1
|
Li F, Zhang HK, Jiang HX, Zhang XY, Chen QX. TUG1 exacerbates cerebral ischemia-reperfusion injury through miR-340-5p-mediated PTEN. J Mol Histol 2024; 55:699-707. [PMID: 39017855 DOI: 10.1007/s10735-024-10224-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 07/07/2024] [Indexed: 07/18/2024]
Abstract
Long non-coding RNAs (LncRNAs) play a substantial role in the process of cerebral ischemia-reperfusion injury (CIRI). The present work aimed to determine the probable mechanism by which LncRNA TUG1 exacerbates CIRI via the miR-340-5p/phosphatase and tensin homolog (PTEN) pathway. After developing a middle cerebral artery occlusion/reperfusion (MCAO/R) model, pcDNA-TUG1 together with miR-340-5p agomir were administrated in vivo. Furthermore, the neurologic defects in rats were assessed by a modified neurological severity score. Moreover, 2,3,5-Triphenyl-2 H-tetrazolium chloride stain-step was performed to determine the brain's infarct size. In addition, western blotting, immunohistochemistry, and qRT-PCR experiments were utilized for gauging the proteomic/genomic expression-profiles. Luciferase reporter assay validated correlations across TUG1, miR-340-5p, together with PTEN. The results indicated relatively reduced miR-340-5p levels in MCAO/R models, while upregulated TUG1 levels. The pcDNA-TUG1-treated rats indicated increasing neurological dysfunction, whereas the miR-340-5p agomir-treated rats showed improvement. Furthermore, miR-340-5p was determined to be the expected and confirmed TUG1 target. All things considered, the findings suggested that PTEN can serve as the target of miR-340-5p. In addition, TUG1 served as a miR-340-5p ceRNA, which promotes PTEN modulation. Furthermore, TUG1 overexpression decreased miR-340-5p's capacity to fend against CIRI. Conclusively, this work proved that in CIRI, targeting the TUG1/miR-340-5p/PTEN regulatory axis is a viable approach for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Fei Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Hui-Kai Zhang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Hong-Xiang Jiang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xin-Yuan Zhang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Qian-Xue Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
2
|
Khoshnam SE, Moalemnia A, Anbiyaee O, Farzaneh M, Ghaderi S. LncRNA MALAT1 and Ischemic Stroke: Pathogenesis and Opportunities. Mol Neurobiol 2024; 61:4369-4380. [PMID: 38087169 DOI: 10.1007/s12035-023-03853-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 11/21/2023] [Indexed: 07/11/2024]
Abstract
Ischemic stroke (IS) stands as a prominent cause of mortality and long-term disability around the world. It arises primarily from a disruption in cerebral blood flow, inflicting severe neural injuries. Hence, there is a pressing need to comprehensively understand the intricate mechanisms underlying IS and identify novel therapeutic targets. Recently, long noncoding RNAs (lncRNAs) have emerged as a novel class of regulatory molecules with the potential to attenuate pathogenic mechanisms following IS. Among these lncRNAs, MALAT1 (metastasis-associated lung adenocarcinoma transcript 1) has been extensively studied due to its involvement in the pathophysiological processes of IS. In this review, we provide an in-depth analysis of the essential role of MALAT1 in the development and progression of both pathogenic and protective mechanisms following IS. These mechanisms include oxidative stress, neuroinflammation, cell death signaling, blood brain barrier dysfunction, and angiogenesis. Furthermore, we summarize the impact of MALAT1 on the susceptibility and severity of IS. This review highlights the potential risks associated with the therapeutic use of MALAT1 for IS, which are attributable to the stimulatory action of MALAT1 on ischemia/reperfusion injury. Ultimately, this review sheds light on the potential molecular mechanisms and associated signaling pathways underlying MALAT1 expression post-IS, with the aim of uncovering potential therapeutic targets.
Collapse
Affiliation(s)
- Seyed Esmaeil Khoshnam
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Arash Moalemnia
- Faculty of Medicine, Dezful University of Medical Sciences, Dezful, Iran
| | - Omid Anbiyaee
- School of Medicine, Cardiovascular Research Center, Namazi Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Shahab Ghaderi
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
3
|
Khan MM, Kirabo A. Long Noncoding RNA MALAT1: Salt-Sensitive Hypertension. Int J Mol Sci 2024; 25:5507. [PMID: 38791545 PMCID: PMC11122212 DOI: 10.3390/ijms25105507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/06/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Hypertension stands as the leading global cause of mortality, affecting one billion individuals and serving as a crucial risk indicator for cardiovascular morbidity and mortality. Elevated salt intake triggers inflammation and hypertension by activating antigen-presenting cells (APCs). We found that one of the primary reasons behind this pro-inflammatory response is the epithelial sodium channel (ENaC), responsible for transporting sodium ions into APCs and the activation of NADPH oxidase, leading to increased oxidative stress. Oxidative stress increases lipid peroxidation and the formation of pro-inflammatory isolevuglandins (IsoLG). Long noncoding RNAs (lncRNAs) play a crucial role in regulating gene expression, and MALAT1, broadly expressed across cell types, including blood vessels and inflammatory cells, is also associated with inflammation regulation. In hypertension, the decreased transcriptional activity of nuclear factor erythroid 2-related factor 2 (Nrf2 or Nfe2l2) correlates with heightened oxidative stress in APCs and impaired control of various antioxidant genes. Kelch-like ECH-associated protein 1 (Keap1), an intracellular inhibitor of Nrf2, exhibits elevated levels of hypertension. Sodium, through an increase in Sp1 transcription factor binding at its promoter, upregulates MALAT1 expression. Silencing MALAT1 inhibits sodium-induced Keap1 upregulation, facilitating the nuclear translocation of Nrf2 and subsequent antioxidant gene transcription. Thus, MALAT1, acting via the Keap1-Nrf2 pathway, modulates antioxidant defense in hypertension. This review explores the potential role of the lncRNA MALAT1 in controlling the Keap1-Nrf2-antioxidant defense pathway in salt-induced hypertension. The inhibition of MALAT1 holds therapeutic potential for the progression of salt-induced hypertension and cardiovascular disease (CVD).
Collapse
Affiliation(s)
- Mohd Mabood Khan
- Department of Medicine, Preston Research Building, Vanderbilt University Medical Centre, Nashville, TN 37232, USA
| | - Annet Kirabo
- Department of Medicine, Preston Research Building, Vanderbilt University Medical Centre, Nashville, TN 37232, USA
| |
Collapse
|
4
|
Gao P, Shi H, Jin X, Guo S, Zhou X, Gao W. Mechanism of astragaloside IV regulating NLRP3 through LOC102555978 to attenuate cerebral ischemia reperfusion induced microglia pyroptosis. Int Immunopharmacol 2024; 131:111862. [PMID: 38513574 DOI: 10.1016/j.intimp.2024.111862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 03/09/2024] [Accepted: 03/11/2024] [Indexed: 03/23/2024]
Abstract
Astragaloside IV(ASⅣ), the main component of Radix Astragali, has been used to treat cerebral ischemia reperfusion injury (CIRI). However, the molecular mechanism of ASIV in CIRI needs to be further elucidated. Long non-coding RNA (lncRNA) is considered to be an important kind of regulatory molecule in CIRI. In this work, the biological effect and molecular mechanism of ASIV in CIRI through lncRNA were analyzed by using rat middle cerebral artery occlusion and reperfusion (MCAO/R) model and primary rat microglia (RM) cells oxygen and glucose deprivation/reoxygenation (OGD/R) model. The neurological deficit score was evaluated, the volume of cerebral infarction was calculated, and pyroptosis related molecules were detected by qPCR and western blot. Then, high-throughput sequencing was performed in sham and MCAO/R groups. The competitive endogenous RNA (ceRNA) networks associated with pyroptosis were constructed by functional enrichment analysis. CCK-8 detection of cell survival rate, qPCR and western blot were used to determine the specific molecular mechanism of ASⅣ through ceRNA in vitro. Results showed thatASⅣ could decrease the neurological deficit score, reduce the volume of cerebral infarction, inhibit inflammatory reaction and pyroptosis in MCAO/R model rats. Next, the ceRNA network was established, including the LOC102555978/miR-3584-5p/NLRP3 regulatory network. In vitro experiments showed that LOC102555978 promotes NLRP3 mediated pyroptosis of RM cells through sponge adsorption of miR-3584-5p, which may provide a potential therapeutic target for post-CIRI inflammation regulation. ASⅣ could inhibit pyroptosis of RM cells by down-regulating LOC102555978. LOC102555978/miR-3584-5p/NLRP3 may be the molecular mechanism of ASⅣ's CIRI protective effect.
Collapse
Affiliation(s)
- Ping Gao
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Haowei Shi
- Department of Neurosurgery, Hebei General Hospital, Shijiazhuang, China
| | - Xiaofei Jin
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Shuhan Guo
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Xiaohong Zhou
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Weijuan Gao
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Hebei University of Chinese Medicine, Shijiazhuang 050200, China.
| |
Collapse
|
5
|
Hamada Y, Takata T, Iwama H, Kawakita R, Nonaka W, Deguchi K, Kobara H, Morishita A, Miyamoto O, Nakamura T, Itano T, Masaki T. Temporal expression profiles of microRNAs associated with acute phase of brain ischemia in gerbil hippocampus. Heliyon 2024; 10:e28875. [PMID: 38576576 PMCID: PMC10990972 DOI: 10.1016/j.heliyon.2024.e28875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 03/22/2024] [Accepted: 03/26/2024] [Indexed: 04/06/2024] Open
Abstract
Neuroprotective therapeutic potential for restoring dysregulated microRNA (miRNA) expression has previously been demonstrated in a gerbil cerebral infarction model. However, since temporal changes in miRNA expression profiles following stroke onset are unknown, miRNAs proving to be useful therapeutic targets have yet to be identified. We evaluated cognitive function, hippocampal neuronal cell death, and microarray-based miRNA expression profiles at 5, 9, 18, 36, and 72 h after 5-min whole brain ischemia in gerbils. A decline in cognitive function occurred in parallel with increased neuronal cell death 36-72 h after ischemia. The Jonckheere-Terpstra test was used to analyze miRNA expression trends 5-72 h after ischemia. The expression levels of 63 miRNAs were significantly upregulated, whereas 32 miRNAs were significantly downregulated, monotonically. Of the 32 monotonically downregulated miRNAs, 18 showed the largest decrease in expression 5-9 h after ischemia. A subset of these dysregulated miRNAs (miR-378a-5p, miR-204-5p, miR-34c-5p, miR-211-5p, miR-34b-3p, and miR-199b-3p) could be associated with brain ischemia and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Yasuhiro Hamada
- Department of Neurology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki, Kagawa, 761-0793, Japan
| | - Tadayuki Takata
- Department of Neurology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki, Kagawa, 761-0793, Japan
| | - Hisakazu Iwama
- Life Science Research Center, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki, Kagawa, 761-0793, Japan
| | - Rie Kawakita
- Department of Neurology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki, Kagawa, 761-0793, Japan
| | - Wakako Nonaka
- Department of Neurology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki, Kagawa, 761-0793, Japan
- Department of General Medicine, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki, Kagawa, 761-0793, Japan
| | - Kazushi Deguchi
- Department of Neurology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki, Kagawa, 761-0793, Japan
| | - Hideki Kobara
- Department of Gastroenterology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki, Kagawa, 761-0793, Japan
| | - Asahiro Morishita
- Department of Gastroenterology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki, Kagawa, 761-0793, Japan
| | - Osamu Miyamoto
- Department of Medical Engineering, Faculty of Health Science and Technology, Kawasaki University of Medical Welfare, 288 Matsushima, Kurashiki, 701-0193, Japan
| | - Takehiro Nakamura
- Department of Physiology 2, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama, 701-0192, Japan
| | - Toshifumi Itano
- Department of Neurology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki, Kagawa, 761-0793, Japan
| | - Tsutomu Masaki
- Department of Gastroenterology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki, Kagawa, 761-0793, Japan
| |
Collapse
|
6
|
Li Y, Fan H, Han X, Sun J, Ni M, Hou X, Fang F, Zhang W, Ma P. Long Non-Coding RNA MALAT1 Protects Against Spinal Cord Injury via Suppressing microRNA-125b-5p Mediated Microglial M1 Polarization, Neuroinflammation, and Neural Apoptosis. Mol Neurobiol 2024; 61:2136-2150. [PMID: 37858031 DOI: 10.1007/s12035-023-03664-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 09/18/2023] [Indexed: 10/21/2023]
Abstract
Our previous studies have discovered that long non-coding RNA (lncRNA) MALAT1 and its target microRNA-125b-5p (miR-125b-5p) are implicated in neurological diseases via regulating neuroinflammation and neuronal injury. This study aimed to further explore the relationship between lncRNA MALAT1 and miR-125b-5p, as well as their effect on microglial activation, neuroinflammation, and neural apoptosis in spinal cord injury (SCI). Primary microglia from Sprague Dawley rats were stimulated with lipopolysaccharide (LPS). Then, microglia were transfected with lncRNA MALAT1 overexpression or knock-down adenovirus-associated virus with or without miR-125b-5p mimic. The culture medium of microglia was incubated with primary neurons. SCI rats were established for in vivo validation. LncRNA MALAT1 expression was reduced by LPS treatment in a dose-dependent manner. LncRNA MALAT1 overexpression suppressed the microglial M1 polarization (decreased iNOS but increased ARG1), neuroinflammation (declined PTGS2, TNF-α, IL-1β, and IL-6), and microglia-induced neural apoptosis (lower TUNEL positive cells and C-caspase3 but higher BCL2) under LPS treatment; its knock-down displayed the opposite trend. Moreover, lncRNA MALAT1 directly bound to and negatively regulated miR-125b-5p. MiR-125b-5p mimic promoted microglial M1 polarization, neuroinflammation, and microglia-induced neural apoptosis following LPS treatment; also, it could attenuate the effect of lncRNA MALAT1. Further in vivo study displayed that lncRNA MALAT1 overexpression elevated the Basso-Beattie-Bresnahan motor function score and improved neural injury. Also, in vivo validation indicated a similar effect of lncRNA MALAT1 on microglial polarization and neuroinflammation as in vitro. LncRNA MALAT1 improves SCI recovery via miR-125b-5p mediated microglial M1 polarization, neuroinflammation, and neural apoptosis.
Collapse
Affiliation(s)
- Yuanlong Li
- Department of Pharmacy, Henan Provincial People's Hospital, Zhengzhou, 450000, Henan, China
- Department of Pharmacy, People's Hospital of Zhengzhou University, Zhengzhou University, No. 7 Weiwu Road, Zhengzhou, 450000, Henan, China
- Department of Pharmacy, People's Hospital of Henan University, School of Clinical Medicine, Henan University, Zhengzhou, 450000, Henan, China
| | - Hua Fan
- School of Clinical Medicine, The First Affiliated Hospital of Henan University of Science and Technology, Henan University of Science and Technology, Luoyang, 471003, Henan, China
| | - Xiong Han
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, 450000, Henan, China
| | - Jun Sun
- Department of Pharmacy, Henan Provincial People's Hospital, Zhengzhou, 450000, Henan, China
- Department of Pharmacy, People's Hospital of Zhengzhou University, Zhengzhou University, No. 7 Weiwu Road, Zhengzhou, 450000, Henan, China
- Department of Pharmacy, People's Hospital of Henan University, School of Clinical Medicine, Henan University, Zhengzhou, 450000, Henan, China
| | - Ming Ni
- Department of Pharmacy, Henan Provincial People's Hospital, Zhengzhou, 450000, Henan, China
- Department of Clinical Pharmacy, Fuwai Central China Cardiovascular Hospital, Zhengzhou, 450000, Henan, China
| | - Xiaodan Hou
- Ward of Heart Failure, Fuwai Central China Cardiovascular Hospital, Zhengzhou, 450000, Henan, China
| | - Fengqin Fang
- Department of Pharmacy, Henan Provincial People's Hospital, Zhengzhou, 450000, Henan, China
- Department of Pharmacy, People's Hospital of Zhengzhou University, Zhengzhou University, No. 7 Weiwu Road, Zhengzhou, 450000, Henan, China
- Department of Pharmacy, People's Hospital of Henan University, School of Clinical Medicine, Henan University, Zhengzhou, 450000, Henan, China
| | - Wei Zhang
- Department of Pharmacy, Henan Provincial People's Hospital, Zhengzhou, 450000, Henan, China
- Department of Pharmacy, People's Hospital of Zhengzhou University, Zhengzhou University, No. 7 Weiwu Road, Zhengzhou, 450000, Henan, China
- Department of Pharmacy, People's Hospital of Henan University, School of Clinical Medicine, Henan University, Zhengzhou, 450000, Henan, China
| | - Peizhi Ma
- Department of Pharmacy, Henan Provincial People's Hospital, Zhengzhou, 450000, Henan, China.
- Department of Pharmacy, People's Hospital of Zhengzhou University, Zhengzhou University, No. 7 Weiwu Road, Zhengzhou, 450000, Henan, China.
- Department of Pharmacy, People's Hospital of Henan University, School of Clinical Medicine, Henan University, Zhengzhou, 450000, Henan, China.
| |
Collapse
|
7
|
Li Z, Pang Y, Hou L, Xing X, Yu F, Gao M, Wang J, Li X, Zhang L, Xiao Y. Exosomal OIP5-AS1 attenuates cerebral ischemia-reperfusion injury by negatively regulating TXNIP protein stability and inhibiting neuronal pyroptosis. Int Immunopharmacol 2024; 127:111310. [PMID: 38103409 DOI: 10.1016/j.intimp.2023.111310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/15/2023] [Accepted: 11/26/2023] [Indexed: 12/19/2023]
Abstract
BACKGROUND Cerebral ischemia-reperfusion injury (CIRI) can cause neuronal apoptosis and lead to irreversible brain injury. Numerous lncRNAs have been reported to play important roles in CIRI, but it is unclear whether these lncRNAs can function through exosomes. METHODS In this study, we utilized the middle cerebral artery occlusion/reperfusion (MCAO/R) animal model and the oxygen-glucose deprivation/ reoxygenation (OGD/R) cell model. RNA sequencing was performed to screen for differentially expressed lncRNAs in M2 microglia-derived exosomes (M2-Exos). RNA pull-down, RNA immunoprecipitation, co-immunoprecipitation and ubiquitination assays were used to explore the molecular mechanism of OIP5-AS1 in alleviating CIRI. RESULTS M2-Exos could alleviate nerve injury and pyroptosis after CIRI in vitro and in vivo. OIP5-AS1 was found to be significantly up-regulated in M2-Exos and down-regulated in OGD/R neurons, MCAO/R mice and ischemic stroke patients. In MCAO/R mice, OIP5-AS1 could reduce cerebral infarct size, cerebral edema and mNSS scores, and inhibit the expression levels of pyroptosis-related proteins in brain tissue. TXNIP was confirmed to be a reliable binding protein of OIP5-AS1. OIP5-AS1 overexpression significantly attenuated MCAO/R-induced upregulation of TXNIP at the protein level, but not at the mRNA level. OIP5-AS1 promoted the TXNIP degradation process and increased the ubiquitination of TXNIP. ITCH could bind to TXNIP. ITCH overexpression or knockdown did not alter the mRNA level of TXNIP, but negatively regulated TXNIP expression at the protein level. ITCH accelerated the degradation and ubiquitination of TXNIP, which could be attenuated by OIP5-AS1 knockdown. OIP5-AS1 could improve neuronal damage and inhibit neuronal pyroptosis through TXNIP. CONCLUSIONS M2-Exo-derived OIP5-AS1 can induce TXNIP ubiquitination and degradation by recruiting ITCH, negatively regulate TXNIP protein stability, inhibit neuronal pyroptosis, and attenuate CIRI.
Collapse
Affiliation(s)
- Zhongchen Li
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng City, Shandong Province 252000, China; Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan City, Shandong Province 250063, China
| | - Yuejiu Pang
- Department of Healthcare Neurology, Provincial Hospital Affiliated to Shandong First Medical University, Jinan City, Shandong Province 250021, China
| | - Lei Hou
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng City, Shandong Province 252000, China
| | - Xiaohui Xing
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng City, Shandong Province 252000, China
| | - Fuhua Yu
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng City, Shandong Province 252000, China
| | - Mingxu Gao
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng City, Shandong Province 252000, China
| | - Jiyue Wang
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng City, Shandong Province 252000, China
| | - Xueyuan Li
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng City, Shandong Province 252000, China.
| | - Liyong Zhang
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng City, Shandong Province 252000, China.
| | - Yilei Xiao
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng City, Shandong Province 252000, China.
| |
Collapse
|
8
|
Li X, Quan P, Si Y, Liu F, Fan Y, Ding F, Sun L, Liu H, Huang S, Sun L, Yang F, Yao L. The microRNA-211-5p/P2RX7/ERK/GPX4 axis regulates epilepsy-associated neuronal ferroptosis and oxidative stress. J Neuroinflammation 2024; 21:13. [PMID: 38191407 PMCID: PMC10773122 DOI: 10.1186/s12974-023-03009-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 12/28/2023] [Indexed: 01/10/2024] Open
Abstract
Ferroptosis is an iron-dependent cell death mechanism involving the accumulation of lipid peroxides. As a critical regulator, glutathione peroxidase 4 (GPX4) has been demonstrated to be downregulated in epilepsy. However, the mechanism of ferroptosis in epilepsy remains unclear. In this study, bioinformatics analysis, analysis of epilepsy patient blood samples and cell and mouse experiments revealed strong associations among epilepsy, ferroptosis, microRNA-211-5p and purinergic receptor P2X 7 (P2RX7). P2RX7 is a nonselective ligand-gated homotrimeric cation channel, and its activation mainly increases neuronal activity during epileptic seizures. In our study, the upregulation of P2RX7 in epilepsy was attributed to the downregulation of microRNA (miR)-211-5p. Furthermore, P2RX7 has been found to regulate GPX4/HO-1 by alleviating lipid peroxidation induced by suppression of the MAPK/ERK signaling pathway in murine models. The dynamic decrease in miR-211-5p expression induces hypersynchronization and both nonconvulsive and convulsive seizures, and forebrain miR-211-5p suppression exacerbates long-lasting pentylenetetrazole-induced seizures. Additionally, in this study, induction of miR-211-5p expression or genetic-silencing of P2RX7 significantly reduced the seizure score and duration in murine models through the abovementioned pathways. These results suggest that the miR-211-5p/P2RX7 axis is a novel target for suppressing both ferroptosis and epilepsy.
Collapse
Affiliation(s)
- Xueying Li
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, Harbin, 150081, China
| | - Pusheng Quan
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, Harbin, 150081, China
- Department of Neurology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Yao Si
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, Harbin, 150081, China
| | - Fei Liu
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, Harbin, 150081, China
| | - Yuwei Fan
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, Harbin, 150081, China
| | - Feifan Ding
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, Harbin, 150081, China
| | - Lina Sun
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, Harbin, 150081, China
| | - Han Liu
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, Harbin, 150081, China
| | - Shuo Huang
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, Harbin, 150081, China
| | - Linlin Sun
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, Harbin, 150081, China.
| | - Fan Yang
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, Harbin, 150081, China.
| | - Lifen Yao
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
9
|
Huang S, Hou D, Zhang L, Pei C, Liang J, Li J, Yang G, Yu D. LncRNA MALAT1 Promoted Neuronal Necroptosis in Cerebral Ischemia-reperfusion Mice by Stabilizing HSP90. Neurochem Res 2023; 48:3457-3471. [PMID: 37470906 DOI: 10.1007/s11064-023-03991-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 07/03/2023] [Accepted: 07/09/2023] [Indexed: 07/21/2023]
Abstract
The objective of this research was to investigate the role of lncRNA MALAT1 and HSP90 in the regulation of neuronal necroptosis in mice with cerebral ischemia-reperfusion (CIR). We used male C57BL/6J mice to establish a middle cerebral artery occlusion (MCAO) model and conducted in vitro experiments using the HT-22 mouse hippocampal neuron cell line. The cellular localization of NeuN and MLKL, as well as the expression levels of neuronal necroptosis factors, MALAT1, and HSP90 were analyzed. Cell viability and necroptosis were assessed, and we also investigated the relationship between MALAT1 and HSP90. The results showed that MALAT1 expression increased after MCAO and oxygen-glucose deprivation/re-oxygenation (OGD/R) treatment in both cerebral tissues and cells compared with the control group. The levels of neuronal necroptosis factors and the co-localization of NeuN and MLKL were also increased in MCAO mice compared with the Sham group. MALAT1 was found to interact with HSP90, and inhibition of HSP90 expression led to decreased phosphorylation levels of neuronal necroptosis factors. Inhibition of MALAT1 expression resulted in decreased co-localization levels of NeuN and MLKL, decreased phosphorylation levels of neuronal necroptosis factors, and reduced necroptosis rate in cerebral tissues. Furthermore, inhibiting MALAT1 expression also led to a shorter half-life of HSP90, increased ubiquitination level, and decreased phosphorylation levels of neuronal necroptosis factors in cells. In conclusion, this study demonstrated that lncRNA MALAT1 promotes neuronal necroptosis in CIR mice by stabilizing HSP90.
Collapse
Affiliation(s)
- Shan Huang
- Department of Neurology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, No. 43 Renmin Avenue, Haikou, 570208, Hainan, China
| | - Dan Hou
- Department of Neurology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, No. 43 Renmin Avenue, Haikou, 570208, Hainan, China
| | - Lei Zhang
- Department of Neurology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, No. 43 Renmin Avenue, Haikou, 570208, Hainan, China
| | - Chaoying Pei
- Department of Neurology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, No. 43 Renmin Avenue, Haikou, 570208, Hainan, China
| | - Ji Liang
- Department of Neurology, The First People's Hospital of Changde, Changde, 415000, Hunan, China
| | - Junqi Li
- Department of Neurology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, No. 43 Renmin Avenue, Haikou, 570208, Hainan, China
| | - Guoshuai Yang
- Department of Neurology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, No. 43 Renmin Avenue, Haikou, 570208, Hainan, China.
| | - Dan Yu
- Department of Neurology, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, No. 43 Renmin Avenue, Haikou, 570208, Hainan, China.
| |
Collapse
|
10
|
Zhang Y, Yuan X, Xu J, Gu H. CircRBM33 induces endothelial dysfunction by targeting the miR-6838-5p/PDCD4 axis affecting blood-brain barrier in mice with cerebral ischemia-reperfusion injury. Clin Hemorheol Microcirc 2023; 85:355-370. [PMID: 37927249 DOI: 10.3233/ch-231776] [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: 11/07/2023]
Abstract
BACKGROUND circRNAs (circRNAs) are involved in the process of cerebral ischemia-reperfusion injury (CI/RI). Our study aims to explore circRBM33 in the endothelial function of the blood-brain barrier (BBB). METHODS The mouse middle cerebral artery occlusion model (MCAO) was established and restored to perfusion, and OGD/R-induced endothelial cells were used to simulate CI/RI. circRBM33, miR-6838-5p and PDCD4, as well as Occludin, ZO-1 and Claudin-5 TJs were evaluated by quantitative PCR and Western blot. The ring structure of circRBM33 was verified by RNAse R and actinomycin D experiments. MTT and LDH Cytotoxicity assay determined viability and toxicity, and flow cytometry determined apoptosis rate. Inflammatory cytokines and the number of microglia in brain tissue were measured by ELISA and IHC. The interaction between genes was verified by RIP and dual luciferase reporter assay. RESULTS circRBM33 was a circrRNA present in the cytoplasm and up-regulated in the brain tissue of MCAO mice and OGD/R-induced endothelial cells. Silenced circRBM33 promoted Occludin, ZO-1, and Claudin-5 expression and cell proliferation, and inhibited cytotoxicity, inflammatory response, and apoptosis. Functionally, circRBM33-absorbed miR-6838-5p was involved in regulating PDCD4, leading to endothelial cell dysfunction, and thus affecting the function of the BBB. CONCLUSIONS circRBM33 by mediating miR-6838-5p/PDCD4 axis induces endothelial dysfunction, thereby affecting the BBB in mice with CI/RI.
Collapse
Affiliation(s)
- Yanbin Zhang
- Department of Neurology, First People's Hospital of Linping District, Hangzhou City, ZheJiang, China
| | - Xiaodong Yuan
- Department of Neurology, First People's Hospital of Linping District, Hangzhou City, ZheJiang, China
| | - Jie Xu
- Department of Neurology, First People's Hospital of Linping District, Hangzhou City, ZheJiang, China
| | - Huafen Gu
- Department of Neurology, First People's Hospital of Linping District, Hangzhou City, ZheJiang, China
| |
Collapse
|
11
|
Hao T, Chen C, Yang S, Zhang Y, Liang F. Mangiferin exerts neuroprotective effects against focal cerebral ischemia in mice by regulating NF-κB signaling pathway. Metab Brain Dis 2023; 38:383-391. [PMID: 36322276 DOI: 10.1007/s11011-022-01066-6] [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: 03/03/2022] [Accepted: 08/08/2022] [Indexed: 11/05/2022]
Abstract
PURPOSE Mangiferin is a natural free radical scavenging antioxidant that induces excitation of the central nervous system. However, the mechanism of neuroprotective effect of mangiferin on focal cerebral ischemia has not been fully investigated. The aim of this study was to investigate the protective effect of mangiferin on focal cerebral ischemia in mice. METHODS Middle cerebral artery occlusion (MCAO) was performed to investigate the effect of mangiferin on focal cerebral ischemia. Mice were randomly divided into 5 groups: sham, MCAO, MCAO + 5 mg/kg mangiferin, MCAO + 20 mg/kg mangiferin and MCAO + 5 mg/kg nimodipine. Neurobehavioral scores, brain edema, brain injury scores, relative infarct size and expression of some inflammatory factors in the brain were evaluated. NF-κB pathway was detected by Western blotting and immunofluorescence. RESULTS The results showed that mangiferin effectively attenuated MCAO-induced brain injury, including improvement of neurological impairment, reduction of brain edema, and reduction of infarct size. Compared with the MCAO group, mangiferin significantly inhibited MCAO-induced neuroinflammation, which can be proved by reduced expression levels of TNF-α, IL-1β, iNOS and COX-2. In addition, we found that phosphorylation of IκBα was inhibited and the expression of NF-κB p65 in the nucleus was reduced after the addition of mangiferin. CONCLUSION Our study suggested that mangiferin exerts neuroprotective effects on focal cerebral ischemia in mice by regulating the NF-κB signaling pathway. Mangiferin may be an effective treatment for cerebral ischemia and other neurological disorders.
Collapse
Affiliation(s)
- Tianpao Hao
- Department of Emergency, The First Affiliated Hospital of Wenzhou Medical University, 325000, Wenzhou, China
| | - Chan Chen
- Department of Geriatric Medicine, The First Affiliated Hospital of Wenzhou Medical University, Southern white elephant town, Ouhai district, 325000, Wenzhou, China
| | - Shen Yang
- Departments of Neurology, Wuhu NO.1 People's Hospital, 241000, Wuhu, NO, China
| | - Yang Zhang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, 325000, Wenzhou, China
| | - Feiyu Liang
- Department of Geriatric Medicine, The First Affiliated Hospital of Wenzhou Medical University, Southern white elephant town, Ouhai district, 325000, Wenzhou, China.
| |
Collapse
|
12
|
Yang K, Zeng L, Ge A, Wang S, Zeng J, Yuan X, Mei Z, Wang G, Ge J. A systematic review of the research progress of non-coding RNA in neuroinflammation and immune regulation in cerebral infarction/ischemia-reperfusion injury. Front Immunol 2022; 13:930171. [PMID: 36275741 PMCID: PMC9585453 DOI: 10.3389/fimmu.2022.930171] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 08/08/2022] [Indexed: 11/15/2022] Open
Abstract
Cerebral infarction/ischemia-reperfusion injury is currently the disease with the highest mortality and disability rate of cardiovascular disease. Current studies have shown that nerve cells die of ischemia several hours after ischemic stroke, which activates the innate immune response in the brain, promotes the production of neurotoxic substances such as inflammatory cytokines, chemokines, reactive oxygen species and − nitrogen oxide, and mediates the destruction of blood-brain barrier and the occurrence of a series of inflammatory cascade reactions. Meanwhile, the expression of adhesion molecules in cerebral vascular endothelial cells increased, and immune inflammatory cells such as polymorphonuclear neutrophils, lymphocytes and mononuclear macrophages passed through vascular endothelial cells and entered the brain tissue. These cells recognize antigens exposed by the central nervous system in the brain, activate adaptive immune responses, and further mediate secondary neuronal damage, aggravating neurological deficits. In order to reduce the above-mentioned damage, the body induces peripheral immunosuppressive responses through negative feedback, which increases the incidence of post-stroke infection. This process is accompanied by changes in the immune status of the ischemic brain tissue in local and systemic systems. A growing number of studies implicate noncoding RNAs (ncRNAs) as novel epigenetic regulatory elements in the dysfunction of various cell subsets in the neurovascular unit after cerebral infarction/ischemia-reperfusion injury. In particular, recent studies have revealed advances in ncRNA biology that greatly expand the understanding of epigenetic regulation of immune responses and inflammation after cerebral infarction/ischemia-reperfusion injury. Identification of aberrant expression patterns and associated biological effects of ncRNAs in patients revealed their potential as novel biomarkers and therapeutic targets for cerebral infarction/ischemia-reperfusion injury. Therefore, this review systematically presents recent studies on the involvement of ncRNAs in cerebral infarction/ischemia-reperfusion injury and neuroimmune inflammatory cascades, and elucidates the functions and mechanisms of cerebral infarction/ischemia-reperfusion-related ncRNAs, providing new opportunities for the discovery of disease biomarkers and targeted therapy. Furthermore, this review introduces clustered regularly interspaced short palindromic repeats (CRISPR)-Display as a possible transformative tool for studying lncRNAs. In the future, ncRNA is expected to be used as a target for diagnosing cerebral infarction/ischemia-reperfusion injury, judging its prognosis and treatment, thereby significantly improving the prognosis of patients.
Collapse
Affiliation(s)
- Kailin Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Liuting Zeng
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Anqi Ge
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Shanshan Wang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Jinsong Zeng
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Xiao Yuan
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Zhigang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Guozuo Wang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Jinwen Ge
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
- Hunan Academy of Chinese Medicine, Changsha, China
- *Correspondence: Jinwen Ge,
| |
Collapse
|
13
|
Shu J, Yang L, Wei W, Zhang L. Identification of programmed cell death-related gene signature and associated regulatory axis in cerebral ischemia/reperfusion injury. Front Genet 2022; 13:934154. [PMID: 35991562 PMCID: PMC9385974 DOI: 10.3389/fgene.2022.934154] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 07/06/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Numerous studies have suggested that programmed cell death (PCD) pathways play vital roles in cerebral ischemia/reperfusion (I/R) injury. However, the specific mechanisms underlying cell death during cerebral I/R injury have yet to be completely clarified. There is thus a need to identify the PCD-related gene signatures and the associated regulatory axes in cerebral I/R injury, which should provide novel therapeutic targets against cerebral I/R injury. Methods: We analyzed transcriptome signatures of brain tissue samples from mice subjected to middle cerebral artery occlusion/reperfusion (MCAO/R) and matched controls, and identified differentially expressed genes related to the three types of PCD(apoptosis, pyroptosis, and necroptosis). We next performed functional enrichment analysis and constructed PCD-related competing endogenous RNA (ceRNA) regulatory networks. We also conducted hub gene analysis to identify hub nodes and key regulatory axes. Results: Fifteen PCD-related genes were identified. Functional enrichment analysis showed that they were particularly associated with corresponding PCD-related biological processes, inflammatory response, and reactive oxygen species metabolic processes. The apoptosis-related ceRNA regulatory network was constructed, which included 24 long noncoding RNAs (lncRNAs), 41 microRNAs (miRNAs), and 4 messenger RNAs (mRNAs); the necroptosis-related ceRNA regulatory network included 16 lncRNAs, 20 miRNAs, and 6 mRNAs; and the pyroptosis-related ceRNA regulatory network included 15 lncRNAs, 18 miRNAs, and 6 mRNAs. Hub gene analysis identified hub nodes in each PCD-related ceRNA regulatory network and seven key regulatory axes in total, namely, lncRNA Malat1/miR-181a-5p/Mapt, lncRNA Malat1/miR-181b-5p/Mapt, lncRNA Neat1/miR-181a-5p/Mapt, and lncRNA Neat1/miR-181b-5p/Mapt for the apoptosis-related ceRNA regulatory network; lncRNA Neat1/miR-181a-5p/Tnf for the necroptosis-related ceRNA regulatory network; lncRNA Malat1/miR-181c-5p/Tnf for the pyroptosis-related ceRNA regulatory network; and lncRNAMalat1/miR-181a-5p for both necroptosis-related and pyroptosis-related ceRNA regulatory networks. Conclusion: The results of this study supported the hypothesis that these PCD pathways (apoptosis, necroptosis, pyroptosis, and PANoptosis) and crosstalk among them might be involved in ischemic stroke and that the key nodes and regulatory axes identified in this study might play vital roles in regulating the above processes. This may offer new insights into the potential mechanisms underlying cell death during cerebral I/R injury and provide new therapeutic targets for neuroprotection.
Collapse
Affiliation(s)
| | | | - Wenshi Wei
- *Correspondence: Wenshi Wei, ; Li Zhang,
| | - Li Zhang
- *Correspondence: Wenshi Wei, ; Li Zhang,
| |
Collapse
|
14
|
Cao Y, Liu J, Lu Q, Huang K, Yang B, Reilly J, Jiang N, Shu X, Shang L. An update on the functional roles of long non‑coding RNAs in ischemic injury (Review). Int J Mol Med 2022; 50:91. [PMID: 35593308 PMCID: PMC9170192 DOI: 10.3892/ijmm.2022.5147] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 05/05/2022] [Indexed: 11/20/2022] Open
Abstract
Ischemic injuries result from ischemia and hypoxia in cells. Tissues and organs receive an insufficient supply of nutrients and accumulate metabolic waste, which leads to the development of inflammation, fibrosis and a series of other issues. Ischemic injuries in the brain, heart, kidneys, lungs and other organs can cause severe adverse effects. Acute renal ischemia induces acute renal failure, heart ischemia induces myocardial infarction and cerebral ischemia induces cerebrovascular accidents, leading to loss of movement, consciousness and possibly, life-threatening disabilities. Existing evidence suggests that long non-coding RNAs (lncRNAs) are regulatory sequences involved in transcription, post-transcription, epigenetic regulation and multiple physiological processes. lncRNAs have been shown to be differentially expressed following ischemic injury, with the severity of the ischemic injury being affected by the upregulation or downregulation of certain types of lncRNA. The present review article provides an extensive summary of the functional roles of lncRNAs in ischemic injury, with a focus on the brain, heart, kidneys and lungs. The present review mainly summarizes the functional roles of lncRNA MALAT1, lncRNA MEG3, lncRNA H19, lncRNA TUG1, lncRNA NEAT1, lncRNA AK139328 and lncRNA CAREL, among which lncRNA MALAT1, in particular, plays a crucial role in ischemic injury and is currently a hot research topic.
Collapse
Affiliation(s)
- Yanqun Cao
- School of Basic Medical Sciences, Shaoyang University, Shaoyang, Hunan 422000, P.R. China
| | - Jia Liu
- School of Basic Medical Sciences, Shaoyang University, Shaoyang, Hunan 422000, P.R. China
| | - Quzhe Lu
- School of Basic Medical Sciences, Shaoyang University, Shaoyang, Hunan 422000, P.R. China
| | - Kai Huang
- School of Basic Medical Sciences, Shaoyang University, Shaoyang, Hunan 422000, P.R. China
| | - Baolin Yang
- Department of Human Anatomy, School of Basic Medicine, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - James Reilly
- Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Glasgow G4 0BA, UK
| | - Na Jiang
- Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Clinical Research Center for Ophthalmic Disease, Nanchang, Jiangxi 330006, P.R. China
| | - Xinhua Shu
- School of Basic Medical Sciences, Shaoyang University, Shaoyang, Hunan 422000, P.R. China
| | - Lei Shang
- Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Clinical Research Center for Ophthalmic Disease, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
15
|
Li MT, Luo CH, Xu PJ, Jin Y. Study on Circulating lncRNA Expression Profile in Patients with Cerebral Infarction. DISEASE MARKERS 2022; 2022:3042105. [PMID: 35585938 PMCID: PMC9110134 DOI: 10.1155/2022/3042105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 04/07/2022] [Accepted: 04/19/2022] [Indexed: 02/05/2023]
Abstract
To analyze the difference of circulating lncRNA expression profile between the healthy control group and cerebral infarction (CI) patients and to study the epigenetic pathogenesis of CI. Forty patients with acute CI admitted to our hospital from December 2016 to December 2017 were selected as CI group, and 40 healthy people in physical examination center were selected as healthy group. In the CI group, blood samples were taken 5 mL at fasting in the morning (within 72 hours of CI), and the blood samples from healthy group were also taken 5 mL at fasting in the morning. The circulating lncRNA expression profile of serum sample was determined by high-throughput technique, and its difference was analyzed. Bioinformatics technology was used to explore its functional mechanism, and GO, KEGG analysis, and gene expression network were established for lncRNA with significant differences. Next, lnc-ZNF32-1 : 1 and lnc-PCGF5-2 : 1 were selected for further validation of serum lncRNA expression in ACI and NC groups, and ceRNA interaction network analysis, diagnostic specificity, and sensitivity of lnc-ZNF32-1 : 1 and lnc-PCGF5-2 : 1 were conducted. The results showed that compared with the healthy group, there were 512 known lncRNA expressed differentially in the serum of patients with CI, of which 371 were upregulated and 141 were downregulated, and 421 known mRNA expressed differentially, of which 245 were upregulated and 176 downregulated. The differentially expressed mRNA was mainly enriched in 53 gene functions, and the target gene was enriched in the pathways such as HTLV-I infection and pathways in cancer. In addition, the results explored that lnc-ZNF32-1 : 1 and lnc-PCGF5-2 : 1 have potential value for CI diagnosis. In conclusion, the expression profile of lncRNA in CI group was significantly different from that in healthy group, indicating that lncRNA might be closely related to the occurrence, development, and prognosis of CI.
Collapse
Affiliation(s)
- Man-tang Li
- Department of Neurology, SSL Central Hospital of Dongguan City, Dongguan Third People's Hospital, Dongguan, 523320 Guangdong Province, China
| | - Cheng-hong Luo
- Department of Neurology, SSL Central Hospital of Dongguan City, Dongguan Third People's Hospital, Dongguan, 523320 Guangdong Province, China
| | - Peng-jie Xu
- Department of Neurology, SSL Central Hospital of Dongguan City, Dongguan Third People's Hospital, Dongguan, 523320 Guangdong Province, China
| | - Ya Jin
- Department of Neurology, SSL Central Hospital of Dongguan City, Dongguan Third People's Hospital, Dongguan, 523320 Guangdong Province, China
| |
Collapse
|
16
|
Li Y, Peng B, Li Y, Huang A, Peng Y, Yu Q, Li Y. MiR-203a-3p/153-3p improves cognitive impairments induced by ischemia/reperfusion via blockade of SRC-mediated MAPK signaling pathway in ischemic stroke. Chem Biol Interact 2022; 358:109900. [DOI: 10.1016/j.cbi.2022.109900] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 01/13/2022] [Accepted: 03/14/2022] [Indexed: 12/17/2022]
|
17
|
Ouyang M, Fang J, Wang M, Huang X, Lan J, Qu Y, Lai W, Xu Q. Advanced glycation end products alter the m 6A-modified RNA profiles in human dermal fibroblasts. Epigenomics 2022; 14:431-449. [PMID: 35285253 DOI: 10.2217/epi-2022-0016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Background: To explore advanced glycation end products (AGEs)-induced m6A modification in fibroblasts and its potential role in photoaging. Methods: We studied m6A modification in AGEs-bovine serum albumin-treated fibroblasts with m6A-mRNA & lncRNA epitranscriptomic microarray and bioinformatics analysis. The m6A modification level was also investigated in skin samples. Results: m6A methylation microarray analysis revealed m6A modification profiles in AGEs-treated fibroblasts. Gene ontology, Kyoto Encyclopedia of Genes and Genomes, protein-protein interaction and competing endogenous RNA network analysis indicated that the genes of differentially methylated mRNAs and lncRNAs were mainly related to inflammation processes. We also found that AGEs-bovine serum albumin dose-dependently increased the m6A level and METTL14 expression in both fibroblasts and sun-exposed skin. Conclusion: Our study provided novel information regarding alterations of m6A modifications in AGEs-induced dermal fibroblasts and potential targets for treatment of photoaging.
Collapse
Affiliation(s)
- Mengting Ouyang
- Department of Dermato-Venereology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Jiaqi Fang
- Department of Dermato-Venereology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Mengyao Wang
- Department of Dermato-Venereology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Xianyin Huang
- Department of Dermato-Venereology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Jingjing Lan
- Department of Dermato-Venereology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Yingying Qu
- Department of Dermato-Venereology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Wei Lai
- Department of Dermato-Venereology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Qingfang Xu
- Department of Dermato-Venereology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| |
Collapse
|