1
|
Bi A, Liu R, Xie M, He B, Yan T, Du Y, Jia Y. Semen Ziziphi Spinosae alleviates cardiomyocyte apoptosis in rats with coronary heart disease via the AMPK/SIRT1/PGC-1α signaling pathway activation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156743. [PMID: 40250033 DOI: 10.1016/j.phymed.2025.156743] [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: 02/01/2025] [Revised: 03/23/2025] [Accepted: 04/08/2025] [Indexed: 04/20/2025]
Abstract
BACKGROUND Coronary heart disease (CHD) represents a significant cardiovascular condition, with its occurrence increasing as a result of alterations in lifestyle and dietary habits. Semen Ziziphi Spinosae (SZS) is commonly utilized for the management of disorders associated with the nervous system, including conditions like depression and insomnia. Recent research has revealed its potential therapeutic properties for cardiovascular issues. Nevertheless, there exists a limited amount of research addressing the mechanisms involved. PURPOSE This research seeks to explore the protective effects that SZS has on cardiac tissue, specifically within the framework of CHD. By conducting this investigation, the study aims to uncover the various mechanisms that play a role in these protective effects. This understanding could yield significant insights into how SZS may result in the preservation and enhancement of cardiac health in patients affected by CHD. STUDY DESIGN The study innovatively combines multiple advanced techniques. It first integrates UPLC-Q-TOF/MS analysis and network pharmacology to identify SZS components. In vitro experiments were conducted using H9c2 rat cardiomyocytes, and in vivo experiments used a CHD model in SD rats. Multiple assays were performed for multi - level and multi - dimensional validation. METHODS In the initial stage, the primary components of SZS and their possible mechanisms for combating CHD were examined through UPLC-Q-TOF/MS analysis in conjunction with network pharmacology approaches. For the in vitro investigation, an ischemia-hypoxia model was established utilizing H9c2 rat cardiomyocytes. The CCK-8 assay was used to assess myocardial injury markers. TUNEL staining and Western blot techniques were employed to confirm the impact of SZS treatment on apoptosis in H9c2 cells. The expression levels of proteins associated with the AMPK/SIRT1/PGC-1α signaling pathway were measured using RT-qPCR and Western blotting, and the results were validated with the AMPK inhibitor, compound C. In the in vivo segment, a model of coronary heart disease (CHD) in SD rats was established through the administration of a high-fat emulsion diet combined with pituitrin injections. Cardiac function in the rats was evaluated through electrocardiograms and echocardiograms. Pathological changes in the heart were observed utilizing TTC and H&E staining. Kits were implemented to measure the serum biochemical indicators in the rats.RT - qPCR and Western blotting were employed to measure the expression levels of proteins related to the AMPK/SIRT1/PGC - 1α signaling pathway. RESULTS The study identified 67 in vitro components, 27 blood - absorbed components, and 12 metabolic components of SZS. Network pharmacology analysis suggested the AMPK/SIRT1/PGC - 1α signaling pathway as a key mechanism. In vitro and in vivo experiments showed that SZS increased cell viability, reduced apoptosis, and activated the AMPK/SIRT1/PGC - 1α signaling pathway. Inhibiting AMPK abolished SZS's effects. SZS also improved cardiac function and reduced myocardial damage in rats with CHD. CONCLUSION This study for the first time highlights that Semen Ziziphi Spinosae plays a beneficial role in cardiovascular health by activating the AMPK/SIRT1/PGC-1α signaling pathway and reducing apoptosis in cardiomyocytes. These findings support its potential application in the treatment of CHD and other cardiac conditions.
Collapse
Affiliation(s)
- Anqi Bi
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Rihong Liu
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Min Xie
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Bosai He
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Tingxu Yan
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Yiyang Du
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China.
| | - Ying Jia
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China.
| |
Collapse
|
2
|
Wang H, Zhang J, Li G, Liu B, Liu M, Tang H, Wen H, He F. Circular RNA transcriptome across various development periods of Paralichthys olivaceus reveal skeletal muscle-specific circchd6 regulating myogenesis. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2025; 55:101518. [PMID: 40334353 DOI: 10.1016/j.cbd.2025.101518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/06/2025] [Accepted: 04/21/2025] [Indexed: 05/09/2025]
Abstract
The Japanese flounder (Paralichthys olivaceus) is greatly influenced in terms of muscle quality and quantity by the development of skeletal muscle. While the mechanisms underlying skeletal muscle development are well-studied, the role of non-coding RNAs, particularly circRNAs, in the skeletal muscle development of Japanese flounder remains unclear. To investigate the expression patterns of circRNAs during different developmental stages (JP1: 7 days, JP2: 90 days, JP3: 24 months (female), JP4: 24 months (male)) in Japanese flounder, we performed transcriptome sequencing analysis. We identified a total of 3523 circRNAs, of which 10.19 % were differentially expressed. These differentially expressed (DE) circRNAs were studied, and their impacts on muscle development were analyzed. The RNA interaction network revealed that skeletal muscle-specific circchd6 targeted novel-miR-508 and further regulated dual specificity tyrosine-phosphorylation regulated kinase 2 (dyrk2). Functional analysis showed that overexpressed circchd6 and dyrk2 promoted myoblast proliferation and differentiation, while novel-miR-508 inhibited both. Our study identified the circchd6-novel-miR-508-dyrk2 axis as a regulatory mechanism and provided new evidence for the use of epigenetic approaches in genetic breeding.
Collapse
Affiliation(s)
- Hao Wang
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266000, China
| | - Jingru Zhang
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266000, China
| | - Guangling Li
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266000, China
| | - Binghua Liu
- Key Lab of Sustainable Development of Marine Fisheries, Ministry of Agriculture and Rural Affairs, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China
| | - Min Liu
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266000, China
| | - Hengtai Tang
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266000, China
| | - Haishen Wen
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266000, China
| | - Feng He
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266000, China.
| |
Collapse
|
3
|
Jiang J, Liu F, Cui D, Xu C, Chi J, Yan T, Guo F. Novel molecular mechanisms of immune evasion in hepatocellular carcinoma: NSUN2-mediated increase of SOAT2 RNA methylation. Cancer Commun (Lond) 2025. [PMID: 40227950 DOI: 10.1002/cac2.70023] [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: 03/01/2024] [Revised: 03/13/2025] [Accepted: 03/17/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a deadly malignancy known for its ability to evade immune surveillance. NOP2/Sun RNA methyltransferase family member 2 (NSUN2), an RNA methyltransferase involved in carcinogenesis, has been associated with immune evasion and energy metabolism reprogramming. This study aimed to examine the molecular mechanisms underlying the involvement of NSUN2 in immune evasion and metabolic reprogramming of HCC. METHODS Single-cell transcriptomic sequencing was applied to examine cellular composition changes, particularly immune cell dynamics, in HCC and adjacent normal tissues. Bulk RNA-seq and proteomics identified key genes and proteins. Methylation sequencing and methylated RNA immunoprecipitation (MeRIP) were carried out to characterize the role of NSUN2 in 5-methylcytosine (m5C) modification of sterol O-acyltransferase 2 (SOAT2). Clinical samples from 30 HCC patients were analyzed using reverse transcription-quantitative polymerase chain reaction and Western blotting. Gene expression was manipulated using CRISPR/Cas9 and lentiviral vectors. In vitro co-culture models and metabolomics were used to study HCC cell-T cell interactions, energy metabolism, and immune evasion. Tumor growth in an orthotopic mouse model was monitored by bioluminescence imaging, with subsequent measurements of tumor weight, volume, and immunohistochemical staining. RESULTS Single-cell transcriptomic analysis identified a marked increase in malignant cells in HCC tissues. Cell communication analysis indicated that tumor cells might promote cancer progression by evading immune clearance. Multi-omics analyses identified NSUN2 as a key regulator in HCC development. MeRIP confirmed that NSUN2 facilitated the m5C modification of SOAT2. Analysis of human HCC tissue samples demonstrated pronounced upregulation of NSUN2 and SOAT2, along with elevated m5C levels in HCC tissues. In vitro experiments uncovered that NSUN2 augmented the reprogramming of energy metabolism and repressed the activity and cytotoxicity of CD8+ T cells, contributing to immune evasion. In vivo studies further substantiated the role of NSUN2 in fostering immune evasion and tumor formation of HCC by modulating the m5C modification of SOAT2. CONCLUSIONS The findings highlight the critical role of NSUN2 in driving HCC progression through the regulation of m5C modification on SOAT2. These findings present potential molecular markers for HCC diagnosis and therapeutic targets for its treatment.
Collapse
Affiliation(s)
- Jinhua Jiang
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P. R. China
- Key Laboratory of Systems Biomedicine, Ministry of Education, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, P. R. China
| | - Feng Liu
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P. R. China
| | - Dan Cui
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P. R. China
| | - Caixia Xu
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P. R. China
| | - Jiachang Chi
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P. R. China
| | - Tinghua Yan
- The First Clinical Medical College of Jinan University, Guangzhou, Guangdong, P. R. China
| | - Fang Guo
- Key Laboratory of Systems Biomedicine, Ministry of Education, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, P. R. China
| |
Collapse
|
4
|
Wang Y, Ling S, Feng H, Hua J, Han Z, Chai R. Recent Advances in the Mutual Regulation of m6A Modification and Non-Coding RNAs in Atherosclerosis. Int J Gen Med 2025; 18:1047-1073. [PMID: 40026815 PMCID: PMC11871936 DOI: 10.2147/ijgm.s508197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 02/20/2025] [Indexed: 03/05/2025] Open
Abstract
Atherosclerosis, a progressive inflammatory disease of the arteries, remains a leading cause of cardiovascular morbidity and mortality worldwide. Recent years have witnessed the pivotal role of N6-methyladenosine (m6A) RNA methylation in regulating various biological processes, including those implicated in atherosclerosis. Current evidence suggested that m6A regulators (writers, erasers, and readers) participated in the modification of multiple non-coding RNAs (ncRNAs), including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), thereby affecting their metabolism and functions. Meanwhile, ncRNAs have also emerged as key modulator of m6A regulators expression in turn. Therefore, understanding the mutual regulation between m6A modifications and ncRNAs is of great significance to identify novel therapeutic targets for atherosclerosis and has great clinical application prospects. This review aims to summarize the recent advances in the reciprocal regulation and provide insights into the interaction between m6A modification and ncRNAs in the context of atherosclerosis.
Collapse
Affiliation(s)
- Yanlu Wang
- Department of Cardiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, 510260, People’s Republic of China
| | - Sisi Ling
- Department of Cardiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, 510260, People’s Republic of China
| | - Hao Feng
- Department of Cardiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, 510260, People’s Republic of China
| | - Junkai Hua
- Department of Cardiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, 510260, People’s Republic of China
| | - Zhiyu Han
- Department of Cardiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, 510260, People’s Republic of China
| | - Renjie Chai
- Department of Cardiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, 510260, People’s Republic of China
| |
Collapse
|
5
|
Wang X, Nie H, Su M, Wu Y, Pang Q, Zhang Y, He C, Song Y. Serum CircNIPSNAP3A is Associated with Metabolic Disorders, Atherosclerosis and Severity of Coronary Artery Disease in a Chinese Population. TOHOKU J EXP MED 2024; 263:123-131. [PMID: 38569888 DOI: 10.1620/tjem.2024.j023] [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] [Indexed: 04/05/2024]
Abstract
The relationships of serum circNIPSNAP3A and circHIPK3 with metabolic disorders, atherosclerosis and severity of coronary artery disease (CAD) remain to be clarified. Three hundred and thirty-eight subjects were categorized into normal coronary artery, atherosclerosis and CAD groups. Clinical data including anthropometric indexes, medical history, and physiological and biochemical parameters were collected. Serum circNIPSNAP3A and circHIPK3 were determined by quantitative real-time PCR. CAD severity was evaluated by clinical manifestation, electrocardiogram and coronary angiography. Both CAD and atherosclerosis groups had a higher serum level of circNIPSNAP3A than the normal coronary artery group (P < 0.05 for all). The subjects with a high percentage (> 66th percentile) of circNIPSNAP3A had higher mean levels of triglycerides, uric acid and homocysteine, and lower mean levels of high-density lipoprotein cholesterol and apolipoprotein AI than those with a low percentage (< 33rd percentile) of circNIPSNAP3A. Notably, circNIPSNAP3A is significantly and independently associated with CAD, and subjects with a high percentage of circNIPSNAP3A had more diseased coronary branches and a higher incidence of acute coronary syndrome than those with a low percentage of circNIPSNAP3A. Regarding circHIPK3, subjects with a medium or high percentage of circHIPK3 had a lower mean level of apolipoprotein AI than those with a low percentage of circHIPK3, but no significant differences in the incidence and severity of CAD among the < 33rd, 33rd-66th, and > 66th percentiles of circHIPK3 were detected. Serum circNIPSNAP3A is related to cardiovascular risk factors and CAD severity, and may be a potential prognostic marker and/or therapeutic target for CAD.
Collapse
Affiliation(s)
- Xue Wang
- Central Laboratory, Clinical Medical College and Affiliated Hospital of Chengdu University
| | - Haiyan Nie
- Clinical Medical College of Chengdu University
| | - Mi Su
- Functional Science Laboratory, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University
| | - Yang Wu
- Clinical Medical College of Chengdu University
| | | | - Youjin Zhang
- Central Laboratory, Clinical Medical College and Affiliated Hospital of Chengdu University
| | - Chuan He
- Department of Cardiology, Clinical Medical College and Affiliated Hospital of Chengdu University
| | - Yongyan Song
- Central Laboratory, Clinical Medical College and Affiliated Hospital of Chengdu University
| |
Collapse
|
6
|
Wen SY, Zhi X, Liu HX, Wang X, Chen YY, Wang L. Is the suppression of CD36 a promising way for atherosclerosis therapy? Biochem Pharmacol 2024; 219:115965. [PMID: 38043719 DOI: 10.1016/j.bcp.2023.115965] [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: 10/07/2023] [Revised: 11/29/2023] [Accepted: 11/30/2023] [Indexed: 12/05/2023]
Abstract
Atherosclerosis is the main underlying pathology of many cardiovascular diseases and is marked by plaque formation in the artery wall. It has posed a serious threat to the health of people all over the world. CD36 acts as a significant regulator of lipid homeostasis, which is closely associated with the onset and progression of atherosclerosis and may be a new therapeutic target. The abnormal overexpression of CD36 facilitates lipid accumulation, foam cell formation, inflammation, endothelial apoptosis, and thrombosis. Numerous natural products and lipid-lowering agents are found to target the suppression of CD36 or inhibit the upregulation of CD36 to prevent and treat atherosclerosis. Here, the structure, expression regulation and function of CD36 in atherosclerosis and its related pharmacological therapies are reviewed. This review highlights the importance of drugs targeting CD36 suppression in the treatment and prevention of atherosclerosis, in order to develop new therapeutic strategies and potential anti-atherosclerotic drugs both preclinically and clinically.
Collapse
Affiliation(s)
- Shi-Yuan Wen
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Xiaoyan Zhi
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Hai-Xin Liu
- School of Traditional Chinese Materia Medica, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Xiaohui Wang
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Yan-Yan Chen
- School of Medicine, Jiangsu University, Zhenjiang, China.
| | - Li Wang
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
7
|
Dergunova LV, Vinogradina MA, Filippenkov IB, Limborska SA, Dergunov AD. Circular RNAs Variously Participate in Coronary Atherogenesis. Curr Issues Mol Biol 2023; 45:6682-6700. [PMID: 37623241 PMCID: PMC10453518 DOI: 10.3390/cimb45080422] [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: 06/29/2023] [Revised: 08/03/2023] [Accepted: 08/11/2023] [Indexed: 08/26/2023] Open
Abstract
Over the past decade, numerous studies have shown that circular RNAs (circRNAs) play a significant role in coronary artery atherogenesis and other cardiovascular diseases. They belong to the class of non-coding RNAs and arise as a result of non-canonical splicing of premature RNA, which results in the formation of closed single-stranded circRNA molecules that lack 5'-end caps and 3'-end poly(A) tails. circRNAs have broad post-transcriptional regulatory activity. Acting as a sponge for miRNAs, circRNAs compete with mRNAs for binding to miRNAs, acting as competing endogenous RNAs. Numerous circRNAs are involved in the circRNA-miRNA-mRNA regulatory axes associated with the pathogenesis of cardiomyopathy, chronic heart failure, hypertension, atherosclerosis, and coronary artery disease. Recent studies have shown that сirc_0001445, circ_0000345, circ_0093887, сircSmoc1-2, and circ_0003423 are involved in the pathogenesis of coronary artery disease (CAD) with an atheroprotective effect, while circ_0002984, circ_0029589, circ_0124644, circ_0091822, and circ_0050486 possess a proatherogenic effect. With their high resistance to endonucleases, circRNAs are promising diagnostic biomarkers and therapeutic targets. This review aims to provide updated information on the involvement of atherogenesis-related circRNAs in the pathogenesis of CAD. We also discuss the main modern approaches to detecting and studying circRNA-miRNA-mRNA interactions, as well as the prospects for using circRNAs as biomarkers and therapeutic targets for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Liudmila V. Dergunova
- Laboratory of Human Molecular Genetics, National Research Center “Kurchatov Institute”, Kurchatov Sq. 2, Moscow 123182, Russia; (M.A.V.); (I.B.F.); (S.A.L.)
| | - Margarita A. Vinogradina
- Laboratory of Human Molecular Genetics, National Research Center “Kurchatov Institute”, Kurchatov Sq. 2, Moscow 123182, Russia; (M.A.V.); (I.B.F.); (S.A.L.)
| | - Ivan B. Filippenkov
- Laboratory of Human Molecular Genetics, National Research Center “Kurchatov Institute”, Kurchatov Sq. 2, Moscow 123182, Russia; (M.A.V.); (I.B.F.); (S.A.L.)
| | - Svetlana A. Limborska
- Laboratory of Human Molecular Genetics, National Research Center “Kurchatov Institute”, Kurchatov Sq. 2, Moscow 123182, Russia; (M.A.V.); (I.B.F.); (S.A.L.)
| | - Alexander D. Dergunov
- Laboratory of Structural Fundamentals of Lipoprotein Metabolism, National Medical Research Center for Therapy and Preventive Medicine, Petroverigsky Street 10, Moscow 101990, Russia;
| |
Collapse
|
8
|
Galindo CL, Khan S, Zhang X, Yeh YS, Liu Z, Razani B. Lipid-laden foam cells in the pathology of atherosclerosis: shedding light on new therapeutic targets. Expert Opin Ther Targets 2023; 27:1231-1245. [PMID: 38009300 PMCID: PMC10843715 DOI: 10.1080/14728222.2023.2288272] [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: 06/28/2023] [Accepted: 11/22/2023] [Indexed: 11/28/2023]
Abstract
INTRODUCTION Lipid-laden foam cells within atherosclerotic plaques are key players in all phases of lesion development including its progression, necrotic core formation, fibrous cap thinning, and eventually plaque rupture. Manipulating foam cell biology is thus an attractive therapeutic strategy at early, middle, and even late stages of atherosclerosis. Traditional therapies have focused on prevention, especially lowering plasma lipid levels. Despite these interventions, atherosclerosis remains a major cause of cardiovascular disease, responsible for the largest numbers of death worldwide. AREAS COVERED Foam cells within atherosclerotic plaques are comprised of macrophages, vascular smooth muscle cells, and other cell types which are exposed to high concentrations of lipoproteins accumulating within the subendothelial intimal layer. Macrophage-derived foam cells are particularly well studied and have provided important insights into lipid metabolism and atherogenesis. The contributions of foam cell-based processes are discussed with an emphasis on areas of therapeutic potential and directions for drug development. EXERT OPINION As key players in atherosclerosis, foam cells are attractive targets for developing more specific, targeted therapies aimed at resolving atherosclerotic plaques. Recent advances in our understanding of lipid handling within these cells provide insights into how they might be manipulated and clinically translated to better treat atherosclerosis.
Collapse
Affiliation(s)
- Cristi L. Galindo
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
| | - Saifur Khan
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
| | - Xiangyu Zhang
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
| | - Yu-Sheng Yeh
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
| | - Ziyang Liu
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
| | - Babak Razani
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
- Pittsburgh VA Medical Center, Pittsburgh, PA
| |
Collapse
|
9
|
Chen W, Xu J, Wu Y, Liang B, Yan M, Sun C, Wang D, Hu X, Liu L, Hu W, Shao Y, Xing D. The potential role and mechanism of circRNA/miRNA axis in cholesterol synthesis. Int J Biol Sci 2023; 19:2879-2896. [PMID: 37324939 PMCID: PMC10266072 DOI: 10.7150/ijbs.84994] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 05/16/2023] [Indexed: 06/17/2023] Open
Abstract
Cholesterol levels are an initiating risk factor for atherosclerosis. Many genes play a central role in cholesterol synthesis, including HMGCR, SQLE, HMGCS1, FDFT1, LSS, MVK, PMK, MVD, FDPS, CYP51, TM7SF2, LBR, MSMO1, NSDHL, HSD17B7, DHCR24, EBP, SC5D, DHCR7, IDI1/2. Especially, HMGCR, SQLE, FDFT1, LSS, FDPS, CYP51, and EBP are promising therapeutic targets for drug development due to many drugs have been approved and entered into clinical research by targeting these genes. However, new targets and drugs still need to be discovered. Interestingly, many small nucleic acid drugs and vaccines were approved for the market, including Inclisiran, Patisiran, Inotersen, Givosiran, Lumasiran, Nusinersen, Volanesorsen, Eteplirsen, Golodirsen, Viltolarsen, Casimersen, Elasomeran, Tozinameran. However, these agents are all linear RNA agents. Circular RNAs (circRNAs) may have longer half-lives, higher stability, lower immunogenicity, lower production costs, and higher delivery efficiency than these agents due to their covalently closed structures. CircRNA agents are developed by several companies, including Orna Therapeutics, Laronde, and CirCode, Therorna. Many studies have shown that circRNAs regulate cholesterol synthesis by regulating HMGCR, SQLE, HMGCS1, ACS, YWHAG, PTEN, DHCR24, SREBP-2, and PMK expression. MiRNAs are essential for circRNA-mediated cholesterol biosynthesis. Notable, the phase II trial for inhibiting miR-122 with nucleic acid drugs has been completed. Suppressing HMGCR, SQLE, and miR-122 with circRNA_ABCA1, circ-PRKCH, circEZH2, circRNA-SCAP, and circFOXO3 are the promising therapeutic target for drug development, specifically the circFOXO3. This review focuses on the role and mechanism of the circRNA/miRNA axis in cholesterol synthesis in the hope of providing knowledge to identify new targets.
Collapse
Affiliation(s)
- Wujun Chen
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong, 266000, China
| | - Jiazhen Xu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong, 266000, China
| | - Yudong Wu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong, 266000, China
| | - Bing Liang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong, 266000, China
| | - Mingzhe Yan
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong, 266000, China
| | - Chuandong Sun
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong, 266000, China
- Department of Liver Disease Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
| | - Dong Wang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong, 266000, China
- Department of Liver Disease Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
| | - Xiaokun Hu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong, 266000, China
- Interventional Medicine Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
| | - Li Liu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong, 266000, China
- Department of Community Health Promotion, Qingdao Municipal Center for Disease Control & Prevention, Qingdao Institute of Preventive Medicine, Qingdao, Shandong, 266033, China
| | - Wenchao Hu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong, 266000, China
- Department of Endocrinology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong, 266000, China
| | - Yingchun Shao
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong, 266000, China
| | - Dongming Xing
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong, 266000, China
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| |
Collapse
|