101
|
The role of long non-coding RNA SNHG12 in neuroprotection following cerebral ischemic injury. Neuroreport 2019; 30:945-952. [DOI: 10.1097/wnr.0000000000001308] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
102
|
Wang Y, Pan WY, Ge JS, Wang XD, Chen W, Luo X, Wang YL. A review of the relationship between long noncoding RNA and post-stroke injury repair. J Int Med Res 2019; 47:4619-4624. [PMID: 31526155 PMCID: PMC6833389 DOI: 10.1177/0300060519867493] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 07/12/2019] [Indexed: 11/15/2022] Open
Abstract
Stroke is a cerebrovascular circulation disorder with sudden onset, which causes disorder of ion balance, inflammation, and acidosis, and that in turn induces ischemia-reperfusion injury, influencing the prognosis of stroke patients. Long noncoding RNAs (lncRNAs) are regulatory sequences involved at the transcriptional, post-transcriptional, and epigenetic levels, have high specific expression in the central nervous system, and effectively regulate the development of the central nervous system and progression of diseases. Stroke induces changes in the expression of many lncRNAs. Therefore, lncRNAs play an important role in the complex pathological process of stroke. Exploring lncRNA could facilitate a comprehensive understanding of the pathological mechanism of stroke and the post-injury molecular regulatory network. However, there are few reports on the role of lncRNA in the pathological development of stroke. In the present review, we discuss the association of lncRNA with post-stroke injury repair.
Collapse
Affiliation(s)
- Yao Wang
- Department of Rehabilitation Medicine, Shenzhen Dapeng New District Nan’ao People’s Hospital, Shenzhen, China
| | - Wei-Yi Pan
- Department of Rehabilitation Medicine, Shenzhen Dapeng New District Nan’ao People’s Hospital, Shenzhen, China
| | - Jun-Sheng Ge
- Department of Rehabilitation Medicine, Shenzhen Dapeng New District Nan’ao People’s Hospital, Shenzhen, China
| | - Xiao-Dong Wang
- Department of Rehabilitation, Shenzhen Second People’s Hospital, The First Affiliated Hospital, Shenzhen University School of Medicine, Shenzhen, China
| | - Wei Chen
- Kerry Rehabilitation Medicine Research Institute, Shenzhen, Guangdong, China
| | - Xun Luo
- Kerry Rehabilitation Medicine Research Institute, Shenzhen, Guangdong, China
| | - Yu-Long Wang
- Department of Rehabilitation, Shenzhen Second People’s Hospital, The First Affiliated Hospital, Shenzhen University School of Medicine, Shenzhen, China
| |
Collapse
|
103
|
Alishahi M, Ghaedrahmati F, Kolagar TA, Winlow W, Nikkar N, Farzaneh M, Khoshnam SE. Long non-coding RNAs and cell death following ischemic stroke. Metab Brain Dis 2019; 34:1243-1251. [PMID: 31055786 DOI: 10.1007/s11011-019-00423-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 04/21/2019] [Indexed: 01/02/2023]
Abstract
Stroke is a major cause of morbidity and mortality worldwide, and extensive efforts have focused on the improvement of therapeutic strategies to reduce cell death following ischemic stroke. Uncovering the cellular and molecular pathophysiological processes in ischemic stroke have been a top priority. Long noncoding RNAs (lncRNAs) are endogenous molecules that play key roles in the pathophysiology of cerebral ischemia, and involved in the neuronal cell death during ischemic stroke. In recent years, a bulk of aberrantly expressed lncRNAs have been screened out in ischemic stroke insulted animals. LncRNAs along with their targets could affect the genetic machinery at molecular levels, and exploring their functions and mechanisms may be a promising option for ischemic stroke treatment. In this review, we summarize the current knowledge for lncRNAs in ischemic stroke, focusing on the role of specific lncRNAs that may underlie cell death to find possible therapeutic targets.
Collapse
Affiliation(s)
- Masoumeh Alishahi
- Department of Biology, Tehran North Branch, Islamic Azad University, Tehran, Iran
| | - Farhoodeh Ghaedrahmati
- Department of Immunology, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - William Winlow
- Dipartimento di Biologia, Università degli Studi di Napoli, Federico II, Via Cintia 26, 80126, Naples, Italy
- Honorary Research Fellow, Institute of Ageing and Chronic Diseases, University of Liverpool, The APEX building, 6 West Derby Street, Liverpool, L7 8TX, UK
| | - Negin Nikkar
- Department of Biology, Faculty of Sciences, Alzahra University, Tehran, Iran
| | - Maryam Farzaneh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Seyed Esmaeil Khoshnam
- Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
104
|
Wu Q, Li T, Zhu D, Lv F, Qin X. Altered expression of long noncoding RNAs in peripheral blood mononuclear cells in patients with impaired leptomeningeal collaterals after acute anterior large vessel occlusions. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:523. [PMID: 31807505 DOI: 10.21037/atm.2019.10.02] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background In the event of acute ischemic stroke (AIS) due to anterior large vessel occlusion (aLVO), leptomeningeal collaterals (LMCs) status is a key factor to define the severity and functional prognosis of this disease. However, the extent of LMCs exhibits substantial variability among the patients, which is genetic determined. Long non-coding RNAs (lncRNAs) expression profiles in human peripheral blood have been found to be altered after AIS. But whether there are specific lncRNAs correlated with LMC status in aLVO has not yet been investigated. Methods Differential lncRNA expression panels in peripheral blood mononuclear cells (PBMCs) were assessed by microarray analysis and individual quantitative real-time polymerase chain reaction (RT-PCR) in three independent sets consist of 134 patients with aLVO and 73 healthy controls (HCs). LMCs Status in those patients was assessed based on baseline computed tomographic angiography (CTA). Results Microarray analysis showed 23 differentially expressed lncRNAs in patients with poor LMCs status. After independent validations by RT-PCR, lncRNA ENST00000422956 was found to be significantly downregulated in patients with poor LMCs status. Receiver-operating characteristic (ROC) analysis revealed the area under the ROC curve (AUC) for ENST00000422956 to predict poor LMCs status was 0.749. Moreover, ENST00000422956 expression level and baseline National Institutes of Health Stroke Scale (NIHSS) score were identified as independent predictors for impaired LMCs, and a significantly positive correlation was observed between ENST00000422956 expression level and LMCs status. Via cis-regulatory analysis, paired box 8 (Pax8) was identified as the target gene for ENST00000422956. Conclusions The dysregulated lncRNA ENST00000422956 in PBMCs was associated with impairment of LMCs in patients with aLVO, suggesting that measurement of circulatory lncRNAs might be included as possible biomarkers for evaluation of LMCs status in AIS. More importantly, this might be the foundation for understand the potential roles of lncRNAs in LMCs formation after ischemic stroke.
Collapse
Affiliation(s)
- Qisi Wu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Ting Li
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Dan Zhu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Fajin Lv
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xinyue Qin
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
105
|
Cai J, Shangguan S, Li G, Cai Y, Chen Y, Ma G, Miao Z, Liu L, Deng Y. Knockdown of lncRNA Gm11974 protect against cerebral ischemic reperfusion through miR-766-3p/NR3C2 axis. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:3847-3853. [PMID: 31556305 DOI: 10.1080/21691401.2019.1666859] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Jiangping Cai
- Department of Neurology, The First Hospital of Quanzhou Affiliated to Fujian Medical University, Quanzhou, People's Republic of China
| | - Shina Shangguan
- Department of Neurology, Shandong University/Affiliated Hospital of Shandong Medical College, Jinan City, China
| | - Guoqian Li
- Department of Neurology, The First Hospital of Quanzhou Affiliated to Fujian Medical University, Quanzhou, People's Republic of China
| | - Yazhen Cai
- Department of Neurology, The First Hospital of Quanzhou Affiliated to Fujian Medical University, Quanzhou, People's Republic of China
| | - Yuanjie Chen
- Department of Neurology, The First Hospital of Quanzhou Affiliated to Fujian Medical University, Quanzhou, People's Republic of China
| | - Gaoting Ma
- Departments of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
| | - Zhongrong Miao
- Departments of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
| | - Lian Liu
- Departments of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
| | - Yiming Deng
- Departments of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
106
|
Bonnet S, Boucherat O, Paulin R, Wu D, Hindmarch CCT, Archer SL, Song R, Moore JB, Provencher S, Zhang L, Uchida S. Clinical value of non-coding RNAs in cardiovascular, pulmonary, and muscle diseases. Am J Physiol Cell Physiol 2019; 318:C1-C28. [PMID: 31483703 DOI: 10.1152/ajpcell.00078.2019] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Although a majority of the mammalian genome is transcribed to RNA, mounting evidence indicates that only a minor proportion of these transcriptional products are actually translated into proteins. Since the discovery of the first non-coding RNA (ncRNA) in the 1980s, the field has gone on to recognize ncRNAs as important molecular regulators of RNA activity and protein function, knowledge of which has stimulated the expansion of a scientific field that quests to understand the role of ncRNAs in cellular physiology, tissue homeostasis, and human disease. Although our knowledge of these molecules has significantly improved over the years, we have limited understanding of their precise functions, protein interacting partners, and tissue-specific activities. Adding to this complexity, it remains unknown exactly how many ncRNAs there are in existence. The increased use of high-throughput transcriptomics techniques has rapidly expanded the list of ncRNAs, which now includes classical ncRNAs (e.g., ribosomal RNAs and transfer RNAs), microRNAs, and long ncRNAs. In addition, splicing by-products of protein-coding genes and ncRNAs, so-called circular RNAs, are now being investigated. Because there is substantial heterogeneity in the functions of ncRNAs, we have summarized the present state of knowledge regarding the functions of ncRNAs in heart, lungs, and skeletal muscle. This review highlights the pathophysiologic relevance of these ncRNAs in the context of human cardiovascular, pulmonary, and muscle diseases.
Collapse
Affiliation(s)
- Sébastien Bonnet
- Pulmonary Hypertension and Vascular Biology Research Group, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Department of Medicine, Université Laval, Quebec City, Quebec, Canada.,Department of Medicine, Université Laval, Quebec City, Quebec, Canada
| | - Olivier Boucherat
- Pulmonary Hypertension and Vascular Biology Research Group, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Department of Medicine, Université Laval, Quebec City, Quebec, Canada.,Department of Medicine, Université Laval, Quebec City, Quebec, Canada
| | - Roxane Paulin
- Pulmonary Hypertension and Vascular Biology Research Group, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Department of Medicine, Université Laval, Quebec City, Quebec, Canada.,Department of Medicine, Université Laval, Quebec City, Quebec, Canada
| | - Danchen Wu
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Charles C T Hindmarch
- Queen's Cardiopulmonary Unit, Translational Institute of Medicine, Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Stephen L Archer
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Rui Song
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Joseph B Moore
- Diabetes and Obesity Center, University of Louisville, Louisville, Kentucky.,The Christina Lee Brown Envirome Institute, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Steeve Provencher
- Pulmonary Hypertension and Vascular Biology Research Group, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Department of Medicine, Université Laval, Quebec City, Quebec, Canada.,Department of Medicine, Université Laval, Quebec City, Quebec, Canada
| | - Lubo Zhang
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Shizuka Uchida
- Diabetes and Obesity Center, University of Louisville, Louisville, Kentucky.,The Christina Lee Brown Envirome Institute, Department of Medicine, University of Louisville, Louisville, Kentucky.,Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky
| |
Collapse
|
107
|
Zou JB, Chai HB, Zhang XF, Guo DY, Tai J, Wang Y, Liang YL, Wang F, Cheng JX, Wang J, Shi YJ. Reconstruction of the lncRNA-miRNA-mRNA network based on competitive endogenous RNA reveal functional lncRNAs in Cerebral Infarction. Sci Rep 2019; 9:12176. [PMID: 31434962 PMCID: PMC6704173 DOI: 10.1038/s41598-019-48435-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 08/06/2019] [Indexed: 02/06/2023] Open
Abstract
Functioning as miRNA sponges, long non-coding RNA (lncRNA) exert its pharmacological action via regulating expression of protein-coding genes. However, the lncRNA-mediated ceRNA in cerebral Infarction (CI) remains unclear. In this study, the expression recordsets of mRNA, lncRNA and miRNA of CI samples were obtained from the NCBI GEO datasets separately. The differentially expressed lncRNAs (DELs), miRNAs (DEMis) and mRNAs (DEMs) were identified by limma package in R platform. A total of 267 DELs, 26 DEMis, and 760 DEMs were identified as differentially expressed profiles, with which we constructed the ceRNA network composed of DELs-DEMis-DEMs. Further, clusterProfiler package in R platform is employed for performing Gene Ontology (GO) and KEGG pathway analysis. An aberrant ceRNA network was constructed according to node degrees in CI, including 28 DELs, 19 DEMs and 12 DEMis, from which we extracted the core network, in which 9 nodes were recognized as kernel genes including Tspan3, Eif4a2, rno-miR-208a-3p, rno-miR-194-5p, Pdpn, H3f3b, Stat3, Cd63 and Sdc4. Finally, with the DELs-DEMis-DEMs ceRNA network provided above, we can improve our understanding of the pathogenesis of CI mediated by lncRNA.
Collapse
Affiliation(s)
- Jun-Bo Zou
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research,Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Hong-Bo Chai
- The first affiliated Hospital of Hunan University of Medicine, Huaihua, 410007, China
| | - Xiao-Fei Zhang
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research,Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Dong-Yan Guo
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research,Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Jia Tai
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research,Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Yu Wang
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research,Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Yu-Lin Liang
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research,Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Fang Wang
- Key laboratory of Modern Prepararation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330000, China
| | - Jiang-Xue Cheng
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research,Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Jing Wang
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research,Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Ya-Jun Shi
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research,Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang, 712046, China.
| |
Collapse
|
108
|
Wang Q, Liu X, Zhu R. Long Noncoding RNAs as Diagnostic and Therapeutic Targets for Ischemic Stroke. Curr Pharm Des 2019; 25:1115-1121. [PMID: 30919772 DOI: 10.2174/1381612825666190328112844] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 03/20/2019] [Indexed: 11/22/2022]
Abstract
LncRNAs (long non-coding RNAs) are endogenous molecules lacking protein-encoding capacity,
which have been identified as key regulators of ischemic stroke. Increasing evidence suggests that lncRNAs play
critical roles in several aspects of ischemic stroke, including atherosclerosis, dyslipidemia, hypertension, and
diabetes mellitus. Hence, lncRNAs may further broaden our understanding of stroke pathogenesis. Altered
lncRNA expression has been found in rodent focal cerebral ischemia models and oxygen–glucose deprived mouse
brain microvascular endothelial cells as well as stroke patients. LncRNAs are considered to be promising biomarkers
for the diagnosis and prognosis of cerebral ischemia. Here, we have reviewed the latest advances in
lncRNA-based therapeutic approaches for ischemic disease. Accordingly, we summarize the current understanding
of lncRNAs and ischemic stroke, focusing on the regulatory role of lncRNAs in ischemic stroke, as well as
their potential as biomarkers and therapeutic targets in cerebral ischemia.
Collapse
Affiliation(s)
- Qianwen Wang
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Xu Liu
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Ruixia Zhu
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| |
Collapse
|
109
|
Huang J, Yang J, Li J, Chen Z, Guo X, Huang S, Gu L, Su L. Association of long noncoding RNA H19 polymorphisms with the susceptibility and clinical features of ischemic stroke in southern Chinese Han population. Metab Brain Dis 2019; 34:1011-1021. [PMID: 31041585 DOI: 10.1007/s11011-019-00417-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 04/07/2019] [Indexed: 01/10/2023]
Abstract
Stroke is the leading cause of death in China. Previous studies have demonstrated that long noncoding RNAs play important roles in ischemic stroke (IS). This study aimed to investigate long noncoding RNA H19 (lncRNA H19) expression in IS cases and the association between lncRNA H19 variants and IS risk and IS-related risk factors. A total of 550 IS cases and 550 controls were recruited for this study. LncRNA H19 expression was detected using quantitative real-time polymerase chain reaction. Genotyping was conducted by the Sequenom MassARRAY technology. LncRNA H19 level in peripheral blood of IS cases was significantly upregulated compared with healthy controls (P = 0.046). No significant association was observed between lncRNA H19 rs217727 and rs4929984 polymorphisms with IS risk in all genetic models, and rs217727-rs4929984 haplotypes are not associated with IS susceptibility. Further meta-analysis also implied that the rs217727 and rs4929984 polymorphisms were not associated with IS in Chinese population. However, rs4929984 is significantly associated with the diastolic blood pressure level of IS patients (additive model: Padj = 0.007; dominant model: Padj = 0.013), whereas rs217727 is associated with international normalized ratio (additive model: Padj = 0.019; recessive model: Padj = 0.004), prothrombin time activity level (additive model: Padj = 0.026; recessive model: Padj = 0.004), and homocysteine level (recessive model: Padj = 0.048) in patients with IS. Our findings suggest that lncRNA H19 level may affect the occurrence of IS, and lncRNA H19 variants may influence blood pressure, coagulation function, and homocysteine metabolism of patients with IS in the southern Chinese Han population.
Collapse
Affiliation(s)
- Jiao Huang
- School of Public Health of Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Jialei Yang
- School of Public Health of Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Jinhong Li
- Department of Internal Neurology, First Affiliated Hospital, Guangxi University of Chinese Medicine, 89-9 Dongge Road, Nanning, 530023, Guangxi, China
| | - Zhaoxia Chen
- School of Public Health of Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Xiaojing Guo
- School of Public Health of Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Siyun Huang
- Department of Internal Neurology, First Affiliated Hospital, Guangxi University of Chinese Medicine, 89-9 Dongge Road, Nanning, 530023, Guangxi, China
| | - Lian Gu
- Department of Internal Neurology, First Affiliated Hospital, Guangxi University of Chinese Medicine, 89-9 Dongge Road, Nanning, 530023, Guangxi, China.
| | - Li Su
- School of Public Health of Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, China.
| |
Collapse
|
110
|
Zhang L, Wang H. Long Non-coding RNA in CNS Injuries: A New Target for Therapeutic Intervention. MOLECULAR THERAPY-NUCLEIC ACIDS 2019; 17:754-766. [PMID: 31437654 PMCID: PMC6709344 DOI: 10.1016/j.omtn.2019.07.013] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 07/17/2019] [Accepted: 07/17/2019] [Indexed: 12/15/2022]
Abstract
CNS injuries, such as traumatic brain injury (TBI), subarachnoid hemorrhage (SAH), intracerebral hemorrhage (ICH), and cerebral ischemic stroke, are important causes of death and long-term disability worldwide. As an important class of pervasive genes involved in many pathophysiological processes, long non-coding RNAs (lncRNAs) have received attention in the past decades. Multiple studies indicate that lncRNAs are abundant in the CNS and have a key role in brain function as well as many neurological disorders, especially in CNS injuries. Several investigations have deciphered that regulation of lncRNAs exert pro-angiogenesis, anti-apoptosis, and anti-inflammation effects in CNS injury via different molecules and pathways, including microRNA (miRNA), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), phosphatidylinositol-4,5-bisphosphate 3-kinase/protein kinase B (PI3K/AKT), Notch, and p53. Thus, lncRNAs show great promise as molecular targets in CNS injuries. In this article, we provide an updated review of the current state of our knowledge about the relationship between lncRNAs and CNS injuries, highlighting the specific roles of lncRNAs in CNS injuries.
Collapse
Affiliation(s)
- Li Zhang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Handong Wang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
111
|
Teng L, Meng R. Long Non-Coding RNA MALAT1 Promotes Acute Cerebral Infarction Through miRNAs-Mediated hs-CRP Regulation. J Mol Neurosci 2019; 69:494-504. [DOI: 10.1007/s12031-019-01384-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Accepted: 07/09/2019] [Indexed: 01/19/2023]
|
112
|
Reichelt-Wurm S, Wirtz T, Chittka D, Lindenmeyer M, Reichelt RM, Beck S, Politis P, Charonis A, Kretz M, Huber TB, Liu S, Banas B, Banas MC. Glomerular expression pattern of long non-coding RNAs in the type 2 diabetes mellitus BTBR mouse model. Sci Rep 2019; 9:9765. [PMID: 31278342 PMCID: PMC6611801 DOI: 10.1038/s41598-019-46180-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 06/11/2019] [Indexed: 11/09/2022] Open
Abstract
The prevalence of type 2 diabetes mellitus (T2DM) and by association diabetic nephropathy (DN) will continuously increase in the next decades. Nevertheless, the underlying molecular mechanisms are largely unknown and studies on the role of new actors like long non-coding RNAs (lncRNAs) barely exist. In the present study, the inherently insulin-resistant mouse strain "black and tan, brachyuric" (BTBR) served as T2DM model. While wild-type mice do not exhibit pathological changes, leptin-deficient diabetic animals develop a severe T2DM accompanied by a DN, which closely resembles the human phenotype. We analyzed the glomerular expression of lncRNAs from wild-type and diabetic BTBR mice (four, eight, 16, and 24 weeks) applying the "GeneChip Mouse Whole Transcriptome 1.0 ST" array. This microarray covered more lncRNA gene loci than any other array before. Over the observed time, our data revealed differential expression patterns of 1746 lncRNAs, which markedly differed from mRNAs. We identified protein-coding and non-coding genes, that were not only co-located but also co-expressed, indicating a potentially cis-acting function of these lncRNAs. In vitro-experiments strongly suggested a cell-specific expression of these lncRNA-mRNA-pairs. Additionally, protein-coding genes, being associated with significantly regulated lncRNAs, were enriched in various biological processes and pathways, that were strongly linked to diabetes.
Collapse
Affiliation(s)
| | - Tobias Wirtz
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Dominik Chittka
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Maja Lindenmeyer
- Nephrological Center, Medical Clinic and Policlinic IV, University Hospital of Munich, Munich, Germany.,III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Robert M Reichelt
- Department of Biochemistry, Genetics and Microbiology, Institute of Microbiology, University of Regensburg, Regensburg, Germany
| | - Sebastian Beck
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Panagiotis Politis
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Aristidis Charonis
- Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Markus Kretz
- Institute of Biochemistry, Genetics and Microbiology, University of Regensburg, Regensburg, Germany
| | - Tobias B Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Shuya Liu
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Bernhard Banas
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Miriam C Banas
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
113
|
Toraih EA, El-Wazir A, Alghamdi SA, Alhazmi AS, El-Wazir M, Abdel-Daim MM, Fawzy MS. Association of long non-coding RNA MIAT and MALAT1 expression profiles in peripheral blood of coronary artery disease patients with previous cardiac events. Genet Mol Biol 2019; 42:509-518. [PMID: 31188931 PMCID: PMC6905438 DOI: 10.1590/1678-4685-gmb-2018-0185] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 11/25/2018] [Indexed: 01/16/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) are implicated in various cellular and pathological processes. Two lncRNAs, myocardial infarction-associated transcript (MIAT) and metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), may be involved in the pathogenesis of coronary artery disease (CAD). Here, we aimed to determine the relative circulating levels of MIAT and MALAT1 in 110 stable CAD patients and 117 controls and to correlate their levels with the clinical and laboratory data. Peripheral blood expression levels were quantified by Real-Time qPCR. The median MIAT expression level in CAD patients was significantly 12-fold higher than controls (p<0.001). Otherwise, the median MALAT1 expression level was comparable in patient and control groups. Both lncRNAs showed significantly higher relative expression levels in patients with positive history of previous cardiac ischemic events, and MIAT showed significantly higher expression in diabetic CAD patients. The area under the curve of MIAT (0.888 ± 0.02 with sensitivity 95.5% and specificity 72.7%), was significantly larger than that of MALAT1 (0.601 ± 0.04 with sensitivity 50% and specificity 63.6%) for detecting the presence of significant CAD. The current findings suggest that lncRNA MIAT could have a diagnostic significance in CAD patients. MALAT1 levels, however, are not sufficiently reliable to have much clinical use in our cases.
Collapse
Affiliation(s)
- Eman A. Toraih
- Genetics Unit, Department of Histology and Cell Biology,
Faculty of Medicine, Suez Canal University, Ismailia, Egypt
- Center of Excellence of Molecular and Cellular Medicine, Suez
Canal University, Ismailia, Egypt
| | - Aya El-Wazir
- Genetics Unit, Department of Histology and Cell Biology,
Faculty of Medicine, Suez Canal University, Ismailia, Egypt
- Center of Excellence of Molecular and Cellular Medicine, Suez
Canal University, Ismailia, Egypt
| | - Saleh A. Alghamdi
- Medical Genetics, Clinical Laboratory Department, College of
Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Ayman S Alhazmi
- Department of Clinical Chemistry, College of Applied Medical
Sciences, Taif University, Taif, Saudi Arabia
| | - Mohammad El-Wazir
- Department of Cardiology, Faculty of Medicine, Suez Canal
University, Ismailia, Egypt
| | - Mohamed M. Abdel-Daim
- Department of Pharmacology, Faculty of Veterinary Medicine,
Suez Canal University, Ismailia, Egypt
| | - Manal S. Fawzy
- Department of Biochemistry, Faculty of Medicine, Northern
Border University, Arar, Saudi Arabia
- Department of Medical Biochemistry and Molecular Biology,
Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
114
|
LncRNA MALAT1 protects human umbilical vein endothelial cells against ox-LDL triggered cell death through regulation of MGP. Mol Cell Toxicol 2019. [DOI: 10.1007/s13273-019-0032-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
115
|
Suppression of lncRNA RMRP ameliorates oxygen-glucose deprivation/re-oxygenation-induced neural cells injury by inhibiting autophagy and PI3K/Akt/mTOR-mediated apoptosis. Biosci Rep 2019; 39:BSR20181367. [PMID: 30926681 PMCID: PMC6591569 DOI: 10.1042/bsr20181367] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 03/18/2019] [Accepted: 03/26/2019] [Indexed: 11/17/2022] Open
Abstract
The aberrant expression of lncRNAs has been inferred to be closely related with the progression of neural ischemia/reperfusion (I/R) injury. RMRP is an lncRNA associated with I/R injury. In order to determine the role of RMRP in I/R injury, the effects of RMRP knockdown on oxygen-glucose deprivation/re-oxygenation (OGD/R)-induced injury in SH-SY5Y cells were evaluated. The effect of OGD/R administration on the expression of RMRP and apoptosis in SH-SY5Y cells, and the effect of RMRP suppression by siRNA on the impairments of cells proliferation and mobility potential due to OGD/R administration were assessed in the current study. At the molecular level, the current study detected the expressions of indicators involved in autophagy and PI3K/Akt/mTOR-mediated apoptosis pathways. The OGD/R administration induced the expression of RMRP and apoptosis in SH-SY5Y cells. After RMRP knockdown, the proliferation potential of SH-SY5Y cells was restored, and apoptosis and cell cycle arrest were inhibited. Moreover, RMRP inhibition also increased the invasion and migration of SH-SY5Y cells which were treated with OGD/R. The effects of RMRP suppression on the phenotypes of SH-SY5Y were associated with the inhibition of LC3II, p-PI3K, p-Akt, and p-mTOR as well as the induction of P62 and Bcl-2. Inhibition of RMRP contributed to the improvement of OGD/R-induced neuronal injury, which might be mediated through the inhibition of autophagy and apoptosis pathways.
Collapse
|
116
|
Li Y, Guo S, Liu W, Jin T, Li X, He X, Zhang X, Su H, Zhang N, Duan C. Silencing of SNHG12 Enhanced the Effectiveness of MSCs in Alleviating Ischemia/Reperfusion Injuries via the PI3K/AKT/mTOR Signaling Pathway. Front Neurosci 2019; 13:645. [PMID: 31293373 PMCID: PMC6603177 DOI: 10.3389/fnins.2019.00645] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 06/05/2019] [Indexed: 12/30/2022] Open
Abstract
Previous studies have reported that the long non-coding RNA SNHG12 (lncRNA SNHG12) plays a critical role in regulating the function of mesenchymal stem cells (MSCs); however, the effect of lncRNA SNHG12 on MSCs in injured brain tissue has rarely been reported. We studied the effect and mechanism of lncRNA SNHG12-modified mesenchymal stem cells (MSCs) in treating brain injuries caused by ischemia/reperfusion (I/R). I/R treated rat brain microvascular endothelial cells (BMECs) were co-cultured with MSCs or I/R pretreated MSCs. Next, BMEC proliferation was detected by using CCK-8 and EdU assays, and cell apoptosis was determined by using flow cytometry and the Hoechst staining method. Autophagy of BMECs was determined using immunofluorescence and expression of associated pathway proteins were measured by western blotting. Moreover, BMEC proliferation, apoptosis, and autophagy were also determined after the BMECs had been co-cultured with shSNHG12-MSCs. In addition, a rat model of middle cerebral artery occlusion (MCAO) was used to further confirm the findings obtained with cells. I/R treatment significantly decreased the proliferation of BMECs, but increased their levels of SNHG12 expression, apoptosis, and autophagy. However, co-culturing of BMECs with MSCs markedly alleviated the reduction in BMEC proliferation and the increases in BMEC apoptosis and autophagy, as well as the phosphorylation of PI3K, AKT, and mTOR proteins in BMECs that had been induced by I/R. Furthermore, shSNHG12 remarkably enhanced the effects of MSCs. In addition, an injection MSCs reduced the infarct areas and rates of cell apoptosis in MACO rats, and reduced the phosphorylation of PI3K, AKT, and mTOR proteins. Moreover, shSNHG12 enhanced the ameliorative effect of MSCs in treating brain injuries in the MACO rats. In conclusion, silencing of SNHG12 enhanced the effects of MSCs in reducing apoptosis and autophagy of BMECs by activating the PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Yuanzhi Li
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Department of Neurosurgery, Affiliated Hengyang Hospital, Southern Medical University (Hengyang Central Hospital), Hengyang, China
| | - Shenquan Guo
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Wenchao Liu
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Tao Jin
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xifeng Li
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xuying He
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xin Zhang
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Hengxian Su
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Nan Zhang
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Chuanzhi Duan
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
117
|
Hu X, Hu X, Huang G. LncRNA MALAT1 is involved in sevoflurane-induced neurotoxicity in developing rats. J Cell Biochem 2019; 120:18209-18218. [PMID: 31190336 DOI: 10.1002/jcb.29127] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 05/09/2019] [Accepted: 05/10/2019] [Indexed: 01/19/2023]
Abstract
OBJECTIVE The purpose of this study is to uncover the effects of long chain noncoding RNA (lncRNA) metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) on sevoflurane-induced neurotoxicity in developing rats. METHODS Sevoflurane neurotoxicity model was established by sevoflurane treatment in 7-day-old Sprague-Dawley rats. The rats were treated with Sevo or MALAT1 small interfering RNA to detect the MALAT1 expression, pathological change, ultrastructure, neuronal apoptosis, expression of apoptosis-related proteins, expression of neurotrophic factors BDNF and NGF, spatial learning and memory function change, as well as neuron cell density of hippocampal tissues. RESULTS MALAT1 was highly expressed in hippocampus tissues of rats. Downregulation of MALAT1 alleviated the pathological change, improved the ultrastructure, inhibited apoptosis of neuronal cells, declined caspase 3 and Bax while elevated Bcl-2, BDNF and NGF, improved capability of spatial learning and memory, and increased density of hippocampal neurons in hippocampal tissues of sevoflurane-induced rats. CONCLUSION Suppression of MALAT1 can reduce the apoptosis of hippocampal neurons induced by sevoflurane anesthesia, improve the capability of spatial learning, and memory function and alleviate the loss of hippocampal nerve cells in developing rats. To a certain extent, it plays the role of protecting brain nerve cells.
Collapse
Affiliation(s)
- Xueyan Hu
- Department of Anesthesiology, Yishui Central Hospital, Linyi, People's Republic of China
| | - Xiaodong Hu
- Department of Anesthesiology, Yishui Central Hospital, Linyi, People's Republic of China
| | - Guirong Huang
- Department of Anesthesiology, Yishui Central Hospital, Linyi, People's Republic of China
| |
Collapse
|
118
|
Deng Z, Cai H, Lin L, Zhu L, Wu W, Yang S, Cai J, Tan J. lncRNA ATXN8OS promotes breast cancer by sequestering miR‑204. Mol Med Rep 2019; 20:1057-1064. [PMID: 31173245 PMCID: PMC6625414 DOI: 10.3892/mmr.2019.10367] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 04/26/2019] [Indexed: 12/25/2022] Open
Abstract
Breast cancer (BC) is a common malignancy among women and the leading cause of female cancer mortality worldwide. In recent years, increasing evidence has shown that long non-coding RNAs (lncRNAs) can act as competing endogenous RNAs (ceRNAs) in human cancer and that they are involved in many biological processes, including proliferation, migration, apoptosis and invasion. In the present study, the biological function and molecular mechanism of ataxin 8 opposite strand (ATXN8OS) in BC tissue and cell lines were investigated. It was found that ATXN8OS was markedly up-regulated in BC tissue and cell lines, and that its level of overexpression was inversely linked with the overall survival rate of patients with BC. Knockdown of ATXN8OS inhibited proliferation, viability and invasion in the human MCF7 and MDA-MB-231 BC cell lines. In addition, microRNA-204 (miR-204) was negatively associated with the expression of ATXN8OS in BC tissues and cell lines. A luciferase assay demonstrated a direct binding site for miR-204 within ATXN8OS, and inhibition of miR-204 stimulated the tumour-promoting effect of ATXN8OS on BC cells. In conclusion, the present study suggested that ATXN8OS acts as a tumour promoter by sequestering miR-204 during the development of BC, therefore providing a mechanistic insight which may facilitate the diagnosis and treatment of BC.
Collapse
Affiliation(s)
- Zhen Deng
- Department of Urology, 900th Hospital of the Joint Logistics Support Force, People's Liberation Army, Fuzhou, Fujian 350000, P.R. China
| | - Huayu Cai
- Department of Urology, 900th Hospital of the Joint Logistics Support Force, People's Liberation Army, Fuzhou, Fujian 350000, P.R. China
| | - Liying Lin
- Department of General Surgery, 900th Hospital of the Joint Logistics Support Force, People's Liberation Army, Fuzhou, Fujian 350000, P.R. China
| | - Lingfeng Zhu
- Department of Urology, 900th Hospital of the Joint Logistics Support Force, People's Liberation Army, Fuzhou, Fujian 350000, P.R. China
| | - Weizhen Wu
- Department of Urology, 900th Hospital of the Joint Logistics Support Force, People's Liberation Army, Fuzhou, Fujian 350000, P.R. China
| | - Shunliang Yang
- Department of Urology, 900th Hospital of the Joint Logistics Support Force, People's Liberation Army, Fuzhou, Fujian 350000, P.R. China
| | - Jinquan Cai
- Department of Urology, 900th Hospital of the Joint Logistics Support Force, People's Liberation Army, Fuzhou, Fujian 350000, P.R. China
| | - Jianming Tan
- Department of Urology, 900th Hospital of the Joint Logistics Support Force, People's Liberation Army, Fuzhou, Fujian 350000, P.R. China
| |
Collapse
|
119
|
LncRNA SNHG12 as a potent autophagy inducer exerts neuroprotective effects against cerebral ischemia/reperfusion injury. Biochem Biophys Res Commun 2019; 514:490-496. [DOI: 10.1016/j.bbrc.2019.04.158] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 04/23/2019] [Indexed: 11/23/2022]
|
120
|
Duan X, Han L, Peng D, Peng C, Xiao L, Bao Q, Peng H. Bioinformatics analysis of a long non‑coding RNA and mRNA regulation network in rats with middle cerebral artery occlusion based on RNA sequencing. Mol Med Rep 2019; 20:417-432. [PMID: 31180537 PMCID: PMC6580035 DOI: 10.3892/mmr.2019.10300] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 03/07/2019] [Indexed: 12/24/2022] Open
Abstract
Long non‑coding RNAs (lncRNAs) have been proven to be critical gene regulators of development and disease. The main aim of the present study was to elucidate the lncRNA‑mRNA regulation network in ischemic stroke induced by middle cerebral artery occlusion (MCAO) using RNA sequencing (RNA‑seq) in rats. lncRNA expression profiles were screened in brain tissues to identify a number of differentially expressed lncRNAs (DELs) and genes (DEGs) by RNA‑seq. Reverse transcription‑quantitative polymerase chain reaction was performed to further confirm the lncRNA expression data. Furthermore, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were used to mine mRNA functions, and a lncRNA‑mRNA network was constructed. Additionally, cis‑ and trans‑regulatory gene analyses of DELs were predicted. A total of 134 DELs (fold change >2, false discovery rate <0.05) and 1,006 DEGs (fold change >2 and P<0.05) were identified. Eighteen lncRNAs were predicted to regulate heme oxygenase 1, mitotic checkpoint serine/threonine kinase B, chemokine ligand 2 and DNA Topoisomerase IIα, amongst other genes. These genes are all associated with a cellular response to inorganic substances, alkaloids, estradiol, reactive oxygen species, metal ions, oxidative stress, and are associated with metabolic pathways, chemokine signaling pathways, malaria, Parkinson's disease, the cell cycle and other GO and KEGG pathway enrichments. The present study identifies novel DELs and an lncRNA‑mRNA regulatory network that may allow for an improved understanding of the molecular mechanism of ischemic stroke induced by MCAO.
Collapse
Affiliation(s)
- Xianchun Duan
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui 230031, P.R. China
| | - Lan Han
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, P.R. China
| | - Daiyin Peng
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, P.R. China
| | - Can Peng
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, P.R. China
| | - Ling Xiao
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, P.R. China
| | - Qiuyu Bao
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, P.R. China
| | - Huasheng Peng
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, P.R. China
| |
Collapse
|
121
|
Akella A, Bhattarai S, Dharap A. Long Noncoding RNAs in the Pathophysiology of Ischemic Stroke. Neuromolecular Med 2019; 21:474-483. [DOI: 10.1007/s12017-019-08542-w] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 05/09/2019] [Indexed: 12/24/2022]
|
122
|
Overexpression of SNHG12 regulates the viability and invasion of renal cell carcinoma cells through modulation of HIF1α. Cancer Cell Int 2019; 19:128. [PMID: 31114448 PMCID: PMC6518781 DOI: 10.1186/s12935-019-0782-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 03/15/2019] [Indexed: 01/07/2023] Open
Abstract
Background Cumulative evidences demonstrated the aberrant overexpression of Small Nucleolar RNA Host Gene 12 (SNHG12) in diverse human cancer. However, the expression status and involvement of SNHG12 in renal cell carcinoma is still elusive. Methods The expression of SNHG12 was determined by q-PCR. The transcriptional regulation was interrogated by luciferase reporter assay. Cell viability was measured with CCK-8 kit. The anchorage-independent was evaluated by soft agar assay. Cell apoptosis was analyzed by Annexin V/7-AAD double staining. The migration and invasion were determined by trans-well assay and wound scratch closure. The in vivo tumor growth was monitored in xenograft mice model. Protein expression was quantified by immunoblotting. Results SNHG12 was aberrantly up-regulated in renal carcinoma both in vivo and in vitro. High expression of SNHG12 associated with poor prognosis. Deficiency of SNHG12 significantly suppressed cell viability, anchorage-independent growth and induced apoptosis. In addition, SNHG12 silencing inhibited migrative and invasive in vitro and xenograft tumor growth in vivo. Mechanistically, SNHG12 modulated HIF1α expression via competing with miR-199a-5p, which consequently contributed to its oncogenic potential. MiR-199a-5p inhibition severely compromised SNHG12 silencing-elicited tumor repressive effects. Conclusion Our data uncovered a crucial role of SNHG12-miR-199a-5p-HIF1α axis in human renal cancer.
Collapse
|
123
|
Wang C, Qu Y, Suo R, Zhu Y. Long non-coding RNA MALAT1 regulates angiogenesis following oxygen-glucose deprivation/reoxygenation. J Cell Mol Med 2019; 23:2970-2983. [PMID: 30784209 PMCID: PMC6433728 DOI: 10.1111/jcmm.14204] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 12/17/2018] [Accepted: 01/06/2019] [Indexed: 12/30/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) have been identified as playing critical roles in multiple diseases. However, little is known regarding their roles and mechanisms in post-stroke angiogenesis. Our studies focused on deciphering the functional roles and the underlying mechanisms of the lncRNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) in the process of angiogenesis following oxygen-glucose deprivation/reoxygenation (OGD/R). We characterized the up-regulation of MALAT1 expression in the process of angiogenesis after hypoxic injury in vivo and in vitro. We further showed that compared with the empty vector, MALAT1 knockdown had significantly reduced the capacity for angiogenesis, which was measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium (MTT), scratching, cell cycle and immunofluorescent staining. Thus, our findings suggest that MALAT1 may mediate proangiogenic function in OGD/R. To further explore the potential mechanisms, we used lentiviruses expressing shMALAT1 and empty vector; the results revealed that shMALAT1 reduced the expression of 15-lipoxygenase 1 (15-LOX1), vascular endothelial growth factor (VEGF) and the phosphorylation of signal transducers and activators of transcription 3 (pSTAT3). Taken together, our results are the first to propose that MALAT1 may regulate angiogenesis through the 15-LOX1/STAT3 signalling pathway, and they may provide a critical target for the treatment of hypoxic injury and an avenue for therapeutic angiogenesis.
Collapse
Affiliation(s)
- Chengya Wang
- Department of NeurologySecond Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - Youyang Qu
- Department of NeurologySecond Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - Rui Suo
- Department of NeurologySecond Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - Yulan Zhu
- Department of NeurologySecond Affiliated Hospital of Harbin Medical UniversityHarbinChina
| |
Collapse
|
124
|
Ren L, Wei C, Li K, Lu Z. LncRNA MALAT1 up-regulates VEGF-A and ANGPT2 to promote angiogenesis in brain microvascular endothelial cells against oxygen-glucose deprivation via targetting miR-145. Biosci Rep 2019; 39:BSR20180226. [PMID: 30038058 PMCID: PMC6400790 DOI: 10.1042/bsr20180226] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Revised: 02/07/2018] [Accepted: 09/07/2018] [Indexed: 01/17/2023] Open
Abstract
Stroke is one of the leading causes of death and long-term disability around the world. Angiogenesis is supposed to protect brain microvascular endothelial cells (BMECs) from oxidative and ischemic stress. Previous studies indicated that interaction between metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) and miR-145 was involved in myocardial ischemia reperfusion, suggesting MALAT1 and miR-145 were also mediated with the progress of angiogenesis and cell migration in oxygen-glucose deprivation (OGD)-induced BMECs. The present study aimed to investigate the functional roles of MALAT1 in regulating miR-145 and its downstream pro-angiogenesis factors, vascular endothelial growth factor (VEGF)-A and Angiopoietin-2 (ANGPT2) during the progress of angiogenesis in OGD-induced BMECs. An in vitro OGD model was employed in mouse BMECs to mimic brain hypoxic and ischemic conditions; MTT was used to determine cell viability. qRT-PCR was used to determine the expression of long non-coding RNA (lncRNA)-MALAT1 and miR-145 under OGD conditions; in vitro tube formation assay was used to investigate angiogenic effect of MALAT1 and miR-145 The relationship between lncRNA-MALAT1/miR-145 and miR-145/VEGF-A/ANGPT2 was evaluated by qRT-PCR and Western blot, and direct binding was assessed using dual luciferase assay. Results showed that the levels of lncRNA-MALAT1 and miR-145 were up-regulated in OGD-induced BMECs. miR-145 functioned as an anti-angiogenic and pro-apoptotic factor in OGD treated BMECs via down-regulating VEGF-A and ANGPT2 directly. While lncRNA-MALAT1 enhanced the expressions of VEGF-A and ANGPT2 by targetting miR-145 to promote angiogenesis and proliferation of BMECs under OGD conditions. Our present study revealed the inhibitory functions of miR-145 on angiogenesis through direct targetting on VEGF-A and ANGPT2 for the first time and proved the protective role of lncRNA-MALAT1 for BMECs under OGD conditions through the direct regulation of miR-145.
Collapse
Affiliation(s)
- Lanfen Ren
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, P.R. China
| | - Chunxia Wei
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, P.R. China
| | - Kui Li
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, P.R. China
| | - Zuneng Lu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, P.R. China
| |
Collapse
|
125
|
Kołat D, Kałuzińska Ż, Bednarek AK, Płuciennik E. The biological characteristics of transcription factors AP-2α and AP-2γ and their importance in various types of cancers. Biosci Rep 2019; 39:BSR20181928. [PMID: 30824562 PMCID: PMC6418405 DOI: 10.1042/bsr20181928] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 02/11/2019] [Accepted: 02/27/2019] [Indexed: 02/07/2023] Open
Abstract
The Activator Protein 2 (AP-2) transcription factor (TF) family is vital for the regulation of gene expression during early development as well as carcinogenesis process. The review focusses on the AP-2α and AP-2γ proteins and their dualistic regulation of gene expression in the process of carcinogenesis. Both AP-2α and AP-2γ influence a wide range of physiological or pathological processes by regulating different pathways and interacting with diverse molecules, i.e. other proteins, long non-coding RNAs (lncRNA) or miRNAs. This review summarizes the newest information about the biology of two, AP-2α and AP-2γ, TFs in the carcinogenesis process. We emphasize that these two proteins could have either oncogenic or suppressive characteristics depending on the type of cancer tissue or their interaction with specific molecules. They have also been found to contribute to resistance and sensitivity to chemotherapy in oncological patients. A better understanding of molecular network of AP-2 factors and other molecules may clarify the atypical molecular mechanisms occurring during carcinogenesis, and may assist in the recognition of new diagnostic biomarkers.
Collapse
Affiliation(s)
- Damian Kołat
- Faculty of Biomedical Sciences and Postgraduate Education, Medical University of Lodz, Lodz, Poland
| | - Żaneta Kałuzińska
- Faculty of Biomedical Sciences and Postgraduate Education, Medical University of Lodz, Lodz, Poland
| | - Andrzej K Bednarek
- Department of Molecular Carcinogenesis, Medical University of Lodz, Lodz, Poland
| | - Elżbieta Płuciennik
- Department of Molecular Carcinogenesis, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
126
|
Chen R, Xu X, Huang L, Zhong W, Cui L. The Regulatory Role of Long Noncoding RNAs in Different Brain Cell Types Involved in Ischemic Stroke. Front Mol Neurosci 2019; 12:61. [PMID: 30967760 PMCID: PMC6440499 DOI: 10.3389/fnmol.2019.00061] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 02/22/2019] [Indexed: 01/01/2023] Open
Abstract
Stroke results in high morbidity and high mortality worldwide, with ischemic stroke accounting for 80% to 85%. As effective treatments for ischemic stroke remain limited because of the narrow therapeutic time window, a better understanding of the pathologic mechanism and new therapeutic intervention targets are needed. Due to the development of next-generation sequencing technologies and the genome-wide analysis of eukaryotic transcriptomes, a large amount of evidence to date demonstrates that long noncoding RNAs (lncRNAs) play a vital role in gene regulation and in ischemic stroke. In recent years, many studies have been focused on the clinical significance of lncRNAs in ischemic stroke, and data shows that the pathological processes underlying ischemic stroke are driven by interactions among different brain cell types, including neurons, glial cells, and vascular cells, which actively participate in the mechanisms of tissue injury and repair. In this mini review article, we provide an overview of the characteristics and underlying regulation mechanisms of lncRNAs relevant to different brain cell types during the course of ischemic stroke. Moreover, we reveal the roles of lncRNAs as potential biomarkers and treatment targets in ischemic stroke.
Collapse
Affiliation(s)
- Runsen Chen
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.,Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Xiangming Xu
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.,Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Lidan Huang
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.,Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Wangtao Zhong
- Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Lili Cui
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
127
|
Ruan W, Li J, Xu Y, Wang Y, Zhao F, Yang X, Jiang H, Zhang L, Saavedra JM, Shi L, Pang T. MALAT1 Up-Regulator Polydatin Protects Brain Microvascular Integrity and Ameliorates Stroke Through C/EBPβ/MALAT1/CREB/PGC-1α/PPARγ Pathway. Cell Mol Neurobiol 2019; 39:265-286. [PMID: 30607811 PMCID: PMC11469806 DOI: 10.1007/s10571-018-00646-4] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 12/19/2018] [Indexed: 02/07/2023]
Abstract
Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is a long non-coding RNA contributing to protect the blood-brain barrier (BBB) after stroke. We searched for small molecules that may up-regulate MALAT1 and focused on polydatin (PD), a natural product, as a possible candidate. PD enhanced MALAT1 gene expression in rat brain microvascular endothelial cells, reducing cell toxicity and apoptosis after oxygen and glucose deprivation (OGD). These effects correlated with reduction of inflammatory factors and enhancement of expression of BBB markers. We found opposite changes after MALAT1 silencing. We determined that C/EBPβ is a key transcription factor for PD-mediated MALAT1 expression. PPARγ activity is involved in MALAT1 protective effects through its coactivator PGC-1α and the transcription factor CREB. This suggests that PD activates the MALAT1/CREB/PGC-1α/PPARγ signaling pathway to protect endothelial cells against ischemia. PD administration to rats subjected to brain ischemia by transient middle cerebral artery occlusion (tMCAO) reduced cerebral infarct volume and brain inflammation, protected cerebrovascular endothelial cells and BBB integrity. These effects correlated with increased expression of MALAT1, C/EBPβ, and PGC-1α. Our results strongly suggest that the beneficial effects of PD involve the C/EBPβ/MALAT1/CREB/PGC-1α/PPARγ pathway, which may provide a novel therapeutic strategy for brain ischemic stroke.
Collapse
Affiliation(s)
- Wenchen Ruan
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, #24 Tong Jia Xiang Street, Nanjing, 210009, People's Republic of China
- College of Basic Medical Sciences, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, 116044, People's Republic of China
| | - Jingwei Li
- Department of Neurology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, People's Republic of China
| | - Yazhou Xu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, #24 Tong Jia Xiang Street, Nanjing, 210009, People's Republic of China
| | - Yunjie Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, #24 Tong Jia Xiang Street, Nanjing, 210009, People's Republic of China
| | - Feng Zhao
- College of Basic Medical Sciences, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, 116044, People's Republic of China
| | - Xu Yang
- College of Basic Medical Sciences, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, 116044, People's Republic of China
| | - Hulin Jiang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, #24 Tong Jia Xiang Street, Nanjing, 210009, People's Republic of China
| | - Luyong Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, #24 Tong Jia Xiang Street, Nanjing, 210009, People's Republic of China
- Center for Drug Screening and Pharmacodynamics Evaluation, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, People's Republic of China
- Key Laboratory of Drug Quality Control and Pharmacovigilance (China Pharmaceutical University), Ministry of Education, Nanjing, 210009, People's Republic of China
| | - Juan M Saavedra
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, 20057, USA
| | - Lei Shi
- College of Basic Medical Sciences, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, 116044, People's Republic of China.
| | - Tao Pang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, #24 Tong Jia Xiang Street, Nanjing, 210009, People's Republic of China.
- Key Laboratory of Drug Quality Control and Pharmacovigilance (China Pharmaceutical University), Ministry of Education, Nanjing, 210009, People's Republic of China.
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, 20057, USA.
| |
Collapse
|
128
|
Comprehensive analysis of differentially expressed profiles of long non-coding RNAs and messenger RNAs in kaolin-induced hydrocephalus. Gene 2019; 697:184-193. [PMID: 30797995 DOI: 10.1016/j.gene.2019.02.041] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 02/04/2019] [Accepted: 02/19/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUNDS The pathophysiology of hydrocephalus induced brain damage remains unclear. Long non-coding RNAs (lncRNAs) have been demonstrated to be implicated in many central nervous system diseases. However, the roles of lncRNAs in hydrocephalus injury are poorly understood. METHODS The present study depicted the expression profiles of lncRNAs and messenger RNAs (mRNAs) in C57BL/6 mice with kaolin-induced hydrocephalus and saline controls using high-throughput RNA sequencing. Afterward, Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were performed to identify potential targets that correlated with hydrocephalus. In addition, co-expression networks and cis- and trans-regulation were predicted using bioinformatics methods. Finally, representative lncRNAs and mRNAs were further validation using quantitative real-time polymerase chain reaction. RESULTS A total of 1575 lncRNAs and 1168 mRNAs were differentially expressed (DE) in hydrocephalus. GO and KEGG analyses indicated several immune and inflammatory response-associated pathways may be important in the hydrocephalus. Besides, functional enrichment analysis based on co-expression network showed several similar pathways, such as chemokine signaling pathway, phagosome, MAPK signaling pathway and complement and coagulation cascade. Cis-regulation prediction revealed 5 novel lncRNAs might regulate their nearby coding genes, and trans-regulation revealed several lncRNAs participate in pathways regulated by transcription factors, including BPTF, FOXM1, NR5A2, P2RX5, and NR6A1. CONCLUSIONS In conclusion, our results provide candidate genes involved in hydrocephalus and suggest a new perspective on the modulation of lncRNAs in hydrocephalus.
Collapse
|
129
|
Abstract
Over many years, extensive efforts have focused on the development and improvement of diagnostic and therapeutic strategies to reduce stroke-associated neurovascular damage, such as blood-brain barrier dysfunction, brain edema, parenchymal inflammation, and neural cell death. However, the only clinically applied pharmacological therapy to date for the treatment of acute ischemic stroke is thrombolysis. Because of the short therapeutic window of current thrombolytic therapy and the activation of various pathophysiological signaling cascades triggered after ischemic stroke, the development of new therapies is urgently required. Noncoding RNAs (ncRNAs) are defined as untranslated regulatory RNA molecules. Although ncRNAs with biological roles have been known for almost 60 years, they have within the past decade emerged as key mediators of posttranscriptional gene expression/function in pathological aspects of ischemic stroke. With properties of relative stability, specificity, and reproducibility, ncRNAs are considered to be promising as biomarkers and better candidates than proteins and genes for early recognition of the onset of disease. In this update, we summarized the current knowledge for three groups of ncRNAs in stroke, focusing on the role of long noncoding RNAs and circular RNAs as biomarkers for stroke and as targets for regulating large sets of genes in related pathways after ischemic stroke.
Collapse
Affiliation(s)
- Xuejing Zhang
- Pittsburgh Institute of Brain Disorders & Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Milton H. Hamblin
- Department of Pharmacology, Tulane University School of Medicine, 1430 Tulane Avenue SL83, New Orleans, LA 70112, USA
| | - Ke-Jie Yin
- Pittsburgh Institute of Brain Disorders & Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
130
|
Zhu W, Tian L, Yue X, Liu J, Fu Y, Yan Y. LncRNA Expression Profiling of Ischemic Stroke During the Transition From the Acute to Subacute Stage. Front Neurol 2019; 10:36. [PMID: 30774621 PMCID: PMC6367239 DOI: 10.3389/fneur.2019.00036] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 01/11/2019] [Indexed: 11/16/2022] Open
Abstract
Ischemic stroke induces profound effects on the peripheral immune system, which may participate the infectious complications. However, the exact function and mechanism of immune reaction in stroke development are not well-elucidated. Recently, several long non-coding RNAs (LncRNAs) are reported to affect ischemic stroke process, especially the immunological response after stroke. In the present study, we investigated the profile of LncRNAs in human ischemic stroke during the transition from the acute to subacute stage, when the state of the peripheral immune system changes from activation to systemic immunosuppression. In this study, we analyzed the RNA-sequencing (RNA-seq) datasets obtained at two time points (24 h and 7 days) from the peripheral blood mononuclear cells of ischemic patients. Vascular risk factor-matched healthy adults were enrolled as controls. A total of 3,009 LncRNAs and 3,982 mRNAs were identified as differentially expressed 24 h after stroke. Furthermore, 2,034 LncRNAs and 1,641 mRNAs were detected to be differentially expressed on day 7. Bioinformatics analyses, including GO, KEGG pathway enrichment analysis, and network analysis, were performed for the identified dysregulated genes. Our study reveals that ischemic stroke can influence the expression of LncRNAs and mRNAs in the peripheral blood at both the acute and subacute stages; the level of LncRNAs in the antigen processing and presentation pathway was clearly upregulated at 24 h and had recovered to normal levels on day 7 after stroke. Moreover, inflammatory mediator regulation of TRP channels and GABAergic synapses were two specifically downregulated pathways on day 7 after stroke. Our findings provide a valuable resource for further study of the role of LncRNAs in peripheral immune system changes following ischemic stroke.
Collapse
Affiliation(s)
- Wenli Zhu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.,Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Lili Tian
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.,Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Xuanye Yue
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Jingyi Liu
- Department of Biochemistry, Smith College, Northampton, MA, United States
| | - Ying Fu
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yaping Yan
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.,Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| |
Collapse
|
131
|
LncRNA SNHG12 inhibits miR-199a to upregulate SIRT1 to attenuate cerebral ischemia/reperfusion injury through activating AMPK signaling pathway. Neurosci Lett 2019; 690:188-195. [DOI: 10.1016/j.neulet.2018.08.026] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 08/20/2018] [Accepted: 08/21/2018] [Indexed: 01/01/2023]
|
132
|
Montaldo P, Kaforou M, Pollara G, Hervás-Marín D, Calabria I, Panadero J, Pedrola L, Lally PJ, Oliveira V, Kage A, Atreja G, Mendoza J, Soe A, Pattnayak S, Shankaran S, Vento M, Herberg J, Thayyil S. Whole Blood Gene Expression Reveals Specific Transcriptome Changes in Neonatal Encephalopathy. Neonatology 2019; 115:68-76. [PMID: 30304723 PMCID: PMC6425817 DOI: 10.1159/000492420] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Accepted: 07/24/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Variable responses to hypothermic neuroprotection are related to the clinical heterogeneity of encephalopathic babies; hence better disease stratification may facilitate the development of individualized neuroprotective therapies. OBJECTIVES We examined if whole blood gene expression analysis can identify specific transcriptome profiles in neonatal encephalopathy. MATERIAL AND METHODS We performed next-generation sequencing on whole blood RNA from 12 babies with neonatal encephalopathy and 6 time-matched healthy term babies. Genes significantly differentially expressed between encephalopathic and control babies were identified. This set of genes was then compared to the host RNA response in septic neonates and subjected to pathway analysis. RESULTS We identified 950 statistically significant genes discriminating perfectly between healthy controls and neonatal encephalopathy. The major pathways in neonatal encephalopathy were axonal guidance signaling (p = 0.0009), granulocyte adhesion and diapedesis (p = 0.003), IL-12 signaling and production in macrophages (p = 0.003), and hypoxia-inducible factor 1α signaling (p = 0.004). There were only 137 genes in common between neonatal encephalopathy and bacterial sepsis sets. CONCLUSION Babies with neonatal encephalopathy have striking differences in gene expression profiles compared with healthy control and septic babies. Gene expression profiles may be useful for disease stratification and for developing personalized neuroprotective therapies.
Collapse
Affiliation(s)
- Paolo Montaldo
- Centre for Perinatal Neuroscience, Imperial College London, London, United .,Neonatal Unit, Università degli Studi della Campania "Luigi Vanvitelli,", Naples,
| | - Myrsini Kaforou
- Paediatric Infectious Diseases, Imperial College London, London, United Kingdom
| | - Gabriele Pollara
- Infection and Immunity, University College London, London, United Kingdom
| | | | | | | | - Laia Pedrola
- Health Research Institute La Fe, Valencia, Spain
| | - Peter J Lally
- Centre for Perinatal Neuroscience, Imperial College London, London, United Kingdom
| | - Vânia Oliveira
- Centre for Perinatal Neuroscience, Imperial College London, London, United Kingdom
| | - Anup Kage
- Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Gaurav Atreja
- Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Josephine Mendoza
- Centre for Perinatal Neuroscience, Imperial College London, London, United Kingdom
| | - Aung Soe
- Medway NHS Foundation Trust, Gillingham, United Kingdom
| | | | - Seetha Shankaran
- Neonatal-Perinatal Division, Wayne State University, Detroit, Michigan, USA
| | - Máximo Vento
- Health Research Institute La Fe, Valencia, Spain
| | - Jethro Herberg
- Paediatric Infectious Diseases, Imperial College London, London, United Kingdom
| | - Sudhin Thayyil
- Centre for Perinatal Neuroscience, Imperial College London, London, United Kingdom
| |
Collapse
|
133
|
Li X, Hou L, Cheng Z, Zhou S, Qi J, Cheng J. Overexpression of GAS5 inhibits abnormal activation of Wnt/β‐catenin signaling pathway in myocardial tissues of rats with coronary artery disease. J Cell Physiol 2018; 234:11348-11359. [PMID: 30511472 DOI: 10.1002/jcp.27792] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 10/31/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Xuexiang Li
- Department of Emergency Internal The Second Affiliated Hospital of Anhui Medical University Hefei China
| | - Linlin Hou
- Department of Cardiovascular The Second Municipal Hospital of Hefei Hefei China
| | - Ziping Cheng
- Department of Cardiovascular The First Affiliated Hospital of Anhui Medical University Hefei China
| | - Shu Zhou
- Department of Emergency Internal The Second Affiliated Hospital of Anhui Medical University Hefei China
| | - Jinwei Qi
- Department of Emergency Internal The Second Affiliated Hospital of Anhui Medical University Hefei China
| | - Jinglin Cheng
- Department of Emergency Internal The Second Affiliated Hospital of Anhui Medical University Hefei China
| |
Collapse
|
134
|
MALAT1 lncRNA Induces Autophagy and Protects Brain Microvascular Endothelial Cells Against Oxygen-Glucose Deprivation by Binding to miR-200c-3p and Upregulating SIRT1 Expression. Neuroscience 2018; 397:116-126. [PMID: 30496821 DOI: 10.1016/j.neuroscience.2018.11.024] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 11/11/2018] [Accepted: 11/15/2018] [Indexed: 01/27/2023]
Abstract
There is growing evidence that long noncoding RNAs (lncRNAs) play important roles in various biological processes. Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is one of the most highly upregulated lncRNAs in cerebral ischemia. However, the molecular mechanism of MALAT1 during cerebral ischemia is still unclear. This experiment is intended to investigate the role of MALAT1 in cerebral ischemia and its relationship with autophagy. Oxygen-glucose deprivation (OGD) in brain microvascular endothelial cells (BMECs) was used to mimic ischemic-like conditions in vitro. Real-time PCR, MTT, LDH assay and western blot were used to evaluate the levels of MALAT1, miR-200c-3p, SIRT1, cell survival and proteins. We found that the expression of MALAT1 and LC3BII were upregulated and p62 was downregulated by OGD. Inhibition of MALAT1 attenuated the autophagy activation and promoted cell death. We further revealed that MALAT1 downregulated the expression of miR-200c-3p by directly binding to miR-200c-3p. Furthermore, miR-200c-3p inhibited the autophagy and survival in BMECs by binding to 3'UTR of SIRT1, whereas MALAT1 overturned the inhibitory effect of miR-200c-3p. In conclusion, our study illuminated a novel Malat1-miR-200c-3p-SIRT1 pathway in the regulation of autophagy, in which, MALAT1 activates autophagy and promotes cell survival by binding to miR-200c-3p and upregulating SIRT1 expression.
Collapse
|
135
|
Yang X, Zi XH. LncRNA SNHG1 alleviates OGD induced injury in BMEC via miR-338/HIF-1α axis. Brain Res 2018; 1714:174-181. [PMID: 30414401 DOI: 10.1016/j.brainres.2018.11.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 11/02/2018] [Accepted: 11/03/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Brain microvascular endothelial cell (BMEC) is an important therapeutic target for the inhibition of brain vascular dysfunction in ischemic stroke. Expression of long non-coding RNA SNHG1 is reportedly upregulated in BMEC after OGD. The present study aims to investigate the potential roles of SNHG1 in OGD-induced injury in BMEC. METHODS Mice primary brain microvascular endothelial cells (BMEC) were cultured under "normal" or "oxygen/glucose-deprived" (OGD) conditions. The expression of SNHG1 and miR-338 after OGD were examined by qPCR. shRNA against SNHG1 was used to knockdown SNHG1 in BMEC. MiR-338-3p mimic and inhibitor were used to change the expression of miR-338 in BMEC. The relationship between SNHG1 and miR-338, and the relationship between miR-338 and HIF-1α were clarified using RNA pull-down and luciferase reporter gene assays, respectively. RESULTS SNHG1 and miR-338 were upregulated in OGD induced BMEC. SNHG1 silence aggravated OGD-induced cell apoptosis by down-regulating Bcl-2, HIF-1α and VEGF-A, and upregulating caspase 3 activity and Bax. MiR-338 was upregulated in SNHG1-silenced BMEC. RNA pull-down assays showed that SNHG1 could be directly bound by miR-338. In addition, miR-338 overexpression reduced cell viability in OGD while miR-338 inhibition protected BMEC against OGD-induced injury. Furthermore, luciferase reporter assay showed that HIF-1α was a direct target of miR-338. CONCLUSIONS SNHG1 exerted protective effects against OGD induced injury via sponging miR-338, thus upregulating HIF-1α/VEGF-A in BMEC.
Collapse
Affiliation(s)
- Xia Yang
- Department of Neurology, The Third Xiangya Hospital of Central South University, Changsha 410013, PR China
| | - Xiao-Hong Zi
- Department of Neurology, The Third Xiangya Hospital of Central South University, Changsha 410013, PR China.
| |
Collapse
|
136
|
Wang SW, Liu Z, Shi ZS. Non-Coding RNA in Acute Ischemic Stroke: Mechanisms, Biomarkers and Therapeutic Targets. Cell Transplant 2018; 27:1763-1777. [PMID: 30362372 PMCID: PMC6300774 DOI: 10.1177/0963689718806818] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Non-coding RNAs (ncRNAs) are a class of functional RNAs that regulate gene expression in a post-transcriptional manner. NcRNAs include microRNAs, long non-coding RNAs and circular RNAs. They are highly expressed in the brain and are involved in the regulation of physiological and pathophysiological processes, including cerebral ischemic injury, neurodegeneration, neural development, and plasticity. Stroke is one of the leading causes of death and physical disability worldwide. Acute ischemic stroke (AIS) occurs when brain blood flow stops, and that stoppage results in reduced oxygen and glucose supply to cells in the brain. In this article, we review the latest progress on ncRNAs in relation to their implications in AIS, as well as their potential as diagnostic and prognostic biomarkers. We also review ncRNAs acting as possible therapeutic targets in future precision medicine. Finally, we conclude with a brief discussion of current challenges and future directions for ncRNAs studies in AIS, which may facilitate the translation of ncRNAs research into clinical practice to improve clinical outcome of AIS.
Collapse
Affiliation(s)
- Sheng-Wen Wang
- 1 Department of Neurosurgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhong Liu
- 2 Department of Neurosurgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhong-Song Shi
- 1 Department of Neurosurgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.,3 RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,4 Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
137
|
Zhao M, Wang J, Xi X, Tan N, Zhang L. SNHG12 Promotes Angiogenesis Following Ischemic Stroke via Regulating miR-150/VEGF Pathway. Neuroscience 2018; 390:231-240. [PMID: 30193860 DOI: 10.1016/j.neuroscience.2018.08.029] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 08/21/2018] [Accepted: 08/26/2018] [Indexed: 12/21/2022]
Abstract
The promotion of angiogenesis is a promising therapeutic strategy for ischemic stroke. Many long noncoding RNAs (lncRNAs) are related to angiogenesis following ischemic stroke. LncRNA small nucleolar RNA host gene 12 (SNHG12) was upregulated in oxygen-glucose deprivation (OGD)-exposed primary brain microvascular endothelial cells and in microvessel from middle cerebral artery occlusion (MCAO) animal brains. However, the role and underlying mechanism of SNHG12 in ischemic stroke especially associated with angiogenesis process remain unknown. The expression of SNHG12 and miR-150 was determined in OGD-stimulated mouse brain microvascular endothelial (bEnd.3) cells. The role and mechanism of SNHG12 in the angiogenesis after ischemic stroke were investigated using gain- and loss-of function approaches both in OGD-exposed bEnd.3 cells and in MCAO mouse models. We found SNHG12 expression was elevated, whereas miR-150 reduced in OGD-exposed bEnd.3 cells. Upregulation of SNHG12 elevated, and SNHG12 knockdown suppressed the capillary-like tube formation, viability, migration, and VEGF expression in OGD-injured bEnd.3 cells. miR-150 mimic reversed, whereas anti-miR-150 further strengthened the effect of SNHG12 upregulation on the angiogenesis in bEnd.3 cells. Furthermore, we found that SNHG12 functioned as a competing endogenous RNA for miR-150 to regulate VEGF expression. Additionally, overexpression of SNHG12 improved the recovery of neurological function, reduced infarct volume and miR-150 expression, increased vascular density and VEGF expression in the infarct border zone of MCAO mice. In conclusion, SNHG12 promotes the angiogenesis following ischemic stroke via miR-150/VEGF pathway, which further clarified the mechanism of angiogenesis after ischemic stroke and provides a target for the treatment of this disease.
Collapse
Affiliation(s)
- Mian Zhao
- The Clinical Laboratory of Xi'an No.1 Hospital, Xi'an 710002, Shaanxi, China
| | - Jun Wang
- The Clinical Laboratory of Xi'an No.1 Hospital, Xi'an 710002, Shaanxi, China
| | - Xinlong Xi
- The Cardiac Intervention Room of Xi'an No.1 Hospital, Xi'an 710002, Shaanxi, China
| | - Nan Tan
- Department of Cadre's Ward of Xi'an No.1 Hospital, Xi'an 710002, Shaanxi, China
| | - Li Zhang
- Department of Clinical Laboratory, Shaanxi Friendship Hospital, Xi'an 710068, Shaanxi, China.
| |
Collapse
|
138
|
Wang Z, Wang R, Wang K, Liu X. Upregulated long noncoding RNA Snhg1 promotes the angiogenesis of brain microvascular endothelial cells after oxygen–glucose deprivation treatment by targeting miR-199a. Can J Physiol Pharmacol 2018; 96:909-915. [PMID: 29883549 DOI: 10.1139/cjpp-2018-0107] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Angiogenesis after ischemic stroke has important clinical significance, which stimulates endogenous recovery mechanisms and improves the neurological outcome. Enhancing angiogenesis may facilitate the function recovery from ischemic stroke. Recent studies have shown that aberrant expression of long noncoding RNAs (lncRNAs) is related to angiogenesis after ischemic stroke. Snhg1, a cancer-related lncRNA, has been reported to be upregulated after stroke. However, little is known about its role in stroke. In this study, we performed in vitro experiments to investigate the effects of Snhg1 on cell survival and angiogenesis and molecular mechanism in ischemic stroke. Oxygen–glucose deprivation/reoxygenation (OGD/R) was used to mimic ischemia/reperfusion injury in vitro. Sngh1 was increased in brain microvascular endothelial cells (BMECs) with the prolongation of exposure to OGD, and promoted BMEC survival under OGD/R condition, and angiogenesis after OGD/R treatment. miR-199a was identified and validated to be a direct target of Snhg1, and function effects of Snhg1 on BMEC survival and angiogenesis depended on miR-199a, which is involved in the regulation of hypoxia inducible factor and vascular endothelial cell growth factor expression. These findings contribute to a better understanding of the pathogenesis of ischemic stroke and facilitate the development of proangiogenesis therapy for this disease.
Collapse
Affiliation(s)
- Zhengfeng Wang
- Department of Neurosurgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Ruihua Wang
- Department of Nuclear Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Kai Wang
- Department of Neurosurgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Xianzhi Liu
- Department of Neurosurgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
139
|
Yang J, Gu L, Guo X, Huang J, Chen Z, Huang G, Kang Y, Zhang X, Long J, Su L. LncRNA ANRIL Expression and ANRIL Gene Polymorphisms Contribute to the Risk of Ischemic Stroke in the Chinese Han Population. Cell Mol Neurobiol 2018; 38:1253-1269. [PMID: 29881905 PMCID: PMC11481959 DOI: 10.1007/s10571-018-0593-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 05/23/2018] [Indexed: 12/13/2022]
Abstract
The aim of the present study was to explore the role of lncRNA ANRIL in the pathogenesis of ischemic stroke (IS) and coronary artery disease (CAD) and to determine the association between ANRIL variants and the genetic susceptibility of IS and CAD in the Chinese Han population. A genetic association study including 550 IS patients, 550 CAD patients, and 550 healthy controls was conducted. The expression levels of lncRNA ANRIL, CDKN2A, and CDKN2B were detected using qRT-PCR. Genotyping was performed by Sequenom MassARRAY on an Agena platform. Our study showed that IS patients had an increased lncRNA ANRIL expression (P = 0.002) and a decreased CDKN2A expression (P < 0.001) compared with normal controls. A significant difference with regard to the genotype distribution of rs2383207 was found between male IS patients and controls (P = 0.011). The minor allele of rs2383207 significantly increased the IS risk under a recessive model (OR = 1.52, 95% CI = 1.05-2.21, P = 0.027). The minor allele of rs1333049 was significantly associated with the risk of IS among the male patients under a recessive model (OR = 1.56, 95% CI = 1.04-2.35, P = 0.031). However, no significant association was found between the ANRIL variants and the risk of CAD (all P > 0.050). In addition, we found a decreased lncRNA ANRIL expression in IS patients who carried the GG genotype of rs1333049 compared with IS patients who carried the CC or CG genotype (P = 0.041). In summary, we found that IS patients had an increased lncRNA ANRIL expression and a decreased CDKN2A expression compared with the controls, which might play an impellent role in pathological processes of IS. The ANRIL variants rs2383207 and rs1333049 were significantly associated with the risk of IS among males but not females in the Chinese Han population.
Collapse
Affiliation(s)
- Jialei Yang
- School of Public Health of Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Lian Gu
- First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi, People's Republic of China
| | - Xiaojing Guo
- School of Public Health of Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Jiao Huang
- School of Public Health of Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Zhaoxia Chen
- School of Public Health of Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Guifeng Huang
- School of Public Health of Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Yiwen Kang
- School of Public Health of Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Xiaoting Zhang
- School of Public Health of Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Jianxiong Long
- School of Public Health of Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China.
| | - Li Su
- School of Public Health of Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China.
| |
Collapse
|
140
|
Zhang L, Luo X, Chen F, Yuan W, Xiao X, Zhang X, Dong Y, Zhang Y, Liu Y. LncRNA SNHG1 regulates cerebrovascular pathologies as a competing endogenous RNA through HIF-1α/VEGF signaling in ischemic stroke. J Cell Biochem 2018; 119:5460-5472. [PMID: 29377234 DOI: 10.1002/jcb.26705] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 01/23/2018] [Indexed: 01/01/2023]
Abstract
Studies have shown that long noncoding ribonucleic acids (lncRNAs) play critical roles in multiple biologic processes. However, the Small Nucleolar RNA Host Gene 1 (SNHG1) function and underlying molecular mechanisms in ischemic stroke have not yet been reported. In the present study, we found that SNHG1 expression was remarkably increased both in isolated cerebral micro-vessels of a middle cerebral artery occlusion (MCAO) mice model, and in oxygen-glucose deprivation (OGD)-cultured mice brain micro-vascular endothelial cells (BMECs), meanwhile, the SNHG1 level was negatively correlated with miR-18a in MCAO mice. Mechanistically, SNHG1 inhibition presents larger brain infarct size and worsens neurological scores in MCAO mice. Consistent with the in vivo findings, SNHG1 inhibition also significantly increased caspase-3 activity and cell apoptosis in OGD-cultured BMECs. Furthermore, we found that SNHG1 functions as a competing endogenous RNA (ceRNA) for miR-18a, thereby regulating the de-repression of its endogenous target HIF-1α and promoting BMEC survival through HIF-1α/VEGF signaling. This study found a neuroprotective effect of SNHG1 mediated by HIF-1α/VEGF signaling through acting as a ceRNA for miR-18a. These findings reveal a novel function of SNHG1, which contributes to an extensive understanding of ischemic stroke and provides novel therapeutic options for this disease.
Collapse
MESH Headings
- Animals
- Apoptosis
- Cells, Cultured
- Disease Models, Animal
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Infarction, Middle Cerebral Artery/genetics
- Infarction, Middle Cerebral Artery/metabolism
- Infarction, Middle Cerebral Artery/pathology
- Male
- Mice
- Mice, Inbred C57BL
- MicroRNAs
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- RNA/metabolism
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Signal Transduction
- Stroke/genetics
- Stroke/metabolism
- Stroke/pathology
- Vascular Endothelial Growth Factor A/genetics
- Vascular Endothelial Growth Factor A/metabolism
Collapse
Affiliation(s)
- Lin Zhang
- Department of Neurology, Xi'an Central Hospital, The Affiliated Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, China
| | - Xianliang Luo
- Department of Medicine, Xi 'an Sixth Hospital, Xi'an, China
| | - Feng Chen
- Department of Neurosurgery, Jinhua Central Hospital, Jinhua, China
| | - Wei Yuan
- Department of Cardiology, Xi'an North Hospital, Xi'an, China
| | - Xinli Xiao
- Institute of neurobiology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Xiaohua Zhang
- Institute of neurobiology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Yaru Dong
- Department of Neurology, Xi'an Central Hospital, Xi'an, China
| | - Yuanxiao Zhang
- Department of Neurology, Xi'an Central Hospital, Xi'an, China
| | - Yong Liu
- Institute of neurobiology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| |
Collapse
|
141
|
Unique MicroRNAs Signature of Lymphocyte of Yang and Yin Syndromes in Acute Ischemic Stroke Patients. Chin J Integr Med 2018; 25:590-597. [PMID: 29926387 DOI: 10.1007/s11655-018-2843-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2016] [Indexed: 01/16/2023]
Abstract
OBJECTIVE To identify the differentially expressed microRNAs (miRNAs) profiles of yang and yin syndromes in patients with acute ischemic stroke, and to provide the molecular basis of the classification of these two syndrome types in acute ischemic stroke patients. METHODS A microarray assay was performed to assess the expression pattern of miRNAs in the lymphocyte of acute ischemic stroke patients. Target genes for the deregulated miRNAs were predicated using the online bioinformatic algorithms and functional annotation via Kyoto encyclopedia of genes and genomes pathway analysis for miRNAs predicted targets was carried out. Based on the predicted target genes of differentially expressed miRNAs, the miRNA-gene-network and miRNA-pathway-network were constructed. RESULTS Yang score based on tongue texture, urine, dejecta, and appearance, etc. showed that clinical symptoms were distinct between yang and yin syndromes. There were significantly higher total leukocyte number and lower total protein level in patients with yang syndrome compared with those in patients with yin syndrome (P<0.05). Comprehensive miRNA analysis identified 36 unique down-regulated miRNAs in yang syndrome group, and 20 unique down-regulated and 2 unique up-regulated miRNAs in yin syndrome group. The key regulatory miRNAs, gene, and pathways in the yang syndrome were hsa-miR-93-5p and -320b, enabled homolog, the metabolic pathways and mitogen-activated protein kinase signaling pathways, respectively, while those in the yin syndrome were hsa-miR-424-5p and -106b-5p, CNOT4, hepatitis B and pathways in cancer, respectively. CONCLUSION These results offered insight into the molecular basis underlying the different pathogenesis of yang or yin syndrome, providing clues for the individualized therapeutic strategies of acute ischemic stroke.
Collapse
|
142
|
Zhang X, Tang X, Hamblin MH, Yin KJ. Long Non-Coding RNA Malat1 Regulates Angiogenesis in Hindlimb Ischemia. Int J Mol Sci 2018; 19:ijms19061723. [PMID: 29891768 PMCID: PMC6032369 DOI: 10.3390/ijms19061723] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 05/28/2018] [Accepted: 06/05/2018] [Indexed: 11/29/2022] Open
Abstract
Angiogenesis is a complex process that depends on the delicate regulation of gene expression. Dysregulation of transcription during angiogenesis often leads to various human diseases. Emerging evidence has recently begun to show that long non-coding RNAs (lncRNAs) may mediate angiogenesis in both physiological and pathological conditions; concurrently, underlying molecular mechanisms are largely unexplored. Previously, our lab identified metastasis associates lung adenocarcinoma transcript 1 (Malat1) as an oxygen-glucose deprivation (OGD)-responsive endothelial lncRNA. Here we reported that genetic deficiency of Malat1 leads to reduced blood vessel formation and local blood flow perfusion in mouse hind limbs at one to four weeks after hindlimb ischemia. Malat1 and vascular endothelial growth factor receptor 2 (VEGFR2) levels were found to be increased in both cultured mouse primary skeletal muscle microvascular endothelial cells (SMMECs) after 16 h OGD followed by 24 h reperfusion and in mouse gastrocnemius muscle that underwent hindlimb ischemia followed by 28 days of reperfusion. Moreover, Malat1 silencing by locked nucleic acid (LNA)-GapmeRs significantly reduced tube formation, cell migration, and cell proliferation in SMMEC cultures. Mechanistically, RNA subcellular isolation and RNA-immunoprecipitation experiments demonstrate that Malat1 directly targets VEGFR2 to facilitate angiogenesis. The results suggest that Malat1 regulates cell-autonomous angiogenesis through direct regulation of VEGFR2.
Collapse
Affiliation(s)
- Xuejing Zhang
- Pittsburgh Institute of Brain Disorders & Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| | - Xuelian Tang
- Pittsburgh Institute of Brain Disorders & Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| | - Milton H Hamblin
- Department of Pharmacology, Tulane University School of Medicine, 1430 Tulane Avenue SL83, New Orleans, LA 70112, USA.
| | - Ke-Jie Yin
- Pittsburgh Institute of Brain Disorders & Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| |
Collapse
|
143
|
Yu B, Wang S. Angio-LncRs: LncRNAs that regulate angiogenesis and vascular disease. Theranostics 2018; 8:3654-3675. [PMID: 30026873 PMCID: PMC6037039 DOI: 10.7150/thno.26024] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 05/16/2018] [Indexed: 12/15/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) represent a large subgroup of RNAs that are longer than 200 nucleotides and have no apparent protein coding potential. They have diverse functions in different biological processes by regulating chromatin remodeling or protein translation. This review summarizes the recent progress of lncRNAs in angiogenesis and vascular diseases. A general overview of lncRNA functional mechanisms will be introduced. A list of lncRNAs, which are termed "Angio-LncRs", including MALAT1, MANTIS, PUNISHER, MEG3, MIAT, SENCR and GATA6-AS, will be discussed regarding their expression, regulation, function and mechanism of action in angiogenesis. Implications of lncRNAs in vascular diseases, such as atherosclerosis, hypertension, vascular retinopathies and tumor angiogenesis will also be discussed.
Collapse
Affiliation(s)
- Bo Yu
- Department of Cell and Molecular Biology, Tulane University, 2000 Percival Stern Hall, 6400 Freret Street, New Orleans, LA, 70118, USA
| | - Shusheng Wang
- Department of Cell and Molecular Biology, Tulane University, 2000 Percival Stern Hall, 6400 Freret Street, New Orleans, LA, 70118, USA
- Department of Ophthalmology, Tulane University School of Medicine, 1430 Tulane Avenue, SL-69, New Orleans, LA 70112, USA
| |
Collapse
|
144
|
Bhattarai S, Aly A, Garcia K, Ruiz D, Pontarelli F, Dharap A. Deep Sequencing Reveals Uncharted Isoform Heterogeneity of the Protein-Coding Transcriptome in Cerebral Ischemia. Mol Neurobiol 2018; 56:1035-1043. [PMID: 29862458 DOI: 10.1007/s12035-018-1147-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 05/22/2018] [Indexed: 11/30/2022]
Abstract
Gene expression in cerebral ischemia has been a subject of intense investigations for several years. Studies utilizing probe-based high-throughput methodologies such as microarrays have contributed significantly to our existing knowledge but lacked the capacity to dissect the transcriptome in detail. Genome-wide RNA-sequencing (RNA-seq) enables comprehensive examinations of transcriptomes for attributes such as strandedness, alternative splicing, alternative transcription start/stop sites, and sequence composition, thus providing a very detailed account of gene expression. Leveraging this capability, we conducted an in-depth, genome-wide evaluation of the protein-coding transcriptome of the adult mouse cortex after transient focal ischemia at 6, 12, or 24 h of reperfusion using RNA-seq. We identified a total of 1007 transcripts at 6 h, 1878 transcripts at 12 h, and 1618 transcripts at 24 h of reperfusion that were significantly altered as compared to sham controls. With isoform-level resolution, we identified 23 splice variants arising from 23 genes that were novel mRNA isoforms. For a subset of genes, we detected reperfusion time-point-dependent splice isoform switching, indicating an expression and/or functional switch for these genes. Finally, for 286 genes across all three reperfusion time-points, we discovered multiple, distinct, simultaneously expressed and differentially altered isoforms per gene that were generated via alternative transcription start/stop sites. Of these, 165 isoforms derived from 109 genes were novel mRNAs. Together, our data unravel the protein-coding transcriptome of the cerebral cortex at an unprecedented depth to provide several new insights into the flexibility and complexity of stroke-related gene transcription and transcript organization.
Collapse
Affiliation(s)
- Sunil Bhattarai
- Laboratory for Stroke Research and Noncoding RNA Biology, JFK Neuroscience Institute, HackensackMeridian Health JFK Medical Center, 65 James Street, Edison, NJ, 08820, USA
| | - Ahmed Aly
- Laboratory for Stroke Research and Noncoding RNA Biology, JFK Neuroscience Institute, HackensackMeridian Health JFK Medical Center, 65 James Street, Edison, NJ, 08820, USA
| | - Kristy Garcia
- Laboratory for Stroke Research and Noncoding RNA Biology, JFK Neuroscience Institute, HackensackMeridian Health JFK Medical Center, 65 James Street, Edison, NJ, 08820, USA
| | - Diandra Ruiz
- Laboratory for Stroke Research and Noncoding RNA Biology, JFK Neuroscience Institute, HackensackMeridian Health JFK Medical Center, 65 James Street, Edison, NJ, 08820, USA
| | - Fabrizio Pontarelli
- Laboratory for Stroke Research and Noncoding RNA Biology, JFK Neuroscience Institute, HackensackMeridian Health JFK Medical Center, 65 James Street, Edison, NJ, 08820, USA
| | - Ashutosh Dharap
- Laboratory for Stroke Research and Noncoding RNA Biology, JFK Neuroscience Institute, HackensackMeridian Health JFK Medical Center, 65 James Street, Edison, NJ, 08820, USA.
| |
Collapse
|
145
|
Chen S, Yin W, Bi K, Lu B. MicroRNA‑497 attenuates cerebral infarction in patients via the TLR4 and CREB signaling pathways. Int J Mol Med 2018; 42:547-556. [PMID: 29620142 DOI: 10.3892/ijmm.2018.3611] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 03/01/2018] [Indexed: 11/05/2022] Open
Abstract
The aim of the present study was to investigate the function and mechanism of microRNA‑497 (miRNA/miR‑149) in the regulation of cerebral infarction. In patients with cerebral infarction, the serum of microRNA‑497 expression was upregulated compared with that in healthy controls. In N2A cells, overexpression of miR‑497 induced cell proliferation, decreased apoptosis and caspase‑3 and caspase‑9 activities, and suppressed Bax protein expression compared with that in the negative control group. Overexpression of miR‑497 reduced inflammation factors, and suppressed the Toll‑like receptor 4 (TLR4), myeloid differentiation primary response protein MyD88 (MyD88) and nuclear factor‑κB (NF‑κB) protein expression of the N2A cells. Next, miR‑497 overexpression suppressed the protein expression of interleukin‑1 receptor associated kinase (IRAK1) and phosphorylated cyclic AMP response element binding protein (p-CREB) in the N2A cells. Following miR‑497 overexpression, TLR4 inhibitor was found to suppress the inflammation factors, suppress the TLR4, MyD88 and NF‑κB protein expression, and reduce the IRAK1 and p‑CREB protein expression of the N2A cells. Lastly, CREB inhibitor also suppressed p‑CREB protein expression, induced cell proliferation, decreased apoptosis and caspase‑3 and caspase‑9 activities, and suppressed Bax protein expression in the N2A cells following miR‑497 overexpression. Taken together, these data demonstrated that miR‑497 attenuated cerebral infarction in patients by regulating the TLR4 and CREB signaling pathways.
Collapse
Affiliation(s)
- Si Chen
- Department of First Neurosurgery, Tangshan Worker Hospital, Tangshan, Hebei 063000, P.R. China
| | - Wenwei Yin
- Department of First Neurosurgery, Tangshan Worker Hospital, Tangshan, Hebei 063000, P.R. China
| | - Kun Bi
- Department of First Neurosurgery, Tangshan Worker Hospital, Tangshan, Hebei 063000, P.R. China
| | - Bin Lu
- Department of First Neurosurgery, Tangshan Worker Hospital, Tangshan, Hebei 063000, P.R. China
| |
Collapse
|
146
|
Ren W, Yang X. Pathophysiology of Long Non-coding RNAs in Ischemic Stroke. Front Mol Neurosci 2018; 11:96. [PMID: 29651234 PMCID: PMC5884949 DOI: 10.3389/fnmol.2018.00096] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 03/12/2018] [Indexed: 12/12/2022] Open
Abstract
Stroke is a neurological disease with high disability and fatality rates, and ischemic stroke accounts for 75% of all stroke cases. The underlying pathophysiologic processes of ischemic stroke include oxidative stress, toxicity of excitatory amino acids, excess calcium ions, increased apoptosis and inflammation. Long non-coding RNAs (lncRNAs) may participate in the regulation of the pathophysiologic processes of ischemic stroke as indicated by altered expression of lncRNAs in blood samples of acute ischemic stroke patients, animal models of focal cerebral ischemia and oxygen-glucose deprivation (OGD) cell models. Because of the potentially important role, lncRNAs might be useful as biomarkers for the diagnosis, treatment and prognosis of ischemic stroke. This article reviews the functions of lncRNAs in different pathophysiology events of ischemic stroke with a focus on specific lncRNAs that may underlie ischemic stroke pathophysiology and that could therefore serve as potential diagnostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Weimin Ren
- Center Laboratory, Jinshan Hospital, Fudan University, Shanghai, China
| | - Xiaobo Yang
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
147
|
Wang G, Wu Y, Zhu Y. Mechanism of MALAT1 preventing apoptosis of vascular endothelial cells induced by oxygen–glucose deficiency and reoxidation. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:798-805. [PMID: 29575939 DOI: 10.1080/21691401.2018.1436065] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Guoping Wang
- Department of Neurology, Anhui Provincial Hospital, Anhui Medical University, Hefei, PR China
- Department of Neurology, Anhui Provincial Hospital, The First Affiliated Hospital of University of Science and Technology of China, Hefei, PR China
| | - Yuanbo Wu
- Department of Neurology, Anhui Provincial Hospital, Anhui Medical University, Hefei, PR China
- Department of Neurology, Anhui Provincial Hospital, The First Affiliated Hospital of University of Science and Technology of China, Hefei, PR China
| | - Yuyou Zhu
- Department of Neurology, Anhui Provincial Hospital, Anhui Medical University, Hefei, PR China
- Department of Neurology, Anhui Provincial Hospital, The First Affiliated Hospital of University of Science and Technology of China, Hefei, PR China
| |
Collapse
|
148
|
Aryankalayil MJ, Chopra S, Levin J, Eke I, Makinde A, Das S, Shankavaram U, Vanpouille-Box C, Demaria S, Coleman CN. Radiation-Induced Long Noncoding RNAs in a Mouse Model after Whole-Body Irradiation. Radiat Res 2018; 189:251-263. [PMID: 29309266 PMCID: PMC5967844 DOI: 10.1667/rr14891.1] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Long noncoding RNAs (lncRNAs) are emerging as key molecules in regulating many biological processes and have been implicated in development and disease pathogenesis. Biomarkers of cancer and normal tissue response to treatment are of great interest in precision medicine, as well as in public health and medical management, such as for assessment of radiation injury after an accidental or intentional exposure. Circulating and functional RNAs, including microRNAs (miRNAs) and lncRNAs, in whole blood and other body fluids are potential valuable candidates as biomarkers. Early prediction of possible acute, intermediate and delayed effects of radiation exposure enables timely therapeutic interventions. To address whether long noncoding RNAs (lncRNAs) could serve as biomarkers for radiation biodosimetry we performed whole genome transcriptome analysis in a mouse model after whole-body irradiation. Differential lncRNA expression patterns were evaluated at 16, 24 and 48 h postirradiation in total RNA isolated from whole blood of mice exposed to 1, 2, 4, 8 and 12 Gy of X rays. Sham-irradiated animals served as controls. Significant alterations in the expression patterns of lncRNAs were observed after different radiation doses at the various time points. We identified several radiation-induced lncRNAs known for DNA damage response as well as immune response. Long noncoding RNA targets of tumor protein 53 (P53), Trp53cor1, Dino, Pvt1 and Tug1 and an upstream regulator of p53, Meg3, were altered in response to radiation. Gm14005 ( Morrbid) and Tmevpg1 were regulated by radiation across all time points and doses. These two lncRNAs have important potential as blood-based radiation biomarkers; Gm14005 ( Morrbid) has recently been shown to play a key role in inflammatory response, while Tmevpg1 has been implicated in the regulation of interferon gamma. Precise molecular biomarkers, likely involving a diverse group of inducible molecules, will not only enable the development and effective use of medical countermeasures but may also be used to detect and circumvent or mitigate normal tissue injury in cancer radiotherapy.
Collapse
Affiliation(s)
| | - Sunita Chopra
- Radiation Oncology Branch, Center for Cancer Research, NMional Cancer Institute, Bethesda, Maryland
| | - Joel Levin
- Radiation Oncology Branch, Center for Cancer Research, NMional Cancer Institute, Bethesda, Maryland
| | - Iris Eke
- Radiation Oncology Branch, Center for Cancer Research, NMional Cancer Institute, Bethesda, Maryland
| | - Adeola Makinde
- Radiation Oncology Branch, Center for Cancer Research, NMional Cancer Institute, Bethesda, Maryland
| | - Shaoli Das
- Radiation Oncology Branch, Center for Cancer Research, NMional Cancer Institute, Bethesda, Maryland
| | - Uma Shankavaram
- Radiation Oncology Branch, Center for Cancer Research, NMional Cancer Institute, Bethesda, Maryland
| | | | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medicine, New York, New York
| | - C. Norman Coleman
- Radiation Oncology Branch, Center for Cancer Research, NMional Cancer Institute, Bethesda, Maryland
- Radiation Research Progrnm, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| |
Collapse
|
149
|
Long Noncoding RNAs: New Players in Ischaemia-Reperfusion Injury. Heart Lung Circ 2018; 27:322-332. [DOI: 10.1016/j.hlc.2017.09.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 09/08/2017] [Accepted: 09/19/2017] [Indexed: 12/22/2022]
|
150
|
Bao MH, Szeto V, Yang BB, Zhu SZ, Sun HS, Feng ZP. Long non-coding RNAs in ischemic stroke. Cell Death Dis 2018; 9:281. [PMID: 29449542 PMCID: PMC5833768 DOI: 10.1038/s41419-018-0282-x] [Citation(s) in RCA: 230] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/27/2017] [Accepted: 12/27/2017] [Indexed: 12/31/2022]
Abstract
Stroke is one of the leading causes of mortality and disability worldwide. Uncovering the cellular and molecular pathophysiological processes in stroke have been a top priority. Long non-coding (lnc) RNAs play critical roles in different kinds of diseases. In recent years, a bulk of aberrantly expressed lncRNAs have been screened out in ischemic stroke patients or ischemia insulted animals using new technologies such as RNA-seq, deep sequencing, and microarrays. Nine specific lncRNAs, antisense non-coding RNA in the INK4 locus (ANRIL), metastasis-associate lung adenocarcinoma transcript 1 (MALAT1), N1LR, maternally expressed gene 3 (MEG3), H19, CaMK2D-associated transcript 1 (C2dat1), Fos downstream transcript (FosDT), small nucleolar RNA host gene 14 (SNHG14), and taurine-upregulated gene 1 (TUG1), were found increased in cerebral ischemic animals and/or oxygen-glucose deprived (OGD) cells. These lncRNAs were suggested to promote cell apoptosis, angiogenesis, inflammation, and cell death. Our Gene Ontology (GO) enrichment analysis predicted that MEG3, H19, and MALAT1 might also be related to functions such as neurogenesis, angiogenesis, and inflammation through mechanisms of gene regulation (DNA transcription, RNA folding, methylation, and gene imprinting). This knowledge may provide a better understanding of the functions and mechanisms of lncRNAs in ischemic stroke. Further elucidating the functions and mechanisms of these lncRNAs in biological systems under normal and pathological conditions may lead to opportunities for identifying biomarkers and novel therapeutic targets of ischemic stroke.
Collapse
Affiliation(s)
- Mei-Hua Bao
- Department of Anatomy, Histology and Embryology, Institute of Neuroscience, Changsha Medical University, Changsha, 410219, China
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Vivian Szeto
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Burton B Yang
- Sunnybrook Research Institute and Department of Laboratory Medicine and Pathology, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Shu-Zhen Zhu
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Hong-Shuo Sun
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| | - Zhong-Ping Feng
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|