level, endothelial cells lose the expression of endothelial cell-specific markers such as CD31/platelet-endothelial cell adhesion molecule, von Willebrand factor, and vascular-endothelial cadherin and gain the expression of mesenchymal cell markers such as α-smooth muscle actin, N-cadherin, vimentin, fibroblast specific protein-1, and collagens. EndMT is induced by numerous different pathways triggered and modulated by multiple different and often redundant mechanisms in a context-dependent manner depending on the pathophysiological status of the cell. EndMT plays an essential role in embryonic development, particularly in atrioventricular valve development; however, EndMT is also implicated in the pathogenesis of several genetically determined and acquired diseases, including malignant, cardiovascular, inflammatory, and fibrotic disorders. Among cardiovascular diseases, aberrant EndMT is reported in atherosclerosis, pulmonary hypertension, valvular disease, fibroelastosis, and cardiac fibrosis. Accordingly, understanding the mechanisms behind the cause and/or effect of EndMT to eventually target EndMT appears to be a promising strategy for treating aberrant EndMT-associated diseases. However, this approach is limited by a lack of precise functional and molecular pathways, causes and/or effects, and a lack of robust animal models and human data about EndMT in different diseases. Here, we review different mechanisms in EndMT and the role of EndMT in various cardiovascular diseases.
Collapse
Affiliation(s)
- Aman Singh
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; (A.S.); (K.S.B.); (H.C.N.); (J.C.F.)
| | - Kriti S. Bhatt
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; (A.S.); (K.S.B.); (H.C.N.); (J.C.F.)
| | - Hien C. Nguyen
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; (A.S.); (K.S.B.); (H.C.N.); (J.C.F.)
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Jefferson C. Frisbee
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; (A.S.); (K.S.B.); (H.C.N.); (J.C.F.)
| | - Krishna K. Singh
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; (A.S.); (K.S.B.); (H.C.N.); (J.C.F.)
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| |
Collapse
|
28
|
Yan M, Wang Z, An Y, Li Z, Li Y, Zhang H, Li C, Wang L, Chen L, Gao C, Wang D, Gao C. OxLDL enhances procoagulant activity of endothelial cells by TMEM16F-mediated phosphatidylserine exposure. Cell Biol Int 2024; 48:848-860. [PMID: 38444077 DOI: 10.1002/cbin.12150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 01/28/2024] [Accepted: 02/20/2024] [Indexed: 03/07/2024]
Abstract
Oxidized low-density lipoprotein (oxLDL), a key component in atherosclerosis and hyperlipidemia, is a risk factor for atherothrombosis in dyslipidemia, yet its mechanism is poorly understood. In this study, we used oxLDL-induced human aortic endothelial cells (HAECs) and high-fat diet (HFD)-fed mice as a hyperlipidemia model. Phosphatidylserine (PS) exposure, cytosolic Ca2+, reactive oxygen species (ROS), and lipid peroxidation were measured by flow cytometer. TMEM16F expression was detected by immunofluorescence, western blot, and reverse transcription polymerase chain reaction. Procoagulant activity (PCA) was measured by coagulation time, intrinsic/extrinsic factor Xase, and thrombin generation. We found that oxLDL-induced PS exposure and the corresponding PCA of HAECs were increased significantly compared with control, which could be inhibited over 90% by lactadherin. Importantly, TMEM16F expression in oxLDL-induced HAECs was upregulated by enhanced intracellular Ca2+ concentration, ROS, and lipid peroxidation, which led to PS exposure. Meanwhile, the knockdown of TMEM16F by short hairpin RNA significantly inhibited PS exposure in oxLDL-induced HAECs. Moreover, we observed that HFD-fed mice dramatically increased the progress of thrombus formation and accompanied upregulated TMEM16F expression by thromboelastography analysis, FeCl3-induced carotid artery thrombosis model, and western blot. Collectively, these results demonstrate that TMEM16F-mediated PS exposure may contribute to prothrombotic status under hyperlipidemic conditions, which may serve as a novel therapeutic target for the prevention of thrombosis in hyperlipidemia.
Collapse
Affiliation(s)
- Meishan Yan
- Department of Medical Laboratory Science and Technology, Harbin Medical University Daqing, Daqing, China
| | - Zelong Wang
- Department of Medical Laboratory Science and Technology, Harbin Medical University Daqing, Daqing, China
| | - Yao An
- Department of Medical Laboratory Science and Technology, Harbin Medical University Daqing, Daqing, China
| | - Zhanni Li
- Department of Medical Laboratory Science and Technology, Harbin Medical University Daqing, Daqing, China
| | - Yun Li
- Hematology Department, Daqing Oil Field General Hospital, Daqing, China
| | - Hongyu Zhang
- Department of Medical Laboratory Science and Technology, Harbin Medical University Daqing, Daqing, China
| | - Caixia Li
- Department of Medical Laboratory Science and Technology, Harbin Medical University Daqing, Daqing, China
| | - Lifeng Wang
- Department of Medical Laboratory Science and Technology, Harbin Medical University Daqing, Daqing, China
| | - Li Chen
- Department of Medical Laboratory Science and Technology, Harbin Medical University Daqing, Daqing, China
| | - Chao Gao
- Department of Medical Laboratory Science and Technology, Harbin Medical University Daqing, Daqing, China
| | - Dongsheng Wang
- Department of Emergency, The Fifth Affiliated Hospital of Harbin Medical University, Daqing, China
| | - Chunyan Gao
- Department of Medical Laboratory Science and Technology, Harbin Medical University Daqing, Daqing, China
| |
Collapse
|
29
|
Ma Y, Lai J, Wan Q, Chen Z, Sun L, Zhang Q, Guan C, Li Q, Wu J. Identification of common mechanisms and biomarkers for dermatomyositis and atherosclerosis based on bioinformatics analysis. Skin Res Technol 2024; 30:e13808. [PMID: 38899746 PMCID: PMC11187814 DOI: 10.1111/srt.13808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND Dermatomyositis (DM) manifests as an autoimmune and inflammatory condition, clinically characterized by subacute progressive proximal muscle weakness, rashes or both along with extramuscular manifestations. Literature indicates that DM shares common risk factors with atherosclerosis (AS), and they often co-occur, yet the etiology and pathogenesis remain to be fully elucidated. This investigation aims to utilize bioinformatics methods to clarify the crucial genes and pathways that influence the pathophysiology of both DM and AS. METHOD Microarray datasets for DM (GSE128470, GSE1551, GSE143323) and AS (GSE100927, GSE28829, GSE43292) were retrieved from the Gene Expression Omnibus (GEO) database. The weighted gene co-expression network analysis (WGCNA) was used to reveal their co-expressed modules. Differentially expression genes (DEGs) were identified using the "limma" package in R software, and the functions of common DEGs were determined by functional enrichment analysis. A protein-protein interaction (PPI) network was established using the STRING database, with central genes evaluated by the cytoHubba plugin, and validated through external datasets. Immune infiltration analysis of the hub genes was conducted using the CIBERSORT method, along with Gene Set Enrichment Analysis (GSEA). Finally, the NetworkAnalyst platform was employed to examine the transcription factors (TFs) responsible for regulating pivotal crosstalk genes. RESULTS Utilizing WGCNA analysis, a total of 271 overlapping genes were pinpointed. Subsequent DEG analysis revealed 34 genes that are commonly found in both DM and AS, including 31 upregulated genes and 3 downregulated genes. The Degree Centrality algorithm was applied separately to the WGCNA and DEG collections to select the 15 genes with the highest connectivity, and crossing the two gene sets yielded 3 hub genes (PTPRC, TYROBP, CXCR4). Validation with external datasets showed their diagnostic value for DM and AS. Analysis of immune infiltration indicates that lymphocytes and macrophages are significantly associated with the pathogenesis of DM and AS. Moreover, GSEA analysis suggested that the shared genes are enriched in various receptor interactions and multiple cytokines and receptor signaling pathways. We coupled the 3 hub genes with their respective predicted genes, identifying a potential key TF, CBFB, which interacts with all 3 hub genes. CONCLUSION This research utilized comprehensive bioinformatics techniques to explore the shared pathogenesis of DM and AS. The three key genes, including PTPRC, TYROBP, and CXCR4, are related to the pathogenesis of DM and AS. The central genes and their correlations with immune cells may serve as potential diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Yirong Ma
- Jiangxi University of Traditional Chinese MedicineNanchangJiangxiChina
| | - Junyu Lai
- Department of cardiovascularAffiliated Hospital of Jiangxi University of Traditional Chinese MedicineNanchangJiangxiChina
| | - Qiang Wan
- Department of cardiovascularAffiliated Hospital of Jiangxi University of Traditional Chinese MedicineNanchangJiangxiChina
| | - Zhengtao Chen
- Department of cardiovascularAffiliated Hospital of Jiangxi University of Traditional Chinese MedicineNanchangJiangxiChina
| | - Liqiang Sun
- Department of cardiovascularAffiliated Hospital of Jiangxi University of Traditional Chinese MedicineNanchangJiangxiChina
| | - Qinhe Zhang
- Jiangxi University of Traditional Chinese MedicineNanchangJiangxiChina
| | - Chengyan Guan
- Jiangxi University of Traditional Chinese MedicineNanchangJiangxiChina
| | - Qiming Li
- Jiangxi University of Traditional Chinese MedicineNanchangJiangxiChina
| | - Jianguang Wu
- Department of cardiovascularAffiliated Hospital of Jiangxi University of Traditional Chinese MedicineNanchangJiangxiChina
| |
Collapse
|
30
|
Zhu L, Bao Y, Liu Z, Liu J, Li Z, Sun X, Zhou A, Wu H. Gualou-Xiebai herb pair ameliorate atherosclerosis in HFD-induced ApoE -/- mice and inhibit the ox-LDL-induced injury of HUVECs by regulating the Nrf2-mediated ferroptosis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 326:117892. [PMID: 38350505 DOI: 10.1016/j.jep.2024.117892] [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: 11/13/2023] [Revised: 01/30/2024] [Accepted: 02/06/2024] [Indexed: 02/15/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Atherosclerosis (AS) is a chronic vascular ailment characterized by inflammatory and lipid deposition in the arterial wall caused by endothelial injury. Ferroptosis is a novel type of cell death, and endothelial ferroptosis is a significant contributor to the progression of AS. Gualou-Xiebai (GLXB) is a renowned Chinese herb pair that serves a crucial function in treating AS. However, whether the underlying mechanism of GLXB plays a role in anti-atherosclerotic effects by inhibiting ferroptosis in endothelial cells has not been determined. AIM OF THE STUDY To explore the influence of GLXB on endothelial ferroptosis and determine its underlying mechanism of action in AS. MATERIALS AND METHODS In ApoE-/- mice, ultrasound was performed in mice fed a high-fat diet (HFD) for 12 weeks to assess the success of AS establishment. Then, ApoE-/- mice were treated with GLXB and Simvastatin (positive control) for 4 weeks. The effects of GLXB on AS pathology were assessed through aorta imaging and hematoxylin-eosin (HE) staining. To confirm the presence of ferroptosis, mitochondrial damage was observed using transmission electron microscope (TEM), along with analysis of free iron and lipid peroxidation levels. In vitro: ox-LDL-induced human vascular endothelial cells (HUVECs) injury and treated with GLXB, the ferroptosis inducer Erastin and an Nrf2 inhibitor ML385. Cell viability was evaluated using the CCK-8 assay in all groups. Flow cytometry was employed to detect lipid peroxidation and intracellular ferrous iron levels. Immunofluorescence staining microscopy verified Nrf2 nuclear translocation. Protein expression were measured by Western blot analysis. RESULTS GLXB improved atherosclerotic aortic lesions and vascular plaques. GLXB inhibited endothelial injury in the aorta by decreasing the levels of inflammatory factors and adhesion factors, and by decreasing the shedding of endothelial cells. GLXB suppressed ferroptosis in ApoE-/- mice by attenuating mitochondrial damage in ECs, increasing the levels of glutathione (GSH) and superoxide dismutase (SOD) in aortic tissues and down-regulating the levels of levels of lipid peroxide (LPO) and malondialdehyde (MDA). Interestingly, Erastin was used to demonstrate in vitro that GLXB inhibition of ferroptosis attenuated ox-LDL-induced injuring effects on HUVECs that were reversed by Erastin. Mechanistically, GLXB activates the Nrf2 signaling pathway to inhibit ferroptosis by increasing downstream anti-ferroptosis target proteins and promoting the interaction between Nrf2 and SLC7A11. More convincingly, ML385 (Nrf2 inhibitor) reversed the anti-ferroptosis effect of GLXB. CONCLUSION GLXB inhibits ferroptosis-mediated endothelial cell injury via activating the Nrf2 signaling pathway and further alleviates AS pathological damage.
Collapse
Affiliation(s)
- Li Zhu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; The Experimental Research Center, Anhui University of Chinese Medicine, Hefei, 230038, China
| | - Youli Bao
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; The Experimental Research Center, Anhui University of Chinese Medicine, Hefei, 230038, China
| | - Zijian Liu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; The Experimental Research Center, Anhui University of Chinese Medicine, Hefei, 230038, China
| | - Jiahui Liu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; The Experimental Research Center, Anhui University of Chinese Medicine, Hefei, 230038, China
| | - Zhenglong Li
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; The Experimental Research Center, Anhui University of Chinese Medicine, Hefei, 230038, China
| | - Xin Sun
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; The Experimental Research Center, Anhui University of Chinese Medicine, Hefei, 230038, China
| | - An Zhou
- The Experimental Research Center, Anhui University of Chinese Medicine, Hefei, 230038, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei, 230012, China.
| | - Hongfei Wu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; The Experimental Research Center, Anhui University of Chinese Medicine, Hefei, 230038, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei, 230012, China.
| |
Collapse
|
31
|
Reppe S, Gundersen S, Sandve GK, Wang Y, Andreassen OA, Medina-Gomez C, Rivadeneira F, Utheim TP, Hovig E, Gautvik KM. Identification of Transcripts with Shared Roles in the Pathogenesis of Postmenopausal Osteoporosis and Cardiovascular Disease. Int J Mol Sci 2024; 25:5554. [PMID: 38791593 PMCID: PMC11121938 DOI: 10.3390/ijms25105554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/11/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
Epidemiological evidence suggests existing comorbidity between postmenopausal osteoporosis (OP) and cardiovascular disease (CVD), but identification of possible shared genes is lacking. The skeletal global transcriptomes were analyzed in trans-iliac bone biopsies (n = 84) from clinically well-characterized postmenopausal women (50 to 86 years) without clinical CVD using microchips and RNA sequencing. One thousand transcripts highly correlated with areal bone mineral density (aBMD) were further analyzed using bioinformatics, and common genes overlapping with CVD and associated biological mechanisms, pathways and functions were identified. Fifty genes (45 mRNAs, 5 miRNAs) were discovered with established roles in oxidative stress, inflammatory response, endothelial function, fibrosis, dyslipidemia and osteoblastogenesis/calcification. These pleiotropic genes with possible CVD comorbidity functions were also present in transcriptomes of microvascular endothelial cells and cardiomyocytes and were differentially expressed between healthy and osteoporotic women with fragility fractures. The results were supported by a genetic pleiotropy-informed conditional False Discovery Rate approach identifying any overlap in single nucleotide polymorphisms (SNPs) within several genes encoding aBMD- and CVD-associated transcripts. The study provides transcriptional and genomic evidence for genes of importance for both BMD regulation and CVD risk in a large collection of postmenopausal bone biopsies. Most of the transcripts identified in the CVD risk categories have no previously recognized roles in OP pathogenesis and provide novel avenues for exploring the mechanistic basis for the biological association between CVD and OP.
Collapse
Affiliation(s)
- Sjur Reppe
- Department of Medical Biochemistry, Oslo University Hospital, 0450 Oslo, Norway
- Unger-Vetlesen Institute, Lovisenberg Diaconal Hospital, 0440 Oslo, Norway
- Department of Plastic and Reconstructive Surgery, Oslo University Hospital, 0424 Oslo, Norway
| | - Sveinung Gundersen
- Center for Bioinformatics, Department of Informatics, University of Oslo, 0313 Oslo, Norway
| | - Geir K. Sandve
- Department of Informatics, University of Oslo, 0373 Oslo, Norway; (G.K.S.)
| | - Yunpeng Wang
- NORMENT, Institute of Clinical Medicine, University of Oslo, 0450 Oslo, Norway; (Y.W.); (O.A.A.)
- Division of Mental Health and Addiction, Oslo University Hospital, 0424 Oslo, Norway
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Ole A. Andreassen
- NORMENT, Institute of Clinical Medicine, University of Oslo, 0450 Oslo, Norway; (Y.W.); (O.A.A.)
- Division of Mental Health and Addiction, Oslo University Hospital, 0424 Oslo, Norway
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA
| | - Carolina Medina-Gomez
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (C.M.-G.); (F.R.)
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (C.M.-G.); (F.R.)
| | - Tor P. Utheim
- Department of Medical Biochemistry, Oslo University Hospital, 0450 Oslo, Norway
- Department of Plastic and Reconstructive Surgery, Oslo University Hospital, 0424 Oslo, Norway
| | - Eivind Hovig
- Department of Informatics, University of Oslo, 0373 Oslo, Norway; (G.K.S.)
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, 0424 Oslo, Norway
| | - Kaare M. Gautvik
- Unger-Vetlesen Institute, Lovisenberg Diaconal Hospital, 0440 Oslo, Norway
| |
Collapse
|
32
|
Dąbkowski K, Kreft E, Sałaga-Zaleska K, Chyła-Danił G, Mickiewicz A, Gruchała M, Kuchta A, Jankowski M. Human In Vitro Oxidized Low-Density Lipoprotein (oxLDL) Increases Urinary Albumin Excretion in Rats. Int J Mol Sci 2024; 25:5498. [PMID: 38791535 PMCID: PMC11122078 DOI: 10.3390/ijms25105498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Hypercholesterolemia-associated oxidative stress increases the formation of oxidized low-density lipoprotein (oxLDL), which can affect endothelial cell function and potentially contribute to renal dysfunction, as reflected by changes in urinary protein excretion. This study aimed to investigate the impact of exogenous oxLDL on urinary excretion of albumin and nephrin. LDL was isolated from a patient with familial hypercholesterolemia (FH) undergoing lipoprotein apheresis (LA) and was oxidized in vitro with Cu (II) ions. Biochemical markers of LDL oxidation, such as TBARS, conjugated dienes, and free ε-amino groups, were measured. Wistar rats were treated with a single intraperitoneal injection of PBS, LDL, or oxLDL (4 mg of protein/kg b.w.). Urine was collected one day before and two days after the injection. We measured blood lipid profiles, urinary protein excretion (specifically albumin and nephrin), and markers of systemic oxidative stress (8-OHdG and 8-iso-PGF2α). The results showed that injection of oxLDL increased urinary albumin excretion by approximately 28% (310 ± 27 μg/24 h vs. 396 ± 26 μg/24 h, p = 0.0003) but had no effect on nephrin excretion. Neither PBS nor LDL had any effect on urinary albumin or nephrin excretion. Additionally, oxLDL did not affect systemic oxidative stress. In conclusion, hypercholesterolemia may adversely affect renal function through oxidatively modified LDL, which interferes with the renal handling of albumin and leads to the development of albuminuria.
Collapse
Affiliation(s)
- Kamil Dąbkowski
- Department of Clinical Chemistry, Medical University of Gdańsk, 80-210 Gdańsk, Poland; (K.D.); (E.K.); (K.S.-Z.); (G.C.-D.); (A.K.)
| | - Ewelina Kreft
- Department of Clinical Chemistry, Medical University of Gdańsk, 80-210 Gdańsk, Poland; (K.D.); (E.K.); (K.S.-Z.); (G.C.-D.); (A.K.)
| | - Kornelia Sałaga-Zaleska
- Department of Clinical Chemistry, Medical University of Gdańsk, 80-210 Gdańsk, Poland; (K.D.); (E.K.); (K.S.-Z.); (G.C.-D.); (A.K.)
| | - Gabriela Chyła-Danił
- Department of Clinical Chemistry, Medical University of Gdańsk, 80-210 Gdańsk, Poland; (K.D.); (E.K.); (K.S.-Z.); (G.C.-D.); (A.K.)
| | - Agnieszka Mickiewicz
- 1st Department of Cardiology, Medical University of Gdańsk, 80-210 Gdańsk, Poland; (A.M.); (M.G.)
| | - Marcin Gruchała
- 1st Department of Cardiology, Medical University of Gdańsk, 80-210 Gdańsk, Poland; (A.M.); (M.G.)
| | - Agnieszka Kuchta
- Department of Clinical Chemistry, Medical University of Gdańsk, 80-210 Gdańsk, Poland; (K.D.); (E.K.); (K.S.-Z.); (G.C.-D.); (A.K.)
| | - Maciej Jankowski
- Department of Clinical Chemistry, Medical University of Gdańsk, 80-210 Gdańsk, Poland; (K.D.); (E.K.); (K.S.-Z.); (G.C.-D.); (A.K.)
| |
Collapse
|
33
|
Yu W, Zhao Y, Ilyas I, Wang L, Little PJ, Xu S. The natural polyphenol fisetin in atherosclerosis prevention: a mechanistic review. J Pharm Pharmacol 2024:rgae053. [PMID: 38733634 DOI: 10.1093/jpp/rgae053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 04/22/2024] [Indexed: 05/13/2024]
Abstract
The incidence and mortality rate of atherosclerotic cardiovascular disease (ASCVD) is increasing yearly worldwide. Recently, a growing body of evidence has unveiled the anti-atherosclerotic properties of fisetin, a natural polyphenol compound. In this article, we reviewed the pharmacologic actions of fisetin on experimental atherosclerosis and its protective effects on disease-relevant cell types such as endothelial cells, macrophages, vascular smooth muscle cells, and platelets. Based on its profound cardiovascular actions, fisetin holds potential for clinical translation and could be developed as a potential therapeutic option for atherosclerosis and its related complications. Large-scale randomized clinical trials are warranted to ascertain the safety and efficacy of fisetin in patients with or high risk for ASCVD.
Collapse
Affiliation(s)
- Wei Yu
- School of Materials Science and Engineering, Hefei University of Technology, Hefei, Anhui, 230009, China
- Anhui Renovo Pharmaceutical Co., Ltd, Hefei, Anhui, 230001, China
- Anhui Guozheng Pharmaceutical Co., Ltd, Hefei, Anhui, 230041, China
| | - Yaping Zhao
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Iqra Ilyas
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Li Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Peter J Little
- Department of Pharmacy, Guangzhou Xinhua University, No. 721, Guangshan Road 1, Tianhe District, Guangzhou, 510520, China
| | - Suowen Xu
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| |
Collapse
|
34
|
Marder M, Remmert C, Perschel JA, Otgonbayar M, von Toerne C, Hauck S, Bushe J, Feuchtinger A, Sheikh B, Moussus M, Meier M. Stem cell-derived vessels-on-chip for cardiovascular disease modeling. Cell Rep 2024; 43:114008. [PMID: 38536819 DOI: 10.1016/j.celrep.2024.114008] [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: 09/13/2023] [Revised: 01/25/2024] [Accepted: 03/12/2024] [Indexed: 04/28/2024] Open
Abstract
The metabolic syndrome is accompanied by vascular complications. Human in vitro disease models are hence required to better understand vascular dysfunctions and guide clinical therapies. Here, we engineered an open microfluidic vessel-on-chip platform that integrates human pluripotent stem cell-derived endothelial cells (SC-ECs). The open microfluidic design enables seamless integration with state-of-the-art analytical technologies, including single-cell RNA sequencing, proteomics by mass spectrometry, and high-resolution imaging. Beyond previous systems, we report SC-EC maturation by means of barrier formation, arterial toning, and high nitric oxide synthesis levels under gravity-driven flow. Functionally, we corroborate the hallmarks of early-onset atherosclerosis with low sample volumes and cell numbers under flow conditions by determining proteome and secretome changes in SC-ECs stimulated with oxidized low-density lipoprotein and free fatty acids. More broadly, our organ-on-chip platform enables the modeling of patient-specific human endothelial tissue and has the potential to become a general tool for animal-free vascular research.
Collapse
Affiliation(s)
- Maren Marder
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Munich, Germany
| | - Caroline Remmert
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Munich, Germany
| | - Julius A Perschel
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Munich, Germany
| | | | | | - Stefanie Hauck
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, Munich, Germany
| | - Judith Bushe
- Core Facility Pathology & Tissue Analytics, Helmholtz Munich, 85764 Neuherberg, Germany
| | - Annette Feuchtinger
- Core Facility Pathology & Tissue Analytics, Helmholtz Munich, 85764 Neuherberg, Germany
| | - Bilal Sheikh
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich, Leipzig, Germany; Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Michel Moussus
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Munich, Germany
| | - Matthias Meier
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Munich, Germany; Centre for Biotechnology and Biomedicine, University of Leipzig, Leipzig, Germany.
| |
Collapse
|
35
|
Lemini C, Silveyra P, Segovia-Mendoza M. Cardiovascular disrupting effects of bisphenols, phthalates, and parabens related to endothelial dysfunction: Review of toxicological and pharmacological mechanisms. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2024; 107:104407. [PMID: 38428705 DOI: 10.1016/j.etap.2024.104407] [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: 07/30/2023] [Accepted: 02/25/2024] [Indexed: 03/03/2024]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death worldwide. CVDs are promoted by the accumulation of lipids and immune cells in the endothelial space resulting in endothelial dysfunction. Endothelial cells are important components of the vascular endothelium, that regulate the vascular flow. The imbalance in the production of vasoactive substances results in the loss of vascular homeostasis, leading the endothelial dysfunction. Thus, endothelial dysfunction plays an essential role in the development of atherosclerosis and can be triggered by different cardiovascular risk factors. On the other hand, the 17β-estradiol (E2) hormone has been related to the regulation of vascular tone through different mechanisms. Several compounds can elicit estrogenic actions similar to those of E2. For these reasons, they have been called endocrine-disrupting compounds (EDCs). This review aims to provide up-to-date information about how different EDCs affect endothelial function and their mechanistic roles in the context of CVDs.
Collapse
Affiliation(s)
- Cristina Lemini
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Patricia Silveyra
- Department of Environmental and Occupational Health, Indiana University Bloomington, School of Public Health, Bloomington, IN, USA
| | - Mariana Segovia-Mendoza
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico.
| |
Collapse
|
36
|
Tomaszewska A, Gonciarz W, Rechcinski T, Chmiela M, Kurdowska AK, Krupa A. Helicobacter pylori components increase the severity of metabolic syndrome and its hepatic manifestations induced by a high fat diet. Sci Rep 2024; 14:5764. [PMID: 38459219 PMCID: PMC10923818 DOI: 10.1038/s41598-024-56308-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 03/05/2024] [Indexed: 03/10/2024] Open
Abstract
The metabolic syndrome, often accompanied by hepatic manifestations, is a high-risk factor for developing cardiovascular disease. Patients with metabolic dysfunction associated with steatohepatic disease (MASDL) are at significant risk of developing coronary artery disease. Atherosclerosis is a systemic inflammatory disorder in which several factors, including dietary or infectious factors, can cause an inflammatory response. Helicobacter pylori (HP) bacteria have been implicated in the progression of proatherogenic vascular endothelial lesions, moreover, our previous study in an experimental in vivo model of Cavia porcellus showed that HP components and high-fat substances acted synergistically in promoting vascular endothelial inflammation, leading to an early onset of a proatherogenic environment. In the present study, our goal was to determine the contribution of HP components to the development of hepatic manifestations of metabolic syndrome in an experimental model. Our results showed that HP infection in animals exposed to a high-fat diet increased oxidative stress and lipid peroxidation, followed by endothelial lipid deposition, impaired endothelial apoptosis, cell lysis, and increased vascular stiffness. Finally, histopathological analysis of liver tissue showed signs of MASLD development in HP-infected animals fed a high-fat diet.
Collapse
Affiliation(s)
- Agata Tomaszewska
- Department of Immunology and Infectious Biology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland.
- Bio-Med-Chem Doctoral School of the University of Lodz and Lodz Institutes of the Polish Academy of Sciences, University of Lodz, Lodz, Poland.
| | - Weronika Gonciarz
- Department of Immunology and Infectious Biology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Tomasz Rechcinski
- 1st Department of Cardiology, Medical University of Lodz, Lodz, Poland
| | - Magdalena Chmiela
- Department of Immunology and Infectious Biology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Anna K Kurdowska
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas, USA
| | - Agnieszka Krupa
- Department of Immunology and Infectious Biology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland.
| |
Collapse
|
37
|
Gupta P, Kumar R. Nitric oxide: A potential etiological agent for vaso-occlusive crises in sickle cell disease. Nitric Oxide 2024; 144:40-46. [PMID: 38316197 DOI: 10.1016/j.niox.2024.01.008] [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: 10/06/2023] [Revised: 12/27/2023] [Accepted: 01/29/2024] [Indexed: 02/07/2024]
Abstract
Nitric oxide (NO), a vasodilator contributes to the vaso-occlusive crisis associated with the sickle cell disease (SCD). Vascular nitric oxide helps in vasodilation, controlled platelet aggregation, and preventing adhesion of sickled red blood cells to the endothelium. It decreases the expression of pro-inflammatory genes responsible for atherogenesis associated with SCD. Haemolysis and activated endothelium in SCD patients reduce the bioavailability of NO which promotes the severity of sickle cell disease mainly causes vaso-occlusive crises. Additionally, NO depletion can also contribute to the formation of thrombus, which can cause serious complications such as stroke, pulmonary embolism etc. Understanding the multifaceted role of NO provides valuable insights into its therapeutic potential for managing SCD and preventing associated complications. Various clinical trials and studies suggested the importance of artificially induced nitric oxide and its supplements in the reduction of severity. Further research on the mechanisms of NO depletion in SCD is needed to develop more effective treatment strategies and improve the management of this debilitating disease.
Collapse
Affiliation(s)
- Parul Gupta
- ICMR-National Institute of Research in Tribal Health, India
| | - Ravindra Kumar
- ICMR-National Institute of Research in Tribal Health, India.
| |
Collapse
|
38
|
Zhang J, Yu H, Pan R, Miao G, Wang Y, Li Z, Yu H, Lu L, Jin X. Airborne magnetite nanoparticles induced early vascular pathologies by disrupting lipid metabolism under high-fat dietary patterns. ENVIRONMENTAL TOXICOLOGY 2024; 39:1175-1186. [PMID: 37860912 DOI: 10.1002/tox.24005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/21/2023] [Accepted: 10/07/2023] [Indexed: 10/21/2023]
Abstract
Magnetite nanoparticles (MNPs) have been extensively detected in the atmospheric environment and implicated as a prominent threat to atherosclerosis, a chronic vascular inflammatory disease. Due to globalization and economic development, the dramatic shift in diet from traditional to high-fat dietary patterns aggravated atherosclerosis progression induced by environmental factors. However, limited knowledge is available regarding vascular risks and underlying mechanisms of airborne MNPs in high-risk populations with high-fat dietary habits. Herein, we demonstrated that MNPs exerted a proatherogenic effect under high-fat dietary patterns, leading to aortic wall thickening, elastic fiber disorganization, macrophage infiltration, and local inflammation. Based on the correlation analysis between MNPs and PM group, we identified that MNPs might be a key PM component in atherogenic toxicity. MNPs exposure disturbed the dynamic process of lipid metabolism, manifested as aortic lipid accumulation, dyslipidemia, and hepatic lipid metabolism disorder, which was modulated by the JAK-STAT pathway. Overall, these findings provide new insight into understanding the cardiovascular risks and mechanisms of MNPs among high-risk populations.
Collapse
Affiliation(s)
- Jingxu Zhang
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, P.R. China
| | - Hongyan Yu
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, P.R. China
| | - Ruonan Pan
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, P.R. China
| | - Gan Miao
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, P.R. China
| | - Yu Wang
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, P.R. China
| | - Ziyuan Li
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, P.R. China
| | - Haiyi Yu
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, P.R. China
| | - Lin Lu
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, P.R. China
| | - Xiaoting Jin
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, P.R. China
| |
Collapse
|
39
|
Solanki K, Bezsonov E, Orekhov A, Parihar SP, Vaja S, White FA, Obukhov AG, Baig MS. Effect of reactive oxygen, nitrogen, and sulfur species on signaling pathways in atherosclerosis. Vascul Pharmacol 2024; 154:107282. [PMID: 38325566 DOI: 10.1016/j.vph.2024.107282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 02/04/2024] [Indexed: 02/09/2024]
Abstract
Atherosclerosis is a chronic inflammatory disease in which fats, lipids, cholesterol, calcium, proliferating smooth muscle cells, and immune cells accumulate in the intima of the large arteries, forming atherosclerotic plaques. A complex interplay of various vascular and immune cells takes place during the initiation and progression of atherosclerosis. Multiple reports indicate that tight control of reactive oxygen species (ROS), reactive nitrogen species (RNS), and reactive sulfur species (RSS) production is critical for maintaining vascular health. Unrestricted ROS and RNS generation may lead to activation of various inflammatory signaling pathways, facilitating atherosclerosis. Given these deleterious consequences, it is important to understand how ROS and RNS affect the signaling processes involved in atherogenesis. Conversely, RSS appears to exhibit an atheroprotective potential and can alleviate the deleterious effects of ROS and RNS. Herein, we review the literature describing the effects of ROS, RNS, and RSS on vascular smooth muscle cells, endothelial cells, and macrophages and focus on how changes in their production affect the initiation and progression of atherosclerosis. This review also discusses the contribution of ROS, RNS, and RSS in mediating various post-translational modifications, such as oxidation, nitrosylation, and sulfation, of the molecules involved in inflammatory signaling.
Collapse
Affiliation(s)
- Kundan Solanki
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Simrol, Indore, India
| | - Evgeny Bezsonov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia; Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Avtsyn Research Institute of Human Morphology, Petrovsky National Research Centre of Surgery, Moscow, Russia; Department of Biology and General Genetics, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia; The Cell Physiology and Pathology Laboratory, Turgenev State University of Orel, Orel, Russia
| | - Alexander Orekhov
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow, Russia
| | - Suraj P Parihar
- Wellcome Centre for Infectious Diseases Research in Africa (CIDRI-Africa) and Institute of Infectious Diseases and Molecular Medicine (IDM), Division of Medical Microbiology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; Department of Biochemistry, Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| | - Shivani Vaja
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Simrol, Indore, India
| | - Fletcher A White
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Alexander G Obukhov
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Mirza S Baig
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Simrol, Indore, India.
| |
Collapse
|
40
|
Shin JJ, Park J, Shin HS, Arab I, Suk K, Lee WH. Roles of lncRNAs in NF-κB-Mediated Macrophage Inflammation and Their Implications in the Pathogenesis of Human Diseases. Int J Mol Sci 2024; 25:2670. [PMID: 38473915 DOI: 10.3390/ijms25052670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/20/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Over the past century, molecular biology's focus has transitioned from proteins to DNA, and now to RNA. Once considered merely a genetic information carrier, RNA is now recognized as both a vital element in early cellular life and a regulator in complex organisms. Long noncoding RNAs (lncRNAs), which are over 200 bases long but do not code for proteins, play roles in gene expression regulation and signal transduction by inducing epigenetic changes or interacting with various proteins and RNAs. These interactions exhibit a range of functions in various cell types, including macrophages. Notably, some macrophage lncRNAs influence the activation of NF-κB, a crucial transcription factor governing immune and inflammatory responses. Macrophage NF-κB is instrumental in the progression of various pathological conditions including sepsis, atherosclerosis, cancer, autoimmune disorders, and hypersensitivity. It orchestrates gene expression related to immune responses, inflammation, cell survival, and proliferation. Consequently, its malfunction is a key contributor to the onset and development of these diseases. This review aims to summarize the function of lncRNAs in regulating NF-κB activity in macrophage activation and inflammation, with a particular emphasis on their relevance to human diseases and their potential as therapeutic targets. The insights gained from studies on macrophage lncRNAs, as discussed in this review, could provide valuable knowledge for the development of treatments for various pathological conditions involving macrophages.
Collapse
Affiliation(s)
- Jae-Joon Shin
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Jeongkwang Park
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Hyeung-Seob Shin
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Imene Arab
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Kyoungho Suk
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Won-Ha Lee
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| |
Collapse
|
41
|
More SA, Deore RS, Pawar HD, Sharma C, Nakhate KT, Rathod SS, Ojha S, Goyal SN. CB2 Cannabinoid Receptor as a Potential Target in Myocardial Infarction: Exploration of Molecular Pathogenesis and Therapeutic Strategies. Int J Mol Sci 2024; 25:1683. [PMID: 38338960 PMCID: PMC10855244 DOI: 10.3390/ijms25031683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 01/22/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
The lipid endocannabinoid system has recently emerged as a novel therapeutic target for several inflammatory and tissue-damaging diseases, including those affecting the cardiovascular system. The primary targets of cannabinoids are cannabinoid type 1 (CB1) and 2 (CB2) receptors. The CB2 receptor is expressed in the cardiomyocytes. While the pathological changes in the myocardium upregulate the CB2 receptor, genetic deletion of the receptor aggravates the changes. The CB2 receptor plays a crucial role in attenuating the advancement of myocardial infarction (MI)-associated pathological changes in the myocardium. Activation of CB2 receptors exerts cardioprotection in MI via numerous molecular pathways. For instance, delta-9-tetrahydrocannabinol attenuated the progression of MI via modulation of the CB2 receptor-dependent anti-inflammatory mechanisms, including suppression of pro-inflammatory cytokines like IL-6, TNF-α, and IL-1β. Through similar mechanisms, natural and synthetic CB2 receptor ligands repair myocardial tissue damage. This review aims to offer an in-depth discussion on the ameliorative potential of CB2 receptors in myocardial injuries induced by a variety of pathogenic mechanisms. Further, the modulation of autophagy, TGF-β/Smad3 signaling, MPTP opening, and ROS production are discussed. The molecular correlation of CB2 receptors with cardiac injury markers, such as troponin I, LDH1, and CK-MB, is explored. Special attention has been paid to novel insights into the potential therapeutic implications of CB2 receptor activation in MI.
Collapse
Affiliation(s)
- Sagar A. More
- Department of Pharmacology, Shri Vile Parle Kelavani Mandal’s Institute of Pharmacy, Dhule 424001, Maharashtra, India; (S.A.M.); (R.S.D.); (H.D.P.); (K.T.N.); (S.S.R.)
| | - Rucha S. Deore
- Department of Pharmacology, Shri Vile Parle Kelavani Mandal’s Institute of Pharmacy, Dhule 424001, Maharashtra, India; (S.A.M.); (R.S.D.); (H.D.P.); (K.T.N.); (S.S.R.)
| | - Harshal D. Pawar
- Department of Pharmacology, Shri Vile Parle Kelavani Mandal’s Institute of Pharmacy, Dhule 424001, Maharashtra, India; (S.A.M.); (R.S.D.); (H.D.P.); (K.T.N.); (S.S.R.)
| | - Charu Sharma
- Department of Internal Medicine, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates;
| | - Kartik T. Nakhate
- Department of Pharmacology, Shri Vile Parle Kelavani Mandal’s Institute of Pharmacy, Dhule 424001, Maharashtra, India; (S.A.M.); (R.S.D.); (H.D.P.); (K.T.N.); (S.S.R.)
| | - Sumit S. Rathod
- Department of Pharmacology, Shri Vile Parle Kelavani Mandal’s Institute of Pharmacy, Dhule 424001, Maharashtra, India; (S.A.M.); (R.S.D.); (H.D.P.); (K.T.N.); (S.S.R.)
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Sameer N. Goyal
- Department of Pharmacology, Shri Vile Parle Kelavani Mandal’s Institute of Pharmacy, Dhule 424001, Maharashtra, India; (S.A.M.); (R.S.D.); (H.D.P.); (K.T.N.); (S.S.R.)
| |
Collapse
|
42
|
Du J, Wu J, Zhang Y, Liu Q, Luo X, Huang X, Wang X, Yang F, Hou J. CTRP13 Mitigates Endothelial Cell Ferroptosis via the AMPK/KLF4 Pathway: Implications for Atherosclerosis Protection. Med Sci Monit 2024; 30:e942733. [PMID: 38273650 PMCID: PMC10826208 DOI: 10.12659/msm.942733] [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: 09/30/2023] [Accepted: 11/11/2023] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND C1q/tumor necrosis factor-related protein 13 (CTRP13) preserves endothelial function and possesses anti-oxidation activity. However, its effects on ferroptosis of human umbilical vein endothelial cells (HUVECs) remain unclear. We investigated the effects of CTRP13 on HUVEC ferroptosis induced by oxidized low-density lipoprotein (ox-LDL) and explored the underlying mechanisms of CTRP13 against ferroptosis via the AMPK/KLF4 pathway. MATERIAL AND METHODS Cell Counting Kit-8 assay was used to evaluate cell viability. Lactate dehydrogenase activity and malondialdehyde content analysis were performed to evaluate the cell membrane integrity and lipid peroxidation. Mito-Tracker, JC-1, and 2',7'-dichlorofluorescein di-acetate were used to evaluate the biological activity of mitochondria, mitochondrial membrane potential, and reactive oxygen species (ROS) in endothelial cells. The ferroptosis indicator expressions, recombinant solute carrier family 7, member 11, glutathione peroxidase 4 (GPX4), and acyl-CoA synthetase long-chain family member 4 were examined using real-time reverse transcription-polymerase chain reaction and Western blot. Immunofluorescence staining detected GPX4 location in endothelial cells. RESULTS The results demonstrate that CTRP13 (450 ng/mL) prevented HUVEC ferroptosis by inhibiting ROS overproduction and mitochondrial dysfunction, and CTRP13 accelerated antioxidant enzyme expression levels, such as heme oxygenase 1, superoxide dismutase 1, and superoxide dismutase 2, compared with the ox-LDL (100 µg/mL) group for 48 h. Additionally, CTRP13 treatment increased p-AMPK/AMPK expression by 47.65% (P<0.05) while decreasing Krüppel-like factor 4 expression by 37.43% (P<0.05) in ox-LDL-induced HUVECs and elucidated the protective effect on endothelial dysfunction from ferroptosis. CONCLUSIONS These findings provide new insights for understanding the effects and mechanism of CTRP13 on preventing endothelial cell ferroptosis.
Collapse
Affiliation(s)
- Jie Du
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, Heilongjiang, PR China
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China
| | - Jianjun Wu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, Heilongjiang, PR China
| | - Youqi Zhang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, Heilongjiang, PR China
| | - Qi Liu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, Heilongjiang, PR China
| | - Xing Luo
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, Heilongjiang, PR China
| | - Xingtao Huang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, Heilongjiang, PR China
| | - Xuedong Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, Heilongjiang, PR China
| | - Fan Yang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, Heilongjiang, PR China
| | - JingBo Hou
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, Heilongjiang, PR China
| |
Collapse
|
43
|
Anees S, Ahmad M, Ashraf S, Bhat AH, Hamid R, Ganie SA. Bioactive fractions from Allium humile alleviate the risk of high fat diet induced atherosclerosis in albino Wistar rats by inhibiting protein kinase C. Fitoterapia 2024; 172:105775. [PMID: 38097019 DOI: 10.1016/j.fitote.2023.105775] [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: 08/23/2023] [Revised: 12/07/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023]
Abstract
Atherosclerosis is a global concern that worsens with age, and plants that are effective medicinal herbs can give a viable alternative. PKC is a key factor in cardiovascular and other disorders; targeting it can reduce the risk of these diseases. We evaluated Allium humile for PKC inhibition and therapeutic efficacy against atherosclerosis. Soxhlet extraction was done to obtain extracts (hexane, ethyl acetate, methanol, ethanol and aqueous) and then tested for DPPH radical scavenging and PKC inhibitory activity. The methanolic extract was more active than the other extracts, so it was subjected to column chromatography, and seventeen fractions were obtained. Only 11, 12, and 15 showed good activity against PKC. Wistar rats were divided into six groups and each group received high fat diet for 30 days. Then the three potent fractions (10 mg/kg) were administered for 15 days along with high fat diet. Fraction II had the highest effectiveness (P < 0.0001) in decreasing lipid levels, lipid peroxidation, reducing IL-6 and TNF-α expression, and raising nitric oxide. This also demonstrated a decrease in PKC activity, as well as a decrease in the formation of the lipoidal layer in the aorta wall and rupture of the intima and media as validated by histological analysis. The two compounds, phytol acetate and cyanidin 3-(6″-o-malonyllaminaribioside) were characterised in fraction II by NMR and HRMS and cyanidin 3-(6″-o-malonyllaminaribioside) inhibited PKC more efficiently. Thus, Allium humile has strong anti-atherogenic activity as well as the ability to inhibit PKC both in vitro and in vivo.
Collapse
Affiliation(s)
- Suhail Anees
- Department of Clinical Biochemistry, University of Kashmir, Srinagar, India
| | - Muzaffar Ahmad
- Department of Biochemistry, University of Kashmir, Srinagar, India
| | - Suhail Ashraf
- Department of Clinical Biochemistry, University of Kashmir, Srinagar, India
| | | | - Rabia Hamid
- Department of Nanotechnology, University of Kashmir, Srinagar, India.
| | - Showkat Ahmad Ganie
- Department of Clinical Biochemistry, University of Kashmir, Srinagar, India.
| |
Collapse
|
44
|
Arabi SM, Chambari M, Bahrami LS, Hadi S, Sahebkar A. Statin Therapy and Flow-Mediated Dilation: A Systematic Review and Dose-Response Meta-Analysis Using the GRADE of Data from Randomized Controlled Trials. Curr Hypertens Rev 2024; 20:90-100. [PMID: 38385489 DOI: 10.2174/0115734021280797240212091416] [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: 10/26/2023] [Revised: 01/14/2024] [Accepted: 01/22/2024] [Indexed: 02/23/2024]
Abstract
INTRODUCTION A previous meta-analysis reported the positive effects of statin therapy on endothelial function. However, the obtained result had several limitations that necessitated updating the information in this field. Therefore, a systematic and meta-analysis review was conducted to determine whether statin therapy could improve endothelial function, as assessed by flow-- mediated dilation (FMD). METHODS MEDLINE, SciVerse Scopus, and Clarivate Analytics Web of Science were searched to identify randomized placebo-controlled trials assessing the impact of statin therapy on FMD. A random-effects model was used for meta-analysis to calculate the mean difference in weight. Meta- regression and subgroup analyses were used to identify sources of heterogeneity. In addition, nonlinear dose-response, quality of evidence, influence analysis, and publication bias evaluation were assessed using standard methods. RESULT Thirty-five trials (41 arms) involving 2178 participants were included in the meta-analysis study. Statin treatment significantly improved FMD [weighted mean difference (WMD): 1.7%, 95% CI: 1.3-2.2, p < 0.001). However, significant heterogeneity was observed (I2=97.9%, p < 0.001). The results of the subgroup analysis showed that health status can contribute to heterogeneity. Non-linear dose-response analysis revealed the most significant improvement in FMD with atorvastatin at a dose of 20 mg/day and simvastatin at 80 mg/day. CONCLUSION Statin therapy significantly improved endothelial function, as assessed by FMD. These changes are clinically significant, but their use should be approached with caution.
Collapse
Affiliation(s)
- Seyyed Mostafa Arabi
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahla Chambari
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Leila Sadat Bahrami
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saeid Hadi
- Department of Health, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
45
|
Tan JJ, Murugan DD, Ling WC, Lee SK, Kang WH. Chronic Administration of Red Yeast Rice Mitigates Endothelial Dysfunction in Spontaneously Hypertensive Rats by Inhibiting Oxidative Stress and Endothelial Nitric Oxide Synthase Uncoupling. Curr Vasc Pharmacol 2024; 22:355-364. [PMID: 38847159 DOI: 10.2174/0115701611295900240529104225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 04/04/2024] [Accepted: 04/18/2024] [Indexed: 10/03/2024]
Abstract
BACKGROUND Hypertension is associated with endothelial dysfunction. An imbalance in the production of Nitric Oxide (NO) and Reactive Oxygen Species (ROS), leading to impaired NO-cyclic Guanosine Monophosphate (cGMP) pathway, contributes to this disorder. Red Yeast Rice (RYR), produced from the fermentation of rice with Monascus purpureus, is a traditional functional food originating from China. Although recognized for its anti-dyslipidemia properties, there has been growing evidence regarding the anti-hypertensive effects of RYR. However, these studies only focused on its direct and short-term effects. AIM This study aims to investigate the vasoprotective effects of chronic oral RYR administration using Spontaneously Hypertensive Rats (SHR). MATERIALS AND METHODS SHR were randomly divided into 3 groups: SHR - Control; SHR - RYR extract (100 mg/kg/day); SHR - lovastatin (10 mg/kg/day). Wistar-Kyoto Rats (WKY) were used as normotensive controls. All animals were treated for 12 weeks by oral gavage. Systolic Blood Pressure (SBP) was measured weekly (tail-cuff method). Vascular reactivity was determined using isolated rat aortic rings in an organ bath. Aortic ROS, NO, tetrahydrobiopterin (BH4), and cGMP levels were evaluated. RESULTS Administration of RYR attenuated SBP elevation and enhanced endothelium-dependent vasodilation in aortic rings. In addition, RYR decreased ROS production and significantly improved the level of vascular NO, BH4, and cGMP. CONCLUSION In an SHR model, treatment with RYR for 12 weeks exerts an SBP lowering effect that can be attributed to improved vascular function via reduction of oxidative stress, decreased endothelial NO Synthase (eNOS) uncoupling and enhanced NO-cGMP pathway.
Collapse
Affiliation(s)
- Jiunn Jye Tan
- M. Kandiah Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Selangor, Malaysia
| | - Dharmani Devi Murugan
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Wei Chih Ling
- M. Kandiah Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Selangor, Malaysia
| | - Siew-Keah Lee
- M. Kandiah Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Selangor, Malaysia
| | - Waye Hann Kang
- M. Kandiah Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Selangor, Malaysia
- M. Kandiah, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman Sungai Long City Campus Jalan Sungai Long Bandar Sungai Long, Cheras, Kajang, Selangor 43000, Malaysia
| |
Collapse
|
46
|
Ma XF, Zhou YR, Zhou ZX, Liu HT, Zhou BB, Deng NH, Zhou K, Tian Z, Wu ZF, Liu XY, Fu MG, Jiang ZS. TRIM65 Suppresses oxLDL-induced Endothelial Inflammation by Interaction with VCAM-1 in Atherogenesis. Curr Med Chem 2024; 31:4898-4911. [PMID: 37608612 DOI: 10.2174/0929867331666230822152350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 05/30/2023] [Accepted: 06/01/2023] [Indexed: 08/24/2023]
Abstract
BACKGROUND AND OBJECTIVE Endothelial cell activation, characterized by increased levels of vascular cell adhesion molecule 1 (VCAM-1), plays a crucial role in the development of atherosclerosis (AS). Therefore, inhibition of VCAM-1-mediated inflammatory response is of great significance in the prevention and treatment of AS. The tripartite motif (TRIM) protein-TRIM65 is involved in the regulation of cancer development, antivirals and inflammation. We aimed to study the functions of TRIM65 in regulating endothelial inflammation by interacting with VCAM-1 in atherogenesis. METHODS AND RESULTS In vitro, we report that human umbilical vein endothelial cells (HUVECs) treated with oxidized low-density lipoprotein (oxLDL) significantly upregulate the expression of TRIM65 in a time- and dose-dependent manner. Overexpression of TRIM65 reduces oxLDL-triggered VCAM-1 protein expression, decreases monocyte adhesion to HUVECs and inhibits the production of the inflammatory cytokines IL-1β, IL-6, IL-8, and TNF-α as well as endothelial oxLDL transcytosis. In contrast, siRNA-mediated knockdown of TRIM65 promotes the expression of VCAM-1, resulting in increased adhesion of monocytes and the release of the inflammatory cytokines IL-1β, IL-6, IL-8, and TNF-α and enhances endothelial oxLDL transcytosis. In vivo, we measured the high expression of TRIM65 in ApoE-/- mouse aortic plaques compared to C57BL/6J mouse aortic plaques. Then, we examined whether the blood levels of VCAM-1 were higher in TRIM65 knockout ApoE-/- mice than in control mice induced by a Western diet. Furthermore, Western blot results showed that the protein expression of VCAM-1 was markedly enhanced in TRIM65 knockout ApoE-/- mouse aortic tissues compared to that of the controls. Immunofluorescence staining revealed that the expression of VCAM-1 was significantly increased in atherosclerotic plaques of TRIM65-/-/ApoE-/- aortic vessels compared to ApoE-/- controls. Mechanistically, TRIM65 specifically interacts with VCAM-1 and targets it for K48-linked ubiquitination. CONCLUSION Our studies indicate that TRIM65 attenuates the endothelial inflammatory response by targeting VCAM-1 for ubiquitination and provides a potential therapeutic target for the inhibition of endothelial inflammation in AS.
Collapse
Affiliation(s)
- Xiao-Feng Ma
- Department of Cardiology, Affiliated Nanhua Hospital, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Yi-Ren Zhou
- Department of Cardiology, Affiliated Nanhua Hospital, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Zhi-Xiang Zhou
- Key Laboratory for Arteriosclerosis of Hunan Province, Institute of Cardiovascular Disease, Hengyang Medical College, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Hui-Ting Liu
- Key Laboratory for Arteriosclerosis of Hunan Province, Institute of Cardiovascular Disease, Hengyang Medical College, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Bo-Bin Zhou
- Key Laboratory for Arteriosclerosis of Hunan Province, Institute of Cardiovascular Disease, Hengyang Medical College, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Nian-Hua Deng
- Key Laboratory for Arteriosclerosis of Hunan Province, Institute of Cardiovascular Disease, Hengyang Medical College, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Kun Zhou
- Key Laboratory for Arteriosclerosis of Hunan Province, Institute of Cardiovascular Disease, Hengyang Medical College, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Zhen Tian
- Key Laboratory for Arteriosclerosis of Hunan Province, Institute of Cardiovascular Disease, Hengyang Medical College, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Ze-Fan Wu
- Key Laboratory for Arteriosclerosis of Hunan Province, Institute of Cardiovascular Disease, Hengyang Medical College, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Xi-Yan Liu
- Key Laboratory for Arteriosclerosis of Hunan Province, Institute of Cardiovascular Disease, Hengyang Medical College, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Ming-Gui Fu
- Department of Basic Medical Science, School of Medicine, University of Missouri Kansas City, Kansas City, MO 64108, USA
| | - Zhi-Sheng Jiang
- Key Laboratory for Arteriosclerosis of Hunan Province, Institute of Cardiovascular Disease, Hengyang Medical College, University of South China, Hengyang City, Hunan Province 421001, PR China
| |
Collapse
|
47
|
Cheng K, Chen X, Luo Y, Sun W, Yang X, Huang S, Wang Y, Wu D. Cumulative Exposure to Oxidized Low-density Lipoprotein is a Potential Predictor for Prognosis in Acute Ischemic Stroke: A Cohort Study. Curr Pharm Des 2024; 30:778-785. [PMID: 38424424 DOI: 10.2174/0113816128280291240220093912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 12/09/2023] [Accepted: 01/17/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND Oxidized Low-Density Lipoprotein (ox-LDL) is crucial in the recrudescence and prognosis of acute ischemic stroke (AIS). We aimed to probe into the influence of cumulative ox-LDL exposure on the 90-day prognosis of AIS. METHODS Patients with AIS were recruited in this research. AIS severity at admission was estimated with infarct volumes and National Institute of Health Stroke Scale (NIHSS) scores. AIS prognosis was assessed using Modified Rankin Scale (mRS) scores at 90 days and the change in NIHSS scores from admission to discharge. Cumulative ox-LDL exposure was defined as ox-LDL level (pg/mL) multiplied by age(y). Multivariate logistic regression analysis was employed to reveal the correlation between exposure factors and the prognosis of AIS. The prognostic prediction ability of cumulative ox-LDL exposure was compared with cumulative LDL exposure by the receiver operating characteristic curve (ROC). RESULTS Higher cumulative ox-LDL exposure was related to worse prognosis, including neurological worsening at discharge (NIHSS increasing more than 2 points) (OR = 3.02, 95% CI, 1.30-6.98, P = 0.01) and poor functional prognosis at 90 days (mRS ≥ 3) (OR = 21.21, 95% CI, 4.72-95.36, P < 0.001). As multivariate regression analysis showed, significantly increased cumulative ox-LDL exposure was relevant to poor functional prognosis at 90 days (OR = 9.92, 95% CI, 1.23-79.76, P = 0.031), but not with neurological worsening at discharge (P = 0.414). ROC curve revealed that cumulative ox-LDL exposure had a higher predictive value (AUC = 0.843, P < 0.001) for functional prognosis of AIS than cumulative LDL exposure (AUC = 0.629, P = 0.023). CONCLUSION Cumulative ox-LDL exposure has a positive correlation with poor prognosis at 90 days of AIS, and has a more accurate predictive ability than cumulative LDL exposure.
Collapse
Affiliation(s)
- Kaili Cheng
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| | - Xiuqi Chen
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| | - Yufan Luo
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| | - Wenbo Sun
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| | - Xiaoli Yang
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| | - Shengwen Huang
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| | - Yuanyuan Wang
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| | - Danhong Wu
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| |
Collapse
|
48
|
Xuan C, Li J, Liu RH, Guo JJ, Zhao C, Zhou TT, Wang Y, He GW, Lun LM. Association between serum gamma-glutamyltransferase and early-onset coronary artery disease: a retrospective case-control study. Ann Med 2023; 55:2289606. [PMID: 38061693 PMCID: PMC10836269 DOI: 10.1080/07853890.2023.2289606] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Serum gamma-glutamyltransferase (GGT) activity has been proposed as a promising predictor of atherosclerosis-related complications and a prognostic marker for cardiovascular diseases. The objective of this study was to investigate the potential correlation between serum levels of GGT and early-onset coronary artery disease (EOCAD). METHODS A retrospective, hospital-based case-control study was conducted, which included 860 patients with EOCAD and gender- and age-matched controls. Serum levels of GGT were measured using the reference measurement procedure on an automatic biochemistry analyser. RESULTS The serum GGT levels of patients with EOCAD (34.90 ± 31.44 U/L) were significantly higher than those of the control group (21.57 ± 16.44 U/L, p < .001). Elevated serum levels of GGT were found to be an independent risk factor for EOCAD, with an odds ratio (OR) of 1.021 (95% confidence interval (CI): 1.014-1.029). Additionally, for every quartile increase in serum GGT levels, the risk of developing EOCAD increased by 1.6-fold. Moreover, serum GGT levels were significantly associated with disease severity, with lower GGT levels observed in patients without significant vascular disease (31.74 ± 24.06 U/L) compared to those with two-vessel disease (33.06 ± 25.00 U/L, p = .002) and three-vessel disease (37.75 ± 36.76 U/L, p = .001). CONCLUSIONS The results of this study suggest that elevated serum GGT levels are associated with the development of EOCAD, and GGT may be implicated in the pathogenesis of the disease. Further large-scale prospective studies are needed to explore the potential relationship between serum GGT levels and the dynamic development of EOCAD.
Collapse
Affiliation(s)
- Chao Xuan
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jing Li
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ru-Hua Liu
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jun-Jie Guo
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Cong Zhao
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ting-Ting Zhou
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yan Wang
- Health Management Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Guo-Wei He
- Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Department of Surgery, Oregon Health and Science University, Portland, OR, USA
| | - Li-Min Lun
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
49
|
Lê S, Cecchin-Albertoni C, Thomas C, Kemoun P, Minty M, Blasco-Baque V. The Role of Dysbiotic Oral Microbiota in Cardiometabolic Diseases: A Narrative Review. Diagnostics (Basel) 2023; 13:3184. [PMID: 37892006 PMCID: PMC10605832 DOI: 10.3390/diagnostics13203184] [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: 08/31/2023] [Revised: 09/25/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
Over the past decade, there have been significant advancements in the high-flow analysis of "omics," shedding light on the relationship between the microbiota and the host. However, the full recognition of this relationship and its implications in cardiometabolic diseases are still underway, despite advancements in understanding the pathophysiology of these conditions. Cardiometabolic diseases, which include a range of conditions from insulin resistance to cardiovascular disease and type 2 diabetes, continue to be the leading cause of mortality worldwide, with a persistently high morbidity rate. While the link between the intestinal microbiota and cardiometabolic risks has been extensively explored, the role of the oral microbiota, the second-largest microbiota in the human body, and specifically the dysbiosis of this microbiota in causing these complications, remains incompletely defined. This review aims to examine the association between the oral microbiota and cardiometabolic diseases, focusing on the dysbiosis of the oral microbiota, particularly in periodontal disease. Additionally, we will dive into the mechanistic aspects of this dysbiosis that contribute to the development of these complications. Finally, we will discuss potential prevention and treatment strategies, including the use of prebiotics, probiotics, and other interventions.
Collapse
Affiliation(s)
- Sylvie Lê
- Département Dentaire, Université Paul Sabatier III (UPS), 3 Chemin des Maraîchers, CEDEX 9, 31062 Toulouse, France; (S.L.); (C.C.-A.); (C.T.); (P.K.); (M.M.)
- Service d’Odontologie Toulouse, CHU Toulouse, 3 Chemin des Maraîchers, CEDEX 9, 31062 Toulouse, France
- Team InCOMM/Intestine ClinicOmics Metabolism & Microbiota, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC)—UMR1297 Inserm, Université Paul Sabatier, 1 Avenue Jean Poulhes, 31432 Toulouse, France
| | - Chiara Cecchin-Albertoni
- Département Dentaire, Université Paul Sabatier III (UPS), 3 Chemin des Maraîchers, CEDEX 9, 31062 Toulouse, France; (S.L.); (C.C.-A.); (C.T.); (P.K.); (M.M.)
- Service d’Odontologie Toulouse, CHU Toulouse, 3 Chemin des Maraîchers, CEDEX 9, 31062 Toulouse, France
- RESTORE Research Center, CNRS, EFS, ENVT, Batiment INCERE, INSERM, Université de Toulouse, 4 bis Avenue Hubert Curien, 31100 Toulouse, France
| | - Charlotte Thomas
- Département Dentaire, Université Paul Sabatier III (UPS), 3 Chemin des Maraîchers, CEDEX 9, 31062 Toulouse, France; (S.L.); (C.C.-A.); (C.T.); (P.K.); (M.M.)
- Service d’Odontologie Toulouse, CHU Toulouse, 3 Chemin des Maraîchers, CEDEX 9, 31062 Toulouse, France
- Team InCOMM/Intestine ClinicOmics Metabolism & Microbiota, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC)—UMR1297 Inserm, Université Paul Sabatier, 1 Avenue Jean Poulhes, 31432 Toulouse, France
| | - Philippe Kemoun
- Département Dentaire, Université Paul Sabatier III (UPS), 3 Chemin des Maraîchers, CEDEX 9, 31062 Toulouse, France; (S.L.); (C.C.-A.); (C.T.); (P.K.); (M.M.)
- Service d’Odontologie Toulouse, CHU Toulouse, 3 Chemin des Maraîchers, CEDEX 9, 31062 Toulouse, France
- RESTORE Research Center, CNRS, EFS, ENVT, Batiment INCERE, INSERM, Université de Toulouse, 4 bis Avenue Hubert Curien, 31100 Toulouse, France
| | - Matthieu Minty
- Département Dentaire, Université Paul Sabatier III (UPS), 3 Chemin des Maraîchers, CEDEX 9, 31062 Toulouse, France; (S.L.); (C.C.-A.); (C.T.); (P.K.); (M.M.)
- Service d’Odontologie Toulouse, CHU Toulouse, 3 Chemin des Maraîchers, CEDEX 9, 31062 Toulouse, France
- Team InCOMM/Intestine ClinicOmics Metabolism & Microbiota, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC)—UMR1297 Inserm, Université Paul Sabatier, 1 Avenue Jean Poulhes, 31432 Toulouse, France
| | - Vincent Blasco-Baque
- Département Dentaire, Université Paul Sabatier III (UPS), 3 Chemin des Maraîchers, CEDEX 9, 31062 Toulouse, France; (S.L.); (C.C.-A.); (C.T.); (P.K.); (M.M.)
- Service d’Odontologie Toulouse, CHU Toulouse, 3 Chemin des Maraîchers, CEDEX 9, 31062 Toulouse, France
- Team InCOMM/Intestine ClinicOmics Metabolism & Microbiota, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC)—UMR1297 Inserm, Université Paul Sabatier, 1 Avenue Jean Poulhes, 31432 Toulouse, France
| |
Collapse
|
50
|
Nakhlband A, Garjani A, Saeedi N, Omidi Y, Ghaffari S, Barar J, Eskandani M. Atherosclerosis preventive effects of marrubiin against (TNF-α)-induced oxidative stress and apoptosis. J Cardiovasc Thorac Res 2023; 15:174-180. [PMID: 38028719 PMCID: PMC10590460 DOI: 10.34172/jcvtr.2023.31704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 08/04/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Atherosclerosis is a complicated cascade of inflammatory processes, oxidative stress, and apoptosis, making it the most prevalent cardiovascular disease. The onset and progression of cardiovascular diseases are greatly influenced by oxidative stress. Targeting oxidative stress is an effective strategy for treating such diseases. Marrubiin is a bioactive furan labdane diterpenoid acts as a strong antioxidant to protect against oxidative damage. This study aimed to investigate the protective effects of marrubiin against oxidative stress and apoptosis in a cellular model of the vascular system. Methods Human umbilical vein endothelial cells were treated with varying concentration of marrubiin and its IC50 value was determined. The antioxidant potential of marrubiin was assessed by measuring the intracellular level of glutathione (GSH) using a colorimetric technique. Since apoptosis plays a significant role in the plaque rupture, the study also evaluated the protective effects of marrubiin on the expression of key genes involved in apoptotic pathways. Results Cells treated with marrubiin showed increased GSH levels compared to cell therapy control cells, indicating marrubiin's ability to counteract the effects of TNF-α's on GSH levels. Furthermore real-time PCR analysis demonstrated that marrubiin upregulated Bcl-xl while downregulating caspase3 and Nox4 in treated cells. These findings suggest that marrubiin protects against apoptosis and oxidative stress. Conclusion Based on our findings, marrubiin is recommended as a preventive/therapeutic treatment for diseases caused by elevated intracellular reactive oxygen species levels in cardiovascular diseases.
Collapse
Affiliation(s)
- Ailar Nakhlband
- Research Center of Psychiatry and Behavioral Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Garjani
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazli Saeedi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Omidi
- Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Samad Ghaffari
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jaleh Barar
- Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Morteza Eskandani
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|