1
|
Qu Y, Xue S, Zheng Y, Du Y, Zhang G, Huang L, Li H, Li H. Upregulated miR‑378a‑3p expression suppresses energy metabolism and promotes apoptosis by targeting a GLUT‑1/ALDOA/PKM2 axis in esophageal carcinoma. Oncol Lett 2023; 26:421. [PMID: 37664650 PMCID: PMC10472027 DOI: 10.3892/ol.2023.14007] [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: 08/24/2022] [Accepted: 07/10/2023] [Indexed: 09/05/2023] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is an aggressive malignancy of the digestive system with increasing incidence and mortality rates. The biological roles of microRNA (miR)-378a-3p in tumor cells remain contested, and the mechanisms underlying the functions, energy metabolism, and cell survival mechanisms in ESCC cells are yet to be fully elucidated. In the present study, miR-378a-3p overexpression and negative control plasmids were transfected into ECA-109 cells using electroporation. Western blotting was used to detect the relative expression of proteins, and flow cytometry was used to detect cell apoptosis. Subsequently, ELISA assays were performed to determine enzyme activity, and an ATP detection kit was used to measure ATP content. Dual-luciferase reporter assays were performed to identify the target genes of miR-378a-3p. The results of the present study demonstrated that miR-378a-3p inhibited the gene expression and enzyme activities of glucose transporter protein 1 (GLUT-1), Aldolase A (ALDOA), and pyruvate kinase M2 (PKM2), all of which are involved in the glycolytic pathway of cells. Energy metabolism was suppressed by miR-378a-3p by reducing ATP content, and this downregulated the expression of Bcl-2 and Survivin. Moreover, increased miR-378a-3p expression promoted cell apoptosis in the early stages by increasing the expression levels and the activity of Bad and Caspase-3, while inhibiting the expression levels of Bcl-2 and Survivin. The results of the present study also demonstrated that GLUT-1/ALDOA/PKM2 were target genes of miR-378a-3p. Notably, miR-378a-3p blocked energy production and promoted the apoptosis of tumor cells via the downregulation of glycolytic enzyme expression and by reducing the mitochondrial membrane potential in ESCC. Bad, Caspase-3, Survivin, and Bcl-2 may be associated with blocking energy production and promoting apoptosis via miR-378a-3p in ESCC cells.
Collapse
Affiliation(s)
- Yuan Qu
- Department of Labour Hygiene and Sanitary Science, College of Public Health, Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| | - Shan Xue
- Medical Research Center, Yuebei People's Hospital, Shantou University, Shaoguan, Guangdong 512025, P.R. China
| | - Yujian Zheng
- Department of Labour Hygiene and Sanitary Science, College of Public Health, Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| | - Yajing Du
- Medical Research Center, Yuebei People's Hospital, Shantou University, Shaoguan, Guangdong 512025, P.R. China
| | - Guoping Zhang
- Tumor Department, Yuebei People's Hospital, Shantou University, Shaoguan, Guangdong 512025, P.R. China
| | - Liting Huang
- Medical Research Center, Yuebei People's Hospital, Shantou University, Shaoguan, Guangdong 512025, P.R. China
| | - Hui Li
- Central Laboratory of Xinjiang Medical University, Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| | - Huiwu Li
- Medical Research Center, Yuebei People's Hospital, Shantou University, Shaoguan, Guangdong 512025, P.R. China
| |
Collapse
|
2
|
Bai X, Qi Z, Zhu M, Lu Z, Zhao X, Zhang L, Song G. The effect of lncRNA MIR155HG-modified MSCs and exosome delivery to synergistically attenuate vein graft intimal hyperplasia. Stem Cell Res Ther 2022; 13:512. [PMID: 36333764 PMCID: PMC9636746 DOI: 10.1186/s13287-022-03197-0] [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: 04/10/2022] [Accepted: 06/28/2022] [Indexed: 11/06/2022] Open
Abstract
Background The mesenchymal stem cells (MSCs) were used to repair tissue injury. However, the treatment effect was not satisfactory. We investigated whether lncRNA MIR155HG could promote survival and migration of MSCs under oxidative stress, which mimics in vivo environments. Furthermore, we studied the protective effect of exosomes secreted by MSCs transfected with MIR155HG on endothelial cells. This study aimed to determine whether exploiting MSCs and exosomes modified with lncRNA MIR155HG would exert synergistic therapeutic effect to attenuate vein graft intimal hyperplasia more effectively. Methods Lentivirus containing lncRNA MIR155HG overexpressing vector was packaged and used to infect MSCs. Then, CCK-8 assay, flow cytometry, Transwell assay, and Elisa assay were used to assess the functional changes of MSCs with overexpressed MIR155HG (OE-MSCs). Furthermore, the associated pathways were screened by Western blot. MIR155HG-MSCs-derived exosomes (OE-exo) were collected and co-cultured with human umbilicus vein endothelial cell (HUVEC). We validated the protective effect of OE-exo on HUVEC. In vivo, both MSCs and exosomes modified with MIR155HG were injected into a vein graft rat model via tail vein. We observed MSCs homing and intimal hyperplasia of vein graft using a fluorescent microscope and histological stain. Results Our study found that lncRNA MIR155HG promoted proliferation, migration, and anti-apoptosis of MSCs. NF-κB pathway took part in the regulation process induced by MIR155HG. OE-exo could enhance the activity and healing ability of HUVEC and reduce apoptosis. In vivo, OE-MSCs had a higher rate of homing to vascular endothelium. The combined treatment with OE-MSCs and OE-exo protected vascular endothelial integrity, reduced inflammatory cell proliferation, and significantly attenuated intimal hyperplasia of vein graft. Conclusion LncRNA MIR155HG could promote the survival and activity of MSCs, and reduce the apoptosis of HUVECs using exosome delivery. Exploiting MSCs and exosomes modified with MIR155HG could attenuate vein graft intimal hyperplasia more effectively and maximize the surgical effect.
Collapse
Affiliation(s)
- Xiao Bai
- grid.452402.50000 0004 1808 3430Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan, 250012 China ,grid.27255.370000 0004 1761 1174Thoracoscopy Institute of Cardiac Surgery, Shandong University, Jinan, China
| | - Zaiwen Qi
- The Fifth People’s Hospital of Jinan, Jinan, China
| | - Mingzhen Zhu
- grid.452402.50000 0004 1808 3430Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan, 250012 China
| | - Zhuangzhuang Lu
- grid.452402.50000 0004 1808 3430Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan, 250012 China
| | - Xin Zhao
- grid.452402.50000 0004 1808 3430Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan, 250012 China
| | - Lining Zhang
- grid.27255.370000 0004 1761 1174Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, 250012 China
| | - Guangmin Song
- grid.452402.50000 0004 1808 3430Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan, 250012 China
| |
Collapse
|
3
|
Valanti EK, Dalakoura-Karagkouni K, Fotakis P, Vafiadaki E, Mantzoros CS, Chroni A, Zannis V, Kardassis D, Sanoudou D. Reconstituted HDL-apoE3 promotes endothelial cell migration through ID1 and its downstream kinases ERK1/2, AKT and p38 MAPK. Metabolism 2022; 127:154954. [PMID: 34875308 DOI: 10.1016/j.metabol.2021.154954] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/15/2021] [Accepted: 11/30/2021] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Atherosclerotic Coronary Artery Disease (ASCAD) is the leading cause of mortality worldwide. Novel therapeutic approaches aiming to improve the atheroprotective functions of High Density Lipoprotein (HDL) include the use of reconstituted HDL forms containing human apolipoprotein A-I (rHDL-apoA-I). Given the strong atheroprotective properties of apolipoprotein E3 (apoE3), rHDL-apoE3 may represent an attractive yet largely unexplored therapeutic agent. OBJECTIVE To evaluate the atheroprotective potential of rHDL-apoE3 starting with the unbiased assessment of global transcriptome effects and focusing on endothelial cell (EC) migration as a critical process in re-endothelialization and atherosclerosis prevention. The cellular, molecular and functional effects of rHDL-apoE3 on EC migration-associated pathways were assessed, as well as the potential translatability of these findings in vivo. METHODS Human Aortic ECs (HAEC) were treated with rHDL-apoE3 and total RNA was analyzed by whole genome microarrays. Expression and phosphorylation changes of key EC migration-associated molecules were validated by qRT-PCR and Western blot analysis in primary HAEC, Human Coronary Artery ECs (HCAEC) and the human EA.hy926 EC line. The capacity of rHDL-apoE3 to stimulate EC migration was assessed by wound healing and transwell migration assays. The contribution of MEK1/2, PI3K and the transcription factor ID1 in rHDL-apoE3-induced EC migration and activation of EC migration-related effectors was assessed using specific inhibitors (PD98059: MEK1/2, LY294002: PI3K) and siRNA-mediated gene silencing, respectively. The capacity of rHDL-apoE3 to improve vascular permeability and hypercholesterolemia in vivo was tested in a mouse model of hypercholesterolemia (apoE KO mice) using Evans Blue assays and lipid/lipoprotein analysis in the serum, respectively. RESULTS rHDL-apoE3 induced significant expression changes in 198 genes of HAEC mainly involved in re-endothelialization and atherosclerosis-associated functions. The most pronounced effect was observed for EC migration, with 42/198 genes being involved in the following EC migration-related pathways: 1) MEK/ERK, 2) PI3K/AKT/eNOS-MMP2/9, 3) RHO-GTPases, 4) integrin. rHDL-apoE3 induced changes in 24 representative transcripts of these pathways in HAEC, increasing the expression of their key proteins PIK3CG, EFNB2, ID1 and FLT1 in HCAEC and EA.hy926 cells. In addition, rHDL-apoE3 stimulated migration of HCAEC and EA.hy926 cells, and the migration was markedly attenuated in the presence of PD98059 or LY294002. rHDL-apoE3 also increased the phosphorylation of ERK1/2, AKT, eNOS and p38 MAPK in these cells, while PD98059 and LY294002 inhibited rHDL-apoE3-induced phosphorylation of ERK1/2, AKT and p38 MAPK, respectively. LY had no effect on rHDL-apoE3-mediated eNOS phosphorylation. ID1 siRNA markedly decreased EA.hy926 cell migration by inhibiting rHDL-apoE3-triggered ERK1/2 and AKT phosphorylation. Finally, administration of a single dose of rHDL-apoE3 in apoE KO mice markedly improved vascular permeability as demonstrated by the reduced concentration of Evans Blue dye in tissues such as the stomach, the tongue and the urinary bladder and ameliorated hypercholesterolemia. CONCLUSIONS rHDL-apoE3 significantly enhanced EC migration in vitro, predominantly via overexpression of ID1 and subsequent activation of MEK1/2 and PI3K, and their downstream targets ERK1/2, AKT and p38 MAPK, respectively, and improved vascular permeability in vivo. These novel insights into the rHDL-apoE3 functions suggest a potential clinical use to promote re-endothelialization and retard development of atherosclerosis.
Collapse
Affiliation(s)
- Eftaxia-Konstantina Valanti
- 4th Department of Internal Medicine, Clinical Genomics and Pharmacogenomics Unit, 'Attikon' Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece; Molecular Biology Division, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Katerina Dalakoura-Karagkouni
- Laboratory of Biochemistry, University of Crete Medical School, Heraklion, Greece; Division of Gene Regulation and Genomics, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology of Hellas, Heraklion, Greece
| | | | - Elizabeth Vafiadaki
- Molecular Biology Division, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | | | - Angeliki Chroni
- Institute of Biosciences and Applications, National Center for Scientific Research "Demokritos", Athens, Greece
| | - Vassilis Zannis
- Molecular Genetics, Boston University Medical School, Boston, USA
| | - Dimitris Kardassis
- Laboratory of Biochemistry, University of Crete Medical School, Heraklion, Greece; Division of Gene Regulation and Genomics, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology of Hellas, Heraklion, Greece
| | - Despina Sanoudou
- 4th Department of Internal Medicine, Clinical Genomics and Pharmacogenomics Unit, 'Attikon' Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece; Molecular Biology Division, Biomedical Research Foundation of the Academy of Athens, Athens, Greece; Center for New Biotechnologies and Precision Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
4
|
Qiu J, Li Y, Wang B, Sun X, Qian D, Ying Y, Zhou J. The Role and Research Progress of Inhibitor of Differentiation 1 in Atherosclerosis. DNA Cell Biol 2022; 41:71-79. [PMID: 35049366 PMCID: PMC8863915 DOI: 10.1089/dna.2021.0745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/29/2021] [Accepted: 09/29/2021] [Indexed: 12/23/2022] Open
Abstract
Inhibitor of differentiation 1 has a helix-loop-helix (HLH) structure, belongs to a class of molecules known as the HLH trans-acting factor family, and plays an important role in advancing the cell cycle, promoting cell proliferation and inhibiting cell differentiation. Recent studies have confirmed that inhibitor of differentiation 1 plays an important role in the endothelial-mesenchymal transition of vascular endothelial cells, angiogenesis, reendothelialization after injury, and the formation and rupture of atherosclerotic plaques. An in-depth understanding of the role of inhibitor of differentiation 1 in atherosclerosis will provide new ideas and strategies for the treatment of related diseases.
Collapse
Affiliation(s)
- Jun Qiu
- Department of Cardiology, Medicine School of Ningbo University, Ningbo, China
- Department of Cardiology, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
- Department of Cardiology, Ningbo Institute of Innovation for Combined Medicine and Engineering (NIIME), Ningbo, China
| | - Youhong Li
- Department of Cardiology, Medicine School of Ningbo University, Ningbo, China
| | - BingYu Wang
- Department of Cardiology, Medicine School of Ningbo University, Ningbo, China
- Department of Cardiology, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
- Department of Cardiology, Ningbo Institute of Innovation for Combined Medicine and Engineering (NIIME), Ningbo, China
| | - XinYi Sun
- Department of Cardiology, Medicine School of Ningbo University, Ningbo, China
- Department of Cardiology, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
- Department of Cardiology, Ningbo Institute of Innovation for Combined Medicine and Engineering (NIIME), Ningbo, China
| | - Dingding Qian
- Department of Cardiology, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
| | - Yuchen Ying
- Department of Cardiology, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
| | - Jianqing Zhou
- Department of Cardiology, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
| |
Collapse
|
5
|
Wang X, Huang Z, Zeng L, Jin X, Yan A, Zhang Y, Tan W. The Role of Survivin and Transcription Factor FOXP1 in Scarring After Glaucoma Surgery. Transl Vis Sci Technol 2022; 11:19. [PMID: 35142784 PMCID: PMC8842717 DOI: 10.1167/tvst.11.2.19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Purpose This study aims to elucidate the role and mechanism of survivin and FOXP1 in scarring after glaucoma surgery and to evaluate the prevention and treatment of excessive wound healing and scar formation in an in vitro model of glaucoma filtration surgery. Methods Human Tenon's capsule fibroblasts (HTFs) were used with TGF-β to establish an in vitro cell model after glaucoma, observe survivin expression in the cell model, and observe HTFs proliferation after treatment with survivin inhibitor YM155 and the expression of α-SMA and collagen type I. In addition, the effects of survivin and cell proliferation in HTFs after knockdown of FOXP1 were observed by Western blot analysis. Results Survivin was upregulated in HTFs after glaucoma surgery, and it could promote the cell proliferation of HTFs. After treatment with its inhibitor YM155, the cell proliferation of HTFs was inhibited, and the expression of α-SMA and collagen type I were decreased. The results showed that in knockdown of FOXP1, the expression of survivin was downregulated, and the cell proliferation of HTFs was significantly reduced. Conclusions This study demonstrates that targeting survivin with an inhibitory YM155 reduced fibrosis and the extracellular matrix (ECM), and it was regulated by the FOXP1 transcription factor. These results suggest that survivin could be a potential target for treating scar formation after glaucoma surgery. Translational Relevance Together with the results from previous survivin and FOXP1 preclinical studies, these data support the evaluation of this gene therapy candidate in clinical trials as a potential durable treatment for antiscarring of glaucoma surgery.
Collapse
Affiliation(s)
- Xiaocong Wang
- Soochow University, Suzhou, Jiangsu, China.,Medical College of Soochow University, Suzhou, Jiangsu, China.,Graduate School of Zunyi Medical University, Zunyi, Guizhou,China.,Department of Ophthalmology, The Third Affiliated Hospital of Zunyi, Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| | - Zhihua Huang
- Graduate School of Zunyi Medical University, Zunyi, Guizhou,China.,The Ninth People's Hospital of Chongqing, Chongqing, China
| | - Lan Zeng
- Graduate School of Zunyi Medical University, Zunyi, Guizhou,China.,Department of Ophthalmology, The Third Affiliated Hospital of Zunyi, Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| | - Xin Jin
- Department of Ophthalmology, The Third Affiliated Hospital of Zunyi, Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| | - Ai Yan
- Department of Ophthalmology, The Third Affiliated Hospital of Zunyi, Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| | - Ying Zhang
- Department of Ophthalmology, The Third Affiliated Hospital of Zunyi, Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| | - Wei Tan
- Soochow University, Suzhou, Jiangsu, China.,Medical College of Soochow University, Suzhou, Jiangsu, China.,Graduate School of Zunyi Medical University, Zunyi, Guizhou,China.,Department of Ophthalmology, The Third Affiliated Hospital of Zunyi, Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| |
Collapse
|
6
|
Xia LZ, Tao J, Chen YJ, Liang LL, Luo GF, Cai ZM, Wang Z. Factors Affecting the Re-Endothelialization of Endothelial Progenitor Cell. DNA Cell Biol 2021; 40:1009-1025. [PMID: 34061680 DOI: 10.1089/dna.2021.0082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The vascular endothelium, which plays an essential role in maintaining the normal shape and function of blood vessels, is a natural barrier between the circulating blood and the vascular wall tissue. The endothelial damage can cause vascular lesions, such as atherosclerosis and restenosis. After the vascular intima injury, the body starts the endothelial repair (re-endothelialization) to inhibit the neointimal hyperplasia. Endothelial progenitor cell is the precursor of endothelial cells and plays an important role in the vascular re-endothelialization. However, re-endothelialization is inevitably affected in vivo and in vitro by factors, which can be divided into two types, namely, promotion and inhibition, and act on different links of the vascular re-endothelialization. This article reviews these factors and related mechanisms.
Collapse
Affiliation(s)
- Lin-Zhen Xia
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, China
| | - Jun Tao
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, China
| | - Yan-Jun Chen
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, China
| | - Ling-Li Liang
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, China
| | - Gui-Fang Luo
- Department of Gynaecology, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Ze-Min Cai
- Pediatrics Department, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Zuo Wang
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, China
| |
Collapse
|