1
|
Guo L, Zhang P, Zhang M, Liang P, Zhou S. LncRNA AGAP2-AS1 stabilizes ATG9A to promote autophagy in endothelial cells - Implications for burn wound healing. Exp Cell Res 2024; 443:114310. [PMID: 39481796 DOI: 10.1016/j.yexcr.2024.114310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 10/24/2024] [Accepted: 10/27/2024] [Indexed: 11/02/2024]
Abstract
Deep second- or mixed-degree burn lesions are difficult to heal due to the impaired dermis supporting of epidermis renewal and nutrition delivery. Early dermis debridement and preservation speed healing and enhance results, emphasizing the need of knowing processes that promote burn-denatured dermis recovery, notably endothelial cell angiogenesis and autophagy. Integrative bioinformatics investigations identified AGAP2-AS1 as a highly elevated lncRNA in burn tissues. Pearson's correlation study connected AGAP2-AS1 to 112 differently co-expressed protein-coding genes involved in burn healing processes such cell cycle and TGF-beta receptor signaling. Experimental validation showed that heat damage elevated AGAP2-AS1 in HUVECs and HDMECs. Functionally, AGAP2-AS1 overexpression in heat-denatured HUVECs and HDMECs increased cell survival, migration, invasion, and angiogenesis. In addition, AGAP2-AS1 overexpression increased endothelial cell autophagy. Additional investigation showed AGAP2-AS1's association with ATG9A, stabilizing it. Post-heat damage, ATG9A knockdown drastically reduced HUVEC and HDMEC survival, migration, invasion, angiogenesis, and autophagy. More notably, ATG9A knockdown drastically reduced the benefits of AGAP2-AS1 overexpression on endothelial cell functions and autophagy. The positive association between AGAP2-AS1 and ATG9A expression in burn tissue samples highlights their crucial roles in endothelial cell response to heat injury, indicating that targeting this axis may aid burn wound healing. The research found that lncRNA AGAP2-AS1 stabilizes ATG9A and promotes autophagy in endothelial cells. These results imply that targeting the AGAP2-AS1/ATG9A axis may improve angiogenesis and tissue regeneration in burn injuries, revealing burn wound healing molecular pathways.
Collapse
Affiliation(s)
- Le Guo
- Department of Burns and Reconstructive Surgery, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Pihong Zhang
- Department of Burns and Reconstructive Surgery, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Minghua Zhang
- Department of Burns and Reconstructive Surgery, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Pengfei Liang
- Department of Burns and Reconstructive Surgery, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Situo Zhou
- Department of Burns and Reconstructive Surgery, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
2
|
Aihemaiti G, Song N, Luo J, Liu F, Toyizibai J, Adili N, Liu C, Ji W, Yang YN, Li X. Targeting lncRNA MALAT1: A Promising Approach to Overcome Metabolic Syndrome. Int J Endocrinol 2024; 2024:1821252. [PMID: 39502508 PMCID: PMC11535177 DOI: 10.1155/2024/1821252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 08/20/2024] [Accepted: 10/08/2024] [Indexed: 11/08/2024] Open
Abstract
Metabolic syndrome (MetS) is a collection of metabolic abnormalities including insulin resistance, atherogenic dyslipidemia, central obesity, and hypertension. Recently, long noncoding RNAs (lncRNAs) have emerged as pivotal regulators of metabolic balance, influencing the genes associated with MetS. Although the prevalence of insulin resistance is rising, leading to an increased risk of type 2 diabetes mellitus (T2DM) and its vascular complications, there is still a notable gap in understanding the role of lncRNAs in the context of clinical diabetes. Among lncRNAs, lung adenocarcinoma metastasis-associated transcript 1 (MALAT1) has been identified as a significant regulator of metabolism-related disorders, including T2DM and cardiovascular disease (CVD). This review explores the mechanism of lncRNA MALAT1 and suggests that targeting it could offer a promising strategy to combat MetS, thereby enhancing the prognosis of MetS.
Collapse
Affiliation(s)
- Gulandanmu Aihemaiti
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Ning Song
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Junyi Luo
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Fen Liu
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Clinical Medical Research Institute of First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Jianaerguli Toyizibai
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Niyaziaili Adili
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Chang Liu
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Wei Ji
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yi-Ning Yang
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Clinical Medical Research Institute of First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Xiaomei Li
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Clinical Medical Research Institute of First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
3
|
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
|
4
|
Zhang P, Gong S, Li S, Yuan Z. PVT1 alleviates hypoxia-induced endothelial apoptosis by enhancing autophagy via the miR-15b-5p/ATG14 and miR-424-5p/ATG14 axis. Biochem Biophys Res Commun 2023; 671:1-9. [PMID: 37290278 DOI: 10.1016/j.bbrc.2023.06.001] [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: 05/05/2023] [Accepted: 06/01/2023] [Indexed: 06/10/2023]
Abstract
Endothelial dysfunction plays a crucial role in the pathogenesis of vascular disease. Long noncoding RNA (lncRNA) and microRNA (miRNA) play important roles in various cellular processes and are involved in several vascular endothelial cells (VECs) biological processes, including cell growth, migration, autophagy, and apoptosis. The functions of plasmacytoma variant translocation 1 (PVT1) in VECs have been progressively investigated in recent years, mainly with regard to proliferation and migration of endothelial cells (ECs). However, the mechanism underlying the regulation of autophagy and apoptosis in human umbilical vein endothelial cells (HUVEC) by PVT1 remains unclear. The present study showed that PVT1 knockdown accelerated apoptosis induced by oxygen and glucose deprivation (OGD) through suppression of cellular autophagy. Bioinformatic prediction of PVT1 target miRNAs revealed that PVT1 interacts with miR-15b-5p and miR-424-5p. The study further showed that miR-15b-5p and miR-424-5p inhibit the functions of autophagy related 14 (ATG14) and suppress cellular autophagy. The results showed that PVT1 can function as a competing endogenous RNA (ceRNA) of miR-15b-5p and miR-424-5p and promote cellular autophagy by competitive binding, which down-regulates apoptosis. The results showed that PVT1 can function as a competing endogenous RNA (ceRNA) of miR-15b-5p and miR-424-5p and promote cellular autophagy through competitive binding, which down-regulates apoptosis. The study provides insight into a novel therapeutic target that may be explored in the future for the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Ping Zhang
- Hengyang Medical College, University of South China, 421001, Hengyang, Hunan, China; The Brain Science Center, Beijing Institute of Basic Medical Sciences, 100850, Beijing, China
| | - Shenghui Gong
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, 100850, Beijing, China
| | - Shuoshuo Li
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, 100850, Beijing, China; School of Life Science, Beijing University of Chinese Medicine, 100105, Beijing, China.
| | - Zengqiang Yuan
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, 100850, Beijing, China.
| |
Collapse
|
5
|
Sun Y, Wang T, Lv Y, Li J, Jiang X, Jiang J, Zhang D, Bian W, Zhang C. MALAT1 promotes platelet activity and thrombus formation through PI3k/Akt/GSK-3β signalling pathway. Stroke Vasc Neurol 2023; 8:181-192. [PMID: 36241224 PMCID: PMC10359792 DOI: 10.1136/svn-2022-001498] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 09/21/2022] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Ischaemic stroke and other cardiovascular illnesses are characterised by abnormalities in the processes of thrombosis and haemostasis, which rely on platelet activity. In platelets, a wide variety of microRNAs (long non-coding RNA, lncRNAs) is found. Due to the absence of nuclear DNA in platelets, lncRNAs may serve as critical post-transcriptional regulators of platelet activities. However, research into the roles of lncRNAs in platelets is limited. OBJECTIVE The purpose of this study is to learn more about the molecular mechanism by which MALAT1 affects platelet activity and thrombus formation. METHODS/RESULTS The CD34+ megakaryocytes used in this research as an in vitro model for human megakaryocytes and platelets. Cell adhesion and spreading are enhanced in the absence and presence of agonists in CD34+ megakaryocytes subjected to MALAT1 knockdown (KD). The adhesion and activity of platelet-like particles produced by MALAT1 KD cells are significantly enhanced at rest and after thrombin activation. Thrombus development on a collagen matrix is also greatly enhanced in the microfluidic whole-blood perfusion model: platelets lacking MALAT1 exhibit elevated accumulation, distributing area and activity. In addition, MALAT1-deficient mice bleed less and form a stable occlusive thrombus more quickly than wild-type mice. PTEN and PDK1 regulated the activity of MALAT1 in platelets to carry out its PI3k/Akt/GSK-3β signalling pathway-related function. CONCLUSION The suppression of MALAT1 expression significantly increases platelet adhesion, spreading, platelet activity, and thrombus formation. lncRNAs may constitute a unique class of platelet function modulators.
Collapse
Affiliation(s)
- Yeying Sun
- College of Pharmacy, Binzhou Medical University, Yantai, Shandong, China
| | - Tao Wang
- College of Pharmacy, Binzhou Medical University, Yantai, Shandong, China
| | - Yan Lv
- College of Life Sciences, Yantai University, Yantai, Shandong, China
| | - Jiahua Li
- College of Pharmacy, Binzhou Medical University, Yantai, Shandong, China
| | - Xiaoli Jiang
- College of Pharmacy, Binzhou Medical University, Yantai, Shandong, China
| | - Jing Jiang
- College of Pharmacy, Binzhou Medical University, Yantai, Shandong, China
| | - Daolai Zhang
- College of Pharmacy, Binzhou Medical University, Yantai, Shandong, China
| | - Weihua Bian
- College of Pharmacy, Binzhou Medical University, Yantai, Shandong, China
| | - Chunxiang Zhang
- College of Pharmacy, Binzhou Medical University, Yantai, Shandong, China
- Department of Cardiology, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
6
|
Tian W, Zhang T, Wang X, Zhang J, Ju J, Xu H. Global research trends in atherosclerosis: A bibliometric and visualized study. Front Cardiovasc Med 2022; 9:956482. [PMID: 36082127 PMCID: PMC9445883 DOI: 10.3389/fcvm.2022.956482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 08/03/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundIncreasing evidence has spurred a considerable evolution of concepts related to atherosclerosis, prompting the need to provide a comprehensive view of the growing literature. By retrieving publications in the Web of Science Core Collection (WoSCC) of Clarivate Analytics, we conducted a bibliometric analysis of the scientific literature on atherosclerosis to describe the research landscape.MethodsA search was conducted of the WoSCC for articles and reviews serving exclusively as a source of information on atherosclerosis published between 2012 and 2022. Microsoft Excel 2019 was used to chart the annual productivity of research relevant to atherosclerosis. Through CiteSpace and VOSviewer, the most prolific countries or regions, authors, journals, and resource-, intellectual-, and knowledge-sharing in atherosclerosis research, as well as co-citation analysis of references and keywords, were analyzed.ResultsA total of 20,014 publications were retrieved. In terms of publications, the United States remains the most productive country (6,390, 31,93%). The most publications have been contributed by Johns Hopkins Univ (730, 3.65%). ALVARO ALONSO produced the most published works (171, 0.85%). With a betweenness centrality of 0.17, ERIN D MICHOS was the most influential author. The most prolific journal was identified as Atherosclerosis (893, 4.46%). Circulation received the most co-citations (14,939, 2.79%). Keywords with the ongoing strong citation bursts were “nucleotide-binding oligomerization (NOD), Leucine-rich repeat (LRR)-containing protein (NLRP3) inflammasome,” “short-chain fatty acids (SCFAs),” “exosome,” and “homeostasis,” etc.ConclusionThe research on atherosclerosis is driven mostly by North America and Europe. Intensive research has focused on the link between inflammation and atherosclerosis, as well as its complications. Specifically, the NLRP3 inflammasome, interleukin-1β, gut microbiota and SCFAs, exosome, long non-coding RNAs, autophagy, and cellular senescence were described to be hot issues in the field.
Collapse
Affiliation(s)
- Wende Tian
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tai Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
- Department of Gastroenterology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xinyi Wang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jie Zhang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Jianqing Ju
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Jianqing Ju,
| | - Hao Xu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Hao Xu,
| |
Collapse
|
7
|
Long non-coding RNA MALAT1 affects intermittent hypoxia-induced endothelial injury by regulating miR-142-3p/HMGB1. Sleep Breath 2022; 26:2015-2024. [DOI: 10.1007/s11325-021-02545-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 11/27/2022]
|
8
|
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: 102] [Impact Index Per Article: 25.5] [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
|
9
|
Li H, Pan Z, Chen Q, Yang Z, Zhang D. SMILR Aggravates the Progression of Atherosclerosis by Sponging miR-10b-3p to Regulate KLF5 Expression. Inflammation 2021; 43:1620-1633. [PMID: 32367412 DOI: 10.1007/s10753-020-01237-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Over the past few decades, long noncoding RNAs (lncRNAs) have been widely accepted to be involved in various diseases, and smooth muscle enriched long noncoding RNA (SMILR) was reported to participate in the proliferation of vascular smooth muscle cells (VSMCs). Nevertheless, the molecular mechanisms of SMILR in atherosclerosis (AS) have not been fully explored. In this study, VSMCs and human mononuclear cells (U937) treated with oxidized low-density lipoprotein (ox-LDL) were used as cell models of AS. We found that the expression of SMILR was upregulated in the serum of AS patients and ox-LDL-induced AS cell models. SMILR knockdown inhibited cell proliferation while increasing cell apoptosis in the AS cell models. In addition, SMILR acted as a sponge for miR-10b-3p, and miR-10b-3p counteracted SMILR-mediated regulation of AS. Moreover, we confirmed that miR-10b-3p could bind with KLF5, and SMILR regulated KLF5 expression by competitively binding miR-10b-3p. Furthermore, miR-10b-3p modulated cell proliferation and apoptosis in AS by targeting KLF5. Finally, miR-10b-3p regulated AS progression in vivo by targeting KLF5. Overall, our study demonstrated that SMILR participated in the progression of AS by targeting the miR-10b-3p/KLF5 axis, which may provide some clues for future studies of AS.
Collapse
Affiliation(s)
- Huaqing Li
- Department of Vascular Surgery, Minhang Hospital, Fudan University, No. 39 Xinling Road, Minhang District, 201199, Shanghai, China
| | - Zhiyu Pan
- Department of Vascular Surgery, Minhang Hospital, Fudan University, No. 39 Xinling Road, Minhang District, 201199, Shanghai, China.
| | - Qian Chen
- Department of Vascular Surgery, Minhang Hospital, Fudan University, No. 39 Xinling Road, Minhang District, 201199, Shanghai, China
| | - Zhen Yang
- Department of Vascular Surgery, Minhang Hospital, Fudan University, No. 39 Xinling Road, Minhang District, 201199, Shanghai, China
| | - Dongbing Zhang
- Department of Vascular Surgery, Minhang Hospital, Fudan University, No. 39 Xinling Road, Minhang District, 201199, Shanghai, China
| |
Collapse
|
10
|
Liang T, Wang Y, Jiao Y, Cong S, Jiang X, Dong L, Zhang G, Xiao D. LncRNA MALAT1 Accelerates Cervical Carcinoma Proliferation by Suppressing miR-124 Expression in Cervical Tumor Cells. JOURNAL OF ONCOLOGY 2021; 2021:8836078. [PMID: 34221014 PMCID: PMC8221887 DOI: 10.1155/2021/8836078] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 03/31/2021] [Accepted: 05/12/2021] [Indexed: 12/20/2022]
Abstract
Emerging studies have clarified the critical role of LncRNA MALAT1 in various pathological progressions. Here, we identified its positive relationship with cervical carcinoma proliferation. Cervical carcinoma has been considered as one of the most malignant tumors among female. Thus, our study was designed to investigate the underlying mechanism of LncRNA MALAT1 on cervical tumor cell proliferation. We observed that miR-124 was the potential target of LncRNA MALAT1 in cervical tumor cell lines (Hela, C-33A, Caski, and SiHa), the expression level of which is negatively correlated with LncRNA MALAT1 in cervical tumor cells, tissues of cervical patients, and mice. Gain- or loss-of-function analyses in cervical tumor cells have further verified the regulatory role of MALAT1 on miR-124. Additionally, the proliferation of cervical carcinoma was inhibited by miR-124 overexpression, whereas it was blocked by LV-MALAT1 transfection. In vivo assays, overexpression of miR-124, or knockdown of MALAT1 exhibited beneficial effects on tumor weight, size, and volume, together with elevating the survival rate, tightly related with the progression of cervical cancer. In conclusion, LncRNA MALAT1 disabled the effects of miR-124 as an inhibitory sponge, accelerating the progression of cervical carcinoma.
Collapse
Affiliation(s)
- Tian Liang
- Department of Obstetrics and Gynaecology, The 1st Affiliated Hospital of Harbin Medical University, 150001 Harbin, China
| | - Yuchen Wang
- Department of Psychiatry, Qiqihar Medical University, 161006 Qiqihar, China
| | - Yu Jiao
- Department of Psychiatry, Qiqihar Medical University, 161006 Qiqihar, China
| | - Shanshan Cong
- Department of Obstetrics and Gynaecology, The 1st Affiliated Hospital of Harbin Medical University, 150001 Harbin, China
| | - Xinyan Jiang
- Department of Obstetrics and Gynaecology, The 1st Affiliated Hospital of Harbin Medical University, 150001 Harbin, China
| | - Lina Dong
- Department of Obstetrics and Gynaecology, The 1st Affiliated Hospital of Harbin Medical University, 150001 Harbin, China
| | - Guangmei Zhang
- Department of Obstetrics and Gynaecology, The 1st Affiliated Hospital of Harbin Medical University, 150001 Harbin, China
| | - Dan Xiao
- Department of Psychiatry, Qiqihar Medical University, 161006 Qiqihar, China
| |
Collapse
|
11
|
Jin L, Hong N, Ai X, Wang J, Li Z, Han Z, Zhang Q, Yu Y, Sun K. LncRNAs as Therapeutic Targets for Autophagy-involved Cardiovascular Diseases: A Review of Molecular Mechanism and T herapy Strategy. Curr Med Chem 2021; 28:1796-1814. [PMID: 32196441 DOI: 10.2174/0929867327666200320161835] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/12/2020] [Accepted: 03/06/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Cardiovascular diseases (CVDs) remain the leading cause of death worldwide. The concept of precision medicine in CVD therapy today requires the incorporation of individual genetic and environmental variability to achieve personalized disease prevention and tailored treatment. Autophagy, an evolutionarily conserved intracellular degradation process, has been demonstrated to be essential in the pathogenesis of various CVDs. Nonetheless, there have been no effective treatments for autophagy- involved CVDs. Long noncoding RNAs (lncRNAs) are noncoding RNA sequences that play versatile roles in autophagy regulation, but much needs to be explored about the relationship between lncRNAs and autophagy-involved CVDs. SUMMARY Increasing evidence has shown that lncRNAs contribute considerably to modulate autophagy in the context of CVDs. In this review, we first summarize the current knowledge of the role lncRNAs play in cardiovascular autophagy and autophagy-involved CVDs. Then, recent developments of antisense oligonucleotides (ASOs) designed to target lncRNAs to specifically modulate autophagy in diseased hearts and vessels are discussed, focusing primarily on structure-activity relationships of distinct chemical modifications and relevant clinical trials. PERSPECTIVE ASOs are promising in cardiovascular drug innovation. We hope that future studies of lncRNA-based therapies would overcome existing technical limitations and help people who suffer from autophagy-involved CVDs.
Collapse
Affiliation(s)
- Lihui Jin
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Nanchao Hong
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Xuefeng Ai
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Jing Wang
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Zhuoyan Li
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Zhenyuan Han
- Department of Oral Pathology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China
| | - Qi Zhang
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Yu Yu
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Kun Sun
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| |
Collapse
|
12
|
PRKAR1B-AS2 Long Noncoding RNA Promotes Tumorigenesis, Survival, and Chemoresistance via the PI3K/AKT/mTOR Pathway. Int J Mol Sci 2021; 22:ijms22041882. [PMID: 33668685 PMCID: PMC7918312 DOI: 10.3390/ijms22041882] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/23/2021] [Accepted: 01/28/2021] [Indexed: 12/19/2022] Open
Abstract
Many long noncoding RNAs have been implicated in tumorigenesis and chemoresistance; however, the underlying mechanisms are not well understood. We investigated the role of PRKAR1B-AS2 long noncoding RNA in ovarian cancer (OC) and chemoresistance and identified potential downstream molecular circuitry underlying its action. Analysis of The Cancer Genome Atlas OC dataset, in vitro experiments, proteomic analysis, and a xenograft OC mouse model were implemented. Our findings indicated that overexpression of PRKAR1B-AS2 is negatively correlated with overall survival in OC patients. Furthermore, PRKAR1B-AS2 knockdown-attenuated proliferation, migration, and invasion of OC cells and ameliorated cisplatin and alpelisib resistance in vitro. In proteomic analysis, silencing PRKAR1B-AS2 markedly inhibited protein expression of PI3K-110α and abrogated the phosphorylation of PDK1, AKT, and mTOR, with no significant effect on PTEN. The RNA immunoprecipitation detected a physical interaction between PRKAR1B-AS2 and PI3K-110α. Moreover, PRKAR1B-AS2 knockdown by systemic administration of 1,2-dioleoyl-sn-glycero-3-phosphatidylcholine nanoparticles loaded with PRKAR1B-AS2–specific small interfering RNA enhanced cisplatin sensitivity in a xenograft OC mouse model. In conclusion, PRKAR1B-AS2 promotes tumor growth and confers chemoresistance by modulating the PI3K/AKT/mTOR pathway. Thus, targeting PRKAR1B-AS2 may represent a novel therapeutic approach for the treatment of OC patients.
Collapse
|
13
|
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
|
14
|
Yang J, Lin X, Wang L, Sun T, Zhao Q, Ma Q, Zhou Y. LncRNA MALAT1 Enhances ox-LDL-Induced Autophagy through the SIRT1/MAPK/NF-κB Pathway in Macrophages. Curr Vasc Pharmacol 2020; 18:652-662. [PMID: 32183682 DOI: 10.2174/1570161118666200317153124] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 02/04/2020] [Accepted: 02/25/2020] [Indexed: 01/07/2023]
Abstract
Atherosclerosis is the main cause of cardiovascular and cerebrovascular diseases. In
advanced atherosclerotic plaque, macrophage apoptosis coupled with inflammatory cytokine secretion
promotes the formation of necrotic cores. It has also been demonstrated that the long-noncoding Ribonucleic
Acid (lnc RNA) metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), with its
potent function on gene transcription modulation, maintains oxidized low-density lipoprotein (ox-LDL)-
induced macrophage autophagy (i.e., helps with cholesterol efflux). It also showed that MALAT1 activated
Sirtuin 1 (SIRT1), which subsequently inhibited the mitogen-activated protein kinase (MAPK)
and nuclear factor kappa-B (NF-κB) signaling pathways. ox-LDL has been used to incubate human
myeloid leukemia mononuclear cells (THP-1)-derived macrophages to establish an in vitro foam cell
model. Quantitative reverse-transcription polymerase chain reaction and Western blot analyses confirmed
the increased expression level of MALAT1 and the autophagy-related protein Microtubuleassociated
protein light chain 3 (LC-3), beclin-1. The small interfering RNA study showed a significant
decrease in autophagy activity and an increase in apoptotic rate when knocking down MALAT1. Further
study demonstrated that MALAT1 inhibited the expression of MAPK and NF-κB (p65) by upregulating
SIRT1.
Collapse
Affiliation(s)
- Jiaqi Yang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Xuze Lin
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Liangshan Wang
- Center for Cardiac Intensive Care, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Tienan Sun
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Qi Zhao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Qian Ma
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Yujie Zhou
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| |
Collapse
|
15
|
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
|
16
|
Luo L, Wang Y, Hu P, Wu J. Long Non-Coding RNA Metastasis Associated Lung Adenocarcinoma Transcript 1 (MALAT1) Promotes Hypertension by Modulating the Hsa-miR-124-3p/Nuclear Receptor Subfamily 3, Group C, Member 2 (NR3C2) and Hsa-miR-135a-5p/NR3C2 Axis. Med Sci Monit 2020; 26:e920478. [PMID: 32222724 PMCID: PMC7139186 DOI: 10.12659/msm.920478] [Citation(s) in RCA: 6] [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 This study was designed to investigate the role of long non-coding RNA (lncRNA) metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) in the proliferation as well as apoptosis of human umbilical vein endothelial cells (HUVECs), to offer a basis for therapy of hypertension. Material/Methods The lncRNA MALAT1 expression, hsa-miR-124-3p, hsa-miR-135a-5p, hsa-miR-135b-5p, and hsa-miR-455-5p in plasma were measured from 230 patients with hypertension and 230 non-hypertensive controls. The mechanism for lncRNA MALAT1 modulating the proliferation and apoptosis of HUVECs was explored by cell transfection, Cell Counting Kit-8 (CCK-8), quantitative real-time polymerase chain reaction (qRT-PCR), western blot, and dual-luciferase reporter assays. Results The expression of hsa-miR-124-3p and hsa-miR-135a-5p was reduced and the expression of lncRNA MALAT1 was increased in the plasma of hypertensive patients. Moreover, the plasma levels of hsa-miR-124-3p and hsa-miR-135a-5p of hypertensive patients were negatively correlated with lncRNA MALAT1 (r=−0.64, −0.72; P<0.01, P<0.01, respectively). The level of nuclear receptor subfamily 3, group C, member 2 (NR3C2) protein was negatively correlated with hsa-miR-124-3p and hsa-miR-135a-5p (r=−0.74, −0.84; P<0.01, P<0.01, respectively). The proliferation of HUVECs was inhibited after the inhibition of MALAT. Additionally, after knocking down MALAT, the levels of hsa-miR-124-3p and hsa-miR-135a-5p in HUVECs were markedly increased, while the expression level of NR3C2 protein was decreased. The apoptotic rate of HUVECs after the transfection of MALAT1 small interfering RNA (si-MALAT1) (3.64±0.21%) was significantly reduced compared to that of transfected si-MALAT1 no template control (NC) (3.76±0.19%) and the control group (10.51±1.24%). Conclusions LncRNA MALAT1 regulates proliferation and apoptosis of HUVECs through the hsa-miR-124-3p/NR3C2 and/or hsa-miR-135a-5p/NR3C2 axis.
Collapse
Affiliation(s)
- Liju Luo
- Department of Geratology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China (mainland)
| | - Yu Wang
- Department of Cardiology, The Affiliated Yueqing Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China (mainland)
| | - Pengfei Hu
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China (mainland)
| | - Jiale Wu
- Department of Geratology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China (mainland)
| |
Collapse
|
17
|
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
|
18
|
Ma J, Xiao Y, Tian B, Chen S, Zhang B, Wu J, Wu Z, Li X, Tang J, Yang D, Zhou Y, Wang H, Su M, Wang W. Long noncoding RNA lnc-ABCA12-3 promotes cell migration, invasion, and proliferation by regulating fibronectin 1 in esophageal squamous cell carcinoma. J Cell Biochem 2019; 121:1374-1387. [PMID: 31512786 DOI: 10.1002/jcb.29373] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 08/20/2019] [Indexed: 12/16/2022]
Abstract
Long noncoding RNAs (lncRNAs) have been shown to play important roles in human cancers, including esophageal squamous cell carcinoma (ESCC). We previously demonstrated that a novel lncRNA, lnc-ABCA12-3, was overexpressed in ESCC tissues. However, the exact function of lnc-ABCA12-3 is unknown. In the current study, we aimed to evaluate the expression of lnc-ABCA12-3 in ESCC and to explore the potential mechanism of lnc-ABCA12-3 in cell migration, invasion, and proliferation. We showed that lnc-ABCA12-3 was upregulated in ESCC tumor tissues and cell lines. The increased expression of lnc-ABCA12-3 was positively associated with advanced tumor-node-metastasis stages and poor prognosis. The knockdown of lnc-ABCA12-3 inhibited the cell migration, invasion, and proliferation abilities of KYSE-510 and Eca-109 cells. We also found that fibronectin 1 (FN1) was upregulated in ESCC tumor tissues. The expression of FN1 messenger RNA was positively correlated with the expression of lnc-ABCA12-3 in ESCC tumor tissues. After lnc-ABCA12-3 knockdown, the expression of FN1 was downregulated. In addition, the overexpression of FN1 restored the abilities of cell migration, invasion and proliferation in Eca-109 cells. Further studies indicated that lnc-ABCA12-3 acted as a competing endogenous RNA for miR-200b-3p to regulate FN1 expression. In conclusion, these results suggest that lnc-ABCA12-3 is a novel oncogene in tumorigenesis and that its high expression is related to a poor prognosis for patients with ESCC. lnc-ABCA12-3 promotes cell migration, invasion, and proliferation via the regulation of FN1 in ESCC. Our data suggest that lnc-ABCA12-3 might serve as a potential prognostic biomarker and therapeutic target for ESCC.
Collapse
Affiliation(s)
- Junliang Ma
- The Second Department of Thoracic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Yuhang Xiao
- Department of Pharmacy, Xiangya Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Bo Tian
- The Second Department of Thoracic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Shaolin Chen
- Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Baihua Zhang
- The Second Department of Thoracic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jie Wu
- The Second Department of Thoracic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Zhining Wu
- The Second Department of Thoracic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xu Li
- The Second Department of Thoracic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jinming Tang
- The Second Department of Thoracic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Desong Yang
- The Second Department of Thoracic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yong Zhou
- The Second Department of Thoracic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Hui Wang
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Min Su
- The Second Department of Thoracic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Wenxiang Wang
- The Second Department of Thoracic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| |
Collapse
|
19
|
Ma Z, Zhang J, Xu X, Qu Y, Dong H, Dang J, Huo Z, Xu G. LncRNA expression profile during autophagy and Malat1 function in macrophages. PLoS One 2019; 14:e0221104. [PMID: 31425535 PMCID: PMC6699732 DOI: 10.1371/journal.pone.0221104] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 07/30/2019] [Indexed: 01/05/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) are a class of functional non-coding transcripts that are longer than 200 nt and regulate gene expression via diverse mechanisms in eukaryotes. In fact, they have emerged as critical epigenetic and transcriptional regulators of autophagy in mammals in response to various stressors. Autophagy not only plays a crucial role in maintaining cellular homeostasis, but it is also essential to immunity, targets intracellular pathogens for degradation, modulates inflammation, and participates in adaptive immune responses. However, the expression profile of lncRNA and its role in regulating autophagy in macrophages have been poorly defined. Here, we used transcriptomic and bioinformatics to analysis LncRNA expression profile during autophagy and functional studies to evaluate the function of the metastasis-associated lung adenocarcinoma transcript-1 (Malat1) lncRNA in macrophages. A total of 1112 putative lncRNAs (240 novel lncRNAs) were identified, including 831 large intergenic, 129 intronic, and 152 anti-sense lncRNA, of which 59 differentially expressed transcripts exhibited a greater than 1.5-fold change under different conditions. The interaction of Malat1 lncRNA with microRNA (mir)-23-3p and lysosomal-associated membrane protein 1 (Lamp1) was found, Malat1 releases inhibition of Lamp1 expression in macrophages through competitive adsorption of mir-23-3p. The results of this study provide a better understanding of lncRNA function in macrophages and a basis for further investigation into the roles and mechanisms of ncRNA in immunology, particularly the functions of Malat1 and mir-23-3p in the pathogenesis of macrophages.
Collapse
Affiliation(s)
- Zhanbing Ma
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan, China
- Department of Medical Genetic and Cell Biology, College of Basic Medicine, Ningxia Medical University, Yinchuan, China
- Key Laboratory of Fertility Preservation and Maintenance (Ningxia Medical University), Ministry of Education, Yinchuan, China
| | - Jing Zhang
- Department of Medical Genetic and Cell Biology, College of Basic Medicine, Ningxia Medical University, Yinchuan, China
- Key Laboratory of Fertility Preservation and Maintenance (Ningxia Medical University), Ministry of Education, Yinchuan, China
| | - Xiangrong Xu
- Department of Medical Laboratory, College of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Yuliang Qu
- Department of Medical Laboratory, College of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Hui Dong
- Key Laboratory of Fertility Preservation and Maintenance (Ningxia Medical University), Ministry of Education, Yinchuan, China
| | - Jie Dang
- Department of Medical Genetic and Cell Biology, College of Basic Medicine, Ningxia Medical University, Yinchuan, China
- Key Laboratory of Fertility Preservation and Maintenance (Ningxia Medical University), Ministry of Education, Yinchuan, China
| | - Zhenghao Huo
- Department of Medical Genetic and Cell Biology, College of Basic Medicine, Ningxia Medical University, Yinchuan, China
- Key Laboratory of Fertility Preservation and Maintenance (Ningxia Medical University), Ministry of Education, Yinchuan, China
| | - Guangxian Xu
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan, China
- Department of Medical Laboratory, College of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| |
Collapse
|
20
|
Barangi S, Hayes AW, Reiter R, Karimi G. The therapeutic role of long non-coding RNAs in human diseases: A focus on the recent insights into autophagy. Pharmacol Res 2019; 142:22-29. [PMID: 30742900 DOI: 10.1016/j.phrs.2019.02.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 02/06/2019] [Accepted: 02/07/2019] [Indexed: 12/15/2022]
Abstract
Long non-coding RNA (lncRNA) is a class of non-coding RNA with ≥200 nucleotides in length which are involved as critical regulators in various cellular processes. LncRNAs contribute to the development and progression of many human diseases. Autophagy is a key catabolic process which helps to maintain the cellular homeostasis through the decay of damaged or unwanted proteins and dysfunctional cytoplasmic organelles. The impairment of the autophagy process has been described in numerous diseases. The autophagy possess can have either a protective or a detrimental role in cells depending on its activation status and other cellular conditions. LncRNAs have been shown to have an important function in the regulation of important biological processes such as autophagy. The relationship between lncRNAs and autophagy has been shown to be involved in the progression and possibly in the prevention of many diseases. In this review, recent findings on the regulatory roles of lncRNAs in the cell autophagy pathway, as well as their relevance to different diseases such as cardiovascular disease, cerebral ischemic stroke and cancer are highlighted.
Collapse
Affiliation(s)
- Samira Barangi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - A Wallace Hayes
- University of South Florida College of Public Health, USA; Michigan State University, East Lansing, MI, USA
| | - Russel Reiter
- University of Texas, Health Science Center at San Antonio, Department of Cellular and Structural Biology, USA
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Pharmaceutical Research Centre, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|