1
|
Cai X, Wang X, Huang Y, Rao X. Molecular mechanism of wedelolactone inhibits high glucose-induced human retinal vascular endothelial cells injury through regulating miR-190 expression. Medicine (Baltimore) 2024; 103:e37388. [PMID: 38788037 PMCID: PMC11124660 DOI: 10.1097/md.0000000000037388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/05/2024] [Indexed: 05/26/2024] Open
Abstract
To investigate the effects and molecular mechanisms of wedelolactone (WEL) on high glucose-induced injury of human retinal vascular endothelial cells (HRECs). The cell injury model was established by incubating HRECs with 30 mmol/L glucose for 24 hour. HRECs were divided into control (Con) group, high glucose (HG) group, HG + WEL-low dose (L) group, HG + WEL-medium dose (M), HG + WEL-high dose (H) group, HG + miR-NC group, HG + miR-190 group, HG + WEL + antimiR-NC group, HG + WEL + antimiR-190 group. The kit detects cellular reactive oxygen species (ROS), superoxide dismutase (SOD), and malondialdehyde (MDA) content; cell apoptosis was analyzed by flow cytometry; miR-190 expression was detected by real-time quantitative PCR (RT-qPCR). Compared with Con group, the levels of ROS and MDA in the HG group were significantly increased (P < .01), the SOD activity and the expression of miR-190 expression were significantly decreased (P < .05), and the apoptosis rate was significantly increased (P < .01). Compared with HG group, the levels of ROS and MDA in HG + WEL-L group, HG + WEL-M group and HG + WEL-H group were significantly decreased (P < .05), SOD activity and miR-190 expression were significantly increased (P < .05), and apoptosis rate was significantly reduced (P < .05). Compared with the HG + miR-NC group, the levels of ROS and MDA in HG + miR-190 group were significantly reduced (P < .01), SOD activity was significantly increased (P < .01), and apoptosis rate was significantly reduced (P < .05). Compared with the HG + WEL + antimiR-NC group, the ROS level and MDA content in the HG + WEL + antimiR-190 group were significantly increased (P < .05), SOD activity was significantly decreased (P < .05), and apoptosis rate was significantly increased (P < .05). Wedelolactone can attenuate high glucose-induced HRECs apoptosis and oxidative stress by up-regulating miR-190 expression.
Collapse
Affiliation(s)
- Xiaojie Cai
- Department of Nursing, People’s Hospital of Chengyang District, Qingdao City, Shandong Province, China
| | - Xiao Wang
- Department of Ultrasound, People’s Hospital of Chengyang District, Qingdao City, Shandong Province, China
| | - Yuping Huang
- Center of Health Management, People’s Hospital of Chengyang District, Qingdao City, Shandong Province, China
| | - Xiaopang Rao
- Department of Ultrasound, People’s Hospital of Chengyang District, Qingdao City, Shandong Province, China
| |
Collapse
|
2
|
Jiang Y, Ma C, Guan Y, Yang W, Yu J, Shi H, Ding Z, Zhang Z. Long noncoding RNA KCNQ1OT1 aggravates cerebral infarction by regulating PTBT1/SIRT1 via miR-16-5p. J Neuropathol Exp Neurol 2024; 83:276-288. [PMID: 38324733 DOI: 10.1093/jnen/nlae005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2024] Open
Abstract
Cerebral infarction (CI) is one of the leading causes of disability and death. LncRNAs are key factors in CI progression. Herein, we studied the function of long noncoding RNA KCNQ1OT1 in CI patient plasma samples and in CI models. Quantitative real-time PCR and Western blotting tested gene and protein expressions. The interactions of KCNQ1OT1/PTBP1 and miR-16-5p were analyzed using dual-luciferase reporter and RNA immunoprecipitation assays; MTT assays measured cell viability. Cell migration and angiogenesis were tested by wound healing and tube formation assays. Pathological changes were analyzed by triphenyltetrazolium chloride and routine staining. We found that KCNQ1OT1 and PTBP1 were overexpressed and miR-16-5p was downregulated in CI patient plasma and in oxygen-glucose deprived (OGD) induced mouse brain microvascular endothelial (bEnd.3) cells. KCNQ1OT1 knockdown suppressed pro-inflammatory cytokine production and stimulated angiogenic responses in OGD-bEnd.3 cells. KCNQ1OT1 upregulated PTBP1 by sponging miR-16-5p. PTBP1 overexpression or miR-16-5p inhibition attenuated the effects of KCNQ1OT1 knockdown. PTBP1 silencing protected against OGD-bEnd.3 cell injury by enhancing SIRT1. KCNQ1OT1 silencing or miR-16-5p overexpression also alleviated ischemic injury in a mice middle cerebral artery occlusion model. Thus, KCNQ1OT1 silencing alleviates CI by regulating the miR-16-5p/PTBP1/SIRT1 pathway, providing a theoretical basis for novel therapeutic strategies targeting CI.
Collapse
Affiliation(s)
- Yuanming Jiang
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Chi Ma
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yuxiu Guan
- Department of Neurology, The Fifth Affiliated Hospital of Harbin Medical University, Daqing, Heilongjiang, China
| | - Wenqi Yang
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Jiaqi Yu
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Hanfei Shi
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Zihang Ding
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Zhuobo Zhang
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
3
|
Alikiaii B, Bagherniya M, Askari G, Rajendram R, Sahebkar A. MicroRNA Profiles in Critically Ill Patients. Curr Med Chem 2024; 31:6801-6825. [PMID: 37496239 DOI: 10.2174/0929867331666230726095222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 05/19/2023] [Accepted: 06/01/2023] [Indexed: 07/28/2023]
Abstract
The use of biomarkers to expedite diagnosis, prognostication, and treatment could significantly improve patient outcomes. The early diagnosis and treatment of critical illnesses can greatly reduce mortality and morbidity. Therefore, there is great interest in the discovery of biomarkers for critical illnesses. Micro-ribonucleic acids (miRNAs) are a highly conserved group of non-coding RNA molecules. They regulate the expression of genes involved in several developmental, physiological, and pathological processes. The characteristics of miRNAs suggest that they could be versatile biomarkers. Assay panels to measure the expression of several miRNAs could facilitate clinical decision-- making for a range of diseases. We have, in this paper, reviewed the current understanding of the role of miRNAs as biomarkers in critically ill patients.
Collapse
Affiliation(s)
- Babak Alikiaii
- Anesthesia and Critical Care Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Bagherniya
- Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Gholamreza Askari
- Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Rajkumar Rajendram
- Department of Medicine, King Abdulaziz Medical City, King Abdulaziz International Medical Research Center, Ministry of National Guard - Health Affairs, Riyadh, Saudi Arabia
- College of Medicine, King Saud bin Abdulaziz University of Health Sciences, Riyadh, Saudi Arabia
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
4
|
Hu T, Li D, Fan T, Zhao X, Chen Z. CircCRIM1/microRNA-141-3p/thioredoxin-binding protein axis mediates neuronal apoptosis after cerebral ischemia-reperfusion. ENVIRONMENTAL TOXICOLOGY 2023; 38:2845-2856. [PMID: 37565716 DOI: 10.1002/tox.23916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/06/2023] [Accepted: 07/21/2023] [Indexed: 08/12/2023]
Abstract
Numerous studies have indicated enrichment of circular RNA (circRNA) in the brain takes on a momentous role in cerebral ischemia-reperfusion (CIR) injury. A recent study discovered a novel circCRIM1, was highly expressed in the middle cerebral artery occlusion-reperfusion (MCAO/R) model. Nevertheless, its specific biological function remained unknown. The study was to explore circCRIM1 in CIR-induced neuronal apoptosis. As measured, circCRIM1 and TXNIP were up-regulated, while miR-141-3p was down-regulated in MCAO/R mouse model and OGD/R SH-SY5Y cells. Depleting circCRIM1 reduced the number of apoptotic neurons in MCAO/R rats, increased the number of Nissl bodies, prevented reactive oxygen species production and oxidative stress imbalance in brain tissues, repressed cleaved caspase-3, Bax, and Cyto C protein levels and increased Bcl-2 levels. Overexpression of circCRIM1 further repressed neuronal activity and accelerated apoptosis in OGD/R model, disrupted redox balance. Depleting circCRIM1 had the opposite effect in OGD/R model. Knocking down miR-141-3p or TXNIP weakened the effects of knocking down circCRIM1 or overexpressing circCRIM1, separately. Mechanistically, circCRIM1 exerted an active role in CIR injury via miR-141-3p to mediate TXNIP. All in all, the circCRIM1/miR-141-3p/TXNIP axis might be a latent therapeutic target for CIR injury.
Collapse
Affiliation(s)
- Teng Hu
- Department of Neurological Intervention, Dalian Municipal Central Hospital, Dalian City, China
| | - Di Li
- Department of Neurological Intervention, Dalian Municipal Central Hospital, Dalian City, China
| | - TiePing Fan
- Department of Neurological Intervention, Dalian Municipal Central Hospital, Dalian City, China
| | - XuSheng Zhao
- Department of Neurological Intervention, Dalian Municipal Central Hospital, Dalian City, China
| | - ZhongJun Chen
- Department of Neurological Intervention, Dalian Municipal Central Hospital, Dalian City, China
| |
Collapse
|
5
|
Rashidi SK, Kalirad A, Rafie S, Behzad E, Dezfouli MA. The role of microRNAs in neurobiology and pathophysiology of the hippocampus. Front Mol Neurosci 2023; 16:1226413. [PMID: 37727513 PMCID: PMC10506409 DOI: 10.3389/fnmol.2023.1226413] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 08/11/2023] [Indexed: 09/21/2023] Open
Abstract
MicroRNAs (miRNAs) are short non-coding and well-conserved RNAs that are linked to many aspects of development and disorders. MicroRNAs control the expression of genes related to different biological processes and play a prominent role in the harmonious expression of many genes. During neural development of the central nervous system, miRNAs are regulated in time and space. In the mature brain, the dynamic expression of miRNAs continues, highlighting their functional importance in neurons. The hippocampus, as one of the crucial brain structures, is a key component of major functional connections in brain. Gene expression abnormalities in the hippocampus lead to disturbance in neurogenesis, neural maturation and synaptic formation. These disturbances are at the root of several neurological disorders and behavioral deficits, including Alzheimer's disease, epilepsy and schizophrenia. There is strong evidence that abnormalities in miRNAs are contributed in neurodegenerative mechanisms in the hippocampus through imbalanced activity of ion channels, neuronal excitability, synaptic plasticity and neuronal apoptosis. Some miRNAs affect oxidative stress, inflammation, neural differentiation, migration and neurogenesis in the hippocampus. Furthermore, major signaling cascades in neurodegeneration, such as NF-Kβ signaling, PI3/Akt signaling and Notch pathway, are closely modulated by miRNAs. These observations, suggest that microRNAs are significant regulators in the complicated network of gene regulation in the hippocampus. In the current review, we focus on the miRNA functional role in the progression of normal development and neurogenesis of the hippocampus. We also consider how miRNAs in the hippocampus are crucial for gene expression mechanisms in pathophysiological pathways.
Collapse
Affiliation(s)
- Seyed Khalil Rashidi
- Department of Medical Biotechnology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Ata Kalirad
- Department of Integrative Evolutionary Biology, Max Planck Institute for Biology Tübingen, Tübingen, Germany
| | - Shahram Rafie
- Department of Neurology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Neuroscience Lab, Golestan Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ebrahim Behzad
- Department of Neurology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Neuroscience Lab, Golestan Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mitra Ansari Dezfouli
- Department of Neurology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Neuroscience Lab, Golestan Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
6
|
Lu W, Wen J. H 2S-mediated inhibition of RhoA/ROCK pathway and noncoding RNAs in ischemic stroke. Metab Brain Dis 2023; 38:163-176. [PMID: 36469178 DOI: 10.1007/s11011-022-01130-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 11/22/2022] [Indexed: 12/11/2022]
Abstract
Ischemic stroke is one of major causes of disability. In the pathological process of ischemic stroke, the up-regulation of Ras homolog gene family, member A (RhoA) and its downstream effector, Ras homolog gene family (Rho)-associated coiled coil-containing kinase (ROCK), contribute to the neuroinflammation, blood-brain barrier (BBB) dysfunction, neuronal apoptosis, axon growth inhibition and astrogliosis. Accumulating evidences have revealed that hydrogen sulphide (H2S) could reduce brain injury in animal model of ischemic stroke via inhibiting the RhoA/ROCK pathway. Recently, noncoding RNAs (ncRNAs) such as circular RNAs (circRNAs), long noncoding RNAs (lncRNAs) and microRNAs (miRNAs) have attracted much attention because of their essential role in adjusting gene expression both in physiological and pathological conditions. Numerous studies have uncovered the role of RhoA/ROCK pathway and ncRNAs in ischemic stroke. In this review, we focused on the role of H2S, RhoA/ROCK pathway and ncRNAs in ischemic stroke and aimed to reveal new strategies for preventing and treating this devastating disease.
Collapse
Affiliation(s)
- Weizhuo Lu
- Medical Branch, Hefei Technology College, Hefei, China
| | - Jiyue Wen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
| |
Collapse
|
7
|
Knockdown of PVT1 Exerts Neuroprotective Effects against Ischemic Stroke Injury through Regulation of miR-214/Gpx1 Axis. BIOMED RESEARCH INTERNATIONAL 2022; 2022:1393177. [PMID: 35978647 PMCID: PMC9377929 DOI: 10.1155/2022/1393177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 07/25/2022] [Indexed: 11/18/2022]
Abstract
Previous studies have reported that lncRNA PVT1 was closely related to ischemic stroke. Here, the role of PVT1 in ischemic stroke and the underlying mechanism were investigated. OGDR-stimulated PC12 cells were used to construct a cell model to mimic ischemic stroke. si-PVT1, miR-214 mimic, inhibitor, or the negative controls were transfected into PC12 cells prior to OGDR treatment. PVT1, miR-214, and Gpx1 expression was measured by qRT-PCR and western blotting assays. Cell proliferation and apoptosis were tested by CCK-8 assay and western blotting. The expression levels of inflammatory factors were determined by ELISA Kit. Results showed that PVT1 was increased significantly in OGDR PC12 cells. PVT1 knockdown significantly enhanced cell viability and attenuated cell apoptosis, ROS generation, and inflammation in OGDR PC12 cells. More importantly, PVT1 or Gpx1 was a target of miR-214. Mechanistically, PVT1 acted as a competing endogenous RNA of miR-214 to regulate the downstream gene Gpx1. In conclusion, PVT1 knockdown attenuated OGDR PC12 cell injury by modulating miR-214/Gpx1 axis. These findings offer a potential novel strategy for ischemic stroke therapy.
Collapse
|
8
|
Rani A, Barter J, Kumar A, Stortz JA, Hollen M, Nacionales D, Moldawer LL, Efron PA, Foster TC. Influence of age and sex on microRNA response and recovery in the hippocampus following sepsis. Aging (Albany NY) 2022; 14:728-746. [PMID: 35094981 PMCID: PMC8833110 DOI: 10.18632/aging.203868] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/20/2022] [Indexed: 11/25/2022]
Abstract
Sepsis, defined as a dysregulated host immune response to infection, is a common and dangerous clinical syndrome. The excessive host inflammatory response can induce immediate and persistent cognitive decline, which can be worse in older individuals. Sex-specific differences in the outcome of infectious diseases and sepsis appear to favor females. We employed a murine model to examine the influence of age and sex on the brain's microRNA (miR) response following sepsis. Young and old mice of both sexes underwent cecal ligation and puncture (CLP) with daily restraint stress. Expression of hippocampal miR was examined in age- and sex-matched controls at 1 and 4 days post-CLP. Few miR were modified in a similar manner across age or sex and these few miR were generally associated with neuroprotection against inflammation. Similar to previous work examining transcription, young females exhibited a better recovery of the miR profile from day 1 to day 4, relative to young males and old females. For young males and all female groups, the initial response mainly involved a decrease in miR expression. In contrast, old males exhibited only upregulated miR on day 1 and day 4 and many of the miR upregulated on day 1 and day 4 were linked to neurodegeneration, increased neuroinflammation, and cognitive impairment. The results emphasize age and sex differences in epigenetic mechanisms that likely contribute to susceptibility or resilience to cognitive impairment due to sepsis.
Collapse
Affiliation(s)
- Asha Rani
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Jolie Barter
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Ashok Kumar
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Julie A Stortz
- Department of Surgery, University of Florida, Gainesville, FL 32611, USA
| | - McKenzie Hollen
- Department of Surgery, University of Florida, Gainesville, FL 32611, USA
| | - Dina Nacionales
- Department of Surgery, University of Florida, Gainesville, FL 32611, USA
| | - Lyle L Moldawer
- Department of Surgery, University of Florida, Gainesville, FL 32611, USA
| | - Philip A Efron
- Department of Surgery, University of Florida, Gainesville, FL 32611, USA
| | - Thomas C Foster
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA.,Genetics and Genomics Program, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
9
|
Zeng T, Zhang S, He Y, Liu Z, Cheng Q. MiR-361-5p promotes oxygen-glucose deprivation/re-oxygenation induced neuronal injury by negatively regulating SQSTM1 in vitro. Metab Brain Dis 2021; 36:2359-2368. [PMID: 34581931 DOI: 10.1007/s11011-021-00845-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 09/17/2021] [Indexed: 02/07/2023]
Abstract
It has been reported that microRNAs (miRNAs) play essential roles in cerebral ischemia and reperfusion (I/R) injury. This study aimed to explore the role of miR-361-5p in oxygen-glucose deprivation/re-oxygenation-induced neuronal injury in vitro. Cerebral I/R injury cell model was established by using PC12 cells exposed to oxygen-glucose deprivation/re-oxygenation (OGD/R). The expression of miR-361-5p and SQSTM1 was evaluated by qRT-PCR or western blot. Neuronal apoptosis was detected by flow cytometry, and cell viability was assessed by CCK-8 assay. The effects of miR-361-5p on the release of LDH and the levels of MDA, SOD, and GSH-Px were investigated by respective detection kits. Dual-luciferase reporter assay and RIP assay were performed to determine the interaction between miR-361-5p and SQSTM1. Rescue experiments were performed to evaluate the function of miR-361-5p and SQSTM1. MiR-361-5p was significantly upregulated, and SQSTM1 was significantly downregulated in OGD/R-stimulated PC12 cells. MiR-361-5p could directly interact with SQSTM1 and negatively regulated it. Inhibition of miR-361-5p efficiently inhibited OGD/R-induced apoptosis and attenuated OGD/R-induced growth defect in PC12 cells. In addition, SQSTM1 overexpression partially attenuates the apoptosis and promoted the viability of OGD/R-treated PC12 cells, which were aggravated by miR-361-5p mimics. Our study demonstrated that miR-361-5p promotes OGD/R-induced neuronal injury via regulating SQSTM1 in PC12 cells.
Collapse
Affiliation(s)
- Tao Zeng
- Department of Neurology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, No. 1 Panfu Road, Yuexiu District, Guangzhou, 510180, Guangdong, People's Republic of China.
| | - Sai Zhang
- Department of Neurology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, No. 1 Panfu Road, Yuexiu District, Guangzhou, 510180, Guangdong, People's Republic of China
| | - Yan He
- Department of Neurology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, No. 1 Panfu Road, Yuexiu District, Guangzhou, 510180, Guangdong, People's Republic of China
| | - Zhenxing Liu
- Department of Neurology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, No. 1 Panfu Road, Yuexiu District, Guangzhou, 510180, Guangdong, People's Republic of China
| | - Qiusheng Cheng
- Department of Neurology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, No. 1 Panfu Road, Yuexiu District, Guangzhou, 510180, Guangdong, People's Republic of China
| |
Collapse
|
10
|
Kimura T, Horikoshi Y, Kuriyagawa C, Niiyama Y. Rho/ROCK Pathway and Noncoding RNAs: Implications in Ischemic Stroke and Spinal Cord Injury. Int J Mol Sci 2021; 22:ijms222111573. [PMID: 34769004 PMCID: PMC8584200 DOI: 10.3390/ijms222111573] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/21/2021] [Accepted: 10/24/2021] [Indexed: 01/18/2023] Open
Abstract
Ischemic strokes (IS) and spinal cord injuries (SCI) are major causes of disability. RhoA is a small GTPase protein that activates a downstream effector, ROCK. The up-regulation of the RhoA/ROCK pathway contributes to neuronal apoptosis, neuroinflammation, blood-brain barrier dysfunction, astrogliosis, and axon growth inhibition in IS and SCI. Noncoding RNAs (ncRNAs), such as microRNAs (miRNAs) and long noncoding RNAs (lncRNAs), were previously considered to be non-functional. However, they have attracted much attention because they play an essential role in regulating gene expression in physiological and pathological conditions. There is growing evidence that ROCK inhibitors, such as fasudil and VX-210, can reduce injury in IS and SCI in animal models and clinical trials. Recently, it has been reported that miRNAs are decreased in IS and SCI, while lncRNAs are increased. Inhibiting the Rho/ROCK pathway with miRNAs alleviates apoptosis, neuroinflammation, oxidative stress, and axon growth inhibition in IS and SCI. Further studies are required to explore the significance of ncRNAs in IS and SCI and to establish new strategies for preventing and treating these devastating diseases.
Collapse
Affiliation(s)
- Tetsu Kimura
- Correspondence: ; Tel.: +81-18-884-6175; Fax: +81-18-884-6448
| | | | | | | |
Collapse
|
11
|
Jia Y, Yi L, Li Q, Liu T, Yang S. LncRNA MALAT1 aggravates oxygen-glucose deprivation/reoxygenation-induced neuronal endoplasmic reticulum stress and apoptosis via the miR-195a-5p/HMGA1 axis. Biol Res 2021; 54:8. [PMID: 33750458 PMCID: PMC7941907 DOI: 10.1186/s40659-021-00331-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 02/26/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND This study aimed to investigate the potential role and molecular mechanism of lncRNA metastasis associated lung adenocarcinoma transcript 1 (MALAT1) in cerebral ischemia/reperfusion injury. RESULTS Using an oxygen-glucose deprivation/reoxygenation (OGD/R) cell model, we determined that the expression of MALAT1 was significantly increased during OGD/R. MALAT1 knockdown reversed OGD/R-induced apoptosis and ER stress. Mechanistically, MALAT1 promoted OGD/R-induced neuronal injury through sponging miR-195a-5p to upregulating high mobility group AT-hook1 (HMGA1). CONCLUSIONS Collectively, these data demonstrate the mechanism underlying the invovlvement of MALAT1 in cerebral ischemia/reperfusion injury, thus providing translational evidence that MALAT1 may serve as a novel biomarker and therapeutic target for ischemic stroke.
Collapse
Affiliation(s)
- Ying Jia
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, No.23 Youzheng Street, Nangang District, Harbin, 150081, Heilongjiang, People's Republic of China
| | - Lian Yi
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, No.23 Youzheng Street, Nangang District, Harbin, 150081, Heilongjiang, People's Republic of China
| | - Qianqian Li
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, No.23 Youzheng Street, Nangang District, Harbin, 150081, Heilongjiang, People's Republic of China
| | - Tingjiao Liu
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, No.23 Youzheng Street, Nangang District, Harbin, 150081, Heilongjiang, People's Republic of China
| | - Shanshan Yang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, No.23 Youzheng Street, Nangang District, Harbin, 150081, Heilongjiang, People's Republic of China.
| |
Collapse
|