1
|
Kumar A, Yap KCH, BharathwajChetty B, Lyu J, Hegde M, Abbas M, Alqahtani MS, Khadlikar S, Zarrabi A, Khosravi A, Kumar AP, Kunnumakkara AB. Regulating the regulators: long non-coding RNAs as autophagic controllers in chronic disease management. J Biomed Sci 2024; 31:105. [PMID: 39716252 DOI: 10.1186/s12929-024-01092-9] [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: 05/31/2024] [Accepted: 10/23/2024] [Indexed: 12/25/2024] Open
Abstract
The increasing prevalence of chronic diseases and their associated morbidities demands a deeper understanding of underlying mechanism and causative factors, with the hope of developing novel therapeutic strategies. Autophagy, a conserved biological process, involves the degradation of damaged organelles or protein aggregates to maintain cellular homeostasis. Disruption of this crucial process leads to increased genomic instability, accumulation of reactive oxygen species (ROS), decreased mitochondrial functions, and suppression of ubiquitination, leading to overall decline in quality of intracellular components. Such deregulation has been implicated in a wide range of pathological conditions such as cancer, cardiovascular, inflammatory, and neurological disorders. This review explores the role of long non-coding RNAs (lncRNAs) as modulators of transcriptional and post-transcriptional gene expression, regulating diverse physiological process like proliferation, development, immunity, and metabolism. Moreover, lncRNAs are known to sequester autophagy related microRNAs by functioning as competing endogenous RNAs (ceRNAs), thereby regulating this vital process. In the present review, we delineate the multitiered regulation of lncRNAs in the autophagic dysfunction of various pathological diseases. Moreover, by highlighting recent findings on the modulation of lncRNAs in different stages of autophagy, and the emerging clinical landscape that recognizes lncRNAs in disease diagnosis and therapy, this review highlights the potential of lncRNAs as biomarkers and therapeutic targets in clinical settings of different stages of autophagic process by regulating ATG and its target genes. This focus on lncRNAs could lead to breakthroughs in personalized medicine, offering new avenues for diagnosis and treatment of complex diseases.
Collapse
Affiliation(s)
- Aviral Kumar
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam, 781039, India
| | - Kenneth Chun-Hong Yap
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Bandari BharathwajChetty
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam, 781039, India
| | - Juncheng Lyu
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Mangala Hegde
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam, 781039, India
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, 61421, Abha, Saudi Arabia
| | - Mohammed S Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, 61421, Abha, Saudi Arabia
- BioImaging Unit, Space Research Centre, Michael Atiyah Building, University of Leicester, Leicester, LE1 7RH, UK
| | - Soham Khadlikar
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam, 781039, India
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering & Natural Sciences, Istinye University, 34396, Istanbul, Türkiye
- Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600 077, India
- Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan, 320315, Taiwan
| | - Arezoo Khosravi
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Istanbul Okan University, 34959, Istanbul, Türkiye
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore.
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam, 781039, India.
| |
Collapse
|
2
|
Zhu L, Liao Y, Jiang B. Role of ROS and autophagy in the pathological process of atherosclerosis. J Physiol Biochem 2024; 80:743-756. [PMID: 39110405 DOI: 10.1007/s13105-024-01039-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 07/25/2024] [Indexed: 12/29/2024]
Abstract
Activation of autophagy and production of reactive oxygen species occur at various stages of atherosclerosis. To clarify the role and mechanism of autophagy and reactive oxygen species in atherosclerosis is of great significance to the prevention and treatment of atherosclerosis. Recent studies have shown that basal autophagy plays an important role in protecting cells from oxidative stress, reducing apoptosis and enhancing atherosclerotic plaque stability. Autophagy deficiency and excessive accumulation of reactive oxygen species can impair the function of endothelial cells, macrophages and smooth muscle cells, trigger autophagic cell death, and lead to instability and even rupture of plaques. However, the main signaling pathways regulating autophagy, the molecular mechanisms of autophagy and reactive oxygen species interaction, how they are initiated and distributed in plaques, and how they affect atherosclerosis progression, remain to be clarified. At present, there is no autophagy inducer used to treat atherosclerosis clinically. Therefore, it is urgent to clarify the mechanism of autophagy and find new targets for autophagy. Antioxidant agents generally have defects such as low reactive oxygen species scavenging efficiency and high cytotoxicity. Highly potent autophagy inducers and reactive oxygen species scavengers still need to be further developed and validated to provide more possibilities for innovative treatments for atherosclerosis.
Collapse
Affiliation(s)
- Liyuan Zhu
- Center of Clinical Pharmacology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yingnan Liao
- Department of Laboratory Medicine, Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical Genetics, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Research Unit for Blindness Prevention, Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Bo Jiang
- Center of Clinical Pharmacology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
3
|
Gu X, Hou J, Rao J, Weng R, Liu S. LncRNA MALAT1 suppresses monocyte-endothelial cell interactions by targeting miR-30b-5p and enhancing ATG5-mediated autophagy. Heliyon 2024; 10:e28882. [PMID: 38601614 PMCID: PMC11004576 DOI: 10.1016/j.heliyon.2024.e28882] [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: 09/27/2023] [Revised: 03/26/2024] [Accepted: 03/26/2024] [Indexed: 04/12/2024] Open
Abstract
Background Monocyte-endothelial cell (EC) interactions are one of the earliest events in the development of atherosclerosis and play a crucial role in atherosclerotic plaque formation. Although attempts have been made to modulate this interaction, the underlying molecular signalling mechanisms remain unclear. This study aimed to investigate the role of long non-coding RNA MALAT1 in monocyte-EC interactions. Methods The expression of MALAT1, ICAM-1, VCAM-1, P-selectin, CCL2 and CXCL1 was evaluated in ApoE-/- mouse aortic tissues and inflamed human umbilical vein endothelial cells (HUVECs). The regulatory impact of MALAT1 on cell adhesion molecules, monocyte-EC adhesion, and autophagy was assessed. The interactions between MALAT1 and microRNAs (miRNAs) were evaluated using dual-luciferase reporter and RNA pull-down assays. Results MALAT1 expression decreased in ApoE-/- mouse aortic tissues and inflammatory HUVECs. MALAT1 overexpression suppressed the expression of ICAM-1, VCAM-1 and CXCL1, and reduced the migration and adhesion of monocytes to ECs. Inhibition of MALAT1 promoted cell adhesion molecule expression and monocyte-EC interactions. Mechanistically, MALAT1 binds directly to miR-30b-5p and decreases its effective expression by functioning as an endogenous sponge, thereby increasing the expression of autophagy-related gene 5 (ATG5) and stimulates endothelial autophagy. Conclusions Our findings suggest that MALAT1 suppresses monocyte-EC interactions by targeting miR-30b-5p and enhancing ATG5-mediated endothelial autophagy. These data imply that MALAT1 may play a protective role at the early stages of the atherosclerotic process.
Collapse
Affiliation(s)
- Xiaodong Gu
- Meizhou Clinical Institute, Shantou University Medical College, Meizhou, 514000, China
- Research Experimental Center, Meizhou People's Hospital (Huangtang Hospital), Meizhou, 514031, China
- Guangdong Engineering Technology Research Center of Molecular Diagnostics for Cardiovascular Diseases, Meizhou, 514000, China
| | - Jingyuan Hou
- Meizhou Clinical Institute, Shantou University Medical College, Meizhou, 514000, China
- Research Experimental Center, Meizhou People's Hospital (Huangtang Hospital), Meizhou, 514031, China
- Guangdong Engineering Technology Research Center of Molecular Diagnostics for Cardiovascular Diseases, Meizhou, 514000, China
| | - Jiawei Rao
- Meizhou Clinical Medical School, Guangdong Medical University, Meizhou, 514000, China
| | - Ruiqiang Weng
- Meizhou Clinical Institute, Shantou University Medical College, Meizhou, 514000, China
- Research Experimental Center, Meizhou People's Hospital (Huangtang Hospital), Meizhou, 514031, China
- Guangdong Engineering Technology Research Center of Molecular Diagnostics for Cardiovascular Diseases, Meizhou, 514000, China
| | - Sudong Liu
- Meizhou Clinical Institute, Shantou University Medical College, Meizhou, 514000, China
- Research Experimental Center, Meizhou People's Hospital (Huangtang Hospital), Meizhou, 514031, China
- Guangdong Engineering Technology Research Center of Molecular Diagnostics for Cardiovascular Diseases, Meizhou, 514000, China
| |
Collapse
|
4
|
Ouyang S, Zhou ZX, Liu HT, Ren Z, Liu H, Deng NH, Tian KJ, Zhou K, Xie HL, Jiang ZS. LncRNA-mediated Modulation of Endothelial Cells: Novel Progress in the Pathogenesis of Coronary Atherosclerotic Disease. Curr Med Chem 2024; 31:1251-1264. [PMID: 36788688 DOI: 10.2174/0929867330666230213100732] [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: 06/13/2022] [Revised: 11/06/2022] [Accepted: 11/17/2022] [Indexed: 02/16/2023]
Abstract
Coronary atherosclerotic disease (CAD) is a common cardiovascular disease and an important cause of death. Moreover, endothelial cells (ECs) injury is an early pathophysiological feature of CAD, and long noncoding RNAs (lncRNAs) can modulate gene expression. Recent studies have shown that lncRNAs are involved in the pathogenesis of CAD, especially by regulating ECs. In this review, we summarize the novel progress of lncRNA-modulated ECs in the pathogenesis of CAD, including ECs proliferation, migration, adhesion, angiogenesis, inflammation, apoptosis, autophagy, and pyroptosis. Thus, as lncRNAs regulate ECs in CAD, lncRNAs will provide ideal and novel targets for the diagnosis and drug therapy of CAD.
Collapse
Affiliation(s)
- Shao Ouyang
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang 421001, China
- Key Laboratory of Heart Failure Prevention & Treatment of Hengyang, Department of Cardiovascular Medicine, Hengyang Medical School, The Second Affiliated Hospital, Clinical Medicine Research Center of Arteriosclerotic Disease of Hunan Province, University of South China, Hunan 421001, China
| | - Zhi-Xiang Zhou
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang 421001, China
| | - Hui-Ting Liu
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang 421001, China
| | - Zhong Ren
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang 421001, China
| | - Huan Liu
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang 421001, China
| | - Nian-Hua Deng
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang 421001, China
| | - Kai-Jiang Tian
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang 421001, China
| | - Kun Zhou
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang 421001, China
| | - Hai-Lin Xie
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang 421001, China
| | - Zhi-Sheng Jiang
- Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang 421001, China
| |
Collapse
|
5
|
Xiong Y, Huang H, Chen F, Tang Y. CircDLGAP4 induces autophagy and improves endothelial cell dysfunction in atherosclerosis by targeting PTPN4 with miR-134-5p. ENVIRONMENTAL TOXICOLOGY 2023; 38:2952-2966. [PMID: 37615249 DOI: 10.1002/tox.23930] [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: 06/09/2023] [Revised: 07/23/2023] [Accepted: 08/01/2023] [Indexed: 08/25/2023]
Abstract
OBJECTIVE Circular RNAs (circRNAs), a new subgroup of non-coding RNAs in the human transcriptome, are crucial in atherosclerosis (AS). Here, a newly identified circRNA circDLGAP4 was demonstrated to be downregulated in oxidized forms of low-density lipoprotein (ox-LDL)-induced HUVECs. METHODS This research adopted ox-LDL to stimulate human umbilical vein endothelial cells (HUVECs) to mimic AS in vitro. To further validate the protective action of circDLGAP4 in AS, a mouse model of AS was constructed with a high-fat diet. Functional assays evaluated circDLGAP4 role in AS in vitro and in vivo. Moreover, mechanism assays evaluated association of circDLGAP4/miR-134-5p/PTPN4. RESULTS CircDLGAP4 was induced to promote cell proliferative behavior and autophagy, inhibit apoptotic and inflammatory activities in ox-LDL-treated HUVECs, and attenuated endothelial barrier function. CircDLGAP4 regulated PTPN4 by directly targeting miR-134-5p. Meanwhile, inhibiting miR-134-5p reduced ox-LDL-induced cell dysfunction. Knockout of PTPN4 reversed circDLGAP4 overexpression or miR-134-5p downregulation in vitro. In addition, reducing circDLGAP4 or overexpressing miR-134-5p increased the red atherosclerotic plaque and lesion area of AS mice, reduced autophagy level, and promoted the release of inflammatory cytokines. CONCLUSION This study extends the role of circRNA in AS by inducing autophagy and improving endothelial dysfunction in AS via the circDLGAP4/miR-134-5p/PTPN4 axis.
Collapse
Affiliation(s)
- Yan Xiong
- Department of Cardiology and Cardiovascular Disease Research Institute, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Hui Huang
- Department of Cardiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Fuli Chen
- Department of Cardiology and Cardiovascular Disease Research Institute, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Yijia Tang
- Department of Cardiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| |
Collapse
|
6
|
Petkovic A, Erceg S, Munjas J, Ninic A, Vladimirov S, Davidovic A, Vukmirovic L, Milanov M, Cvijanovic D, Mitic T, Sopic M. LncRNAs as Regulators of Atherosclerotic Plaque Stability. Cells 2023; 12:1832. [PMID: 37508497 PMCID: PMC10378138 DOI: 10.3390/cells12141832] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/11/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Current clinical data show that, despite constant efforts to develop novel therapies and clinical approaches, atherosclerotic cardiovascular diseases (ASCVD) are still one of the leading causes of death worldwide. Advanced and unstable atherosclerotic plaques most often trigger acute coronary events that can lead to fatal outcomes. However, despite the fact that different plaque phenotypes may require different treatments, current approaches to prognosis, diagnosis, and classification of acute coronary syndrome do not consider the diversity of plaque phenotypes. Long non-coding RNAs (lncRNAs) represent an important class of molecules that are implicated in epigenetic control of numerous cellular processes. Here we review the latest knowledge about lncRNAs' influence on plaque development and stability through regulation of immune response, lipid metabolism, extracellular matrix remodelling, endothelial cell function, and vascular smooth muscle function, with special emphasis on pro-atherogenic and anti-atherogenic lncRNA functions. In addition, we present current challenges in the research of lncRNAs' role in atherosclerosis and translation of the findings from animal models to humans. Finally, we present the directions for future lncRNA-oriented research, which may ultimately result in patient-oriented therapeutic strategies for ASCVD.
Collapse
Affiliation(s)
- Aleksa Petkovic
- Clinical-Hospital Centre "Dr Dragiša Mišović-Dedinje", 11000 Belgrade, Serbia
| | - Sanja Erceg
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia
| | - Jelena Munjas
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia
| | - Ana Ninic
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia
| | - Sandra Vladimirov
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia
| | - Aleksandar Davidovic
- Intern Clinic, Clinical Ward for Cardiovascular Diseases, Clinical-Hospital Centre Zvezdara, 11000 Belgrade, Serbia
- Department for Internal Medicine, Faculty of Dentistry, University of Belgrade, 11000 Belgrade, Serbia
| | - Luka Vukmirovic
- Intern Clinic, Clinical Ward for Cardiovascular Diseases, Clinical-Hospital Centre Zvezdara, 11000 Belgrade, Serbia
| | - Marko Milanov
- Intern Clinic, Clinical Ward for Cardiovascular Diseases, Clinical-Hospital Centre Zvezdara, 11000 Belgrade, Serbia
| | - Dane Cvijanovic
- Intern Clinic, Clinical Ward for Cardiovascular Diseases, Clinical-Hospital Centre Zvezdara, 11000 Belgrade, Serbia
| | - Tijana Mitic
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK
| | - Miron Sopic
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia
| |
Collapse
|
7
|
Liu S, Xu S, Liu S, Chen H. Importance of DJ-1 in autophagy regulation and disease. Arch Biochem Biophys 2023:109672. [PMID: 37336341 DOI: 10.1016/j.abb.2023.109672] [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: 04/17/2023] [Revised: 05/28/2023] [Accepted: 06/16/2023] [Indexed: 06/21/2023]
Abstract
Autophagy is a highly conserved biological process that has evolved across evolution. It can be activated by various external stimuli including oxidative stress, amino acid starvation, infection, and hypoxia. Autophagy is the primary mechanism for preserving cellular homeostasis and is implicated in the regulation of metabolism, cell differentiation, tolerance to starvation conditions, and resistance to aging. As a multifunctional protein, DJ-1 is commonly expressed in vivo and is associated with a variety of biological processes. Its most widely studied role is its function as an oxidative stress sensor that inhibits the production of excessive reactive oxygen species (ROS) in the mitochondria and subsequently the cellular damage caused by oxidative stress. In recent years, many studies have identified DJ-1 as another important factor regulating autophagy; it regulates autophagy in various ways, most commonly by regulating the oxidative stress response. In particular, DJ-1-regulated autophagy is involved in cancer progression and plays a key role in alleviating neurodegenerative diseases(NDS) and defective reperfusion diseases. It could serve as a potential target for the regulation of autophagy and participate in disease treatment as a meaningful modality. Therefore, exploring DJ-1-regulated autophagy could provide new avenues for future disease treatment.
Collapse
Affiliation(s)
- Shiyi Liu
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, 330006, PR China; Second Clinical Medical College, Nanchang University, Nanchang, 330006, PR China
| | - Sheng Xu
- Second Clinical Medical College, Nanchang University, Nanchang, 330006, PR China
| | - Song Liu
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, 330006, PR China
| | - Heping Chen
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, 330006, PR China.
| |
Collapse
|
8
|
Chang WW, Zhang L, Wen LY, Huang Q, Tong X, Tao YJ, Chen GM. Association of tag single nucleotide polymorphisms (SNPs) at lncRNA MALAT1 with type 2 diabetes mellitus susceptibility in the Chinese Han population: A case-control study. Gene X 2023; 851:147008. [DOI: 10.1016/j.gene.2022.147008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/10/2022] [Accepted: 10/19/2022] [Indexed: 11/05/2022] Open
|
9
|
Yan X, Shang X, Feng Z, Chen B, Wu Y, Zhou Y, Li Y, Zhang L. Triterpenoid saponins of Ilex pubescens against TNF-α induced inflammation and apoptosis in human umbilical vein endothelial cells via autophagy pathway. J Pharm Pharmacol 2022; 74:1749-1757. [PMID: 36206186 DOI: 10.1093/jpp/rgac074] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 09/05/2022] [Indexed: 11/14/2022]
Abstract
OBJECTIVES Triterpenoid saponins of Ilex pubescens (IPTS), the main active components of Ilex pubescens, has a therapeutic effect on atherosclerosis (AS). The ingredients in IPTS that could be intracellularly transported by human umbilical vein endothelial cells (HUVECs) may play an essential role in AS. This study attempted to explore its mechanism from the perspectives of HUVECs' inflammation, apoptosis, and autophagy. METHODS By using a tumour necrosis factor-α (TNF-α)-induced HUVECs injury model, cell viability and the expression of intercellular adhesion molecule 1 (ICAM1), matrix metalloproteinase 9 (MMP9), cleave-caspase-3 and cleave-caspase-9, in combination with the results of flow cytometry, JC-1 and Hoechst 33258 staining were investigated to evaluate the anti-inflammatory and anti-apoptotic impact effects of IPTS on HUVECs. Afterwards, the expression of microtubule-associated proteins light chain 3II (LC3II) and sequestosome 1 (p62) was determined to test the effect of IPTS on autophagy. Finally, by adding an autophagy inhibitor 3-methyladenine (3-MA), we investigated whether IPTS exerts anti-inflammatory and anti-apoptotic effects through the autophagy pathway. KEY FINDINGS We firstly demonstrated that pretreatment with IPTS could increase the cell viability, maintain the cell morphology and reduce TNF-α-induced inflammation and apoptosis of HUVECs. Moreover, IPTS pretreatment was proved to raise the expression of LC3II /LC3I while decreasing the expression of p62, which indicated that IPTS could activate HUVECs' autophagy. IPTS has been shown for the first time to exert anti-inflammatory and anti-apoptotic effects through autophagy and thereby resisting TNF-α-induced inflammatory injury of HUVECs. CONCLUSIONS This study preliminarily confirmed that IPTS ameliorated HUVECs' inflammation and apoptosis by increasing autophagy.
Collapse
Affiliation(s)
- Xuemei Yan
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.,Urumqi Hospital of Traditional Chinese Medicine, Urumqi 830000, China
| | - Xueying Shang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.,Guangzhou Baiyunshan Chenliji Pharmaceutical Co., Guangzhou 510000, China
| | - Zhiqiang Feng
- Department of Pharmaceutical Analysis, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Bingying Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yurong Wu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yuan Zhou
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yu Li
- School of Nursing, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Lei Zhang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| |
Collapse
|
10
|
Mameli E, Martello A, Caporali A. Autophagy at the interface of endothelial cell homeostasis and vascular disease. FEBS J 2022; 289:2976-2991. [PMID: 33934518 DOI: 10.1111/febs.15873] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 03/16/2021] [Accepted: 04/09/2021] [Indexed: 12/19/2022]
Abstract
Autophagy is an essential intracellular process for cellular quality control. It enables cell homeostasis through the selective degradation of harmful protein aggregates and damaged organelles. Autophagy is essential for recycling nutrients, generating energy to maintain cell viability in most tissues and during adverse conditions such as hypoxia/ischaemia. The progressive understanding of the mechanisms modulating autophagy in the vasculature has recently led numerous studies to link intact autophagic responses with endothelial cell (EC) homeostasis and function. Preserved autophagic flux within the ECs has an essential role in maintaining their physiological characteristics, whereas defective autophagy can promote endothelial pro-inflammatory and atherogenic phenotype. However, we still lack a good knowledge of the complete molecular repertoire controlling various aspects of endothelial autophagy and how this is associated with vascular diseases. Here, we provide an overview of the current state of the art of autophagy in ECs. We review the discoveries that have so far defined autophagy as an essential mechanism in vascular biology and analyse how autophagy influences ECs behaviour in vascular disease. Finally, we emphasise opportunities for compounds to regulate autophagy in ECs and discuss the challenges of exploiting them to resolve vascular disease.
Collapse
Affiliation(s)
- Eleonora Mameli
- University/BHF Centre for Cardiovascular Science, QMRI, University of Edinburgh, UK
| | | | - Andrea Caporali
- University/BHF Centre for Cardiovascular Science, QMRI, University of Edinburgh, UK
| |
Collapse
|
11
|
Yang J, Liu Z. Mechanistic Pathogenesis of Endothelial Dysfunction in Diabetic Nephropathy and Retinopathy. Front Endocrinol (Lausanne) 2022; 13:816400. [PMID: 35692405 PMCID: PMC9174994 DOI: 10.3389/fendo.2022.816400] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 03/28/2022] [Indexed: 12/15/2022] Open
Abstract
Diabetic nephropathy (DN) and diabetic retinopathy (DR) are microvascular complications of diabetes. Microvascular endothelial cells are thought to be the major targets of hyperglycemic injury. In diabetic microvasculature, the intracellular hyperglycemia causes damages to the vascular endothelium, via multiple pathophysiological process consist of inflammation, endothelial cell crosstalk with podocytes/pericytes and exosomes. In addition, DN and DR diseases development are involved in several critical regulators including the cell adhesion molecules (CAMs), the vascular endothelial growth factor (VEGF) family and the Notch signal. The present review attempts to gain a deeper understanding of the pathogenesis complexities underlying the endothelial dysfunction in diabetes diabetic and retinopathy, contributing to the development of new mechanistic therapeutic strategies against diabetes-induced microvascular endothelial dysfunction.
Collapse
Affiliation(s)
- Jing Yang
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center For Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Zhangsuo Liu
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center For Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
12
|
Ghafouri-Fard S, Shoorei H, Mohaqiq M, Majidpoor J, Moosavi MA, Taheri M. Exploring the role of non-coding RNAs in autophagy. Autophagy 2022; 18:949-970. [PMID: 33525971 PMCID: PMC9196749 DOI: 10.1080/15548627.2021.1883881] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 01/13/2021] [Accepted: 01/27/2021] [Indexed: 12/12/2022] Open
Abstract
As a self-degradative mechanism, macroautophagy/autophagy has a role in the maintenance of energy homeostasis during critical periods in the development of cells. It also controls cellular damage through the eradication of damaged proteins and organelles. This process is accomplished by tens of ATG (autophagy-related) proteins. Recent studies have shown the involvement of non-coding RNAs in the regulation of autophagy. These transcripts mostly modulate the expression of ATG genes. Both long non-coding RNAs (lncRNAs) and microRNAs (miRNAs) have been shown to modulate the autophagy mechanism. Levels of several lncRNAs and miRNAs are altered in this process. In the present review, we discuss the role of lncRNAs and miRNAs in the regulation of autophagy in diverse contexts such as cancer, deep vein thrombosis, spinal cord injury, diabetes and its complications, acute myocardial infarction, osteoarthritis, pre-eclampsia and epilepsy.Abbreviations: AMI: acute myocardial infarction; ATG: autophagy-related; lncRNA: long non-coding RNA; miRNA: microRNA.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Shoorei
- Department of Anatomical Sciences, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Mahdi Mohaqiq
- Wake Forest Institute for Regenerative Medicine, School of Medicine, Wake Forest University, Winston-Salem, NC, USA
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Amin Moosavi
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Mohammad Taheri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Prerna K, Dubey VK. Beclin1-mediated interplay between autophagy and apoptosis: New understanding. Int J Biol Macromol 2022; 204:258-273. [PMID: 35143849 DOI: 10.1016/j.ijbiomac.2022.02.005] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 01/04/2023]
Abstract
The definition for autophagy holds a 'single' meaning as a conserved cellular process that constitutes a recycling pathway for damaged organelles and long-lived proteins to maintain nutrient homeostasis and mediate quality control within the cell. But this process of autophagy may behave ambiguously depending on the physiological stress as the stress progresses in the cellular microenvironment; the 'single' meaning of the autophagy changes from the 'cytoplasmic turnover process' to 'tumor suppressive' and a farther extent, 'tumor promoter' process. In a tumorigenic state, the chemotherapy-mediated resistance and intolerance due to upregulated autophagy in cancer cells have become a significant concern. This concern has provided insight to the scientific community to enter into the arena of cross-talk between autophagy and apoptosis. Recent findings and ongoing research have provided insights on some of the key regulators of this cross-talk; one of them is Beclin1 and their involvement in the physiological and the pathophysiological processes; however, reconciliation of these two forms of death remains an arena to be explored extensively. This review sheds light on the interplay between autophagy and apoptosis, emphasizing one of the key players, Beclin1, and its importance in health and diseases.
Collapse
Affiliation(s)
- Kumari Prerna
- School of Biochemical Engineering, Indian Institute of Technology (BHU) Varanasi, UP-221005, India
| | - Vikash Kumar Dubey
- School of Biochemical Engineering, Indian Institute of Technology (BHU) Varanasi, UP-221005, India.
| |
Collapse
|
14
|
Zhao X, Wang C, Liu M, Meng F, Liu K. LncRNA FENDRR Servers as a Possible Marker of Essential Hypertension and Regulates Human Umbilical Vein Endothelial Cells Dysfunction via miR-423-5p/Nox4 Axis. Int J Gen Med 2022; 15:2529-2540. [PMID: 35282648 PMCID: PMC8906997 DOI: 10.2147/ijgm.s338147] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 01/05/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose Essential hypertension (EH) is an intricate non-communicable infirmity and lncRNAs are validated as essential mediators in EH. The study aimed to propose the expression pattern of FENDRR and miR-423-5p, substantiate the potential mechanism of FENDRR/miR-423-5p/Nox4 axis in EH. Patients and Methods The expression of FENDRR and miR-423-5p was evaluated by qRT-PCR and the clinical significance was explored by the ROC curve. Pearson correlation indicated the relationship between FENDRR and miR-423-5p. The function of FENDRR and miR-423-5p on HUVECs was clarified by CCK-8 assay, Transwell assay, and flow cytometry. Western blot was used to assess the relative protein expression of Nox4. Results FENDRR was highly expressed and miR-423-5p was lowly expressed in EH patients and a negative correlation between them was determined. FENDRR might serve as a predictive diagnosis in differentiating EH patients. Knockdown of FENDRR or overexpression of miR-423-5p showed expansionary effects in cell proliferation, cell migration, and inhibiting cell apoptosis. Meanwhile, miR-423-5p was determined as a target of FENDRR and mediated the function of FENDRR on HUVECs. Moreover, Nox4 is a down-streaming target gene of miR-423-5p. The protein expression of Nox4 was regulated by the alternation of miR-423-5p expression. Conclusion FENDRR played an energetic role in EH and contributed to HUVECs dysfunction by restricting cell proliferation, suppressing cell migration, and accelerating cell apoptosis by manipulating the miR-423-5p/Nox4 axis.
Collapse
Affiliation(s)
- Xiaojian Zhao
- Department of Hypertension, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Chen Wang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Min Liu
- Department of Hypertension, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Fansen Meng
- Department of Hypertension, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Kai Liu
- Department of Hypertension, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
- Correspondence: Kai Liu, Department of Hypertension, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, No. 7 Weiwu Road, Zhengzhou, Henan, 450003, People’s Republic of China, Tel/Fax +86-371-65964376, Email
| |
Collapse
|
15
|
Tang X, Zhang Y, Liu X, Li X, Zhao H, Cui H, Shi Y, Chen Y, Xu H, Meng Z, Zhao L, Chen H, Wang Z, Zhu M, Lin Y, Yang B, Zhang Y. Aloe-emodin derivative produces anti-atherosclerosis effect by reinforcing AMBRA1-mediated endothelial autophagy. Eur J Pharmacol 2022; 916:174641. [PMID: 34800465 DOI: 10.1016/j.ejphar.2021.174641] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 10/15/2021] [Accepted: 11/12/2021] [Indexed: 11/29/2022]
Abstract
Atherosclerosis is an inflammatory disease of high lethality associated with endothelial dysfunction. Due to the pathophysiological complexity and our incomplete understanding of the mechanisms for the development and progression of atherosclerosis, effective means for the prevention and treatment of atherosclerosis still need further exploration. This study was designed to investigate the potential effects and underlying mechanisms of aloe-emodin derivative (AED) on atherosclerosis. High fat diet (HFD) treated ApoE-/- mice were used as an animal model of atherosclerosis. Intragastric administration of aloe-emodin (AE) or AED for 12 weeks markedly reduced the atherosclerotic plaque in aorta with decreased plaque area, lipid accumulation, macrophage infiltration, collagen content and metabolic abnormalities. By comparison, AED produced more potent anti-atherosclerosis effects than AE at the same dose. AED enhanced production of autophagy flux in cultured human aortic endothelial cells (HAECs). Moreover, AED increased the expression of activating molecule in Beclin1-regulated autophagy 1 (AMBRA1), a key protein involved in autophagosome formation. Furthermore, knockdown of AMBRA1 blocked the promotion effect of AED on autophagy in HAECs. Taken together, AED facilitates endothelial autophagy via AMBRA1 during the progression of atherosclerosis, suggesting the potential application of this compound for atherosclerosis treatment.
Collapse
Affiliation(s)
- Xueqing Tang
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Yue Zhang
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, PR China
| | - Xin Liu
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Xiaohan Li
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Hongrui Zhao
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Hao Cui
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Yang Shi
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Yongchao Chen
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Honglin Xu
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Ziyu Meng
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Limin Zhao
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Hui Chen
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Zhixia Wang
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Mengying Zhu
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Yuan Lin
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Baofeng Yang
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone, Chinese Academy of Medical Sciences, 2019RU070, PR China; Department of Pharmacology and Therapeutics, Melbourne School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences University of Melbourne, Melbourne, Australia.
| | - Yong Zhang
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China; Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Harbin, PR China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone, Chinese Academy of Medical Sciences, 2019RU070, PR China.
| |
Collapse
|
16
|
Zhao M, Lian A, Zhong L, Guo R. The regulatory mechanism between lysosomes and mitochondria in the aetiology of cardiovascular diseases. Acta Physiol (Oxf) 2022; 234:e13757. [PMID: 34978753 DOI: 10.1111/apha.13757] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 11/16/2021] [Accepted: 01/01/2022] [Indexed: 11/28/2022]
Abstract
Coordinated action among various organelles maintains cellular functions. For instance, mitochondria and lysosomes are the main organelles contributing to cellular metabolism and provide energy for cardiomyocyte contraction. They also provide essential signalling platforms in the cell that regulate many key processes such as autophagy, apoptosis, oxidative stress, inflammation and cell death. Often, abnormalities in mitochondrial or lysosomal structures and functions bring about cardiovascular diseases (CVDs). Although the communication between mitochondria and lysosomes throughout the cardiovascular system is intensely studied, the regulatory mechanisms have not been completely understood. Thus, we summarize the most recent studies related to mitochondria and lysosomes' role in CVDs and their potential connections and communications under cardiac pathophysiological conditions. Further, we discuss limitations and future perspectives regarding diagnosis, therapeutic strategies and drug discovery in CVDs.
Collapse
Affiliation(s)
- Mengxue Zhao
- College of Life Sciences Institute of Life Science and Green Development Hebei University Baoding China
| | - Andrew Lian
- College of Osteopathic Medicine of the Pacific Western University of Health Sciences Pomona California USA
| | - Li Zhong
- College of Life Sciences Institute of Life Science and Green Development Hebei University Baoding China
- College of Osteopathic Medicine of the Pacific Western University of Health Sciences Pomona California USA
| | - Rui Guo
- College of Life Sciences Institute of Life Science and Green Development Hebei University Baoding China
- The Key Laboratory of Zoological Systematics and Application College of Life Sciences Hebei University Baoding China
| |
Collapse
|
17
|
Cai Z, Wu Y, Ju G, Wang G, Liu B. Role of BCAR4 in prostate cancer cell autophagy. Transl Androl Urol 2022; 10:4253-4261. [PMID: 34984190 PMCID: PMC8661267 DOI: 10.21037/tau-21-929] [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: 09/22/2021] [Accepted: 11/04/2021] [Indexed: 11/06/2022] Open
Abstract
Background Increased autophagy of prostate cancer (PC) cells contributes to their resistance to chemotherapy. Recently, we reported that a long non-coding RNA (lncRNA)-breast-cancer anti-estrogen resistance 4 (BCAR4)-is highly expressed in PC and contributes to castration resistance through activation of GLI2 signaling. However, the role of BCAR4 in the regulation of PC cell autophagy is unknown and is the subject of the current study. Methods BCAR4 and Beclin-1 levels and the alteration in autophagy pathway genes were assessed in PC using a public database and in our own clinical specimens. The correlation between BCAR4 and Beclin-1 levels in PC and PC cell lines was determined and their regulatory relationship was assessed by overexpression and knockout assay. The final effect on autophagy was measured by microtubule-associated protein 1A/1B-light chain 3 (LC3) levels. The mechanism that underlies the control of Beclin-1 by BCAR4 was analyzed by cancer database and gain-of-function and loss-of-function approaches. Results BCAR4 and Beclin-1 were both upregulated in PC and were positively correlated. BCAR4 directly activated Beclin-1 at transcriptional level, which subsequently increased the ratio of LC3 II to LC3I to augment PC cell autophagy. Beclin-1 did not control levels of BCAR4. Mechanically, BCAR4 and Beclin-1 shared several targeting microRNAs, among which miR-15 and miR-146 appeared to be the mediators of the effects of BACR4 on Beclin-1. Conclusions BCAR4 may enhance PC cell autophagy through altering miRNA-regulated Beclin-1 expression in PC.
Collapse
Affiliation(s)
- Zhiping Cai
- Department of Urology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Yapei Wu
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Guanqun Ju
- Department of Urology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Gangmin Wang
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Bing Liu
- Department of Urology, Changzheng Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
18
|
Zhang Y, Tang Y, Yan J. LncRNA-XIST Promotes Proliferation and Migration in ox-LDL Stimulated Vascular Smooth Muscle Cells through miR-539-5p/SPP1 Axis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9911982. [PMID: 35028010 PMCID: PMC8752241 DOI: 10.1155/2022/9911982] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 11/05/2021] [Indexed: 12/22/2022]
Abstract
Long noncoding RNAs (lncRNAs) are untranslated transcripts greater than 200 nucleotides in length. Despite not being translated, they play a role in the regulation of transcription, translation, and other cellular processes and have been identified as key regulator in the progression of atherosclerosis. This study focused on the lncRNA X-inactive specific transcript (XIST), which participates in the regulation of X chromosome inactivation. XIST is produced by the XIST gene and is located on human chromosome Xql3.2. We also focused on discovering the possible role and mechanism of lncRNA XIST in oxidized low-density lipoprotein- (ox-LDL-) stimulated vascular smooth muscle cells (VSMCs), which could further help evalute its possible a role in the progression of atherosclerosis. XIST was overexpressed in ox-LDL-stimulated VSMCs, while the expression of miR-539-5p was decreased. XIST knockdown hindered the proliferation and migration of ox-LDL-treated VSMCs. XIST inhibits the miR-539-5p expression through direct interaction. Besides, miR-539-5p inhibitors can partially reverse the effect of XIST depletion on the proliferation and migration of VSMCs induced by ox-LDL stimulation. Further mechanistic analysis showed that secreted phosphoprotein 1 (SPP1) is the target of miR-539-5p, and XIST acts as a competing endogenous RNA for miR-539-5p to enhance the expression of SPP1. In addition, miR-539-5p inhibitor exerts its proliferation and migration effects by activating the miR-539-5p/SPP1 axis in VSMCs stimulated by ox-LDL. In conclusion, our study findings show that XIST inhibition can inhibit the proliferation and migration of atherosclerosis vascular smooth muscle cells, which provides a new theoretical basis for atherosclerosis treatment.
Collapse
Affiliation(s)
- Yi Zhang
- Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yong Tang
- Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jianhua Yan
- Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
19
|
Abstract
Regulatory RNAs like microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) control vascular and immune cells' phenotype and thus play a crucial role in atherosclerosis. Moreover, the mutual interactions between miRNAs and lncRNAs link both types of regulatory RNAs in a functional network that affects lesion formation. In this review, we deduce novel concepts of atherosclerosis from the analysis of the current data on regulatory RNAs' role in endothelial cells (ECs) and macrophages. In contrast to arterial ECs, which adopt a stable phenotype by adaptation to high shear stress, macrophages are highly plastic and quickly change their activation status. At predilection sites of atherosclerosis, such as arterial bifurcations, ECs are exposed to disturbed laminar flow, which generates a dysadaptive stress response mediated by miRNAs. Whereas the highly abundant miR-126-5p promotes regenerative proliferation of dysadapted ECs, miR-103-3p stimulates inflammatory activation and impairs endothelial regeneration by aberrant proliferation and micronuclei formation. In macrophages, miRNAs are essential in regulating energy and lipid metabolism, which affects inflammatory activation and foam cell formation.Moreover, lipopolysaccharide-induced miR-155 and miR-146 shape inflammatory macrophage activation through their oppositional effects on NF-kB. Most lncRNAs are not conserved between species, except a small group of very long lncRNAs, such as MALAT1, which blocks numerous miRNAs by providing non-functional binding sites. In summary, regulatory RNAs' roles are highly context-dependent, and therapeutic approaches that target specific functional interactions of miRNAs appear promising against cardiovascular diseases.
Collapse
Affiliation(s)
- Andreas Schober
- Institute for Cardiovascular Prevention, University Hospital, Ludwig-Maximilians-University, Munich, Germany.
| | - Saffiyeh Saboor Maleki
- Institute for Cardiovascular Prevention, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Maliheh Nazari-Jahantigh
- Institute for Cardiovascular Prevention, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
20
|
Emerging role of long non-coding RNAs in endothelial dysfunction and their molecular mechanisms. Biomed Pharmacother 2021; 145:112421. [PMID: 34798473 DOI: 10.1016/j.biopha.2021.112421] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/10/2021] [Accepted: 11/10/2021] [Indexed: 02/06/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) are the novel class of transcripts involved in transcriptional, post-transcriptional, translational, and post-translational regulation of physiology and the pathology of diseases. Studies have evidenced that the impairment of endothelium is a critical event in the pathogenesis of atherosclerosis and its complications. Endothelial dysfunction is characterized by an imbalance in vasodilation and vasoconstriction, oxidative stress, proinflammatory factors, and nitric oxide bioavailability. Disruption of the endothelial barrier permeability, the first step in developing atherosclerotic lesions is a consequence of endothelial dysfunction. Though several factors interfere with the normal functioning of the endothelium, intrinsic epigenetic mechanisms governing endothelial function are regulated by lncRNAs and perturbations contribute to the pathogenesis of the disease. This review comprehensively addresses the biogenesis of lncRNA and molecular mechanisms underlying and regulation in endothelial function. An insight correlating lncRNAs and endothelial dysfunction-associated diseases can positively impact the development of novel biomarkers and therapeutic targets in endothelial dysfunction-associated diseases and treatment strategies.
Collapse
|
21
|
Mo L, Jiang HB, Tian GR, Lu GJ. The proliferation and migration of atherosclerosis-related HVSMCs were inhibited by downregulation of lncRNA XIST via regulation of the miR-761/BMP9 axis. Kaohsiung J Med Sci 2021; 38:18-29. [PMID: 34595819 DOI: 10.1002/kjm2.12456] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 07/08/2021] [Accepted: 08/18/2021] [Indexed: 11/12/2022] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease that can be caused by the proliferation and migration of human vascular smooth muscle cells (HVSMCs). Here, we found that lncRNA XIST was related to the abnormal proliferation and migration of HVSMCs, and thus, the mechanism by which XIST regulated HVSMCs was further investigated. HVSMCs were treated with oxidized low-density lipoprotein (ox-LDL, 100 μg/ml) as AS models. CCK8 assays, flow cytometry, Transwell assays and wound healing assays were applied to evaluate cell viability, cell cycle analysis, and cell migration, respectively. A dual-luciferase reporter assay was employed to verify the binding relationships between XIST and miR-761, miR-761, and BMP9. Ox-LDL induced the proliferation and migration of HVSMCs, upregulated the expression of XIST, downregulated miR-761 expression, and activated the BMP9/ALK1/endoglin pathway. Luciferase assays revealed that XIST sponged miR-761. XIST knockdown ameliorated ox-LDL-mediated effects in HVSMCs, which were largely abolished by miR-761 silencing. BMP9 was targeted-inhibited by miR-761. MiR-761 overexpression alleviated ox-LDL-mediated effects in HVSMCs. However, BMP9 overexpression abolished miR-761-mediated effects in HVSMCs treated with ox-LDL. Our findings suggested that XIST knockdown suppressed the proliferation and migration of HVSMCs by promoting miR-761, which targeted-inhibited the BMP9/ALK1/endoglin pathway.
Collapse
Affiliation(s)
- Long Mo
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Heng-Bo Jiang
- Department of Cardiology, The Affiliated Hospital of Yongzhou Vocation & Technology College of Hunan Province, Yongzhou, Hunan Province, China
| | - Gui-Ru Tian
- Department of Cardiology, People's Hospital of Sangzhi County of Hunan Province, Zhangjiajie, Hunan Province, China
| | - Gui-Jing Lu
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| |
Collapse
|
22
|
Qian W, Zheng ZQ, Nie JG, Liu LJ, Meng XZ, Sun H, Xiao FM, Kang T. LncRNA SNHG12 alleviates hypertensive vascular endothelial injury through miR-25-3p/SIRT6 pathway. J Leukoc Biol 2021; 110:651-661. [PMID: 33464650 DOI: 10.1002/jlb.1a0820-501r] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/26/2020] [Accepted: 12/12/2020] [Indexed: 01/08/2023] Open
Abstract
The objective of this study was to find the role of LncRNA SNHG12 in the regulation of hypertensive vascular endothelial injury. LncRNA SNHG12 and miR-25-3p expression were detected by quantitative RT-PCR. Protein levels of Sirtuin 6 (SIRT6), endothelial cell (EC) senescence markers p16 and p21, and EC marker CD31 were measured by Western blot. The apoptosis of HUVECs was detected by flow cytometry. The binding between LncRNA SNHG12 and miR-25-3p was verified by dual luciferase reporter gene assay and RNA pull-down assay. As a result, LncRNA SNHG12 was down-regulated in aortic primary ECs isolated from Ang II-induced hypertensive mice and 1 kidney/deoxycorticosterone acetate/salt-induced hypertensive mice. In Ang II-treated HUVECs, the expression level of SNHG12 was reduced and the overexpression of SNHG12 inhibited EC senescence markers p16 and p21 expressions, the apoptosis of HUVECs, and caspase-3 activity. Further investigation confirmed that LncRNA SNHG12 bound to miR-25-3p, and negatively regulated miR-25-3p expression. MiR-25-3p directly targeted SIRT6 and negatively regulated SIRT6 expression. In addition, SNHG12 overexpression inhibited Ang II-induced HUVECs injury through regulating miR-25-3p. Finally, in vivo experiments showed LncRNA SNHG12 overexpression alleviated vascular endothelial injury in Ang II-induced hypertensive mice. In conclusion, LncRNA SNHG12 alleviates vascular endothelial injury induced by hypertension through miR-25-3p/SIRT6 pathway.
Collapse
Affiliation(s)
- Wei Qian
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ze-Qi Zheng
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jun-Gang Nie
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Li-Juan Liu
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiang-Zhu Meng
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Hong Sun
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Feng-Ming Xiao
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ting Kang
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
23
|
Xie Q, Peng J, Guo Y, Li F. MicroRNA-33-5p inhibits cholesterol efflux in vascular endothelial cells by regulating citrate synthase and ATP-binding cassette transporter A1. BMC Cardiovasc Disord 2021; 21:433. [PMID: 34517822 PMCID: PMC8438969 DOI: 10.1186/s12872-021-02228-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 08/24/2021] [Indexed: 12/16/2022] Open
Abstract
Background A high level of total cholesterol is associated with several lipid metabolism disorders, including atherosclerosis and cardiovascular diseases. ATP-binding cassette (ABC) transporter A1 (ABCA1) and miR-33-5p play crucial roles in atherosclerosis by controlling cholesterol efflux. While citrate is a precursor metabolite for lipid and cholesterol synthesis, little is known about the association between citrate synthase (CS) and cholesterol efflux. This study investigated the role of the miR-33-5p/ABCA1/CS axis in regulating cholesterol efflux in vascular endothelial cells (VECs). Materials and methods VECs were treated with oxidized low-density lipoprotein cholesterol (ox-LDL), or pretreated with plasmids overexpressing CS, ABCA1, siRNAs against CS and ABCA1, and an miR-33-5p inhibitor. Cell apoptosis, cellular senescence-associated β-galactosidase activity, inflammation, and cholesterol efflux were detected. Results Treatment with ox-LDL decreased ABCA1 and CS levels and increased miR-33-5p expression and apoptosis in dose-dependent manners. In contrast, treatment with the miR-33-5p inhibitor and ABCA1 and CS overexpression plasmids inhibited the above-mentioned ox-LDL-induced changes. In addition, treatment with ox-LDL decreased cholesterol efflux, induced aging, and promoted the production of inflammatory cytokines (i.e., IL-6 and tumor necrosis factor TNF-α), as well as the expression of Bax and Caspase 3 proteins in VECs. All these changes were rescued by miR-33-5p inhibition and ABCA1 and CS overexpression. The inhibition of ABCA1 and CS by siRNAs eliminated the effects mediated by the miR-33-5p inhibitor, and knockdown of CS eliminated the effects of ABCA1 on VECs. Conclusions This study demonstrated the crucial roles played by the miR-33-5p/ABCA1/CS axis in regulating cholesterol efflux, inflammation, apoptosis, and aging in VECs, and also suggested the axis as a target for managing lipid metabolism disorders. Supplementary Information The online version contains supplementary material available at 10.1186/s12872-021-02228-7.
Collapse
Affiliation(s)
- Qiong Xie
- Department of Cardiology, Hunan Provincial People's Hospital, The First Hospital Affiliated With Hunan Normal University, Changsha, 410005, Hunan, People's Republic of China
| | - Jianqiang Peng
- Department of Cardiology, Hunan Provincial People's Hospital, The First Hospital Affiliated With Hunan Normal University, Changsha, 410005, Hunan, People's Republic of China
| | - Ying Guo
- Department of Cardiology, Hunan Provincial People's Hospital, The First Hospital Affiliated With Hunan Normal University, Changsha, 410005, Hunan, People's Republic of China
| | - Feng Li
- Departments of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, middle Ren-Min Road No. 139, Changsha, 410011, Hunan, People's Republic of China.
| |
Collapse
|
24
|
Malekmohammad K, Bezsonov EE, Rafieian-Kopaei M. Role of Lipid Accumulation and Inflammation in Atherosclerosis: Focus on Molecular and Cellular Mechanisms. Front Cardiovasc Med 2021; 8:707529. [PMID: 34552965 PMCID: PMC8450356 DOI: 10.3389/fcvm.2021.707529] [Citation(s) in RCA: 164] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 07/20/2021] [Indexed: 12/18/2022] Open
Abstract
Atherosclerosis is a chronic lipid-driven and maladaptive inflammatory disease of arterial intima. It is characterized by the dysfunction of lipid homeostasis and signaling pathways that control the inflammation. This article reviews the role of inflammation and lipid accumulation, especially low-density lipoprotein (LDL), in the pathogenesis of atherosclerosis, with more emphasis on cellular mechanisms. Furthermore, this review will briefly highlight the role of medicinal plants, long non-coding RNA (lncRNA), and microRNAs in the pathophysiology, treatment, and prevention of atherosclerosis. Lipid homeostasis at various levels, including receptor-mediated uptake, synthesis, storage, metabolism, efflux, and its impairments are important for the development of atherosclerosis. The major source of cholesterol and lipid accumulation in the arterial wall is proatherogenic modified low-density lipoprotein (mLDL). Modified lipoproteins, such as oxidized low-density lipoprotein (ox-LDL) and LDL binding with proteoglycans of the extracellular matrix in the intima of blood vessels, cause aggregation of lipoprotein particles, endothelial damage, leukocyte recruitment, foam cell formation, and inflammation. Inflammation is the key contributor to atherosclerosis and participates in all phases of atherosclerosis. Also, several studies have shown that microRNAs and lncRNAs have appeared as key regulators of several physiological and pathophysiological processes in atherosclerosis, including regulation of HDL biogenesis, cholesterol efflux, lipid metabolism, regulating of smooth muscle proliferation, and controlling of inflammation. Thus, both lipid homeostasis and the inflammatory immune response are closely linked, and their cellular and molecular pathways interact with each other.
Collapse
Affiliation(s)
| | - Evgeny E. Bezsonov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Institute of Human Morphology, Moscow, Russia
- Institute for Atherosclerosis Research, Moscow, Russia
- Department of Biology and General Genetics, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Mahmoud Rafieian-Kopaei
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
25
|
Zhou H, Jiang F, Leng Y. Propofol Ameliorates ox-LDL-Induced Endothelial Damage Through Enhancing Autophagy via PI3K/Akt/m-TOR Pathway: A Novel Therapeutic Strategy in Atherosclerosis. Front Mol Biosci 2021; 8:695336. [PMID: 34250023 PMCID: PMC8267008 DOI: 10.3389/fmolb.2021.695336] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 05/19/2021] [Indexed: 11/18/2022] Open
Abstract
Objective: Atherosclerosis (AS) represents a common age-associated disease, which may be accelerated by oxidized low-density lipoprotein (ox-LDL)-induced endothelial cell injury. This study aimed to investigate the effects of Propofol on ox-LDL-induced endothelial damage and the underlying molecular mechanisms. Methods: Human umbilical vein endothelial cells (HUVECs) were exposed to ox-LDL to induce endothelial damage. HUVECs were pretreated with 0, 5, 25 and 100°μM Propofol, followed by exposure to 100°μg/ml ox-LDL for 24°h. Cell viability was assessed by cell counting kit-8 (CCK-8) assay. The expression of autophagy- and apoptosis-related proteins was detected via western blot. Autophagosome was investigated under a transmission electron microscope. After co-treatment with autophagy inhibitor Bafilomycin A1 or si-Beclin-1, cell apoptosis was detected by flow cytometry. Furthermore, under cotreatment with PI3K activator 740Y-P, PI3K/Akt/m-TOR pathway- and autophagy-related proteins were examined by western blot. Results: With a concentration-dependent manner, Propofol promoted the viability of HUVECs exposed to ox-LDL, and increased LC3-II/I ratio and Beclin-1 expression, and decreased P62 expression. The formation of autophagosome was enhanced by Propofol. Furthermore, Propofol treatment elevated Bcl-2/Bax ratio and lowered Caspase-3 expression. Bafilomycin A1 or si-Beclin-1 distinctly ameliorated the inhibitory effects of Propofol on apoptosis in ox-LDL-exposed HUVECs. Moreover, Propofol lowered the activation of PI3K/Akt/m-TOR pathway in HUVECs under exposure to ox-LDL. However, its inhibitory effects were weakened by 740Y-P. Conclusion: Collectively, this study revealed that Propofol could ameliorate ox-LDL-induced endothelial damage through enhancing autophagy via PI3K/Akt/m-TOR pathway, which might offer a novel therapeutic strategy in AS.
Collapse
Affiliation(s)
- Hongyi Zhou
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China.,Department of Anesthesiology, Tongzhou Maternal and Child Health Hospital of Beijing, Beijing, China
| | - Fan Jiang
- Department of General Medicine, Beijing Luhe Hospital, Capital Medical University, Beijin, China
| | - Yufang Leng
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China.,Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
26
|
Tsilimigras DI, Bibli SI, Siasos G, Oikonomou E, Perrea DN, Filis K, Tousoulis D, Sigala F. Regulation of Long Non-Coding RNAs by Statins in Atherosclerosis. Biomolecules 2021; 11:623. [PMID: 33922114 PMCID: PMC8143454 DOI: 10.3390/biom11050623] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/12/2021] [Accepted: 04/20/2021] [Indexed: 01/04/2023] Open
Abstract
Despite increased public health awareness, atherosclerosis remains a leading cause of mortality worldwide. Significant variations in response to statin treatment have been noted among different populations suggesting that the efficacy of statins may be altered by both genetic and environmental factors. The existing literature suggests that certain long noncoding RNAs (lncRNAs) might be up- or downregulated among patients with atherosclerosis. LncRNA may act on multiple levels (cholesterol homeostasis, vascular inflammation, and plaque destabilization) and exert atheroprotective or atherogenic effects. To date, only a few studies have investigated the interplay between statins and lncRNAs known to be implicated in atherosclerosis. The current review characterizes the role of lncRNAs in atherosclerosis and summarizes the available evidence related to the effect of statins in regulating lncRNAs.
Collapse
Affiliation(s)
- Diamantis I. Tsilimigras
- First Propaedeutic Department of Surgery, Division of Vascular Surgery, Hippokration Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.F.); (F.S.)
| | - Sofia-Iris Bibli
- Centre for Molecular Medicine, Institute for Vascular Signalling, Goethe University, 60323 Frankfurt am Main, Germany;
| | - Gerasimos Siasos
- First Department of Cardiology, Hippokration Hospital, National and Kapodistrian University of Athens Medical School, 11527 Athens, Greece; (G.S.); (E.O.); (D.T.)
| | - Evangelos Oikonomou
- First Department of Cardiology, Hippokration Hospital, National and Kapodistrian University of Athens Medical School, 11527 Athens, Greece; (G.S.); (E.O.); (D.T.)
| | - Despina N. Perrea
- Laboratory for Experimental Surgery and Surgical Research “N.S. Christeas”, National and Kapodistrian University of Athens, 15772 Athens, Greece;
| | - Konstantinos Filis
- First Propaedeutic Department of Surgery, Division of Vascular Surgery, Hippokration Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.F.); (F.S.)
| | - Dimitrios Tousoulis
- First Department of Cardiology, Hippokration Hospital, National and Kapodistrian University of Athens Medical School, 11527 Athens, Greece; (G.S.); (E.O.); (D.T.)
| | - Fragiska Sigala
- First Propaedeutic Department of Surgery, Division of Vascular Surgery, Hippokration Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.F.); (F.S.)
| |
Collapse
|
27
|
MicroRNA-216a Promotes Endothelial Inflammation by Smad7/I κB α Pathway in Atherosclerosis. DISEASE MARKERS 2020; 2020:8864322. [PMID: 33282009 PMCID: PMC7688351 DOI: 10.1155/2020/8864322] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 10/14/2020] [Accepted: 10/28/2020] [Indexed: 01/23/2023]
Abstract
Background The endothelium is the first line of defence against harmful microenvironment risks, and microRNAs (miRNAs) involved in vascular inflammation may be promising therapeutic targets to modulate atherosclerosis progression. In this study, we aimed to investigate the mechanism by which microRNA-216a (miR-216a) modulated inflammation activation of endothelial cells. Methods. A replicative senescence model of human umbilical vein endothelial cells (HUVECs) was established, and population-doubling levels (PDLs) were defined during passages. PDL8 HUVECs were transfected with miR-216a mimics/inhibitor or small interfering RNA (siRNA) of SMAD family member 7 (Smad7). Real-time PCR and Western blot assays were performed to detect the regulatory role of miR-216a on Smad7 and NF-κB inhibitor alpha (IκBα) expression. The effect of miR-216a on adhesive capability of HUVECs to THP-1 cells was examined. MiR-216a and Smad7 expression in vivo were measured using human carotid atherosclerotic plaques of the patients who underwent carotid endarterectomy (n = 41). Results Luciferase assays showed that Smad7 was a direct target of miR-216a. Smad7 mRNA expression, negatively correlated with miR-216a during endothelial aging, was downregulated in senescent PDL44 cells, compared with young PDL8 HUVECs. MiR-216a markedly increased endothelial inflammation and adhesive capability to monocytes in PDL8 cells by promoting the phosphorylation and degradation of IκBα and then activating NF-κB signalling pathway. The effect of miR-216a on endothelial cells was consistent with that blocked Smad7 by siRNAs. When inhibiting endogenous miR-216a, the Smad7/IκBα expression was rescued, which led to decreased endothelial inflammation and monocytes recruitment. In human carotid atherosclerotic plaques, Smad7 level was remarkably decreased in high miR-216a group compared with low miR-216a group. Moreover, miR-216a was negatively correlated with Smad7 and IκBα levels and positively correlated with interleukin 1 beta (IL1β) expression in vivo. Conclusion In summary, our findings suggest a new mechanism of vascular endothelial inflammation involving Smad7/IκBα signalling pathway in atherosclerosis.
Collapse
|
28
|
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
|
29
|
Lu S, Liang Q, Huang Y, Meng F, Liu J. Definition and review on a category of long non-coding RNA: Atherosclerosis-associated circulating lncRNA (ASCLncRNA). PeerJ 2020; 8:e10001. [PMID: 33240586 PMCID: PMC7666546 DOI: 10.7717/peerj.10001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 08/29/2020] [Indexed: 12/18/2022] Open
Abstract
Atherosclerosis (AS) is one of the most common cardiovascular system diseases which seriously affects public health in modern society. Finding potential biomarkers in the complicated pathological progression of AS is of great significance for the prevention and treatment of AS. Studies have shown that long noncoding RNAs (lncRNAs) can be widely involved in the regulation of many physiological processes, and have important roles in different stages of AS formation. LncRNAs can be secreted into the circulatory system through exosomes, microvesicles, and apoptotic bodies. Recently, increasing studies have been focused on the relationships between circulating lncRNAs and AS development. The lncRNAs in circulating blood are expected to be new non-invasive diagnostic markers for monitoring the progression of AS. We briefly reviewed the previously reported lncRNA transcripts which related to AS development and detectable in circulating blood, including ANRIL, SENCR, CoroMarker, LIPCAR, HIF1α-AS1, LncRNA H19, APPAT, KCNQ1OT1, LncPPARδ, LincRNA-p21, MALAT1, MIAT, and UCA1. Further researches and a definition of atherosclerosis-associated circulating lncRNA (ASCLncRNA) were also discussed.
Collapse
Affiliation(s)
- Shanshan Lu
- Department of Histology and Embryology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Qin Liang
- Department of Histology and Embryology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Yanqing Huang
- Department of Histology and Embryology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Fanming Meng
- Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Junwen Liu
- Department of Histology and Embryology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China.,China-Africa Research Center of Infectious Diseases, School of Basic Medical Sciences, Central South University, Changsha, Hunan Province, China
| |
Collapse
|
30
|
Ren K, Xu XD, Yu XH, Li MQ, Shi MW, Liu QX, Jiang T, Zheng XL, Yin K, Zhao GJ. LncRNA-modulated autophagy in plaque cells: a new paradigm of gene regulation in atherosclerosis? Aging (Albany NY) 2020; 12:22335-22349. [PMID: 33154191 PMCID: PMC7695379 DOI: 10.18632/aging.103786] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/14/2020] [Indexed: 12/25/2022]
Abstract
The development of atherosclerosis is accompanied by the functional deterioration of plaque cells, which leads to the escalation of endothelial inflammation, abnormal vascular smooth muscle cell phenotype switching and the accumulation of lipid-laden macrophages within vascular walls. Autophagy, a highly conserved homeostatic mechanism, is critical for the delivery of cytoplasmic substrates to lysosomes for degradation. Moderate levels of autophagy prevent atherosclerosis by safeguarding plaque cells against apoptosis, preventing inflammation, and limiting the lipid burden, whereas excessive autophagy exacerbates cell damage and inflammation and thereby accelerates the formation of atherosclerotic plaques. Increasing lines of evidence suggest that long noncoding RNAs can be either beneficial or detrimental to atherosclerosis development by regulating the autophagy level. This review summarizes the research progress related to 1) the significant role of autophagy in atherosclerosis and 2) the effects of the lncRNA-mediated modulation of autophagy on the plaque cell fate, inflammation levels, proliferative capacity, and cholesterol metabolism and subsequently on atherogenesis.
Collapse
Affiliation(s)
- Kun Ren
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan City People’s Hospital, Qingyuan, Guangdong, China.,Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Xiao-Dan Xu
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xiao-Hai Yu
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Meng-Qi Li
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan City People’s Hospital, Qingyuan, Guangdong, China
| | - Meng-Wen Shi
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Qi-Xian Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Ting Jiang
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan City People’s Hospital, Qingyuan, Guangdong, China
| | - Xi-Long Zheng
- Department of Biochemistry and Molecular Biology, Libin Cardiovascular Institute of Alberta, University of Calgary, Health Sciences Center, Calgary, AB, Canada.,Key Laboratory of Molecular Targets and Clinical Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Kai Yin
- The Second Affiliated Hospital of Guilin Medical University, Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, China
| | - Guo-Jun Zhao
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan City People’s Hospital, Qingyuan, Guangdong, China
| |
Collapse
|
31
|
Meng Q, Pu L, Luo X, Wang B, Li F, Liu B. Regulatory Roles of Related Long Non-coding RNAs in the Process of Atherosclerosis. Front Physiol 2020; 11:564604. [PMID: 33192561 PMCID: PMC7604474 DOI: 10.3389/fphys.2020.564604] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 08/17/2020] [Indexed: 12/19/2022] Open
Abstract
Atherosclerosis (AS) is the main cause of coronary heart disease, cerebral infarction, and peripheral vascular disease, which comprise serious hazards to human health. Atherosclerosis is characterized by the deposition of lipids on the interior walls of blood vessels, causing an inflammatory response of immune cells, endothelial cells, and smooth muscle cells, and a proliferation cascade reaction. Despite years of research, the underlying pathogenesis of AS is not fully defined. Recent advances in our understanding of the molecular mechanisms by which non-coding RNA influences the initiation and progression of AS have shown that long non-coding RNAs (lncRNAs) regulate important stages in the atherosclerotic process. In this review, we summarize current knowledge of lncRNAs, which influence the development of AS. We review the regulatory processes of lncRNAs on core stages of atherosclerotic progression, including lipid metabolism, inflammation, vascular cell proliferation, apoptosis, adhesion and migration, and angiogenesis. A growing body of evidence suggests that lncRNAs have great potential as new therapeutic targets for the treatment of vascular diseases.
Collapse
Affiliation(s)
- Qingyu Meng
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, China
| | - Luya Pu
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, China
| | - Xizi Luo
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, China
| | - Baisen Wang
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, China
| | - Fan Li
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, China.,The Key Laboratory for Bionics Engineering, Ministry of Education, Jilin University, Changchun, China.,Engineering Research Center for Medical Biomaterials of Jilin Province, Jilin University, Changchun, China.,Key Laboratory for Health Biomedical Materials of Jilin Province, Jilin University, Changchun, China.,State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang, China
| | - Bin Liu
- Cardiovascular Disease Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
32
|
Zhang H, Lu B. The Roles of ceRNAs-Mediated Autophagy in Cancer Chemoresistance and Metastasis. Cancers (Basel) 2020; 12:cancers12102926. [PMID: 33050642 PMCID: PMC7600306 DOI: 10.3390/cancers12102926] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 09/28/2020] [Accepted: 10/07/2020] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Chemoresistance and metastasis are the main causes of treatment failure in cancers. Autophagy contribute to the survival and metastasis of cancer cells. Competing endogenous RNA (ceRNA), particularly long non-coding RNAs and circular RNA (circRNA), can bridge the interplay between autophagy and chemoresistance or metastasis in cancers via sponging miRNAs. This review aims to discuss on the function of ceRNA-mediated autophagy in the process of metastasis and chemoresistance in cancers. ceRNA network can sequester the targeted miRNA expression to indirectly upregulate the expression of autophagy-related genes, and thereof participate in autophagy-mediated chemoresistance and metastasis. Our clarification of the mechanism of autophagy regulation in metastasis and chemoresistance may greatly improve the efficacy of chemotherapy and survival in cancer patients. The combination of the tissue-specific miRNA delivery and selective autophagy inhibitors, such as hydroxychloroquine, is attractive to treat cancer patients in the future. Abstract Chemoresistance and metastasis are the main causes of treatment failure and unfavorable outcome in cancers. There is a pressing need to reveal their mechanisms and to discover novel therapy targets. Autophagy is composed of a cascade of steps controlled by different autophagy-related genes (ATGs). Accumulating evidence suggests that dysregulated autophagy contributes to chemoresistance and metastasis via competing endogenous RNA (ceRNA) networks including lncRNAs and circRNAs. ceRNAs sequester the targeted miRNA expression to indirectly upregulate ATGs expression, and thereof participate in autophagy-mediated chemoresistance and metastasis. Here, we attempt to summarize the roles of ceRNAs in cancer chemoresistance and metastasis through autophagy regulation.
Collapse
Affiliation(s)
- Huilin Zhang
- Department of Surgical Pathology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou 310002, Zhejiang Province, China;
| | - Bingjian Lu
- Department of Surgical Pathology and Center for Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou 310002, Zhejiang Province, China
- Correspondence: ; Tel.: +86-571-89991702
| |
Collapse
|
33
|
Zareba L, Fitas A, Wolska M, Junger E, Eyileten C, Wicik Z, De Rosa S, Siller-Matula JM, Postula M. MicroRNAs and Long Noncoding RNAs in Coronary Artery Disease: New and Potential Therapeutic Targets. Cardiol Clin 2020; 38:601-617. [PMID: 33036721 DOI: 10.1016/j.ccl.2020.07.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Noncoding RNAs (ncRNAs), including long noncoding RNAs and microRNAs, play an important role in coronary artery disease onset and progression. The ability of ncRNAs to simultaneously regulate many target genes allows them to modulate various key processes involved in atherosclerosis, including lipid metabolism, smooth muscle cell proliferation, autophagy, and foam cell formation. This review focuses on the therapeutic potential of the most important ncRNAs in coronary artery disease. Moreover, various other promising microRNAs and long noncoding RNAs that attract substantial scientific interest as potential therapeutic targets in coronary artery disease and merit further investigation are presented.
Collapse
Affiliation(s)
- Lukasz Zareba
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Banacha 1B Str., Warsaw 02-097, Poland
| | - Alex Fitas
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Banacha 1B Str., Warsaw 02-097, Poland
| | - Marta Wolska
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Banacha 1B Str., Warsaw 02-097, Poland
| | - Eva Junger
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Banacha 1B Str., Warsaw 02-097, Poland
| | - Ceren Eyileten
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Banacha 1B Str., Warsaw 02-097, Poland
| | - Zofia Wicik
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Banacha 1B Str., Warsaw 02-097, Poland; Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, Alameda da Universidade, s/n-Anchieta, São Paulo 09606-045, Brazil
| | - Salvatore De Rosa
- Division of Cardiology, Department of Medical and Surgical Sciences, "Magna Graecia" University, Viale Europa, Catanzaro 88100, Italy
| | - Jolanta M Siller-Matula
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Banacha 1B Str., Warsaw 02-097, Poland; Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Spitalgasse 23, Vienna 1090, Austria
| | - Marek Postula
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Banacha 1B Str., Warsaw 02-097, Poland; Longevity Center, Warsaw, Poland.
| |
Collapse
|
34
|
Chi K, Zhang J, Sun H, Liu Y, Li Y, Yuan T, Zhang F. Knockdown of lncRNA HOXA-AS3 Suppresses the Progression of Atherosclerosis via Sponging miR-455-5p. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:3651-3662. [PMID: 32982172 PMCID: PMC7490108 DOI: 10.2147/dddt.s249830] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 08/07/2020] [Indexed: 12/22/2022]
Abstract
Background Atherosclerosis can lead to multiple cardiovascular diseases, especially myocardial infarction. Long noncoding RNAs (lncRNAs) are involved in multiple diseases, including atherosclerosis. LncRNA HOXA-AS3 was found to be notably upregulated in atherosclerosis. However, the biological function of HOXA-AS3 during the occurrence and development of atherosclerosis remains unclear. Materials and Methods Human vascular endothelial cells (HUVECs) were treated with oxidized low-density lipoprotein (oxLDL) to mimic atherosclerosis in vitro. Gene and protein expressions in HUVECs were detected by RT-qPCR and Western blot, respectively. Cell proliferation was tested by CCK-8 and Ki67 staining. Cell apoptosis and cycle were measured by flow cytometry. Additionally, the correlation between HOXA-AS3 and miR-455-5p was confirmed by dual luciferase report assay and RNA pull-down. Finally, in vivo model of atherosclerosis was established to confirm the function of HOXA-AS3 during the development of atherosclerosis in vivo. Results LncRNA HOXA-AS3 was upregulated in oxLDL-treated HUVECs. In addition, oxLDL-induced growth inhibition of HUVECs was significantly reversed by knockdown of HOXA-AS3. Consistently, oxLDL notably induced G1 arrest in HUVECs, while this phenomenon was greatly reversed by HOXA-AS3 siRNA. Furthermore, downregulation of HOXA-AS3 notably inhibited the progression of atherosclerosis through mediation of miR-455-5p/p27 Kip1 axis. Besides, silencing of HOXA-AS3 notably relieved the symptom of atherosclerosis in vivo. Conclusion Downregulation of HOXA-AS3 significantly suppressed the progression of atherosclerosis via regulating miR-455-5p/p27 Kip1 axis. Thus, HOXA-AS3 might serve as a potential target for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Kui Chi
- Department of Vascular Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Jinwen Zhang
- Department of Vascular Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Huanhuan Sun
- Department of Vascular Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Yang Liu
- Department of Vascular Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Ye Li
- Department of Anesthesiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Tao Yuan
- Department of Vascular Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China
| | - Feng Zhang
- Department of Vascular Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, People's Republic of China
| |
Collapse
|
35
|
Zhang S, Zhu X, Li G. E2F1/SNHG7/miR-186-5p/MMP2 axis modulates the proliferation and migration of vascular endothelial cell in atherosclerosis. Life Sci 2020; 257:118013. [DOI: 10.1016/j.lfs.2020.118013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/16/2020] [Accepted: 06/22/2020] [Indexed: 02/08/2023]
|
36
|
Abstract
Purpose of Review To summarize recent insights into long non-coding RNAs (lncRNAs) involved in atherosclerosis. Because atherosclerosis is the main underlying pathology of cardiovascular diseases (CVD), the world’s deadliest disease, finding novel therapeutic strategies is of high interest. Recent Findings LncRNAs can bind to proteins, DNA, and RNA regulating disease initiation and plaque growth as well as plaque stability in different cell types such as endothelial cells (ECs), vascular smooth muscle cells (VSMCs), and macrophages. A number of lncRNAs have been implicated in cholesterol homeostasis and foam cell formation such as LASER, LeXis, and CHROME. Among others, MANTIS, lncRNA-CCL2, and MALAT1 were shown to be involved in vascular inflammation. Further regulations include, but are not limited to, DNA damage response in ECs, phenotypic switch of VSMCs, and various cell death mechanisms. Interestingly, some lncRNAs are closely correlated with response to statin treatment, such as NEXN-AS1 or LASER. Additionally, some lncRNAs may serve as CVD biomarkers. Summary LncRNAs are a potential novel therapeutic target to treat CVD, but research of lncRNA in atherosclerosis is still in its infancy. With increasing knowledge of the complex and diverse regulations of lncRNAs in the heterogeneous environment of atherosclerotic plaques, lncRNAs hold promise for their clinical translation in the near future.
Collapse
Affiliation(s)
- Tatjana Josefs
- Department of Physiology, Amsterdam Cardiovascular Science, VU University, Amsterdam UMC, Postbus 7057, 1007 MB, Amsterdam, The Netherlands
| | - Reinier A Boon
- Department of Physiology, Amsterdam Cardiovascular Science, VU University, Amsterdam UMC, Postbus 7057, 1007 MB, Amsterdam, The Netherlands. .,Institute for Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany. .,German Center for Cardiovascular Research (DZHK), Frankfurt am Main, Germany.
| |
Collapse
|
37
|
Chen J, Wang L, Liu WH, Shi J, Zhong Y, Liu SJ, Liu SM. Aspirin protects human coronary artery endothelial cells by inducing autophagy. Physiol Int 2020; 107:294-305. [PMID: 32750030 DOI: 10.1556/2060.2020.00029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 05/13/2020] [Indexed: 11/19/2022]
Abstract
Although the use of aspirin has substantially reduced the risks of cardiovascular events and death, its potential mechanisms have not been fully elucidated. In a previous study, we found that aspirin triggers cellular autophagy. In the present study, we aimed to determine the protective effects of aspirin on human coronary artery endothelial cells (HCAECs) and explore its underlying mechanisms. HCAECs were treated with oxidized low-density lipoprotein (ox-LDL), angiotensin II (Ang-II), or high glucose (HG) with or without aspirin stimulation. The expression levels of endothelial nitric oxide (NO) synthase (eNOS), p-eNOS, LC3, p62, phosphor-nuclear factor kappa B (p-NF-κB), p-p38 mitogen-activated protein kinase (p-p38 MAPK), and Beclin-1 were detected via immunoblotting analysis. Concentrations of soluble intercellular adhesion molecule-1 (sICAM-1) and soluble vascular cell adhesion molecule-1 (sVCAM-1) were measured via ELISA. NO levels were determined using the Griess reagent. Autophagic flux was tracked by tandem mRFP-GFP-tagged LC3. Results showed that aspirin increased eNOS level and reduced injury to the endothelial cells (ECs) caused by ox-LDL, Ang-II, and HG treatment in a dose-dependent manner. Aspirin also increased the LC3II/LC3I ratio, decreased p62 expression, and enhanced autophagic flux (autophagosome and autolysosome puncta) in the HCAECs. p-NF-κB and p-p38 mitogen-activated protein kinase inhibition, sVCAM-1 and sICAM-1 secretion, and eNOS activity promotion by aspirin treatment were found to be dependent on Beclin-1. These results suggested that aspirin can protect ECs from ox-LDL-, Ang-II-, and HG-induced injury by activating autophagy in a Beclin-1-dependent manner.
Collapse
Affiliation(s)
- J Chen
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, PR China
| | - L Wang
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, PR China
| | - W H Liu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, PR China
| | - J Shi
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, PR China
| | - Y Zhong
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, PR China
| | - S J Liu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, PR China
| | | |
Collapse
|
38
|
Qin X, Guo J. MicroRNA-328-3p Protects Vascular Endothelial Cells Against Oxidized Low-Density Lipoprotein Induced Injury via Targeting Forkhead Box Protein O4 (FOXO4) in Atherosclerosis. Med Sci Monit 2020; 26:e921877. [PMID: 32329448 PMCID: PMC7195608 DOI: 10.12659/msm.921877] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background Endothelial cell (EC) injury is underlies for the pathogenesis of atherosclerosis (AS). MicroRNAs (miRNAs) have been indicated play important role in modulating AS occurrence and development. However, how miR-328-3p modulates EC injury molecular level for AS remains unclear. Material/Methods MiR-328-3p and forkhead box protein O4 (FOXO4) expression were detected using quantitative real-time polymerase chain reaction (qRT-PCR) and western blot. Cell viability was analyzed by Cell Counting Kit-8 (CCK-8) assay. Flow cytometry was utilized to analyze the apoptotic rate. The migration and invasion abilities were measured by Transwell assay. Western blot was applied to examine the expression of C-caspase 3, Beclin, LC3-I, and LC3-II. The levels of interleukin (IL)-1β, IL-10, and tumor necrosis factor-α (TNF-α) were tested by enzyme-linked immunosorbent assay (ELISA) assay and western blot. Results MiR-328-3p expression was downregulated in oxidized low-density lipoprotein (ox-LDL) induced human umbilical vein endothelial cells (HUVECs). Overexpressed miR-328-3p obviously alleviated ox-LDL induced inhibition on cell viability, migration and invasion, stimulation on apoptosis, autophagy as well as inflammation in HUVECs. FOXO4 was elevated in ox-LDL HUVECs, and functional assay indicated that FOXO4 aggravated ox-LDL induced HUVECs impairment. In addition, FOXO4 was a target of miR-328-3p in HUVECs; rescue experiments suggested miR-328-3p could protect HUVECs against ox-LDL induced injury via regulating FOXO4. Conclusions MiR-328-3p protected vascular endothelial cells against ox-LDL induced injury via targeting FOXO4, suggesting a novel insight for atherosclerosis treatment.
Collapse
Affiliation(s)
- Xiaowei Qin
- Department of Cardiac Function, The Affiliated Hospital of Kangda College of Nanjing Medica University/The First People's Hospital of Lianyungang/ The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, Jiangsu, China (mainland)
| | - Jiantao Guo
- Department of Cardiac Surgery, First hospital of Jilin University, Changchun, Jilin, China (mainland)
| |
Collapse
|
39
|
Autophagy in endothelial cells regulates their haematopoiesis-supporting ability. EBioMedicine 2020; 53:102677. [PMID: 32114389 PMCID: PMC7047195 DOI: 10.1016/j.ebiom.2020.102677] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 01/22/2020] [Accepted: 01/30/2020] [Indexed: 12/17/2022] Open
Abstract
Background Endothelial cells (ECs) function as an instructive platform to support haematopoietic stem cell (HSC) homeostasis. Our recent studies found that impaired bone marrow (BM) ECs are responsible for the defective haematopoiesis in patients with poor graft function (PGF), which is characterised by pancytopenia post-allotransplant. Although activated autophagy was reported to benefit ECs, whether EC autophagy plays a critical role in supporting HSCs and its effect on PGF patients post-allotransplant remain unclear. Methods To evaluate whether the autophagy status of ECs modulates their ability to support haematopoiesis, human umbilical vein endothelial cells (HUVECs) and primary BM ECs derived from healthy donors were subjected to knockdown or overexpression of Beclin-1 (an autophagy-related protein). Moreover, BM ECs derived from PGF patients were studied. Findings Beclin-1 knockdown significantly reduced the haematopoiesis-supporting ability of ECs by suppressing autophagy, which could be restored by activating autophagy via Beclin-1 upregulation. Moreover, autophagy positively regulated haematopoiesis-related genes in HUVECs. Subsequently, a prospective case-control study demonstrated that defective autophagy reduced Beclin-1 expression and the colony-forming unit (CFU) plating efficiency in BM ECs from PGF patients compared to matched patients with good graft function. Rapamycin, an autophagy activator, quantitatively and functionally improved BM ECs from PGF patients in vitro and enhanced their ability to support HSCs by activating the Beclin-1 pathway. Interpretation Our results suggest that the autophagy status of ECs modulates their ability to support haematopoiesis by regulating the Beclin-1 pathway. Defective autophagy in BM ECs may be involved in the pathogenesis of PGF post-allotransplant. Rapamycin provides a promising therapeutic approach for PGF patients. Funding Please see funding sources.
Collapse
|
40
|
Yan Y, Song D, Wu J, Wang J. Long Non-Coding RNAs Link Oxidized Low-Density Lipoprotein With the Inflammatory Response of Macrophages in Atherogenesis. Front Immunol 2020; 11:24. [PMID: 32082313 PMCID: PMC7003668 DOI: 10.3389/fimmu.2020.00024] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 01/07/2020] [Indexed: 12/15/2022] Open
Abstract
Atherosclerosis is characterized as a chronic inflammatory response to cholesterol deposition in arteries. Low-density lipoprotein (LDL), especially the oxidized form (ox-LDL), plays a crucial role in the occurrence and development of atherosclerosis by inducing endothelial cell (EC) dysfunction, attracting monocyte-derived macrophages, and promoting chronic inflammation. However, the mechanisms linking cholesterol accumulation with inflammation in macrophage foam cells are poorly understood. Long non-coding RNAs (lncRNAs) are a group of non-protein-coding RNAs longer than 200 nucleotides and are found to regulate the progress of atherosclerosis. Recently, many lncRNAs interfering with cholesterol deposition or inflammation were identified, which might help elucidate their underlying molecular mechanism or be used as novel therapeutic targets. In this review, we summarize and highlight the role of lncRNAs linking cholesterol (mainly ox-LDL) accumulation with inflammation in macrophages during the process of atherosclerosis.
Collapse
Affiliation(s)
- Youyou Yan
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
| | - Dandan Song
- Department of Clinical Laboratory, Second Hospital of Jilin University, Changchun, China
| | - Junduo Wu
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| | - Junnan Wang
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
41
|
Huang W, Huang F, Lei Z, Luo H. LncRNA SNHG11 Promotes Proliferation, Migration, Apoptosis, and Autophagy by Regulating hsa-miR-184/AGO2 in HCC. Onco Targets Ther 2020; 13:413-421. [PMID: 32021286 PMCID: PMC6969695 DOI: 10.2147/ott.s237161] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 12/19/2019] [Indexed: 12/22/2022] Open
Abstract
Background The most common malignant tumor of the digestive system is HCC. However, the mechanism and pathogenesis of HCC occurrence and progress are still unknown. LncRNA is closely related to the occurrence and progress of HCC. It is important to investigate the effect and role of lncRNA in HCC. Materials and Methods LncRNA microarray assay was used to screen the differential expression profile of lncRNA. SNHG11, miR-184 and GO2 expression was analyzed by RT-PCR. The ability of SNHG11 to serve as a sponge for miRNA and the fact that miR-184 directly targets mRNA were revealed by dual luciferase assay and RIP. Apoptosis and autophagy related proteins were detected by Western blot. Cell proliferation, invasion, migration, and apoptosis were detected by CCK-8 assay, wound healing assay, transwell assay, and flow cytometry. Results LncRNA microarray assay and RT-PCR results revealed that the expression of SNHG11 was increased in HCC tumor tissues and also upregulated in HCC cells. SNHG11 had a connection with poor survival rate in HCC. In addition, dual luciferase assay and RIP results revealed that SNHG11 serves as a sponge for miR-184 and miR-184 directly targets AGO2. Pearson correlation analysis showed that SNHG11 with miR-184 and miR-184 with AGO2 were negative correlations, and SNHG11 with AGO2 was a positive correlation. Cell function assay and Western blot showed SNHG4/miR-184/AGO2 regulatory loop was critical for HCC cell proliferation, migration, apoptosis, and autophagy. Conclusion Our study demonstrated that the expression of SNHG11 is higher in HCC; moreover, SNHG11 promotes proliferation, migration, apoptosis, and autophagy by regulating AGO2 via miR-184 in HCC. Our verification of the role of SNHG11 may provide a novel biomarker for the diagnosis, therapy, and prognosis of HCC.
Collapse
Affiliation(s)
- Wei Huang
- Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, People's Republic of China
| | - Feizhou Huang
- Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, People's Republic of China
| | - Zhao Lei
- Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, People's Republic of China
| | - Hongwu Luo
- Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, People's Republic of China
| |
Collapse
|
42
|
Liu H, Shi C, Deng Y. MALAT1 affects hypoxia-induced vascular endothelial cell injury and autophagy by regulating miR-19b-3p/HIF-1α axis. Mol Cell Biochem 2020; 466:25-34. [PMID: 31933110 DOI: 10.1007/s11010-020-03684-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Accepted: 01/04/2020] [Indexed: 12/13/2022]
Abstract
Cardiovascular disease has become the leading cause of death in the world. Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) plays an important role in cardiovascular disease, such as stroke. However, the role of MALAT1 in hypoxia (HYP)-induced vascular endothelial cells (VECs) remains unclear. In the present study, HYP-treated human umbilical vein endothelial cells (HUVECs) were utilized to simulate HYP-induced VEC injury. It was found that after HYP treatment, the levels of MALAT1 and hypoxia-induced factor-1 (HIF-1α) in HUVECs were upregulated, while the level of miR-19b-3p was downregulated. Knockdown of MALAT1 with siRNA significantly reduced the HIF-1α level induced by HYP. In addition, MALAT1 knockdown inhibited HYP-induced HUVECs apoptosis, autophagy and inflammation. The overexpression of HIF-1α overcame the effect of MALAT1 knockdown. Mechanism analysis showed that MALAT1-targeted miR-19b-3p and then regulated downstream HIF-1α. MALAT1 knockdown increased the level of miR-19b-3p in cells, and increased miR-19b-3p further inhibited the expression of HIF-1α, thereby reducing the HYP-induced HUVECs apoptosis, autophagy and inflammation. Taken together, these results suggest that MALAT1 may be a potential target for mitigating HYP-induced endothelial cell injury.
Collapse
Affiliation(s)
- Huzi Liu
- Department of Cardiothoracic Surgery, The Second Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, 030001, China
| | - Chunli Shi
- Department of Outpatient, Shanxi Cardiovascular Hospital (Institute), The Affiliated Cardiovascular Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, 030024, China
| | - Yongzhi Deng
- Department of Cardiovascular Surgery, Shanxi Cardiovascular Hospital (Institute), The Affiliated Cardiovascular Hospital of Shanxi Medical University, Shanxi Medical University, No. 18, Yifen Street, Wanbailin District, Taiyuan, 030024, Shanxi, China.
| |
Collapse
|
43
|
Jia Z, An L, Lu Y, Xu C, Wang S, Wang J, Teng X. Oxidized Low Density Lipoprotein-Induced Atherogenic Response of Human Umbilical Vascular Endothelial Cells (HUVECs) was Protected by Atorvastatin by Regulating miR-26a-5p/Phosphatase and Tensin Homolog (PTEN). Med Sci Monit 2019; 25:9836-9843. [PMID: 31865360 PMCID: PMC6938650 DOI: 10.12659/msm.918405] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background Atherosclerosis is a chronic and multifactorial disease, and it is the main reason of coronary heart disease, cerebral infarction, and peripheral vascular disease, which leads to the formation of lesions in arterial blood vessels. Our study aimed to explore the protective effect and its underlying mechanism of atorvastatin (ATV) on oxidized low-density lipoprotein (ox-LDL)-induced atherosclerosis. Material/Methods Human umbilical vascular endothelial cells (HUVECs) were cultured and pretreated with ox-LDL to establish an in vitro atherosclerotic cell model. Cell Counting Kit-8 (CCK-8) assay, TUNEL staining, and Transwell assay were used to detect the cell activity, apoptosis, and migration in HUVECs. Quantitative real-time polymerase chain reaction (qRT-PCR) and western blot were applied to measure the mRNA and protein expressions of adhesion-related genes in HUVECs. Results Pretreated with 100 mg/L ox-LDL resulted in a 57.23% decrease of cell viability and 81.09% increase of apoptotic injury in HUVECs compare to the control. Meanwhile, ox-LDL pretreatment increased the cell migration and the expression of miR-26a-5p in HUVECs. ATV treatment could effectively reverse the cellular damage induced by ox-LDL, decrease the release of adhesion-related molecules, and downregulate the expression of miR-26a-5p by 44.79% in HUVECs. Moreover, phosphatase and tensin homolog (PTEN) was demonstrated to be the target gene of miR-26a-5p. Conclusions Our results highlight that ATV protects against ox-LDL-induced downregulation of cell viability, upregulation of cell apoptosis, migration, as well as the release of adhesion-related molecules in HUVECs through the miR-26a-5p/PTEN axis. This study provides new insights into the underlying mechanism of ATV therapeutic potential in atherosclerosis, and also provides a new strategy for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Zhuowen Jia
- Department of Geriatric Cardiology, Heilongjiang Provincial Hospital, Harbin, Heilongjiang, China (mainland)
| | - Liping An
- Department of Geriatric Cardiology, Heilongjiang Provincial Hospital, Harbin, Heilongjiang, China (mainland)
| | - Yanhong Lu
- Department of Geriatric Cardiology, Heilongjiang Provincial Hospital, Harbin, Heilongjiang, China (mainland)
| | - Chaorui Xu
- Department of Geriatric Cardiology, Heilongjiang Provincial Hospital, Harbin, Heilongjiang, China (mainland)
| | - Sha Wang
- Department of Geriatric Cardiology, Heilongjiang Provincial Hospital, Harbin, Heilongjiang, China (mainland)
| | - Jipeng Wang
- Department of Geriatric Cardiology, Heilongjiang Provincial Hospital, Harbin, Heilongjiang, China (mainland)
| | - Xiane Teng
- Department of Geriatric Cardiology, Heilongjiang Provincial Hospital, Harbin, Heilongjiang, China (mainland)
| |
Collapse
|
44
|
Yan Y, Song D, Song X, Song C. The role of lncRNA MALAT1 in cardiovascular disease. IUBMB Life 2019; 72:334-342. [PMID: 31856403 DOI: 10.1002/iub.2210] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 11/24/2019] [Indexed: 12/13/2022]
Abstract
Cardiovascular disease (CVD) is the first leading cause of death worldwide. Understanding the molecular mechanism of signaling pathways involved in pathology of CVD is benefit for targeted therapeutics. Recently, long non-coding RNAs (lncRNAs) are found and involved in regulation of pathology of CVD at different levels. Among them, MALAT1 attracted more attention as it was profoundly expressed in endothelial cells or cardiomyocytes in response to the risk factors of CVD, such as hypoxia, high glucose, cytokine, and oxidative stress. In this review, we summarize recent progresses in research on the molecular mechanism of MALAT1 on regulating the pathophysiological processes of CVD as well as its potential therapeutic applications.
Collapse
Affiliation(s)
- Youyou Yan
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China.,Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
| | - Dandan Song
- Department of Clinical Laboratory, Second Hospital of Jilin University, Changchun, China
| | - Xianjing Song
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| | - Chunli Song
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
45
|
Jin D, Guo J, Wu Y, Du J, Yang L, Wang X, Di W, Hu B, An J, Kong L, Pan L, Su G. m 6A mRNA methylation initiated by METTL3 directly promotes YAP translation and increases YAP activity by regulating the MALAT1-miR-1914-3p-YAP axis to induce NSCLC drug resistance and metastasis. J Hematol Oncol 2019; 12:135. [PMID: 31818312 PMCID: PMC6902496 DOI: 10.1186/s13045-019-0830-6] [Citation(s) in RCA: 342] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 11/13/2019] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND METTL3 is an RNA methyltransferase that mediates m6A modification and is implicated in mRNA biogenesis, decay, and translation. However, the biomechanism through which METTL3 regulates MALAT1-miR-1914-3p-YAP axis activity to induce NSCLC drug resistance and metastasis is not very clear. METHODS The expression of mRNA was analyzed by qPCR assays. Protein levels were analyzed by western blotting and immunofluorescent staining. Cellular proliferation was detected by CCK8 assays. Cell migration and invasion were analyzed by wound healing and transwell assays, respectively. Promoter activities and gene transcription were analyzed by luciferase reporter assays. Finally, m6A modification was analyzed by MeRIP. RESULTS METTL3 increased the m6A modification of YAP. METTL3, YTHDF3, YTHDF1, and eIF3b directly promoted YAP translation through an interaction with the translation initiation machinery. Moreover, the RNA level of MALAT1 was increased due to a higher level of m6A modification mediated by METTL3. Meanwhile, the stability of MALAT1 was increased by METTL3/YTHDF3 complex. Additionally, MALAT1 functions as a competing endogenous RNA that sponges miR-1914-3p to promote the invasion and metastasis of NSCLC via YAP. Furthermore, the reduction of YAP m6A modification by METTL3 knockdown inhibits tumor growth and enhances sensitivity to DDP in vivo. CONCLUSION Results indicated that the m6A mRNA methylation initiated by METTL3 promotes YAP mRNA translation via recruiting YTHDF1/3 and eIF3b to the translation initiation complex and increases YAP mRNA stability through regulating the MALAT1-miR-1914-3p-YAP axis. The increased YAP expression and activity induce NSCLC drug resistance and metastasis.
Collapse
Affiliation(s)
- Dan Jin
- grid.452240.5Clinical Medical Laboratory, Binzhou Medical University Hospital, Binzhou, 256603 People’s Republic of China
| | - Jiwei Guo
- Cancer research institute, Binzhou Medical University Hospital, Binzhou, 256603, People's Republic of China.
| | - Yan Wu
- grid.452240.5Cancer research institute, Binzhou Medical University Hospital, Binzhou, 256603 People’s Republic of China
| | - Jing Du
- grid.452240.5Cancer research institute, Binzhou Medical University Hospital, Binzhou, 256603 People’s Republic of China
| | - Lijuan Yang
- grid.452240.5Cancer research institute, Binzhou Medical University Hospital, Binzhou, 256603 People’s Republic of China
| | - Xiaohong Wang
- grid.452240.5Department of Thyroid and Breast Surgery, Binzhou Medical University Hospital, Binzhou, 256603 People’s Republic of China
| | - Weihua Di
- grid.452240.5Department of Pain, Binzhou Medical University Hospital, Binzhou, 256603 People’s Republic of China
| | - Baoguang Hu
- grid.452240.5Department of Gastrointestinal Surgery, Binzhou Medical University Hospital, Binzhou, 256603 People’s Republic of China
| | - Jiajia An
- grid.452240.5Department of Clinical Laboratory, Binzhou Medical University Hospital, Binzhou, 256603 People’s Republic of China
| | - Lingqun Kong
- grid.452240.5Department of Hepatobiliary Surgery, Binzhou Medical University Hospital, Binzhou, 256603 People’s Republic of China
| | - Lei Pan
- grid.452240.5Department of Respiratory and Critical Care Medicine, Binzhou Medical University Hospital, Binzhou, 256603 People’s Republic of China
| | - Guoming Su
- Department of Nursing, Binzhou Polytechnic University, Binzhou, 256603 People’s Republic of China
| |
Collapse
|
46
|
Yang X, Li Z, Zhang L, Wu X, Kang Q, Li L. Retracted Article: Knockdown of long non-coding RNA OIP5-AS1 suppresses cell proliferation and migration in ox-LDL-induced human vascular smooth muscle cells (hVMSCs) through targeting miR-152-3p/PAPPA axis. RSC Adv 2019; 9:32499-32509. [PMID: 35702340 PMCID: PMC9097493 DOI: 10.1039/c9ra06614d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 09/26/2019] [Indexed: 12/22/2022] Open
Abstract
Emerging evidence has demonstrated that long non-coding RNA Opa-interacting protein 5 antisense RNA 1 (OIP5-AS1) is associated with cellular behaviors among malignant tumors. However, the role of OIP5-AS1 in atherosclerosis remains largely undefined. The aim of this study was to explore the expression and role of OIP5-AS1 in a cell model of atherosclerosis, as well as the underlying mechanism. We found that expression of OIP5-AS1 was upregulated in human vascular smooth muscle cells (hVMSCs) under oxidized low density lipoprotein (ox-LDL) administration, and knockdown of OIP5-AS1 suppressed cell viability (CCK-8) and proliferating cell nuclear antigen (PCNA) protein level in ox-LDL-treated hVMSCs, as well as inhibited cell migration rate (wound healing assay) and protein expression of matrix metalloproteinase (MMP)-2 and MMP-9. Mechanically, OIP5-AS1 functioned as competing endogenous RNA (ceRNA) to positively regulate PAPPA expression through sponging miRNA-152-3p (miR-152), and pregnancy-associated plasma protein A (PAPPA) was identified as a downstream target gene for miR-152. Moreover, expression of miR-152 was downregulated and PAPPA was upregulated in ox-LDL-treated hVMSCs. Similarly to OIP5-AS1 knockdown, miR-215 overexpression could inhibit cell proliferation and migration of hVMSCs administrated by ox-LDL, which was abated by PAPPA upregulation. Moreover, miR-215 downregulation partially reversed the suppressive role of OIP5-AS1 knockdown as well. In conclusion, knockdown of OIP5-AS1 suppressed ox-LDL-treated hVMSC proliferation and migration presumably through targeting miR-152/PAPPA axis, suggesting a novel OIP5-AS1/miR-152/PAPPA pathway in atherogenesis.
Collapse
Affiliation(s)
- Xiangya Yang
- Department of Cardial Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University Luoyang China
| | - Zhongrui Li
- Department of Nursing, Luoyang Central Hospital Affiliated to Zhengzhou University No. 288 Middle Zhongzhou Road Luoyang 471000 China +86-379-6389201
| | - Lei Zhang
- Department of Cardial Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University Luoyang China
| | - Xiaoshan Wu
- Department of Cardial Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University Luoyang China
| | - Qixin Kang
- Department of Cardial Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University Luoyang China
| | - Li Li
- Department of Nursing, Luoyang Central Hospital Affiliated to Zhengzhou University No. 288 Middle Zhongzhou Road Luoyang 471000 China +86-379-6389201
| |
Collapse
|