1
|
Lin Y, Liu Y, Feng J, Ye F, Hu L, Cao Y. Metabolomics analysis of acute lung injury induced by aortic dissection in mice. Ann Med Surg (Lond) 2025; 87:497-505. [PMID: 40110328 PMCID: PMC11918661 DOI: 10.1097/ms9.0000000000002885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 11/07/2024] [Indexed: 03/22/2025] Open
Abstract
Thoracic aortic aneurysm/dissection (TAA/D) is a complicated vascular disorder with galloping development and high mortality. Phenotype switching plays an important role in the pathological process of TAA/D. Previous studies indicated the potential correlation between the expression level of lncRNA RP11-465L10.10 and MMP9 involved in the development of TAA/D. Here, our results showed that RP11-465L10.10 and MMP9 were highly increased in TAD patient tissues, which was consistent in Ang II-induced vascular smooth muscle cell (VSMC) phenotype switching. However, the effects and underlying mechanism of RP11-465L10.10 on VSMC phenotypic switching remain uncertain. Therefore, the expression of SM22α, cell proliferation, and migration were investigated when ectopically expressed RP11-465L10.10 in VSMCs. The results showed that RP11-465L10.10 overexpression decreased SM22α expression and facilitated VSMC proliferation, migration, and MMP9 expression. Furthermore, the NF-κB signaling pathway was enriched in transcriptome data analysis, indicating that NF-κB signaling may be involved in RP11-465L10.10-induced VSMC phenotype switching and MMP9 expression. In conclusion, this study demonstrated that RP11-465L10.10 induces VSMC phenotype switching and MMP9 expression via the NF-κB signaling pathway, suggesting that RP11-465L10.10 might be a potential therapeutic target for TAA/D treatment.
Collapse
Affiliation(s)
- Yang Lin
- Department of Cardiovascular Surgery of the GaoZhou People's Hospital, Gaozhou, Guangdong, China
| | - Yi Liu
- Department of Cardiovascular Surgery of the GaoZhou People's Hospital, Gaozhou, Guangdong, China
| | - Jianfei Feng
- Department of Cardiovascular Surgery of the GaoZhou People's Hospital, Gaozhou, Guangdong, China
| | - Fuyong Ye
- Department of Cardiovascular Surgery of the GaoZhou People's Hospital, Gaozhou, Guangdong, China
| | - Lian Hu
- Department of Cardiovascular Surgery of the GaoZhou People's Hospital, Gaozhou, Guangdong, China
| | - Yong Cao
- Department of Cardiovascular Surgery of the GaoZhou People's Hospital, Gaozhou, Guangdong, China
| |
Collapse
|
2
|
Ma CN, Shi SR, Zhang XY, Xin GS, Zou X, Li WL, Guo SD. Targeting PDGF/PDGFR Signaling Pathway by microRNA, lncRNA, and circRNA for Therapy of Vascular Diseases: A Narrow Review. Biomolecules 2024; 14:1446. [PMID: 39595622 PMCID: PMC11592287 DOI: 10.3390/biom14111446] [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: 09/24/2024] [Revised: 11/05/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Despite the significant progress in diagnostic and therapeutic strategies, vascular diseases, such as cardiovascular diseases (CVDs) and respiratory diseases, still cannot be successfully eliminated. Vascular cells play a key role in maintaining vascular homeostasis. Notably, a variety of cells produce and secrete platelet-derived growth factors (PDGFs), which promote mitosis and induce the division, proliferation, and migration of vascular cells including vascular smooth muscle cells (SMCs), aortic SMCs, endothelial cells, and airway SMCs. Therefore, PDGF/PDGR receptor signaling pathways play vital roles in regulating the homeostasis of blood vessels and the onset and development of CVDs, such as atherosclerosis, and respiratory diseases including asthma and pulmonary arterial hypertension. Recently, accumulating evidence has demonstrated that microRNA, long-chain non-coding RNA, and circular RNA are involved in the regulation of PDGF/PDGFR signaling pathways through competitive interactions with target mRNAs, contributing to the occurrence and development of the above-mentioned diseases. These novel findings are useful for laboratory research and clinical studies. The aim of this article is to conclude the recent progresses in this field, particular the mechanisms of action of these non-coding RNAs in regulating vascular remodeling, providing potential strategies for the diagnosis, prevention, and treatment of vascular-dysfunction-related diseases, particularly CVDs and respiratory diseases.
Collapse
Affiliation(s)
- Chao-Nan Ma
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China; (C.-N.M.); (S.-R.S.); (X.-Y.Z.)
| | - Shan-Rui Shi
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China; (C.-N.M.); (S.-R.S.); (X.-Y.Z.)
| | - Xue-Ying Zhang
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China; (C.-N.M.); (S.-R.S.); (X.-Y.Z.)
| | - Guo-Song Xin
- School of Pharmacy, Engineering Research Center for Medicine, Harbin University of Commerce, Harbin 150076, China; (G.-S.X.); (X.Z.)
| | - Xiang Zou
- School of Pharmacy, Engineering Research Center for Medicine, Harbin University of Commerce, Harbin 150076, China; (G.-S.X.); (X.Z.)
| | - Wen-Lan Li
- School of Pharmacy, Engineering Research Center for Medicine, Harbin University of Commerce, Harbin 150076, China; (G.-S.X.); (X.Z.)
| | - Shou-Dong Guo
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China; (C.-N.M.); (S.-R.S.); (X.-Y.Z.)
- School of Pharmacy, Engineering Research Center for Medicine, Harbin University of Commerce, Harbin 150076, China; (G.-S.X.); (X.Z.)
| |
Collapse
|
3
|
Hussain MS, Afzal O, Gupta G, Altamimi ASA, Almalki WH, Alzarea SI, Kazmi I, Kukreti N, Gupta S, Sulakhiya K, Singh SK, Dua K. Probing the links: Long non-coding RNAs and NF-κB signalling in atherosclerosis. Pathol Res Pract 2023; 249:154773. [PMID: 37647827 DOI: 10.1016/j.prp.2023.154773] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/16/2023] [Accepted: 08/16/2023] [Indexed: 09/01/2023]
Abstract
Atherosclerosis is a chronic inflammatory disease that involves the accumulation of lipids and immune cells in the arterial wall. NF-kB signaling is a key regulator of inflammation and is known to play a critical role in atherosclerosis. Recent studies have shown that lncRNAs can regulate NF-kB and contribute to the development and progression of atherosclerosis. Preliminary findings reveal significant alterations in the expression of specific lncRNAs in atherosclerotic lesions compared to healthy arterial tissue. Experimental evidence suggests that these dysregulated lncRNAs can influence the NF-kB pathway. By unravelling the crosstalk between lncRNAs and NF-kB signaling, this review aims to enhance our understanding of the molecular mechanisms underlying atherosclerosis. Identifying novel therapeutic targets and diagnostic markers may lead to developing interventions and management strategies for this prevalent cardiovascular disease. This review summarizes the current knowledge on the role of lncRNAs in NF-kB signaling in atherosclerosis and highlights their potential as therapeutic targets for this disease.
Collapse
Affiliation(s)
- Md Sadique Hussain
- School of Pharmaceutical Sciences, Jaipur National University, Jagatpura, 302017 Jaipur, Rajasthan, India
| | - Obaid Afzal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Road, Jagatpura 302017, Jaipur, India; Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India.
| | | | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al-Jouf, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Neelima Kukreti
- School of Pharmacy, Graphic Era Hill University, Dehradun 248007, India
| | - Saurabh Gupta
- Chameli Devi Institute of Pharmacy, Department of Pharmacology, Indore, Madhya Pradesh, India
| | - Kunjbihari Sulakhiya
- Neuro Pharmacology Research Laboratory (NPRL), Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, Madhya Pradesh, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia; Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| |
Collapse
|
4
|
Xun M, Zhang J, Wu M, Chen Y. Long non-coding RNAs: The growth controller of vascular smooth muscle cells in cardiovascular diseases. Int J Biochem Cell Biol 2023; 157:106392. [PMID: 36828237 DOI: 10.1016/j.biocel.2023.106392] [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: 10/05/2022] [Revised: 02/13/2023] [Accepted: 02/19/2023] [Indexed: 02/25/2023]
Abstract
The active proliferation and migration of vascular smooth muscle cells supports the healing of vessel damage while their abnormal aggression or destitution contribute to the aberrant intima-medial structure and function in various cardiovascular diseases, so the understanding of the proliferation disorders of vascular smooth muscle cell and the related mechanism is the basis of effective intervention and control for cardiovascular diseases. Recently, long non-coding RNAs have stood out as upstream switchers for multiple proliferative signaling pathways and molecules, and many of them have been shown to conduce to the dysregulated proliferation and apoptosis of vascular smooth muscle cells under various pathogenic stimuli. This article discusses the long non-coding RNAs disclosed and linked to atherosclerosis, pulmonary hypertension, and aneurysms, and focuses upon their modulation of vascular smooth muscle cell population affecting three deadly cardiovascular diseases.
Collapse
Affiliation(s)
- Min Xun
- Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang 421001, China
| | - Jie Zhang
- Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang 421001, China
| | - Meichun Wu
- Hengyang Medical School, University of South China, Hengyang 421001, China; School of Nursing, University of South China, Hengyang 421001, China
| | - Yuping Chen
- Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang 421001, China; Hengyang Medical School, University of South China, Hengyang 421001, China.
| |
Collapse
|
5
|
Shirazi-Tehrani E, Chamasemani A, Firouzabadi N, Mousaei M. ncRNAs and polyphenols: new therapeutic strategies for hypertension. RNA Biol 2022; 19:575-587. [PMID: 35438046 PMCID: PMC9037439 DOI: 10.1080/15476286.2022.2066335] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Polyphenols have gained significant attention in protecting several chronic diseases, such as cardiovascular diseases (CVDs). Accumulating evidence indicates that polyphenols have potential protective roles for various CVDs. Hypertension (HTN) is among the hazardous CVDs accounting for nearly 8.5 million deaths worldwide. HTN is a complex and multifactorial disease and a combination of genetic susceptibility and environmental factors play major roles in its development. However, the underlying regulatory mechanisms are still elusive. Polyphenols have shown to cause favourable and beneficial effects in the management of HTN. Noncoding RNAs (ncRNAs) as influential mediators in modulating the biological properties of polyphenols, have shown significant footprints in CVDs. ncRNAs control basic functions in virtually all cell types relevant to the cardiovascular system and, thus, a direct link with blood pressure (BP) regulation is highly probable. Recent evidence suggests that a number of ncRNAs, including main small ncRNAs, microRNAs (miRNAs) and long ncRNAs (lncRNAs), play crucial roles with respect to the antihypertensive effects of polyphenols. Indeed, targeting lncRNAs by polyphenols will be a novel and promising strategy in the management of HTN. Herein, we reviewed the effects of polyphenols in HTN. Additionally, we emphasized on the potential effects of polyphenols on regulations of main ncRNAs, which imply the role of polyphenols in regulating ncRNAs in order to exert protective effects and thus proposing them as new targets for HTN treatment.Abbreviations : CVD: cardiovascular disease; BP: blood pressure; HTN: hypertension, lncRNAs: long noncoding RNAs; p38-MAPK: p38-mitogenactivated protein kinase; OPCs: oligomeric procyanidins; GTP: guanosine triphosphate; ROS: reactive oxygen species; cGMP: cyclic guanosine monophosphate; SGC: soluble guanylate cyclase; PI3K: phosphatidylinositol 3-kinase; cGMP: Cyclic GMP; eNOS: endothelial NO synthase; ERK ½: extracellular signal-regulated kinase ½; L-Arg: L-Arginine; MAPK: mitogen-activated protein kinases; NO: Nitric oxide; P: Phosphorus; PDK1: Phosphoinositide-dependent kinase 1; PI3-K: Phosphatidylinositol 3-kinase; PIP2: Phosphatidylinositol diphosphate; ncRNAs: non-protein-coding RNA; miRNAs: microRNAs; OPCs: oligomeric procyanidins; RES: resveratrol; GE: grape extract; T2DM: type 2 diabetes mellitus; IL: interleukin; TNF-α: tumour necrosis factor-alpha; NF-κB: nuclear factor NF-kappa-B; ALP: alkaline phosphatase; PARP1: poly [ADP-ribose] polymerase 1; HIF1a: Hypoxia-inducible-factor 1A; NFATc2: nuclear factor of activated T cells 2; PAD: peripheral artery disease; SHR: spontaneously hypertensive rat; RAAS: renin-angiotensin-aldosterone system; AT1R: angiotensin type-1 receptor; Nox: NADPH oxidase; HO-1: haem oxygenase-1; JAK/STAT: Janus kinase/signal transducers/activators of the transcription; PNS: panax notoginseng saponin; snoRNA: small nucleolar RNA; hnRNA: heterogeneous nuclear RNA; VSMCs: vascular smooth muscle cells; irf7: interferon regulatory factor 7; limo2: LIM only domain 2; GWAS: genome-wide association study; GAS5: Growth arrest-specific 5; Asb3, Ankyrin repeat and SPCS box containing 3; Chac2: cation transport regulator homolog 2; Pex11b: peroxisomal membrane 11B; Sp5: Sp5 transcription factor; EGCG: epigallocatechin gallate; ApoE: Apo lipoprotein E; ERK-MAP kinase: extracellular signal-regulated kinases-mitogen-activated protein kinase; PAH: pulmonary artery hypertension; PAP: pulmonary arterial pressure; HIF1a: hypoxia-inducible-factor 1A; NFATc2: nuclear factor of activated T cells 2; HMEC-1: Human microvascular endothelial cells; stat2: signal transducers and activators of transcription 2; JNK: c-Jun N-terminal kinase; iNOS: inducible NO synthase. SNP: single nucleotide polymorphism; CAD: coronary artery disease.
Collapse
Affiliation(s)
- Elham Shirazi-Tehrani
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alireza Chamasemani
- Department of Cardiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Negar Firouzabadi
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Marzieh Mousaei
- Archaea Centre, Department of Biology, University of Copenhagen, Copenhagen N, Denmark
| |
Collapse
|
6
|
Lin Y, Huang H, Yu Y, Zhu F, Xiao W, Yang Z, Shao L, Shen Z. Long non-coding RNA RP11-465L10.10 promotes vascular smooth muscle cells phenotype switching and MMP9 expression via the NF-κB pathway. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1776. [PMID: 35071470 PMCID: PMC8756256 DOI: 10.21037/atm-21-6402] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/17/2021] [Indexed: 01/02/2023]
Abstract
Background Thoracic aortic aneurysm/dissection (TAA/D) are complicated vascular disorders with rapid development and high mortality. Vascular smooth muscle cells (VSMCs) phenotype switching plays an important role in the pathological process of TAA/D. Previous studies have indicated a potential correlation between long non-coding RNA (lncRNA) RP11-465L10.10 and matrix metallopeptidase 9 (MMP9) involved in the development of TAA/D. This study aims to investigate the role of lncRNA RP11-465L10.10 in VSMCs phenotype switching and the molecular mechanism in regulating MMP9 expression. Methods The expression of RP11-465L10.10 in vascular tissues and in VMSCs was detected by RT-qPCR. To investigate the role of RP11-465L10.10 on VSMCs phenotype switching, an RP11-465L10.10-overexpressed lentiviral vector was constructed and transfected into VSMCs. Through EdU staining, migration assay, flow cytometry analysis, the roles of RP11-465L10.10 were estimated. Bioinformatics indicated that RP11-465L10.10 upregulating MMP9 expression via NF-κB signaling, and SN50 (a specific inhibitor of NF-κB pathway) was used to inhibit the NF-κB pathway activation, then the expression of MMP9 was detected in RP11-465L10.10 overexpressed VMSCs. Results In this study, we found RP11-465L10.10 and MMP9 were highly increased in TAD patient tissues, which was consistent in angiotensin II-induced VSMCs phenotype switching. RP11-465L10.10 overexpression facilitated VSMCs phenotype switching and MMP9 expression. Mechanismly, NF-κB signal pathway was involved in RP11-465L10.10 induced VSMCs phenotype switching and MMP9 expression by transcriptome data analysis and experimental confirm. Conclusion This study demonstrated that RP11-465L10.10 induces VSMCs phenotype switching and MMP9 expression via the NF-κB signal pathway, suggesting that RP11-465L10.10 might be a potential therapeutic target for TAA/D treatment.
Collapse
Affiliation(s)
- Yang Lin
- Department of Cardiovascular Surgery, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Haoyue Huang
- Department of Cardiovascular Surgery, the First Affiliated Hospital of Soochow University, Suzhou, China.,Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - You Yu
- Department of Cardiovascular Surgery, the First Affiliated Hospital of Soochow University, Suzhou, China.,Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Feng Zhu
- Department of Cardiovascular Surgery, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Weizhang Xiao
- Department of Cardiovascular Surgery, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ziying Yang
- Department of Cardiovascular Surgery, the First Affiliated Hospital of Soochow University, Suzhou, China.,Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Lianbo Shao
- Department of Cardiovascular Surgery, the First Affiliated Hospital of Soochow University, Suzhou, China.,Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Zhenya Shen
- Department of Cardiovascular Surgery, the First Affiliated Hospital of Soochow University, Suzhou, China.,Institute for Cardiovascular Science, Soochow University, Suzhou, China
| |
Collapse
|
7
|
Long Non-coding RNAs: Potential Players in Cardiotoxicity Induced by Chemotherapy Drugs. Cardiovasc Toxicol 2021; 22:191-206. [PMID: 34417760 DOI: 10.1007/s12012-021-09681-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 07/24/2021] [Indexed: 10/20/2022]
Abstract
One of the most important side effects of chemotherapy is cardiovascular complications, such as cardiotoxicity. Many factors are involved in the pathogenesis of cardiotoxicity; one of the most important of which is long non-coding RNAs (lncRNAs). lncRNA has 200-1000 nucleotides. It is involved in important processes such as cell proliferation, regeneration and apoptosis; today it is used as a prognostic and diagnostic factor. A, various drugs by acting on lncRNAs can affect cells. Therefore, by accurately identifying IncRNAs function, we can play an effective role in preventing the development of cardiotoxicity-induced chemotherapy drugs, and use them as a therapeutic strategy to improve clinical symptoms and increase patient survival.
Collapse
|
8
|
Circ_UBR4 knockdown alleviates oxidized low-density lipoprotein-provoked growth and migration of human vascular smooth muscle cells by acting on the miR-637/FOXO4 pathway. J Cardiovasc Pharmacol 2021; 78:534-543. [PMID: 34225339 DOI: 10.1097/fjc.0000000000001098] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 06/12/2021] [Indexed: 11/27/2022]
Abstract
ABSTRACT Excessive proliferation and migration of human vascular smooth muscle cells (HVSMCs) induced by oxidized low-density lipoprotein (ox-LDL) are important pathological features of atherosclerosis (AS). Emerging evidence indicates that circular RNAs (circRNAs) deregulation is involved in this pathological process. The objective of this study was to explore the role of circRNA ubiquitin protein ligase E3 component n-recognin 4 (circ_UBR4) in ox-LDL-treated HVSMCs. The expression of circ_UBR4, microRNA-637 (miR-637) and forkhead box O4 (FOXO4) mRNA was detected by quantitative real-time PCR (qPCR). Cell cycle progression was examined by flow cytometry assay. Cell viability was examined by MTT assay. Cell migration was examined by transwell assay. The protein levels of proliferating cell nuclear antigen (PCNA), matrix metalloproteinase 2 (MMP2) and FOXO4 were measured by western blot. The relationship between miR-637 and circ_UBR4 or FOXO4 was confirmed by dual-luciferase reporter assay. The results presented that the expression of circ_UBR4 was increased in AS serum samples and ox-LDL-treated HVSMCs. Cell cycle progression, cell proliferation and cell migration were promoted by ox-LDL, while circ_UBR4 knockdown inhibited HVSMCs proliferation and migration. MiR-637 was a target of circ_UBR4, and FOXO4 was a target of miR-637. Circ_UBR4 positively regulated FOXO4 expression by targeting miR-637. Circ_UBR4 knockdown-inhibited HVSMCs proliferation and migration were recovered by miR-637 inhibition, and miR-637 restoration-inhibited HVSMCs proliferation and migration were recovered by FOXO4 overexpression. In conclusion, circ_UBR4 knockdown inhibited ox-LDL-induced excessive proliferation and migration of HVSMCs by regulating FOXO4 via targeting miR-637.
Collapse
|
9
|
Si X, Chen Q, Zhang J, Zhou W, Chen L, Chen J, Deng N, Li W, Liu D, Wang L, Shi L, Sun W, Song H, Zhong L. MicroRNA-23b prevents aortic aneurysm formation by inhibiting smooth muscle cell phenotypic switching via FoxO4 suppression. Life Sci 2021; 288:119092. [PMID: 33737086 DOI: 10.1016/j.lfs.2021.119092] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 01/10/2021] [Accepted: 01/14/2021] [Indexed: 12/21/2022]
Abstract
AIMS Phenotypic switching of vascular smooth muscle cells (VSMCs) is essential for the formation of abdominal aortic aneurysms (AAAs). MicroRNA-23b (miR-23b) has recently been shown to play a vital role in maintaining the VSMC contractile phenotype; however, little is known about the role of miR-23b in the formation of AAAs. Here, we investigated whether miR-23b prevents AAA formation by inhibiting VSMC phenotypic switching. MATERIALS AND METHODS We administered angiotensin II (Ang II, 1000 ng/kg/min) or vehicle to 10-12-week-old male apolipoprotein E knockout (ApoE-/-) or C57BL/6J mice via subcutaneous osmotic minipumps for 4 weeks. KEY FINDINGS The expression of miR-23b was significantly reduced in the aorta during the early onset of AAA in angiotensin II-treated ApoE-/- mice and in human AAA samples. In vitro experiments showed that the suppression of SMC contractile marker gene expression induced by Ang II was accelerated by miR-23b inhibitors but inhibited by mimics. In vivo studies revealed that miR-23b deficiency in Ang II-treated C57BL/6J mice aggravated the formation of AAAs in these mice compared with control mice; the opposite results were observed in miR-23b-overexpressing mice. Mechanistically, miR-23b knockdown significantly increased the expression of the transcription factor forkhead box O4 (FoxO4) during VSMC phenotypic switching induced by Ang II. In addition, a luciferase reporter assay showed that FoxO4 is a target of miR-23b in VSMCs. SIGNIFICANCE Our study revealed a pivotal role for miR-23b in protecting against aortic aneurysm formation by maintaining the VSMC contractile phenotype.
Collapse
Affiliation(s)
- Xiaoyun Si
- Department of Cardiology, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Road, Guiyang 550004, Guizhou Province, China; Geriatrics Department, Zhuhai People's Hospital (Zhuhai Hospital affiliated with Jinan University), Zhuhai 519000, China
| | - Qixian Chen
- Department of Pulmonary and Critical Care Medicines, Zhuhai People's Hospital (Zhuhai Hospital affiliated with Jinan University), Zhuhai 519000, China
| | - Jiechang Zhang
- Department of Cardiology, Zhuhai People's Hospital (Zhuhai Hospital affiliated with Jinan University), Zhuhai 519000, China
| | - Wei Zhou
- Department of Cardiology, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Road, Guiyang 550004, Guizhou Province, China
| | - Lijun Chen
- Department of Hematology and Rheumatology, Zhuhai People's Hospital (Zhuhai Hospital affiliated with Jinan University), Zhuhai 519000, China
| | - Jingjing Chen
- Department of Cardiology, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Road, Guiyang 550004, Guizhou Province, China
| | - Na Deng
- Department of Cardiology, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Road, Guiyang 550004, Guizhou Province, China
| | - Wei Li
- Department of Cardiology, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Road, Guiyang 550004, Guizhou Province, China
| | - Danan Liu
- Department of Cardiology, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Road, Guiyang 550004, Guizhou Province, China
| | - Long Wang
- Department of Cardiology, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Road, Guiyang 550004, Guizhou Province, China
| | - Linyan Shi
- Department of Cardiology, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Road, Guiyang 550004, Guizhou Province, China
| | - Weihong Sun
- Department of Cardiology, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Road, Guiyang 550004, Guizhou Province, China
| | - Haoyu Song
- Department of Cardiology, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, University Town, Ganzhou Development District, Ganzhou 341000, China.
| | - Lintao Zhong
- Department of Cardiology, Zhuhai People's Hospital (Zhuhai Hospital affiliated with Jinan University), Zhuhai 519000, China.
| |
Collapse
|
10
|
Yang X, Yang Y, Guo J, Meng Y, Li M, Yang P, Liu X, Aung LHH, Yu T, Li Y. Targeting the epigenome in in-stent restenosis: from mechanisms to therapy. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 23:1136-1160. [PMID: 33664994 PMCID: PMC7896131 DOI: 10.1016/j.omtn.2021.01.024] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Coronary artery disease (CAD) is one of the most common causes of death worldwide. The introduction of percutaneous revascularization has revolutionized the therapy of patients with CAD. Despite the advent of drug-eluting stents, restenosis remains the main challenge in treating patients with CAD. In-stent restenosis (ISR) indicates the reduction in lumen diameter after percutaneous coronary intervention, in which the vessel's lumen re-narrowing is attributed to the aberrant proliferation and migration of vascular smooth muscle cells (VSMCs) and dysregulation of endothelial cells (ECs). Increasing evidence has demonstrated that epigenetics is involved in the occurrence and progression of ISR. In this review, we provide the latest and comprehensive analysis of three separate but related epigenetic mechanisms regulating ISR, namely, DNA methylation, histone modification, and non-coding RNAs. Initially, we discuss the mechanism of restenosis. Furthermore, we discuss the biological mechanism underlying the diverse epigenetic modifications modulating gene expression and functions of VSMCs, as well as ECs in ISR. Finally, we discuss potential therapeutic targets of the small molecule inhibitors of cardiovascular epigenetic factors. A more detailed understanding of epigenetic regulation is essential for elucidating this complex biological process, which will assist in developing and improving ISR therapy.
Collapse
Affiliation(s)
- Xi Yang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Road No. 59 Haier, Qingdao 266100, Shandong, People’s Republic of China
| | - Yanyan Yang
- Department of Immunology, School of Basic Medicine, Qingdao University, No. 308 Ningxia Road, Qingdao 266071, People’s Republic of China
| | - Junjie Guo
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Road No. 59 Haier, Qingdao 266100, Shandong, People’s Republic of China
| | - Yuanyuan Meng
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao 266000, People’s Republic of China
| | - Min Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao 266021, People’s Republic of China
| | - Panyu Yang
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao 266000, People’s Republic of China
| | - Xin Liu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Road No. 59 Haier, Qingdao 266100, Shandong, People’s Republic of China
| | - Lynn Htet Htet Aung
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao 266021, People’s Republic of China
| | - Tao Yu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao 266000, People’s Republic of China
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao 266021, People’s Republic of China
| | - Yonghong Li
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Road No. 59 Haier, Qingdao 266100, Shandong, People’s Republic of China
| |
Collapse
|
11
|
Qu C, Liu X, Guo Y, Fo Y, Chen X, Zhou J, Yang B. MiR-128-3p inhibits vascular smooth muscle cell proliferation and migration by repressing FOXO4/MMP9 signaling pathway. Mol Med 2020; 26:116. [PMID: 33238881 PMCID: PMC7687681 DOI: 10.1186/s10020-020-00242-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 11/12/2020] [Indexed: 12/12/2022] Open
Abstract
Background MicroRNAs (miRNAs) have been identified as important participants in the development of atherosclerosis (AS). The present study explored the role of miR-128-3p in the dysfunction of vascular smooth muscle cells (VSMCs) and the underlying mechanism. Methods Human VSMCs and ApoE knockout (ApoE−/−) C57BL/6J mice were used to establish AS cell and animal models, respectively. Expression levels of miR-128-3p, forkhead box O4 (FOXO4) and matrix metallopeptidase 9 (MMP9) were detected using qRT-PCR and Western blot, respectively. CCK-8, BrdU, and Transwell assays as well as flow cytometry analysis were performed to detect the proliferation, migration and apoptosis of VSMCs. Levels of inflammatory cytokines and lipids in human VSMCs, mice serum and mice VSMCs were also determined. The binding site between miR-128-3p and 3′UTR of FOXO4 was confirmed using luciferase reporter gene assay. Results MiR-128-3p was found to be decreased in AS patient serum, ox-LDL-treated VSMCs, AS mice serum and VSMCs of AS mice. Transfection of miR-128-3p mimics suppressed the proliferation and migration of VSMCs, accompanied by the promoted apoptosis and the decreased levels of inflammatory cytokines. Further experiments confirmed the interaction between miR-128-3p and FOXO4. Augmentation of FOXO4 or MMP9 reversed the effects of miR-128-3p. Besides, miR-128-3p inhibited triglyceride (TG), total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C) but increased high-density lipoprotein cholesterol (HDL-C) in the serum of AS mice. Conclusion MiR-128-3p repressed the proliferation and migration of VSMCs through inhibiting the expressions of FOXO4 and MMP9.
Collapse
Affiliation(s)
- Chuan Qu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Jiefang Road No.238, Wuhan, 430060, Hubei, China
| | - Xin Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Jiefang Road No.238, Wuhan, 430060, Hubei, China
| | - Yan Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Jiefang Road No.238, Wuhan, 430060, Hubei, China
| | - Yuhong Fo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Jiefang Road No.238, Wuhan, 430060, Hubei, China
| | - Xiuhuan Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Jiefang Road No.238, Wuhan, 430060, Hubei, China
| | - Jining Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Jiefang Road No.238, Wuhan, 430060, Hubei, China
| | - Bo Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Jiefang Road No.238, Wuhan, 430060, Hubei, China.
| |
Collapse
|
12
|
Yuan Y, Xu L, Geng Z, Liu J, Zhang L, Wu Y, He D, Qu P. The role of non-coding RNA network in atherosclerosis. Life Sci 2020; 265:118756. [PMID: 33189816 DOI: 10.1016/j.lfs.2020.118756] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/29/2020] [Accepted: 11/10/2020] [Indexed: 12/26/2022]
Abstract
Atherosclerosis is the primary culprit of cardiovascular and cerebrovascular diseases. Also, atherogenesis and the development of atherosclerosis involve endothelial cells, monocytes/macrophages, smooth myocytes, and others. Increasingly, studies have found that non-coding RNA (ncRNA) which can regulate apoptosis, pyroptosis, autophagy, proliferation, and monocyte migration participates in atherogenesis and progress of atherosclerosis by the above. The ncRNA networks may be essential in regulating the complicated process of atherosclerosis. Accordingly, this review delves into the regulatory roles of ncRNA, which were introduced previously. The answer above is particularly crucial to explain further the regulatory mechanism of ncRNA in cardiovascular disorders. Furthermore, we discuss the possibility and related research of ncRNAs as a biomarker and therapeutic target for the prevention, diagnosis, and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Yuchan Yuan
- Institute of Heart and Vessel Diseases, The Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian 116023, People's Republic of China
| | - Ling Xu
- Department of clinical laboratory, Xinhua Hospital Affiliated to Dalian University, Dalian 116021, People's Republic of China
| | - Zhaohong Geng
- Department of Cardiology, The Second Affiliated Hospital of Dalian Medical University, Dalian 116023, People's Republic of China
| | - Jingjing Liu
- Institute of Heart and Vessel Diseases, The Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian 116023, People's Republic of China
| | - Lijiao Zhang
- Department of Cardiology, The Second Affiliated Hospital of Dalian Medical University, Dalian 116023, People's Republic of China
| | - Yuhang Wu
- Institute of Heart and Vessel Diseases, The Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian 116023, People's Republic of China
| | - Dan He
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing 100191, People's Republic of China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, People's Republic of China.
| | - Peng Qu
- Institute of Heart and Vessel Diseases, The Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian 116023, People's Republic of China; Department of Cardiology, The Second Affiliated Hospital of Dalian Medical University, Dalian 116023, People's Republic of China.
| |
Collapse
|
13
|
Long noncoding RNAs as novel players in the pathogenesis of hypertension. Hypertens Res 2020; 43:597-608. [PMID: 32020084 DOI: 10.1038/s41440-020-0408-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 01/12/2020] [Accepted: 01/12/2020] [Indexed: 02/08/2023]
Abstract
Long noncoding RNAs (lncRNAs) are non-(protein)-coding RNAs longer than ~200 nucleotides and have been reported to be involved in multiple human diseases by regulating gene expression. A growing body of evidence has demonstrated that lncRNAs are also widely implicated in mechanisms of hypertension, including regulation of the proliferation, migration, and apoptosis of VSMCs; the production of iNOS and NO; and the angiogenic function of endothelial cells. Several lncRNAs were also differentially expressed in the renal and cardiac tissues of hypertensive rats and even in placental samples from preeclampsia patients. In particular, several circulating lncRNAs have been identified as novel biomarkers of hypertension. In this review, we summarize the current studies of lncRNAs in the pathogenesis of hypertension in order to aid in better understanding the molecular mechanism of hypertension and provide a basis to explore new therapeutic targets.
Collapse
|
14
|
Liu W, Li Y, Luo B. Current perspective on the regulation of FOXO4 and its role in disease progression. Cell Mol Life Sci 2020; 77:651-663. [PMID: 31529218 PMCID: PMC11104957 DOI: 10.1007/s00018-019-03297-w] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 08/21/2019] [Accepted: 09/09/2019] [Indexed: 12/12/2022]
Abstract
Forkhead box O4 (FOXO4) is a member of the FOXO family that regulates a number of genes involved in metabolism, cell cycle, apoptosis, and cellular homeostasis via transcriptional activity. It also mediates cell responses to oxidative stress and treatment with antitumor agents. The expression of FOXO4 is repressed by microRNAs in multiple cancer cells, while FOXO4 function is regulated by post-translational modifications and interaction with other proteins. The deregulation of FOXO4 is closely linked to the progression of several types of cancer, senescence, and other diseases. In this review, we present recent findings on the regulation of FOXO4 in physiological and pathological conditions and provide an overview of the complex role of FOXO4 in disease development and response to therapy.
Collapse
Affiliation(s)
- Wen Liu
- Department of Pathogenic Biology, Faculty of Medicine, Qingdao University, Qingdao, China
| | - Yong Li
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Faculty of Medicine, Qingdao University, Qingdao, China
| | - Bing Luo
- Department of Pathogenic Biology, Faculty of Medicine, Qingdao University, Qingdao, China.
| |
Collapse
|
15
|
Khachigian LM. Transcription Factors Targeted by miRNAs Regulating Smooth Muscle Cell Growth and Intimal Thickening after Vascular Injury. Int J Mol Sci 2019; 20:ijms20215445. [PMID: 31683712 PMCID: PMC6861964 DOI: 10.3390/ijms20215445] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/20/2019] [Accepted: 10/15/2019] [Indexed: 12/21/2022] Open
Abstract
Neointima formation after percutaneous coronary intervention (PCI) is a manifestation of “phenotype switching” by vascular smooth muscle cells (SMC), a process that involves de-differentiation from a contractile quiescent phenotype to one that is richly synthetic. In response to injury, SMCs migrate, proliferate, down-regulate SMC-specific differentiation genes, and later, can revert to the contractile phenotype. The vascular response to injury is regulated by microRNAs (or miRNAs), small non-coding RNAs that control gene expression. Interactions between miRNAs and transcription factors impact gene regulatory networks. This article briefly reviews the roles of a range of miRNAs in molecular and cellular processes that control intimal thickening, focusing mainly on transcription factors, some of which are encoded by immediate-early genes. Examples include Egr-1, junB, KLF4, KLF5, Elk-1, Ets-1, HMGB1, Smad1, Smad3, FoxO4, SRF, Rb, Sp1 and c-Myb. Such mechanistic information could inform the development of strategies that block SMC growth, neointima formation, and potentially overcome limitations of lasting efficacy following PCI.
Collapse
Affiliation(s)
- Levon M Khachigian
- Vascular Biology and Translational Research, School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney NSW 2052, Australia.
| |
Collapse
|
16
|
Li M, Duan L, Li Y, Liu B. Long noncoding RNA/circular noncoding RNA-miRNA-mRNA axes in cardiovascular diseases. Life Sci 2019; 233:116440. [PMID: 31047893 DOI: 10.1016/j.lfs.2019.04.066] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 04/23/2019] [Accepted: 04/29/2019] [Indexed: 02/01/2023]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death worldwide. Non-coding RNAs including long non-coding RNAs (lncRNAs), circular RNAs (circRNAs) and microRNAs (miRNAs) have been reported to participate in pathological developments of CVDs through various mechanisms. Among them, the networks among lncRNAs/circRNAs, miRNAs, and mRNAs have recently attracted attention. Understanding the molecular mechanism could aid the discovery of therapeutic targets or strategies in CVDs including atherosclerosis, myocardial infarction (MI), hypertrophy, heart failure (HF) and cardiomyopathy. In this review, we summarize the latest research involving the lncRNA/circRNA-miRNA-mRNA axis in CVDs, with emphasis on the molecular mechanism.
Collapse
Affiliation(s)
- Ming Li
- Department of Gastroenterology, The Second Hospital of Jilin University, 218 Ziqiang Road, Changchun 130041, China
| | - Liwei Duan
- Department of Gastroenterology, The Second Hospital of Jilin University, 218 Ziqiang Road, Changchun 130041, China
| | - Yangxue Li
- Department of Cardiology, The Second Hospital of Jilin University, 218 Ziqiang Road, Changchun 130041, China
| | - Bin Liu
- Department of Cardiology, The Second Hospital of Jilin University, 218 Ziqiang Road, Changchun 130041, China.
| |
Collapse
|
17
|
Liu X, Ma BD, Liu S, Liu J, Ma BX. Long noncoding RNA LINC00341 promotes the vascular smooth muscle cells proliferation and migration via miR-214/FOXO4 feedback loop. Am J Transl Res 2019; 11:1835-1842. [PMID: 30972207 PMCID: PMC6456561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Accepted: 02/13/2019] [Indexed: 06/09/2023]
Abstract
Increasing evidences have indicated the vital roles of long noncoding RNA (lncRNA) in the atherosclerosis. However, whether lncRNA LINC00341 play pivotal roles in the vascular smooth muscle cells (VSMCs) is still unclear. This work presents the authentic functions of LINC00341 on the proliferation and migration of VSMCs and unveils the underlying mechanism. Functional experiment data demonstrated that LINC00341 expression was increased in the ox-LDL induced VSMCs with dose-dependent and time-dependent mode. Moreover, the knockdown of LINC00341 suppressed the proliferation and migration ability of VSMCs. Mechanically, we found that LINC00341 promoted the FOXO4 protein expression via sponging miR-214, which, in return, resulting in the transcription activation of LINC00341. In conclusion, the results conclude that LINC00341 promotes the proliferation and migration of VSMCs and confirm the positive feedback loop of LINC00341/miR-214/FOXO4 axis.
Collapse
Affiliation(s)
- Xiu Liu
- Department of Cardiology, Yantai Affiliated Hospital of Binzhou Medical UniversityYantai, Shandong, China
| | - Bing-Dong Ma
- Department of Hand Microsurgery, Affiliated Hospital of Binzhou Medical UniversityBinzhou, Shandong, China
| | - Shan Liu
- Department of Intervention, Yantai Affiliated Hospital of Binzhou Medical UniversityYantai, Shandong, China
| | - Ju Liu
- Medical Research Center, Shandong Provincial Qianfoshan HospitalJinan, Shandong, China
| | - Bao-Xin Ma
- Department of Cardiology, Affiliated Hospital of Binzhou Medical UniversityBinzhou, Shandong, China
| |
Collapse
|
18
|
Li Q, Gao C, Deng H, Song Q, Yuan L. Benzo[a]pyrene induces pyroptotic and autophagic death through inhibiting PI3K/Akt signaling pathway in HL-7702 human normal liver cells. J Toxicol Sci 2019; 44:121-131. [PMID: 30726812 DOI: 10.2131/jts.44.121] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Benzo(α)pyrene (BaP) possesses a forceful hepatotoxicity, and is ubiquitous in foods and ambient air. Our previous study found that BaP induced pyroptotic and autophagic death in HL-7702 human liver cells; the relevant mechanisms, however, remain unknown. This work was therefore to unravel the effects of the PI3K/Akt signaling pathway on pyroptotic and autophagic death triggered by BaP. Cells were treated with or without LY294002 (PI3K/Akt inhibitor) and IGF-1 (PI3K/Akt activator) before BaP exposure, and the results showed that compared with the control, the protein expression of p-Akt was markedly decreased by BaP (p < 0.05). IGF-1 did not subvert this inhibitive effect of BaP, while LY294002 enhanced it. Furthermore, the protein expression of pyroptosis (Cleaved Caspase-1, NO, IL-1β, IL-18), as well as LDH and the relative electrical conductivity were significantly augmented by BaP. The levels of these indices were increased by LY294002 pretreatment, and decreased by IGF-1. Similarly, LY294002 enhanced BaP-induced increase in the key protein expression of autophagy (Beclin-1 and LC3II), while IGF-1 weakened it. Finally, the phosphorylation of FOXO4 was clearly (p < 0.01) inhibited by BaP, and LY294002 suppressed this inhibitive effect of BaP, while IGF-1 strengthened it. In conclusion, BaP was able to induce pyroptotic and autophagic death via blocking the PI3K/Akt signaling pathway in HL-7702 liver cells.
Collapse
Affiliation(s)
- Qingshu Li
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, China
| | - Chunxia Gao
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, China
| | - Hong Deng
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, China
| | - Quancai Song
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, China
| | - Li Yuan
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, China
| |
Collapse
|