1
|
Xia S, Ye Y, Liu J, Qiu H, Lin M, He Z, Huang L, Wang M, Luo Y. The Role of MALAT1 in Regulating the Proangiogenic Functions, Invasion, and Migration of Trophoblasts in Selective Fetal Growth Restriction. Biomolecules 2024; 14:988. [PMID: 39199376 PMCID: PMC11352967 DOI: 10.3390/biom14080988] [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: 07/02/2024] [Revised: 08/01/2024] [Accepted: 08/09/2024] [Indexed: 09/01/2024] Open
Abstract
Epigenetic regulation is an important entry point to study the pathogenesis of selective fetal growth restriction (sFGR), and an understanding of the role of long noncoding RNAs (lncRNAs) in sFGR is lacking. Our study aimed to investigate the potential role of a lncRNA, metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), in sFGR using molecular biology experiments and gain- or loss-of-function assays. We found that the levels of MALAT1, ERRγ, and HSD17B1 were downregulated and that of miR-424 was upregulated in the placental shares of the smaller twins. Moreover, angiogenesis was impaired in the placental share of the smaller fetus and MALAT1 could regulate the paracrine effects of trophoblasts on endothelium angiogenesis and proliferation by regulating miR-424. In trophoblasts, MALAT1 could competitively bind to miR-424 to regulate the expression of ERRγ and HSD17B1, thus regulating trophoblast invasion and migration. MALAT1 overexpression could decrease apoptosis and promote proliferation, alleviating cell damage induced by hypoxia. Taken together, the downregulation of MALAT1 can reduce the expression of ERRγ and HSD17B1 by competitively binding to miR-424, impairing the proangiogenic effect of trophoblasts, trophoblast invasion and migration, and the ability of trophoblasts to compensate for hypoxia, which may be involved in the pathogenesis of sFGR through various aspects.
Collapse
Affiliation(s)
- Shuting Xia
- Department of Obstetrics & Gynecology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China; (S.X.); (Y.Y.); (J.L.); (H.Q.); (M.L.); (Z.H.); (L.H.); (M.W.)
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou 510080, China
| | - Yingnan Ye
- Department of Obstetrics & Gynecology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China; (S.X.); (Y.Y.); (J.L.); (H.Q.); (M.L.); (Z.H.); (L.H.); (M.W.)
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou 510080, China
| | - Jialiu Liu
- Department of Obstetrics & Gynecology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China; (S.X.); (Y.Y.); (J.L.); (H.Q.); (M.L.); (Z.H.); (L.H.); (M.W.)
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou 510080, China
| | - Hanfei Qiu
- Department of Obstetrics & Gynecology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China; (S.X.); (Y.Y.); (J.L.); (H.Q.); (M.L.); (Z.H.); (L.H.); (M.W.)
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou 510080, China
| | - Minhuan Lin
- Department of Obstetrics & Gynecology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China; (S.X.); (Y.Y.); (J.L.); (H.Q.); (M.L.); (Z.H.); (L.H.); (M.W.)
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou 510080, China
| | - Zhiming He
- Department of Obstetrics & Gynecology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China; (S.X.); (Y.Y.); (J.L.); (H.Q.); (M.L.); (Z.H.); (L.H.); (M.W.)
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou 510080, China
| | - Linhuan Huang
- Department of Obstetrics & Gynecology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China; (S.X.); (Y.Y.); (J.L.); (H.Q.); (M.L.); (Z.H.); (L.H.); (M.W.)
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou 510080, China
| | - Malie Wang
- Department of Obstetrics & Gynecology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China; (S.X.); (Y.Y.); (J.L.); (H.Q.); (M.L.); (Z.H.); (L.H.); (M.W.)
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou 510080, China
| | - Yanmin Luo
- Department of Obstetrics & Gynecology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China; (S.X.); (Y.Y.); (J.L.); (H.Q.); (M.L.); (Z.H.); (L.H.); (M.W.)
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou 510080, China
| |
Collapse
|
2
|
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
|
3
|
Hu J, Huang K, Bao F, Zhong S, Fan Q, Li W. Low-dose lipopolysaccharide inhibits spinal cord injury-induced neuronal apoptosis by regulating autophagy through the lncRNA MALAT1/Nrf2 axis. PeerJ 2023; 11:e15919. [PMID: 37663283 PMCID: PMC10470450 DOI: 10.7717/peerj.15919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 07/27/2023] [Indexed: 09/05/2023] Open
Abstract
Background Spinal cord injury (SCI) is a neurological disease associated with a high disability rate. Low-dose lipopolysaccharide (LPS) has been reported to activate cross-immune tolerance and alleviate the effects of various traumatic stimuli. The present study aimed to explore the effect of LPS on SCI and the potential molecular mechanism. Methods Male Sprague-Dawley (SD) rats were used to established an in vivo SCI model and were intraperitoneally injected with lentivirus particles encoding a MALAT1 small interfering RNA (siRNA) on day 10 prior to SCI and with 0.2 mg/kg LPS 72 h prior to SCI. Basso, Beattie, and Bresnahan (BBB) scoring; HE staining; and TUNEL assay were used to assess neurological function and pathophysiological changes. Western blot and immunohistochemistry (IHC) were used to detect cell autophagy and Nrf2 nuclear translocation. PC12 cells were exposed to oxygen-glucose deprivation/reoxygenation (OGD/R) to establish an in vitro SCI model. In vitro SCI model cells were pretreated with LPS and transfected with siMALAT1 or MALAT1 overexpression plasmid aimed at knocking down MALAT1 or overexpressing MALAT1. The cell counting kit-8 (CCK-8) assay was used to measure the toxicity of LPS towards PC12 cells. Flow cytometry and immunofluorescence analysis were performed to investigate cell apoptosis and Nrf2 nuclear translocation. Results SCI rats preconditioned with low-dose LPS had higher BBB scores, reduced SCI injury, increased MALAT1 expression and activated autophagy and Nrf2 nuclear translocation in the in vivo SCI model. In the in vitro SCI model, low-dose LPS treatment suppressed the apoptotic ratio of PC12 cells, increased MALAT1 expression, activated autophagy, and promoted Nrf2 nuclear translocation. Silencing MALAT1 exacerbated OGD/R injury in vitro and weakened the protective effect of low-dose LPS. Overexpression of MALAT1 inhibits OGD/R-induced apoptosis by inducing autophagy and promoting Nrf2 nuclear translocation. This was also been confirmed in animal experiments, silencing MALAT1 blocked the promotion of Nrf2 by low-dose LPS and the alleviated of SCI apoptosis. Conclusions Low-dose LPS exhibited a protective role on SCI by activating autophagy and suppressing nerve cell apoptosis via the lncRNA MALAT1/Nrf2 axis.
Collapse
Affiliation(s)
- Jianhua Hu
- Orthopedic Surgery, The First People’s Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
- Faculty of Medical Science, Kunming University of Science and Technology, Kunming, China
| | - Kun Huang
- Orthopedic Surgery, The First People’s Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Feilong Bao
- Faculty of Medical Science, Kunming University of Science and Technology, Kunming, China
- Yunnan Key Laboratory of Digital Orthopaedics, Kunming, China
| | - Shixiao Zhong
- Faculty of Medical Science, Kunming University of Science and Technology, Kunming, China
- Yunnan Key Laboratory of Digital Orthopaedics, Kunming, China
| | - Qianbo Fan
- Faculty of Medical Science, Kunming University of Science and Technology, Kunming, China
- Yunnan Key Laboratory of Digital Orthopaedics, Kunming, China
| | - Weichao Li
- Orthopedic Surgery, The First People’s Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
- Faculty of Medical Science, Kunming University of Science and Technology, Kunming, China
- Yunnan Key Laboratory of Digital Orthopaedics, Kunming, China
| |
Collapse
|
4
|
Davis MJ, Earley S, Li YS, Chien S. Vascular mechanotransduction. Physiol Rev 2023; 103:1247-1421. [PMID: 36603156 PMCID: PMC9942936 DOI: 10.1152/physrev.00053.2021] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 09/26/2022] [Accepted: 10/04/2022] [Indexed: 01/07/2023] Open
Abstract
This review aims to survey the current state of mechanotransduction in vascular smooth muscle cells (VSMCs) and endothelial cells (ECs), including their sensing of mechanical stimuli and transduction of mechanical signals that result in the acute functional modulation and longer-term transcriptomic and epigenetic regulation of blood vessels. The mechanosensors discussed include ion channels, plasma membrane-associated structures and receptors, and junction proteins. The mechanosignaling pathways presented include the cytoskeleton, integrins, extracellular matrix, and intracellular signaling molecules. These are followed by discussions on mechanical regulation of transcriptome and epigenetics, relevance of mechanotransduction to health and disease, and interactions between VSMCs and ECs. Throughout this review, we offer suggestions for specific topics that require further understanding. In the closing section on conclusions and perspectives, we summarize what is known and point out the need to treat the vasculature as a system, including not only VSMCs and ECs but also the extracellular matrix and other types of cells such as resident macrophages and pericytes, so that we can fully understand the physiology and pathophysiology of the blood vessel as a whole, thus enhancing the comprehension, diagnosis, treatment, and prevention of vascular diseases.
Collapse
Affiliation(s)
- Michael J Davis
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Scott Earley
- Department of Pharmacology, University of Nevada, Reno, Nevada
| | - Yi-Shuan Li
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
| | - Shu Chien
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
- Department of Medicine, University of California, San Diego, California
| |
Collapse
|
5
|
Lo YL, Lin HC, Tseng WH. Tumor pH-functionalized and charge-tunable nanoparticles for the nucleus/cytoplasm-directed delivery of oxaliplatin and miRNA in the treatment of head and neck cancer. Acta Biomater 2022; 153:465-480. [PMID: 36115656 DOI: 10.1016/j.actbio.2022.09.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 09/06/2022] [Accepted: 09/09/2022] [Indexed: 11/01/2022]
Abstract
Prospective tumor pH-responsive and charge-convertible nanoparticles have been utilized to reduce side effects and improve the active tumor-targeting ability and nuclear/cytoplasmic localization of chemo- and gene therapeutics for the treatment of head and neck cancer (HNC). Oxaliplatin (Oxa) is a third-generation platinum compound that prevents DNA replication. miR-320 may regulate cancer cell apoptosis, resistance, and progression. Innovative nanoparticles incorporating miR-320 and Oxa were modified with a ligand, cell-penetrating peptide, and nucleus-targeted peptide. The nanoparticles were coated with a charge/size-tunable shield to prevent peptide degradation and decoated at acidic tumor sites to expose peptides for active targeting. Results indicated that the designed nanoparticles exhibited a uniform size and satisfactory drug encapsulation efficiency. The nanoparticles displayed the pH-responsive release and uptake of Oxa and miR-320 into human tongue squamous carcinoma SAS cells. The nanoparticles successfully delivered Oxa and miR-320 to the nucleus and cytoplasm, respectively. This work is the first to demonstrate the concurrent intracellular modulation of the NRP1/Rac1, PI3K/Akt/mTOR, GSK-3β/FOXM1/β-catenin, P-gp/MRPs, KRAS/Erk/Oct4/Yap1, and N-cadherin/Vimentin/Slug pathways to inhibit the growth, progression, and multidrug resistance of cancer cells. In SAS-bearing mice, co-treatment with Oxa- and miR-320-loaded nanoparticles exhibited superior antitumor efficacy and remarkably decreased Oxa-associated toxicities. The nucleus/cytoplasm-localized nanoparticles with a tumor pH-sensitive and size/charge-adjustable coating may be a useful combinatorial spatiotemporal nanoplatform for nucleic acids and chemotherapeutics to achieve maximum therapeutic safety and efficacy against HNC. STATEMENT OF SIGNIFICANCE: Innovative nanoparticles incorporating miR-320 and oxaliplatin were modified with a ligand, cell-penetrating peptide, and nucleus-targeted peptide. The tumor pH-sensitive and charge/size-adjustable shield of polyglutamic acid-PEG protected against peptide degradation during systemic circulation. This work represents the first example of the concurrent intracellular modulation of the NRP1/Rac1, PI3K/Akt/mTOR, GSK-3β/FOXM1/β-catenin, P-gp/MRPs, KRAS/Erk/Oct4/Yap1, and N-cadherin/Vimentin/Slug pathways to inhibit cancer cell growth, cancer cell progression, and multidrug resistance simultaneously. The versatile nanoparticles with a tumor pH-functionalized coating could deliver chemotherapeutics and miRNA to the nucleus/cytoplasm. The nanoparticles successfully reduced chemotherapy-associated toxicities and maximized the antitumor efficacy of combinatorial therapy against head and neck cancer.
Collapse
Affiliation(s)
- Yu-Li Lo
- Department and Institute of Pharmacology, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; Faculty of Pharmacy, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan.
| | - Hua-Ching Lin
- Division of Colorectal Surgery, Surgical Department, Chen-Hsin General Hospital, Taipei, Taiwan; Department of Healthcare Information and Management, Ming Chuan University, Taoyuan, Taiwan
| | - Wei-Hsuan Tseng
- Department and Institute of Pharmacology, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| |
Collapse
|
6
|
Cui Y, Xu H, Yang Y, Zhao D, Wen Y, Lv C, Qiu H, Wang C. The regulation of miR-320a/XBP1 axis through LINC00963 for endoplasmic reticulum stress and autophagy in diffuse large B-cell lymphoma. Cancer Cell Int 2021; 21:305. [PMID: 34112145 PMCID: PMC8194177 DOI: 10.1186/s12935-021-01992-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 05/22/2021] [Indexed: 02/06/2023] Open
Abstract
Background This study incorporates fundamental research referring to considerable amounts of gene-sequencing data and bioinformatics tools to analyze the pathological mechanisms of diffuse large B-cell lymphoma (DLBCL). Methods A lncRNA-miRNA-mRNA ceRNA network of DLBCL was constructed through database analysis combining GTEx and TCGA. qPCR was used to detect the expression of LINC00963 and miR-320a in DLBCL cell lines. After LINC00963 or miR-320a overexpression in vitro, western blot was performed to assess the protein levels of UPR sensors (GRP78, p-IRE1, IRE1, active ATF6, ATF4 and XBP1), along with apoptosis markers (Bcl-2, Bax, caspase 3) and autophagy indicators (Beclin1, LC3II, LC3I and p62). Additionally, the expression of LC3 was analyzed through immunofluorescence (IF) assay. Results Following LINC00963 overexpression in vitro, SUDHL4 cell line showed a marked increase in the level of UPR-related GRP78, p-IRE1 and spliced XBP-1/XBP-1(s), apoptosis-related Bax and cleaved caspase 3, as well as autophagy-related Beclin1 and LC3II, whereas miR-320a mimic greatly diminished the effects of LINC00963 overexpression. Moreover, LINC00963 targeted miR-320a while miR-320a bound to the 3’UTR of XBP1. It was also found that LINC00963 overexpression resulted in significantly delayed tumor growth in a xenograft model of DLBCL. Conclusion Mechanistically, LINC00963/miR-320a regulated XBP1-apoptosis pathway
and autophagy, implying the therapeutic potential of
this pathway for selective targeting. The data presented here illustrated
the mechanism of LINC00963/miR-320a/XBP1 in DLBCL for
the first time. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-01992-y.
Collapse
Affiliation(s)
- Yuying Cui
- School of Basic Medicine, Jiamusi University, Jiamusi, 154007, Heilongjiang, China
| | - Hui Xu
- School of Basic Medicine, Jiamusi University, Jiamusi, 154007, Heilongjiang, China
| | - Yu Yang
- School of Basic Medicine, Jiamusi University, Jiamusi, 154007, Heilongjiang, China
| | - Dongmei Zhao
- School of Public Health, Jiamusi University, Jiamusi, 154007, Heilongjiang, China.,School of Clinical Medical, Jiamusi University, Jiamusi, 154007, Heilongjiang, China
| | - Yu Wen
- School of Basic Medicine, Jiamusi University, Jiamusi, 154007, Heilongjiang, China
| | - Chao Lv
- School of Basic Medicine, Jiamusi University, Jiamusi, 154007, Heilongjiang, China
| | - Hongbin Qiu
- School of Basic Medicine, Jiamusi University, Jiamusi, 154007, Heilongjiang, China.
| | - Chennan Wang
- School of Basic Medicine, Jiamusi University, Jiamusi, 154007, Heilongjiang, China.
| |
Collapse
|
7
|
Exercise Improves Endothelial Function via the lncRNA MALAT1/miR-320a Axis in Obese Children and Adolescents. Cardiol Res Pract 2021; 2021:8840698. [PMID: 34123418 PMCID: PMC8189819 DOI: 10.1155/2021/8840698] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 02/25/2021] [Accepted: 05/20/2021] [Indexed: 01/10/2023] Open
Abstract
Background Endothelial dysfunction commonly occurs in obese children and adolescents, leading to an increased risk of cardiovascular diseases. Exercise has significant protective effects against endothelial dysfunction through regulating some noncoding RNAs. This study aimed to investigate the relationship of long noncoding RNA MALAT1 and microRNA-320a (miR-320a) with the exercise-induced improvement of endothelial dysfunction in obese children and adolescents. Methods Sixty obese children and adolescents were included in this study, including 40 cases that received 12-week exercise training and 20 cases that received only diet control. The anthropometric and blood indices before and after exercise were recorded and compared, and the endothelial dysfunction was evaluated by examining the levels of markers, including VCAM-1, ICAM-1, and E-selectin, using an ELISA assay. The expression levels of noncoding RNAs were assessed using real-time quantitative PCR, and their correlation with patients' recorded indices and endothelial dysfunction markers was analyzed. Results The 12-week exercise training significantly decreased the levels of VCAM-1, ICAM-1, and E-selectin and could inhibit MALAT1 but promote miR-320a expression in obese children and adolescents. The expression of MALAT1 and miR-320a was correlated with the changes in the anthropometric and blood indices of obese children and adolescents, and their correlations with endothelial dysfunction markers were obtained. Conclusion All the data revealed that exercise has significantly protective effects against endothelial dysfunction and can regulate the expression of the MALAT1/miR-320a axis. MALAT1 and miR-320a were correlated with endothelial dysfunction markers, indicating that the MALAT1/miR-320a axis may be related with the alleviating effects of exercise on endothelial function in obese children and adolescents.
Collapse
|
8
|
Zhao W, Yin Y, Cao H, Wang Y. LncRNA MALAT1/miR-320a axis is associated with exercise-induced improvement of endothelial dysfunction in obese children. Microvasc Res 2021:104194. [PMID: 34062189 DOI: 10.1016/j.mvr.2021.104194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 03/15/2021] [Accepted: 05/26/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Endothelial dysfunction commonly occurs in obese children, leading to increased risk of cardiovascular diseases. Exercise has protective effects against endothelial dysfunction through regulating some noncoding RNAs. This study aimed to investigate the relationship of long noncoding RNA MALAT1 and microRNA-320a (miR-320a) with the exercise-induced improvement of endothelial dysfunction in obese children. METHODS Sixty obese children were included in this study, and 40 cases received a 12-week exercise training. The morphological and blood indices before and after exercise were recorded and compared, and the endothelial dysfunction was evaluated by examining the levels of VCAM-1, ICAM-1, E-selectin, IL-6 and TNF-α using ELISA kits. The expression of noncoding RNAs was assessed using Real-Time quantitative PCR. Endothelial cells were used to explore the effects of MALAT1 and miR-320a on endothelial function. RESULTS The 12-well exercise training decreased the levels of VCAM-1, ICAM-1 and E-selectin, and inhibited MALAT1 but promoted miR-320a expression in obese children. The expression of MALAT1 and miR-320a was correlated with the changes of morphological and blood indices in obese children, and their correlations with endothelial dysfunction markers were obtained. Additionally, MALAT1 overexpression or miR-320a reduction led to inhibited proliferation and increased inflammation in HUVECs. CONCLUSION All the data revealed that exercise has significantly protective effects against endothelial dysfunction, and can regulate the expression of the MALAT1/miR-320a axis. MALAT1 and miR-320a were correlated with endothelial dysfunction, indicating that the MALAT1/miR-320a axis may be related with the alleviating effects of exercise on endothelial function in obese children.
Collapse
Affiliation(s)
- Wei Zhao
- Department of Neonatology, Weifang People's Hospital, Weifang 261041, Shandong, China
| | - Yane Yin
- Department of Neonatology, Weifang People's Hospital, Weifang 261041, Shandong, China
| | - Huiling Cao
- Department of Neonatology, Weifang People's Hospital, Weifang 261041, Shandong, China
| | - Yandong Wang
- Department of Pediatrics, Weifang People's Hospital, Weifang 261041, Shandong, China.
| |
Collapse
|
9
|
Lu Y, Xi J, Zhang Y, Chen W, Zhang F, Li C, Wang Z. SNHG1 Inhibits ox-LDL-Induced Inflammatory Response and Apoptosis of HUVECs via Up-Regulating GNAI2 and PCBP1. Front Pharmacol 2020; 11:703. [PMID: 32536864 PMCID: PMC7266976 DOI: 10.3389/fphar.2020.00703] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 04/29/2020] [Indexed: 12/30/2022] Open
Abstract
Dysfunction of human endothelial cells is an important trigger for atherosclerosis. Oxidative low-density lipoprotein (ox-LDL) usually was used to stimulate the dysfunction of human umbilical vein endothelial cells (HUVECs). LncRNA SNHG1 (small nucleolar RNA host gene 1) is a cerebral infarction-associated gene. The present study was designed to investigate the role of SNHG1 in ox-LDL-induced HUVECs. Cell viability was evaluated by CCK-8 and MTT assay. Cell apoptosis was detected by flow cytometry analysis. Cell inflammatory response was evaluated by detecting LDH, IL-6, IL-1β levels. The results revealed that up-regulation of SNHG1 attenuated ox-LDL-induced cell injury and inflammatory response in HUVECs. Next, mechanism assays including RNA immunoprecipitation (RIP) assay, luciferase reporter assay, and RNA pull-down assay, helped us to identify the interaction between miR-556-5 and SNHG1. GNAI2 (G protein subunit alpha i2) and PCBP1 (poly(rC) binding protein 1) were identified as the downstream targets of miR-556-5p. SNHG1 regulated dysfunctions of ox-LDL-induced HUVECs via sponging miR-556-5p and up-regulating GNAI2 and PCBP1. SNHG1 attenuated cell injury and inflammatory response in ox-LDL-induced HUVECs via up-regulating both GNAI2 and PCBP1 at a miR-556-5p dependent way.
Collapse
Affiliation(s)
- Yuan Lu
- Department of Cardiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Jue Xi
- Department of Endocrinology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yao Zhang
- Department of Cardiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Wensu Chen
- Department of Cardiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Fengyun Zhang
- Department of Cardiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Chenzong Li
- Department of Cardiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Zhirong Wang
- Department of Cardiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
10
|
Luo L, Wang Y, Hu P, Wu J. Long Non-Coding RNA Metastasis Associated Lung Adenocarcinoma Transcript 1 (MALAT1) Promotes Hypertension by Modulating the Hsa-miR-124-3p/Nuclear Receptor Subfamily 3, Group C, Member 2 (NR3C2) and Hsa-miR-135a-5p/NR3C2 Axis. Med Sci Monit 2020; 26:e920478. [PMID: 32222724 PMCID: PMC7139186 DOI: 10.12659/msm.920478] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background This study was designed to investigate the role of long non-coding RNA (lncRNA) metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) in the proliferation as well as apoptosis of human umbilical vein endothelial cells (HUVECs), to offer a basis for therapy of hypertension. Material/Methods The lncRNA MALAT1 expression, hsa-miR-124-3p, hsa-miR-135a-5p, hsa-miR-135b-5p, and hsa-miR-455-5p in plasma were measured from 230 patients with hypertension and 230 non-hypertensive controls. The mechanism for lncRNA MALAT1 modulating the proliferation and apoptosis of HUVECs was explored by cell transfection, Cell Counting Kit-8 (CCK-8), quantitative real-time polymerase chain reaction (qRT-PCR), western blot, and dual-luciferase reporter assays. Results The expression of hsa-miR-124-3p and hsa-miR-135a-5p was reduced and the expression of lncRNA MALAT1 was increased in the plasma of hypertensive patients. Moreover, the plasma levels of hsa-miR-124-3p and hsa-miR-135a-5p of hypertensive patients were negatively correlated with lncRNA MALAT1 (r=−0.64, −0.72; P<0.01, P<0.01, respectively). The level of nuclear receptor subfamily 3, group C, member 2 (NR3C2) protein was negatively correlated with hsa-miR-124-3p and hsa-miR-135a-5p (r=−0.74, −0.84; P<0.01, P<0.01, respectively). The proliferation of HUVECs was inhibited after the inhibition of MALAT. Additionally, after knocking down MALAT, the levels of hsa-miR-124-3p and hsa-miR-135a-5p in HUVECs were markedly increased, while the expression level of NR3C2 protein was decreased. The apoptotic rate of HUVECs after the transfection of MALAT1 small interfering RNA (si-MALAT1) (3.64±0.21%) was significantly reduced compared to that of transfected si-MALAT1 no template control (NC) (3.76±0.19%) and the control group (10.51±1.24%). Conclusions LncRNA MALAT1 regulates proliferation and apoptosis of HUVECs through the hsa-miR-124-3p/NR3C2 and/or hsa-miR-135a-5p/NR3C2 axis.
Collapse
Affiliation(s)
- Liju Luo
- Department of Geratology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China (mainland)
| | - Yu Wang
- Department of Cardiology, The Affiliated Yueqing Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China (mainland)
| | - Pengfei Hu
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China (mainland)
| | - Jiale Wu
- Department of Geratology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China (mainland)
| |
Collapse
|