201
|
Wang P, Liu X, Han G, Dai S, Ni Q, Xiao S, Huang J. Downregulated lncRNA UCA1 acts as ceRNA to adsorb microRNA-498 to repress proliferation, invasion and epithelial mesenchymal transition of esophageal cancer cells by decreasing ZEB2 expression. Cell Cycle 2019; 18:2359-2376. [PMID: 31387451 PMCID: PMC6738578 DOI: 10.1080/15384101.2019.1648959] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 07/04/2019] [Accepted: 07/07/2019] [Indexed: 12/24/2022] Open
Abstract
Objective: Esophageal cancer (EC) is one of the most general malignant tumors in humans. There were few studies researching the connections between lncRNA UCA1 and EC. This study is to research the effect of lncRNA UCA1 adsorbing microRNA-498 (miR-498) as a ceRNA to regulate ZEB2 expression on epithelial mesenchymal transition (EMT), invasion and migration of EC cells. Methods: UCA1, miR-498 and ZEB2 expression in EC tissues and cells was detected by RT-qPCR or western blot analysis. EC cells were transfected with siRNA-UCA1, miR-498 mimics or their controls to determine cell colony, proliferation, cycle distribution, apoptosis, migration and invasion by colony formation assay, CCK-8 assay, flow cytometry, and Transwell assay, respectively. The protein expression of PCNA, c-Myc, E-cadherin, N-cadherin, MMP-2 and MMP-9 was detected by Western blot analysis. The growth rate and weight of transplanted tumor in nude mice were observed. Results: There were overly expressed UCA1 and ZEB2 and lowly expressed miR-498 in EC tissues and cells. LncRNA UCA1 acted as ceRNA to inhibit miR-498 expression and thereby increasing ZEB2 expression. With down-regulated UCA1 and up-regulated miR-498, ZEB2 expression, cell proliferation, colony formation, invasion, migration ability, EMT, tumor growth rate and weight in nude mice were apparently reduced. Conclusion: This study demonstrates that inhibited UCA1 up-regulated miR-498 and down-regulated ZEB2, thereby repressing proliferation activity, invasion, migration, and EMT of EC cells.
Collapse
Affiliation(s)
- Peng Wang
- Department of Oncology, Taizhou People’s Hospital, Taizhou, PR. China
| | - Xinfa Liu
- Anesthesiology Department, Taizhou People’s Hospital, Taizhou, PR. China
| | - Gaohua Han
- Department of Oncology, Taizhou People’s Hospital, Taizhou, PR. China
| | - Shengbin Dai
- Department of Oncology, Taizhou People’s Hospital, Taizhou, PR. China
| | - Qingtao Ni
- Department of Oncology, Taizhou People’s Hospital, Taizhou, PR. China
| | - Shujun Xiao
- Department of Gerontology, Taizhou People’s Hospital, Taizhou, PR. China
| | - Junxing Huang
- Department of Oncology, Taizhou People’s Hospital, Taizhou, PR. China
| |
Collapse
|
202
|
Deng Y, Chen D, Gao F, Lv H, Zhang G, Sun X, Liu L, Mo D, Ma N, Song L, Huo X, Yan T, Zhang J, Miao Z. Exosomes derived from microRNA-138-5p-overexpressing bone marrow-derived mesenchymal stem cells confer neuroprotection to astrocytes following ischemic stroke via inhibition of LCN2. J Biol Eng 2019; 13:71. [PMID: 31485266 PMCID: PMC6714399 DOI: 10.1186/s13036-019-0193-0] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 06/30/2019] [Indexed: 12/16/2022] Open
Abstract
Background MicroRNAs (miRNAs) are implicated in the progression of ischemic stroke (IS) and bone marrow-derived mesenchymal stem cells (BMSCs)-derived exosomes play a role in IS therapy. Herein we hypothesized that the BMSCs-derived exosomes containing overexpressed miR-138-5p could protect the astrocytes following IS involved with lipocalin 2 (LCN2). Methods The differentially expressed gene related to IS was initially identified by bioinformatics analysis. miR-138-5p was predicted to regulate LCN2. The expression of miR-138-5p and LCN2 was altered in the oxygen-glucose deprivation (OGD)-induced astrocytes. Furthermore, the cell behaviors and inflammatory responses were evaluated both in astrocytes alone and astrocytes co-cultured with exosomes derived from BMSCs overexpressing miR-138-5p to explore the involvement of miR-138-5p and LCN2 in IS. Besides, middle cerebral artery occlusion (MCAO) mouse model was established to explore the effect of BMSCs-derived exosomal miR-138-5p in IS in vivo. Results LCN2 was highly expressed in IS. Besides, LCN2 was a target gene of miR-138-5p. BMSCs-derived exosomes could be endocytosed by astrocytes via co-culture. Overexpression of miR-138-5p promoted the proliferation and inhibited apoptosis of astrocytes injured by OGD, accompanied by the reduced expression of inflammatory factors, which was achieved by down-regulating LCN2. More importantly, BMSCs delivered miR-138-5p to the astrocytes via exosomes and BMSCs-derived exosomal miR-138-5p alleviated neuron injury in IS mice. Conclusion BMSCs-derived exosomal miR-138-5p reduces neurological impairment by promoting proliferation and inhibiting inflammatory responses of astrocytes following IS by targeting LCN2, which may provide a novel target for IS treatment.
Collapse
Affiliation(s)
- Yiming Deng
- 1Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, No. 6, Tiantan Xili, Fengtai District, Beijing, 100050 People's Republic of China.,2China National Clinical Research Center for Neurological Diseases, Beijing, 100070 People's Republic of China.,3Center of Stroke, Beijing Institute for Brain Disorders, Beijing, 100069 China
| | - Duanduan Chen
- 4School of Life Science, Beijing Institute of Technology, Beijing, 100081 China
| | - Feng Gao
- 1Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, No. 6, Tiantan Xili, Fengtai District, Beijing, 100050 People's Republic of China.,2China National Clinical Research Center for Neurological Diseases, Beijing, 100070 People's Republic of China.,3Center of Stroke, Beijing Institute for Brain Disorders, Beijing, 100069 China
| | - Hong Lv
- 5Departments of Clinical Laboratory, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050 People's Republic of China
| | - Guojun Zhang
- 5Departments of Clinical Laboratory, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050 People's Republic of China
| | - Xuan Sun
- 1Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, No. 6, Tiantan Xili, Fengtai District, Beijing, 100050 People's Republic of China.,2China National Clinical Research Center for Neurological Diseases, Beijing, 100070 People's Republic of China.,3Center of Stroke, Beijing Institute for Brain Disorders, Beijing, 100069 China
| | - Lian Liu
- 1Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, No. 6, Tiantan Xili, Fengtai District, Beijing, 100050 People's Republic of China.,2China National Clinical Research Center for Neurological Diseases, Beijing, 100070 People's Republic of China.,3Center of Stroke, Beijing Institute for Brain Disorders, Beijing, 100069 China
| | - Dapeng Mo
- 1Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, No. 6, Tiantan Xili, Fengtai District, Beijing, 100050 People's Republic of China.,2China National Clinical Research Center for Neurological Diseases, Beijing, 100070 People's Republic of China.,3Center of Stroke, Beijing Institute for Brain Disorders, Beijing, 100069 China
| | - Ning Ma
- 1Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, No. 6, Tiantan Xili, Fengtai District, Beijing, 100050 People's Republic of China.,2China National Clinical Research Center for Neurological Diseases, Beijing, 100070 People's Republic of China.,3Center of Stroke, Beijing Institute for Brain Disorders, Beijing, 100069 China
| | - Ligang Song
- 1Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, No. 6, Tiantan Xili, Fengtai District, Beijing, 100050 People's Republic of China.,2China National Clinical Research Center for Neurological Diseases, Beijing, 100070 People's Republic of China.,3Center of Stroke, Beijing Institute for Brain Disorders, Beijing, 100069 China
| | - Xiaochuan Huo
- 1Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, No. 6, Tiantan Xili, Fengtai District, Beijing, 100050 People's Republic of China.,2China National Clinical Research Center for Neurological Diseases, Beijing, 100070 People's Republic of China.,3Center of Stroke, Beijing Institute for Brain Disorders, Beijing, 100069 China
| | - Tianyi Yan
- 4School of Life Science, Beijing Institute of Technology, Beijing, 100081 China
| | - Jingbo Zhang
- 1Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, No. 6, Tiantan Xili, Fengtai District, Beijing, 100050 People's Republic of China.,2China National Clinical Research Center for Neurological Diseases, Beijing, 100070 People's Republic of China.,3Center of Stroke, Beijing Institute for Brain Disorders, Beijing, 100069 China
| | - Zhongrong Miao
- 1Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, No. 6, Tiantan Xili, Fengtai District, Beijing, 100050 People's Republic of China.,2China National Clinical Research Center for Neurological Diseases, Beijing, 100070 People's Republic of China.,3Center of Stroke, Beijing Institute for Brain Disorders, Beijing, 100069 China
| |
Collapse
|
203
|
Zhang YP, Liu KL, Yang Z, Lu BS, Qi JC, Han ZW, Yin YW, Zhang M, Chen DM, Wang XW, Li W, Xin H. The involvement of FBP1 in prostate cancer cell epithelial mesenchymal transition, invasion and metastasis by regulating the MAPK signaling pathway. Cell Cycle 2019; 18:2432-2446. [PMID: 31448674 DOI: 10.1080/15384101.2019.1648956] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Prostate cancer (PCa) is a frequently occurring malignancy in males, and epithelial mesenchymal transition (EMT) plays a critical role in PCa metastasis. Thus, developing biomarkers inhibiting EMT may provide significance for treatment of PCa. Hence, the aim of the current study was to investigate the mechanism by which FBP1 gene silencing influences PCa cell EMT, invasion and metastasis by mediating the MAPK pathway. PCa cell lines exhibiting the highest FBP1 expression were selected and treated with plasmids of siRNA-FBP1 sequence 1 and 2, pcDNA3.1-Flag-FBP1 (over-expression plasmid of FBP1), U0126 (an inhibitor of the ERK signaling pathway) and PD98059 (an inhibitor of the MEK signaling pathway). Cell proliferation, migration and invasion were detected by MTT assay, wound healing assay and Transwell assay, respectively. The mRNA and protein expression of related factors of EMT and MAPK signaling were determined by RT-qPCR and western blot analysis, respectively. Xenograft tumor growth after inoculation of DU145 cells was regularly analyzed in the nude mice. The positive expression of EMT markers was determined by immunohistochemistry. DU-145 and PC-3 cells displaying the highest FBP1 expression were selected for further analysis. The PCa cells treated with siRNA-FBP1 exhibited increased proliferation, migration rate and invasion, in addition to facilitated xenograft tumor growth. Notably, siRNA-FBP1 was identified to accelerate PCa cell EMT by elevating the expression of Vimentin and N-cadherin while diminishing E-cadherin expression via activation of the MAPK signaling pathway. The aforementioned results were reversed in PCa cells treated by pcDNA3.1-Flag-FBP1. Evidence has been provided in this study that FBP1 gene silencing activates the MAPK pathway, which ultimately promotes cell EMT, invasion and metastasis in PCa.
Collapse
Affiliation(s)
- Yan-Ping Zhang
- Department of Urology, The Second Hospital of Hebei Medical University , Shijiazhuang , P.R. China
| | - Kai-Long Liu
- Department of Urology, The Second Hospital of Hebei Medical University , Shijiazhuang , P.R. China
| | - Zhan Yang
- Department of Urology, The Second Hospital of Hebei Medical University , Shijiazhuang , P.R. China
| | - Bao-Sai Lu
- Department of Urology, The Second Hospital of Hebei Medical University , Shijiazhuang , P.R. China
| | - Jin-Chun Qi
- Department of Urology, The Second Hospital of Hebei Medical University , Shijiazhuang , P.R. China
| | - Zhen-Wei Han
- Department of Urology, The Second Hospital of Hebei Medical University , Shijiazhuang , P.R. China
| | - Yue-Wei Yin
- Department of Urology, The Second Hospital of Hebei Medical University , Shijiazhuang , P.R. China
| | - Ming Zhang
- Department of Urology, The Second Hospital of Hebei Medical University , Shijiazhuang , P.R. China
| | - De-Min Chen
- Department of Urology, The Second Hospital of Hebei Medical University , Shijiazhuang , P.R. China
| | - Xiao-Wei Wang
- Department of Urology, The Second Hospital of Hebei Medical University , Shijiazhuang , P.R. China
| | - Wei Li
- Department of Urology, The Second Hospital of Hebei Medical University , Shijiazhuang , P.R. China
| | - Hong Xin
- Department of Obstetrics, The Second Hospital of Hebei Medical University , Shijiazhuang , P.R. China
| |
Collapse
|
204
|
Zhang P, Chen F, Jia Q, Hu D. Upregulation of microRNA‐133a and downregulation of connective tissue growth factor suppress cell proliferation, migration, and invasion in human glioma through the JAK/STAT signaling pathway. IUBMB Life 2019; 71:1857-1875. [PMID: 31381269 DOI: 10.1002/iub.2126] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 06/29/2019] [Indexed: 12/31/2022]
Affiliation(s)
- Peng Zhang
- Department of NeurosurgeryHenan Provincial People's Hospital Zhengzhou Henan People's Republic of China
- People's Hospital of Zhengzhou University
- Medical College of Henan University
| | - Fang‐Zhou Chen
- Department of NeurosurgeryThe Second Affiliated Hospital of Shandong First Medical University Taian Shandong People's Republic of China
| | - Qing‐Bin Jia
- Department of NeurosurgeryLiaocheng People's Hospital Liaocheng Shandong People's Republic of China
| | - Dian‐Feng Hu
- Department of NeurosurgeryLiaocheng People's Hospital Liaocheng Shandong People's Republic of China
| |
Collapse
|
205
|
Xue X, Luo L. LncRNA HIF1A-AS1 contributes to ventricular remodeling after myocardial ischemia/reperfusion injury by adsorption of microRNA-204 to regulating SOCS2 expression. Cell Cycle 2019; 18:2465-2480. [PMID: 31354024 DOI: 10.1080/15384101.2019.1648960] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Objectives: Long non-coding RNAs (lncRNAs) serve pivotal roles in heart disease, while the role of lncRNA hypoxia-inducible factor 1α-antisense RNA 1 (HIF1A-AS1) is rarely mentioned. Therefore, the objective of this study was to investigate the mechanism of lncRNA HIF1A-AS1 regulating suppressor of cytokine signaling 2 (SOCS2) expression by adsorption of microRNA-204 (miR-204) on ventricular remodeling after myocardial ischemia-reperfusion (I/R) injury in mice. Methods: The mouse model of I/R was established by left coronary artery occlusion. The expression of HIF1A-AS1, miR-204 and SOCS2 was determined. The mice were injected with HIF1A-AS1-siRNA, miR-204 mimics or their controls to investigate their effects on cardiac function and ventricular remodeling of mice after I/R injury. The binding relationship between HIF1A-AS1 and miR-204 as well as between miR-204 and SOCS2 were verified. Results: HIF1A-AS1 and SOCS2 were upregulated and miR-204 was downregulated in myocardial tissues in mice after I/R injury. LVEDD, LVEDS, LVEDP, LVMI and RVMI expression reduced while LVEF, LVFS, +dp/dt max and - dp/dt max increased through knockdown HIF1A-AS1 and upregulated miR-204. The expression of BNP, cTnI, LDH, CK, TNF-α, IL-1β, IL-6 and β-MHC reduced, and the expression of α-MHC increased when HIF1A-AS1 was poorly expressed and miR-204 was highly expressed. Silencing HIF1A-AS1 and upregulating miR-204 inhibited apoptosis of cells. LncRNA HIF1A-AS1 could act as ceRNA to adsorb miR-204 to suppress miR-204 expression and elevate SOCS2 expression. Conclusion: Our study provides evidence that downregulation of HIF1A-AS1 and upregulation of miR-204 could alleviate ventricular remodeling and improve cardiac function in mice after myocardial I/R injury via regulating SOCS2.
Collapse
Affiliation(s)
- Xiang Xue
- Cardiovascular Medicine Department, Changzhou No.7 People' s Hospital , Changzhou , Jiangsu , China
| | - Libo Luo
- Cardiovascular Medicine Department, Changzhou No.7 People' s Hospital , Changzhou , Jiangsu , China
| |
Collapse
|
206
|
Li WQ, Zhang JP, Wang YY, Li XZ, Sun L. MicroRNA-422a functions as a tumor suppressor in non-small cell lung cancer through SULF2-mediated TGF-β/SMAD signaling pathway. Cell Cycle 2019; 18:1727-1744. [PMID: 31204561 PMCID: PMC6649599 DOI: 10.1080/15384101.2019.1632135] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
MicroRNAs (miRNAs) have been demonstrated to participate in a variety of human cancers by functioning as post-transcriptional regulators of oncogenes or antioncogenes including non-small cell lung cancer (NSCLC). The aim of the current study was to identify the role of miR-422a in NSCLC via sulfatase 2 (SULF2) to further elucidate the mechanism of NSCLC. Initially, the expression of miR-422a and SULF2 was determined in NSCLC tissues and cells. The role of miR-422a in NSCLC was identified in relation with a miR-422a mimic or inhibitor, siRNA against SULF2 and TGF-β1. The regulatory effects of miR-422a were examined following detection of the related epithelial mesenchymal transition (EMT)-related genes, and the apoptosis-related genes and evaluation of their cellular biological functions. The expression pattern of miR-422a, SULF2, and the TGF-β/SMAD pathway-related genes was detected to elucidate the mechanism by which miR-422a influences the progression of NSCLC. Finally, xenograft tumors in nude mice were observed for tumorigenicity evaluation purposes. Our results showed that miR-422a was poorly expressed while SULF2 was highly expressed in NSCLC. Dual luciferase reporter gene assay further verified that miR-422a targeted SULF2. Altogether, this study demonstrated that miR-422a downregulated SULF2 to inhibit the TGF-β/SMAD pathway. NSCLC cell proliferation, migration, invasion, colony formation, EMT and tumorigenesis were all inhibited while apoptosis was promoted upon restoration of miR-422a or silencing of SULF2. However, the activation of the TGF-β/SMAD pathway was determined to reverse the tumor-suppressive effects of si-SULF2. miR-422a restoration, which ultimately inhibited the progression of NSCLC by suppressing the TGF-β/SMAD pathway via SULF2.
Collapse
Affiliation(s)
- Wei-Qiang Li
- a Department of Thoracic Surgery , Beijing Luhe Hospital, Capital Medical University , Beijing , P. R. China
| | - Jian-Peng Zhang
- a Department of Thoracic Surgery , Beijing Luhe Hospital, Capital Medical University , Beijing , P. R. China
| | - Yan-Yu Wang
- a Department of Thoracic Surgery , Beijing Luhe Hospital, Capital Medical University , Beijing , P. R. China
| | - Xin-Zhen Li
- a Department of Thoracic Surgery , Beijing Luhe Hospital, Capital Medical University , Beijing , P. R. China
| | - Lin Sun
- a Department of Thoracic Surgery , Beijing Luhe Hospital, Capital Medical University , Beijing , P. R. China
| |
Collapse
|
207
|
Liu DC, Song LL, Liang Q, Hao L, Zhang ZG, Han CH. Long noncoding RNA LEF1-AS1 silencing suppresses the initiation and development of prostate cancer by acting as a molecular sponge of miR-330-5p via LEF1 repression. J Cell Physiol 2019; 234:12727-12744. [PMID: 30613973 DOI: 10.1002/jcp.27893] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 11/14/2018] [Indexed: 02/06/2023]
Abstract
Prostate cancer (PCa) is one of the major cancers affecting males with high mortality around the world. Recent studies have found that some long noncoding RNAs play a critical part in the cellular processes of PCa. In our study, aberrant expressed lymphoid enhancer-binding factor-1 antisense RNA 1 (LEF1-AS1), microRNA-330-5p (miR-330-5p), and lymphoid enhancer-binding factor-1 (LEF1) were screened out from a microarray database, the role of the novel noncoding RNA regulatory circuitry in the initiation and development of PCa was investigated. LEF1-AS1 and LEF1 were highly expressed while miR-330-5p was poorly expressed in PCa. Following that, the PCa PC-3 cell line was adopted for subsequently experiments, in which the expression of LEF1-AS1 and miR-330-5p was subsequently altered by means of exogenous transfection. After that, the effects of up- or downregulation of LEF1-AS1 and miR-330-5p on epithelial-mesenchymal transition (EMT) and the cell ability for proliferation, invasion, migration in vitro, and tumorigenesis and lymph node metastasis (LNM) in vivo were evaluated. RNA crosstalk revealed that LEF1-AS1 bound to miR-330-5p and LEF1 was the target gene of miR-330-5p. Silenced LEF1-AS1 or elevated miR-330-5p exhibited inhibited EMT processes, reduced ability of proliferation, invasion and migration, coupling with decreased tumorigenesis and LNM in nude mice. The key findings of this study collectively propose downregulation of LEF1-AS1 competing with miR-330-5p to inhibit EMT, invasion and migration of PCa by LEF1 repression.
Collapse
Affiliation(s)
- Da-Chuang Liu
- Department of Urology, Xuzhou Central Hospital, Xuzhou, P. R. China
- Xuzhou Institute of Medical Science, Xuzhou, P. R. China
| | - Lin-Lin Song
- Department of Respiratory, Xuzhou Central Hospital, Xuzhou, P. R. China
| | - Qing Liang
- Department of Urology, Xuzhou Central Hospital, Xuzhou, P. R. China
| | - Lin Hao
- Department of Urology, Xuzhou Central Hospital, Xuzhou, P. R. China
| | - Zhi-Guo Zhang
- Department of Urology, Xuzhou Central Hospital, Xuzhou, P. R. China
| | - Cong-Hui Han
- Department of Urology, Xuzhou Central Hospital, Xuzhou, P. R. China
- Xuzhou Institute of Medical Science, Xuzhou, P. R. China
| |
Collapse
|
208
|
Huang X, Qiao F, Xue P. The protective role of microRNA-140-5p in synovial injury of rats with knee osteoarthritis via inactivating the TLR4/Myd88/NF-κB signaling pathway. Cell Cycle 2019; 18:2344-2358. [PMID: 31345099 PMCID: PMC6738526 DOI: 10.1080/15384101.2019.1647025] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Objective: Recently, many studies have revealed the effect of microRNAs (miRNAs) in knee osteoarthritis (KOA). This study aims to explore the role of miR-140-5p in protective effects and mechanisms of synovial injury of rats with KOA via regulating the TLR4/Myd88/NF-κB signaling pathway. Methods: The models of KOA Wistar rats were established by operation of anterior cruciate ligament transection. Rats were injected with agomir NC or miR-140-5p agomir. MiR-140-5p expression in KOA synovial tissues and synoviocytes was evaluated by reverse transcription quantitative polymerase chain reaction (RT-qPCR). The synoviocytes were transfected with mimics NC sequence and miR-140-5p mimics sequence. The expression of TLR4/Myd88/NF-κB signaling pathway-related proteins was measured by RT-qPCR and western blot analysis. The proliferation and apoptosis of synoviocytes in rats with KOA were evaluated by a string of experiments. The expression levels of inflammatory factors in KOA synovial tissues and synoviocytes were detected. Results: MiR-140-5p was down-regulated in KOA synovial tissues and synoviocytes. Upregulation of miR-140-5p could inhibit the inflammation reaction and the apoptosis of synoviocytes as well as promote proliferation of synoviocytes of rats with KOA. Furthermore, upregulated miR-140-5p could inactivate the TLR4/Myd88/NF-κB signaling pathway in rats with KOA. Conclusion: This study suggests that upregulated miR-140-5p could protect synovial injury by restraining inflammation reaction and apoptosis of synoviocytes in KOA rats via TLR4/Myd88/NF-κB signaling pathway inactivation.
Collapse
Affiliation(s)
- Xiaoqiang Huang
- Orthopaedics Department, Honghui Hospital, Xi'an Jiaotong University , Xi'an , PR China
| | - Feng Qiao
- Orthopaedics Department of Integrated Traditional Chinese and Western Medicine, Honghui Hospital, Xi'an Jiaotong University , Xi'an , PR China
| | - Peng Xue
- Orthopaedics Department of Integrated Traditional Chinese and Western Medicine, Honghui Hospital, Xi'an Jiaotong University , Xi'an , PR China
| |
Collapse
|
209
|
Yin Z, Ma T, Huang B, Lin L, Zhou Y, Yan J, Zou Y, Chen S. Macrophage-derived exosomal microRNA-501-3p promotes progression of pancreatic ductal adenocarcinoma through the TGFBR3-mediated TGF-β signaling pathway. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:310. [PMID: 31307515 PMCID: PMC6631643 DOI: 10.1186/s13046-019-1313-x] [Citation(s) in RCA: 244] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 07/04/2019] [Indexed: 12/13/2022]
Abstract
Background Exosomes from cancer cells or immune cells, carrying bio-macromolecules or microRNAs (miRNAs), participate in tumor pathogenesis and progression by modulating microenvironment. Our study aims to investigate the role of these microRNA-501-3p (miR-501-3p) containing exosomes derived from tumor-associated macrophage (TAM) in the progression of pancreatic ductal adenocarcinoma (PDAC). Methods Firstly, the function of TAM recruitment in PDAC tissues was assessed, followed by identification of the effects of M2 macrophage-derived exosomes on PDAC cell activities and tumor formation and metastasis in mice. In silico analysis was conducted to predict differentially expressed genes and regulatory miRNAs related to PDAC treated with macrophages, which determined miR-501-3p and TGFBR3 for subsequent experiments. Next, gain- and loss-of-function experiments were performed to examine their role in PDAC progression with the involvement of the TGF-β signaling pathway. Results TAM recruitment in PDAC tissues was associated with metastasis. Highly expressed miR-501-3p was observed in PDAC tissues and TAM-derived exosomes. Both M2 macrophage-derived exosomes and miR-501-3p promoted PDAC cell migration and invasion, as well as tumor formation and metastasis in nude mice. MiR-501-3p was verified to target TGFBR3. PDAC cells presented with down-regulated TGFBR3, which was further decreased in response to M2 macrophage treatment. TGF-β signaling pathway activation was implicated in the promotion of miR-501-3p in PDAC development. The suppression of macrophage-derived exosomal miR-501-3p resulted in the inhibition of tumor formation and metastasis in vivo. Conclusion M2 macrophage-derived exosomal miR-501-3p inhibits tumor suppressor TGFBR3 gene and facilitates the development of PDAC by activating the TGF-β signaling pathway, which provides novel targets for the molecular treatment of PDAC. Electronic supplementary material The online version of this article (10.1186/s13046-019-1313-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zi Yin
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, No. 106, Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, People's Republic of China.
| | - Tingting Ma
- Department of Obstetrics and Gynecology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Bowen Huang
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, No. 106, Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, People's Republic of China
| | - Lehang Lin
- Medical Research Center, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Yu Zhou
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, No. 106, Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, People's Republic of China
| | - Jinhai Yan
- Pathology Department, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, People's Republic of China
| | - Yiping Zou
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, No. 106, Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, People's Republic of China
| | - Sheng Chen
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, No. 106, Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, People's Republic of China.
| |
Collapse
|
210
|
Shu C, Shu Y, Gao Y, Chi H, Han J. Inhibitory effect of AQP1 silencing on adhesion and angiogenesis in ectopic endometrial cells of mice with endometriosis through activating the Wnt signaling pathway. Cell Cycle 2019; 18:2026-2039. [PMID: 31251110 DOI: 10.1080/15384101.2019.1637202] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The development mechanism of endometriosis remains unknown. Water channel aquaporin-1 (AQP1) enhances water flux across cell membranes, which is highly expressed and associated with cell migration, metastasis and angiogenesis in some human cancers. In this study, the role of the Wnt signaling pathway mediated by AQP1 in endometriosis was investigated, in a bid to provide new therapeutic targets for endometriosis. Microarray expression profiles were screened to acquired differentially expressed genes related to endometriosis. Mouse models with endometriosis were established and grouped. The level of endometriosis was evaluated by measurement of the volume of ectopic region. The expression of AQP1, pathway-related factors (Wnt1 and Wnt4), adhesion molecules (VCAM-1 and ICAM-1), invasive factors (MMP-2, MMP-9, TIMP-1 and TIMP-2), angiogenic factors (VEGF-A, VEGFR1 and VEGFR2) and apoptotic factors (Caspase-3, Caspase-9, Bax and BcL-2) was measured by RT-qPCR and western blot analysis. Furthermore, the role of AQP1 in adhesion, invasion, angiogenesis, and apoptosis of ectopic endometrial cells was determined by transfection of si-AQP1 plasmid. AQP1 was robustly expressed in endometriosis. AQP1 gene silencing alleviated the progression of endometriosis by activating the Wnt signaling pathway in mice with endometriosis. Specifically, silencing of AQP1 gene inhibited ectopic endometrial cell adhesion and invasion abilities, suppressed angiogenesis while promoted apoptosis. Collectively, the present study highlights the role of AQP1 in the regulation of the Wnt signaling pathway in endometriosis mouse models, suggesting that AQP1 could represent a new target aimed at improving the survival of patients with endometriosis.
Collapse
Affiliation(s)
- Chang Shu
- a Department of Obstetrics and Gynecology, The First Hospital of Jilin University , Changchun , P.R. China
| | - Yang Shu
- a Department of Obstetrics and Gynecology, The First Hospital of Jilin University , Changchun , P.R. China
| | - Yongmei Gao
- a Department of Obstetrics and Gynecology, The First Hospital of Jilin University , Changchun , P.R. China
| | - Hui Chi
- a Department of Obstetrics and Gynecology, The First Hospital of Jilin University , Changchun , P.R. China
| | - Jun Han
- b Department of Neonatology, The First Hospital of Jilin University , Changchun , P.R. China
| |
Collapse
|
211
|
Li J, Li L, Yuan H, Huang XW, Xiang T, Dai S. Up-regulated lncRNA GAS5 promotes chemosensitivity and apoptosis of triple-negative breast cancer cells. Cell Cycle 2019; 18:1965-1975. [PMID: 31282278 DOI: 10.1080/15384101.2019.1635870] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Up to accomplishment of this study, the role of long non-coding RNAs (lncRNAs) in breast cancer has been investigated in several researches. Nevertheless, its association with the chemosensitivity of cancer was little known. Therefore, this study is focused on lncRNA GAS5 and its influence in the chemosensitivity of triple-negative breast cancer (TNBC). Expression of GAS5 in TNBC tissues and cells was detected by reverse transcription quantitative polymerase chain reaction (RT-qPCR) and its methylation was evaluated using methylation-specific polymerase chain reaction (MSP). Moreover, in order to define the contributory role of GAS5 in TNBC, GAS5 expression, proliferation, and apoptosis of TNBC cells were detected by a series of experiment. Finally, the effects of GAS5 in vivo were investigated by measuring tumor formation in nude mice. GAS5 was poorly expressed in TNBC tissues and cells, which could regulate the progression of TNBC. The methylation of CpG island in the promoter region of GAS5 in MDA-MB-231 and MDA-MB-468 cells was decreased, while GAS5 expression in cells was increased. Overexpressed GAS5 reduced the inhibitory concentration (IC50) value and the cell proliferation of TNBC, and promoted their apoptosis, so as to delay the progression of TNBC. Our study provides evidence that up-regulated GAS5 suppressed the progression of TNBC and promoted chemosensitivity and apoptosis of TNBC cells. Thus, GAS5 may be a potential candidate for the treatment of TNBC.
Collapse
Affiliation(s)
- Juntao Li
- a Departmnet of Breast and Thyroid Surgery, People's Hospital of Ganzhou City , Ganzhou , Jiangxi Province , PR. China
| | - Lin Li
- b Departmnet of Pharmacy Intravenous Admixture Services, People's Hospital of Ganzhou City , Ganzhou , Jiangxi Province , PR. China
| | - Huozhong Yuan
- a Departmnet of Breast and Thyroid Surgery, People's Hospital of Ganzhou City , Ganzhou , Jiangxi Province , PR. China
| | - Xing-Wei Huang
- a Departmnet of Breast and Thyroid Surgery, People's Hospital of Ganzhou City , Ganzhou , Jiangxi Province , PR. China
| | - Tianxin Xiang
- c Department of Infectious Diseases, The First Affiliated Hospital of Nanchang Universicty , Nanchang , Jiangxi Province , PR. China
| | - Sujuan Dai
- d Departmnet of Pathology, People's Hospital of Ganzhou City , Ganzhou , Jiangxi Province , PR. China
| |
Collapse
|
212
|
Zhu J, Lin X, Yan C, Yang S, Zhu Z. microRNA-98 protects sepsis mice from cardiac dysfunction, liver and lung injury by negatively regulating HMGA2 through inhibiting NF-κB signaling pathway. Cell Cycle 2019; 18:1948-1964. [PMID: 31234706 DOI: 10.1080/15384101.2019.1635869] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Recently, MicroRNA-98 (miR-98) works as a biomarker in some diseases, such as lung cancer, Schizophrenia, and breast cancer, but there still lack of studies on the function of miR-98 during sepsis. Thus, our study is conducted to figure out the function of miR-98 for the regulation of cardiac dysfunction, liver and lung injury in sepsis mice. Cecum ligation and puncture was used to establish the sepsis mice model. Next, miR-Con and agomiR-98 were injected into the tail vein of mice 48 h after modeling. Then, expression of miR-98, HMGA2, NF-κB, inflammatory factors, apoptosis-related proteins in myocardial, liver and lung tissues of septic mice were determined. Moreover, other indices that were associated with cardiac dysfunction, liver and lung injury in septic mice were detected. Finally, bioinformatics analysis and luciferase activity assay were utilized to validate the binding site between miR-98 and HMGA2. miR-98 was poorly expressed, while HMGA2, NF-κB pathway-related proteins were highly expressed in myocardial, liver, and lung tissues of mice with sepsis. Upregulated miR-98 inhibited HMGA2, NF-κB, TNF-α, IL-6, Bcl-2 and increased IL-10, Cleaved caspase-3 and Bax expression in myocardial, liver, and lung tissues of septic mice. Upregulation of miR-98 decreased LVEDP, CTn-I, BNP, ALT, AST, TBIL, LDH, and PaCO2 while increased +dp/dt max, -dp/dt max, pH and PaO2 in sepsis mice. miR-98 was a direct target gene of HMGA2. Our study provides evidence that miR-98 protects sepsis mice from cardiac dysfunction, liver and lung injury by negatively mediating HMGA2 via the inhibition of the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Jingfa Zhu
- a Department of Emergency, Quanzhou First Hospital Affiliated to Fujian Medical University , Quanzhou , China
| | - Xingyu Lin
- b Department of Emergency, Fujian Medical University Union Hospital , Fuzhou , China
| | - Cairong Yan
- a Department of Emergency, Quanzhou First Hospital Affiliated to Fujian Medical University , Quanzhou , China
| | - Shaodong Yang
- a Department of Emergency, Quanzhou First Hospital Affiliated to Fujian Medical University , Quanzhou , China
| | - Zhixia Zhu
- a Department of Emergency, Quanzhou First Hospital Affiliated to Fujian Medical University , Quanzhou , China
| |
Collapse
|
213
|
Shi G, Zhang H, Yu Q, Hu C, Ji Y. GATA1 gene silencing inhibits invasion, proliferation and migration of cholangiocarcinoma stem cells via disrupting the PI3K/AKT pathway. Onco Targets Ther 2019; 12:5335-5354. [PMID: 31456644 PMCID: PMC6620705 DOI: 10.2147/ott.s198750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 05/12/2019] [Indexed: 12/14/2022] Open
Abstract
Background/aims: Intrahepatic cholangiocarcinoma (CCA) is the second most prevalent type primary liver malignancy, accompanied by an increasing global incidence and mortality rate. Research has documented the contribution of the GATA binding protein-1 (GATA1) in the progression of liver cancer. Here, we aim to investigate the role of GATA1 in CCA stem cells via the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) pathway. Methods: Initially, microarray-based gene expression profiling was employed to identify the differentially expressed genes associated with CCA. Subsequently, an investigation was conducted to explore the potential biological significance behind the silencing of GATA1 and the regulatory mechanism between GATA1 and PI3K/AKT pathway. CCA cell lines QBC-939 and RBE were selected and treated with siRNA against GATA1 or/and a PI3K/AKT pathway inhibitor LY294002. In vivo experiment was also conducted to confirm in vitro findings. Results: GATA1 exhibited higher expression in CCA samples and was predicted to affect the progression of CCA through blockade of the PI3K/AKT pathway. siRNA-mediated downregulation of GATA1 and LY294002 treatment resulted in reduced proliferation, migration and invasion abilities of CCA stem cells, together with impeded tumor growth, and led to increased cell apoptosis and primary cilium expression. Additionally, the siRNA-mediated GATA1 downregulation had an inhibitory effect on the PI3K/AKT pathway. LY294002 was manifested to enhance the inhibitory effects of GATA1 inhibition on CCA progression. These in vitro findings were reproduced in vivo on siRNA against GATA1 or LY294002 injected nude mice. Conclusion: Altogether, the present study highlighted that downregulation of GATA1 via blockade of the PI3K/AKT pathway could inhibit the CCA stem cell proliferation, migration and invasion, and tumor growth, and promote cell apoptosis, primary cilium expression.
Collapse
Affiliation(s)
- Guang Shi
- Department of Hematology and Oncology, the Second Hospital of Jilin University, Changchun 130041, People's Republic of China
| | - Hong Zhang
- Department of Clinical Medicine, Changchun Medical College, Changchun 130031, People's Republic of China
| | - Qiong Yu
- Department of Hematology and Oncology, the Second Hospital of Jilin University, Changchun 130041, People's Republic of China
| | - Chunmei Hu
- Department of Hematology and Oncology, the Second Hospital of Jilin University, Changchun 130041, People's Republic of China
| | - Youbo Ji
- Department of Pain, the Second Hospital of Jilin University, Changchun 130041, People's Republic of China
| |
Collapse
|
214
|
Zeng Z, Gao ZL, Zhang ZP, Jiang HB, Yang CQ, Yang J, Xia XB. Downregulation of CKS1B restrains the proliferation, migration, invasion and angiogenesis of retinoblastoma cells through the MEK/ERK signaling pathway. Int J Mol Med 2019; 44:103-114. [PMID: 31115482 PMCID: PMC6559318 DOI: 10.3892/ijmm.2019.4183] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 04/23/2019] [Indexed: 02/06/2023] Open
Abstract
Retinoblastoma (RB) is a common neoplasm that is exhibited in individuals globally. Increasing evidence demonstrated that cyclin‑dependent kinase regulatory subunit 1B (CKS1B) may be involved in the pathogenesis of various tumor types, including multiple myeloma and breast cancer. In the present study, the hypothesis that CKS1B downregulation would effectively inhibit the proliferation, invasion and angiogenesis of RB cells through the mitogen‑activated protein kinase kinase (MEK)/extracellular signal‑regulated kinase (ERK) signaling pathway was examined. Initial investigation of the expression profile of CKS1B in RB and adjacent retina tissues was performed using reverse transcription‑quantitative polymerase chain reaction and western blot analysis. A total of three RB cell lines, SO‑RB50, Y79 and HXO‑RB44, were examined for selection of the cell line with the highest expression of CKS1B, and human normal retinal vascular endothelial cells (ACBRI‑181) were also evaluated. CKS1B short hairpin RNA (shRNA) sequences (shRNA CKS1B‑1, shRNA CKS1B‑2 and shRNA CKS1B‑3) and negative control shRNA sequences were constructed and transfected into cells at the third generation to evaluate the role of shCKS1B and the MEK/ERK signaling pathway in RB. Furthermore, the effect of shCKS1B on cell proliferation, migration, invasion, apoptosis and angiogenesis was investigated. CKS1B was determined to be highly expressed in RB tissue, compared with adjacent retina tissue. SO‑RB50 and HXO‑RB44 cells treated with shRNA CKS1B‑1 and shRNA CKS1B‑2 were selected for the present experiments. Activation of the MEK/ERK signaling pathway increases the expression of MEK, ERK, B‑cell lymphoma 2, proliferating cell nuclear antigen, cyclin D1, vascular endothelia growth factor and basic fibroblast growth factor, enhances cell proliferation, migration, invasion and lumen formation, and decreases apoptosis. Following silencing CKS1B, the aforementioned conditions were reversed. The key observations of the present study demonstrated that shCKS1B can inhibit the proliferation, invasion and angiogenesis of RB cells by suppressing the MEK/ERK signaling pathway. Thus, CKS1B represents a potential research target in the development of therapeutics for RB.
Collapse
Affiliation(s)
- Zhou Zeng
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, Hunan 410008
| | - Zhao-Lin Gao
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, Hunan 410008
| | - Zhi-Pei Zhang
- Department of Ophthalmology, The First People's Hospital of Changde City, Changde, Hunan 415000, P.R. China
| | - Hai-Bo Jiang
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, Hunan 410008
| | - Chang-Quan Yang
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, Hunan 410008
| | - Jie Yang
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, Hunan 410008
| | - Xiao-Bo Xia
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, Hunan 410008
| |
Collapse
|
215
|
Shang J, Gao ZY, Zhang LY, Wang CY. Over-expression of JAZF1 promotes cardiac microvascular endothelial cell proliferation and angiogenesis via activation of the Akt signaling pathway in rats with myocardial ischemia-reperfusion. Cell Cycle 2019; 18:1619-1634. [PMID: 31177938 PMCID: PMC6619954 DOI: 10.1080/15384101.2019.1629774] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Myocardial ischemia-reperfusion (I/R) injury is caused by endothelial dysfunction and enhanced oxidative stress. The overexpression of JAZF1, a zinc finger protein, has been reported to promote cell proliferation and suppress myogenic differentiation in type 2 diabetes. However, the involvement of JAZF1 in myocardial I/R injury remains to be unclear. The current study aims to investigate the role by which JAZF1 influences cardiac microvascular endothelial cells (CMECs) in a rat model of myocardial I/R injury. A total of 50 rats were established as a myocardial I/R model to isolate CMECs, with alterations in JAZF1 expression. After that, the gain- or loss-function of JAZF1 on the proliferation, apoptosis and tube formation ability of CMECs were evaluated by a series of in vitro experiments. Results indicated that JAZF1 was down-regulated in CMECs of rats with myocardial I/R injury. After treatment with JAZF1, the levels of VEGF, Bcl-2, PDGF and p-Akt/Akt were all increased; however, the expression of Bax, caspase-3, caspase-9, p-Bad/Bad, c-caspase-3/caspase-3, c-caspase-9/caspase-9, and p-FKHR/FKHR exhibited decreased levels; CMEC proliferation and angiogenesis were increased, while cell apoptosis was attenuated. CMECs transfected with JAZF1 shRNA exhibited the contrary tendencies. The key findings of this study suggest that the over-expression of JAZF1 alleviates myocardial I/R injury by enhancing proliferation and angiogenesis of CMECs and in turn inhibiting apoptosis of CMECs via the activation of the Akt signaling pathway.
Collapse
Affiliation(s)
- Jie Shang
- a Department of Electrocardiogram , Yantai Yuhuangding Hospital , Yantai , P. R. China
| | - Zhi-Yong Gao
- b Department of Rehabilitation , Yantai Yuhuangding Hospital , Yantai , P. R. China
| | - Li-Yan Zhang
- c Department of Cardiovascular Medicine , Longkou Nanshan Health Valley Tumor Hospital , Longkou , P.R. China
| | - Chun-Yu Wang
- a Department of Electrocardiogram , Yantai Yuhuangding Hospital , Yantai , P. R. China
| |
Collapse
|
216
|
Qi X, Yu XJ, Wang XM, Song TN, Zhang J, Guo XZ, Li GJ, Shao M. Knockdown of KCNQ1OT1 Suppresses Cell Invasion and Sensitizes Osteosarcoma Cells to CDDP by Upregulating DNMT1-Mediated Kcnq1 Expression. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 17:804-818. [PMID: 31454677 PMCID: PMC6716066 DOI: 10.1016/j.omtn.2019.06.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 06/13/2019] [Accepted: 06/14/2019] [Indexed: 12/17/2022]
Abstract
Osteosarcoma is a malignant bone tumor, with a high incidence worldwide. The involvement of long non-coding RNAs (lncRNAs) in cancers and their molecular association with the progression of osteosarcoma have been previously discussed. We conducted the present study to examine the effect of lncRNA KCNQ1 opposite strand/antisense transcript 1 (KCNQ1OT1) on osteosarcoma cell invasion and chemosensitivity to cisplatin (CDDP). After determination of the expression of Kcnq1 in osteosarcoma tissues and cells, the plasmids with overexpression or knockdown KCNQ1OT1 were introduced into the cells to aid the identification of cell proliferation, migration, invasion, chemosensitivity to CDDP, and apoptosis. Then, the interaction between KCNQ1OT1 and the Kcnq1/DNA methyltransferase 1 (DNMT1) axis was evaluated by measuring the level of Kcnq1 promoter region methylation and DNMT1 enrichment of the Kcnq1 promoter region. Low Kcnq1 expression and high KCNQ1OT1 expression were shown in osteosarcoma tissues and cells. Kcnq1 was negatively mediated by KCNQ1OT1 via DNMT1. The overexpression of Kcnq1 or knockdown of KCNQ1OT1 inhibited the proliferation, migration, and invasion, and it promoted the chemosensitivity to CDDP and apoptosis of MG-63 cells and its CDDP-resistant cell lines. Moreover, the same trend was observed in the cells following methylation inhibitor treatment. Collectively, knockdown of KCNQ1OT1 can inhibit the osteosarcoma progression through the Kcnq1/DNMT1 axis.
Collapse
Affiliation(s)
- Xu Qi
- Department of Neurology, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, P.R. China
| | - Xiao-Jun Yu
- The 1st Department of Orthopedics, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, P.R. China
| | - Xu-Ming Wang
- The 1st Department of Orthopedics, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, P.R. China
| | - Tie-Nan Song
- The 1st Department of Orthopedics, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, P.R. China
| | - Jie Zhang
- The 1st Department of Orthopedics, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, P.R. China
| | - Xin-Zhen Guo
- The 1st Department of Orthopedics, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, P.R. China
| | - Guo-Jun Li
- The 1st Department of Orthopedics, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, P.R. China
| | - Ming Shao
- The 1st Department of Orthopedics, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, P.R. China.
| |
Collapse
|
217
|
Li XJ, Li ZF, Xu YY, Han Z, Liu ZJ. microRNA-374 inhibits proliferation and promotes apoptosis of mouse melanoma cells by inactivating the Wnt signalling pathway through its effect on tyrosinase. J Cell Mol Med 2019; 23:4991-5005. [PMID: 31207106 PMCID: PMC6653165 DOI: 10.1111/jcmm.14348] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 03/16/2019] [Accepted: 04/01/2019] [Indexed: 01/05/2023] Open
Abstract
Melanoma is one of the most malignant skin tumours with constantly increasing incidence worldwide. Previous studies have demonstrated that microRNA‐374 (miR‐374) is a novel biomarker for cancer therapy. Therefore, this study explores whether miR‐374 targeting tyrosinase (TYR) affects melanoma and its underlying mechanism. We constructed subcutaneous melanoma models to carry out the following experiments. The cells were transfected with a series of miR‐374 mimics, miR‐374 inhibitors or siRNA against TYR. Dual luciferase reporter gene assay was used for the verification of the targeting relationship between miR‐374 and TYR. Reverse transcription quantitative polymerase chain reaction and western blot analysis were conducted to determine the expression of miR‐374, TYR, β‐catenin, B‐cell leukaemia 2 (Bcl‐2), Bcl‐2 associated X protein (Bax), Low‐density lipoprotein receptor‐related protein 6 (LRP6), Leucine‐rich repeat G protein‐coupled receptor 5 (LGR5) and CyclinD1. Cell proliferation, migration, invasion, cell cycle distribution and apoptosis were evaluated using cell counting kit‐8 assay, scratch test, transwell assay and flow cytometry respectively. TYR was proved as a putative target of miR‐374 as the evidenced by the result. It was observed that up‐regulated miR‐374 or down‐regulated TYR increased expression of Bax and decreased expressions of TYR, β‐catenin, LRP6, Bcl‐2, CyclinD1 and LGR5, along with diminished cell proliferation, migration, invasion and enhanced apoptosis. Meanwhile, cells with miR‐374 inhibitors showed an opposite trend. These findings indicated that up‐regulated miR‐374 could inhibit the expression of TYR to suppress cell proliferation, migration, invasion and promote cell apoptosis in melanoma cells by inhibiting the Wnt signalling pathway.
Collapse
Affiliation(s)
- Xiao-Jing Li
- Department of Dermatology, Affiliated Hospital of Hebei Engineering University, Handan, P. R. China
| | - Zhi-Feng Li
- Department of Dermatology, Affiliated Hospital of Hebei Engineering University, Handan, P. R. China
| | - Yan-Yan Xu
- Department of Dermatology, Affiliated Hospital of Hebei Engineering University, Handan, P. R. China
| | - Zhao Han
- Department of Dermatology, Affiliated Hospital of Hebei Engineering University, Handan, P. R. China
| | - Zhi-Jun Liu
- Department of Dermatology, Affiliated Hospital of Hebei Engineering University, Handan, P. R. China
| |
Collapse
|
218
|
Li G, An J, Han X, Zhang X, Wang W, Wang S. Hypermethylation of microRNA‐149 activates SDF‐1/CXCR4 to promote osteogenic differentiation of mesenchymal stem cells. J Cell Physiol 2019; 234:23485-23494. [PMID: 31206187 DOI: 10.1002/jcp.28917] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 05/12/2019] [Accepted: 05/14/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Guangjie Li
- The First Hospital of Lanzhou University Lanzhou China
- Lanzhou University Second Hospital Lanzhou China
| | - Jiangdong An
- Lanzhou University Second Hospital Lanzhou China
| | - Xingwen Han
- The First Hospital of Lanzhou University Lanzhou China
| | | | - Wenjin Wang
- The First Hospital of Lanzhou University Lanzhou China
| | - Shuanke Wang
- Lanzhou University Second Hospital Lanzhou China
| |
Collapse
|
219
|
Qian WH, Liu YY, Li X, Pan Y. MicroRNA-141 ameliorates alcoholic hepatitis‑induced intestinal injury and intestinal endotoxemia partially via a TLR4-dependent mechanism. Int J Mol Med 2019; 44:569-581. [PMID: 31173169 PMCID: PMC6605973 DOI: 10.3892/ijmm.2019.4221] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 04/23/2019] [Indexed: 12/20/2022] Open
Abstract
Alcoholic hepatitis (AH) is a fatal inflammatory syndrome with no effective treatments. Intestinal injury and intestinal endotoxemia (IETM) contribute greatly in the development of AH. MicroRNAs (miRNAs/miRs) have been reported to affect intestinal injury. The present study aims to investigate the role of miR-141 in intestinal injury and IETM of AH. An AH model was successfully established in mice and they were the injected with a series of miR-141 mimic, miR-141 inhibitor or toll like receptor 4 monoclonal antibody (TLR4mAb; an inhibitor of the Toll-like receptor TLR pathway). After that, the intestinal tissues and intestinal epithelial cells were isolated from differently treated AH mice. The expression of miR-141 and TLR pathway-associated genes and the levels of inflammatory factors were determined. Furthermore, a target prediction program and a luciferase reporter assay were employed to examine whether miR-141 targets TLR4. Finally, MTT and transwell assays were carried out to detect cell viability and cell permeability. Intestinal tissues from AH mice treated with miR-141 mimic or TLR4mAb exhibited lower levels of inflammatory factors and reduced expression of the TLR pathway-associated genes, suggesting a decreased inflammatory response as well as inactivation of the TLR pathway by miR-141. The luciferase reporter assay suggested that miR-141 negatively regulated TLR4. Intestinal epithelial cells treated with miR-141 mimic or TLR4mAb demonstrated enhanced viability and reduced permeability. Opposite results were observed in AH mice treated with a miR-141 inhibitor. Collectively, the results of the present study demonstrated that miR-141 could ameliorate intestinal injury and repress the progression of IETM through targeting TLR4 and inhibiting the TLR pathway.
Collapse
Affiliation(s)
- Wei-He Qian
- Department of Clinical Laboratory, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu 223302, P.R. China
| | - Yuan-Yuan Liu
- Department of Endocrinology, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Xiang Li
- Department of Clinical Laboratory, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu 223302, P.R. China
| | - Yan Pan
- Department of Clinical Laboratory, Lianshui County People's Hospital, Huai'an, Jiangsu 223400, P.R. China
| |
Collapse
|
220
|
Hu ZG, Zheng CW, Su HZ, Zeng YL, Lin CJ, Guo ZY, Zhong FD, Yuan GD, He SQ. MicroRNA-329-mediated PTTG1 downregulation inactivates the MAPK signaling pathway to suppress cell proliferation and tumor growth in cholangiocarcinoma. J Cell Biochem 2019; 120:9964-9978. [PMID: 30582202 DOI: 10.1002/jcb.28279] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 11/19/2018] [Indexed: 12/14/2022]
Abstract
Cholangiocarcinoma (CCA) is a severe malignancy usually producing a poor prognosis and high mortality rate. MicroRNAs (miRNAs) have been reported in association with CCA; however, the role miR-329 plays in the CCA condition still remains unclear. Therefore, this study was conducted to explore the underlying mechanism of which miR-329 is influencing the progression of CCA. This work studied the differential analysis of the expression chips of CCA obtained from the Gene Expression Omnibus database. Next, to determine both the expression and role of pituitary tumor transforming gene-1 (PTTG1) in CCA, the miRNAs regulating PTTG1 were predicted. In the CCA cells that had been intervened with miR-329 upregulation or inhibition, along with PTTG1 silencing, expression of miR-329, PTTG1, p-p38/p38, p-ERK5/ERK5, proliferating cell nuclear antigen (PCNA), Cyclin D1, Bcl-2-associated X protein (Bax), B-cell CLL/lymphoma 2 (Bcl-2), and caspase-3 were determined. The effects of both miR-329 and PTTG1 on cell proliferation, cell-cycle distribution, and apoptosis were also assayed. The miR-329 was likely to affect the CCA development through regulation of the PTTG1-mediated mitogen-activated protein kinase (MAPK) signaling pathway. The miR-329 targeted PTTG1, leading to inactivation of the MAPK signaling pathway. Upregulation of miR-329 and silencing of PTTG1 inhibited the CCA cell proliferation, induced cell-cycle arrest, and subsequently promoted apoptosis with elevations in Bax, cleaved caspase-3, and total caspase-3, but showed declines in PCNA, Cyclin D1, and Bcl-2. Moreover, miR-329 was also found to suppress the tumor growth by downregulation of PTTG1. To summarize, miR-329 inhibited the expression of PTTG1 to inactivate the MAPK signaling pathway, thus suppressing the CCA progression, thereby providing a therapeutic basis for the CCA treatment.
Collapse
Affiliation(s)
- Zhi-Gao Hu
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, People's Republic of China
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Chao-Wen Zheng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Hui-Zhao Su
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Yong-Lian Zeng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Cheng-Jie Lin
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, People's Republic of China
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Zhen-Ya Guo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Fu-Di Zhong
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Guan-Dou Yuan
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Song-Qing He
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| |
Collapse
|
221
|
Lei JJ, Li HQ, Mo ZH, Liu KJ, Zhu LJ, Li CY, Chen WL, Zhang L. Long noncoding RNA CDKN2B-AS1 interacts with transcription factor BCL11A to regulate progression of cerebral infarction through mediating MAP4K1 transcription. FASEB J 2019; 33:7037-7048. [PMID: 30870006 DOI: 10.1096/fj.201802252r] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
The effective therapeutic approach of cerebral infarction is limited because of its underlying complexity. Recently, multiple long noncoding RNAs (lncRNAs) have been identified in the pathogenesis of cerebral infarction. Here, the current study aims to explore the interaction among lncRNA cyclin-dependent kinase inhibitor-2B-antisense RNA 1 (CDKN2B-AS1), transcription factor B-cell lymphoma/leukemia 11A (BCL11A), and MAPKK kinase kinase 1 (MAP4K1) and further investigate whether they affect cerebral infarction progression. The expression of CDKN2B-AS1, BCL11A, and MAP4K1 was altered in lymphocytes extracted from patients with cerebral infarction. In order to identify their roles in regulatory T (Treg) cells, the proliferation and apoptosis of the CD4+CD25+ Treg cells were examined, and levels of IL-4, IL-10, and TGF-β were determined. Also, the RNA crosstalk among CDKN2B-AS1, BCL11A, and MAP4K1 was validated. Finally, we established a rat model of middle cerebral arterial occlusion to evaluate the neurologic impairment and cerebral infarction volume. The results revealed that lymphocytes in patients with cerebral infarction presented with the up-regulated expression of CDKN2B-AS1. Moreover, BCL11A could specifically bind to CDKN2B-AS1 and MAP4K1 promoter so as to inhibit MAP4K1. Moreover, it was observed that down-regulated CDKN2B-AS1 inhibited CD4+CD25+ Treg-cell proliferation, reduced levels of IL-4, IL-10, and TGF-β and cerebral infarction volume, and elevated MAP4K1 expression. Collectively, our study provides evidence that CDKN2B-AS1 silencing could increase MAP4K1 expression to inhibit the CD4+CD25+ Treg-cell proliferation by reducing enrichment of transcription factor BCL11A, thereby protecting against cerebral infarction progression, highlighting a promising therapeutic strategy for treating cerebral infarction.-Lei, J.-J., Li, H.-Q., Mo, Z.-H., Liu, K.-J., Zhu, L.-J., Li, C.-Y., Chen, W.-L., Zhang, L. Long noncoding RNA CDKN2B-AS1 interacts with transcription factor BCL11A to regulate progression of cerebral infarction through mediating MAP4K1 transcription.
Collapse
Affiliation(s)
- Jun-Jie Lei
- Department of Neurology, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China; and
| | - Hui-Qing Li
- Department of Neurology, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China; and
| | - Zhi-Huai Mo
- Department of Neurology, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China; and
| | - Ke-Jia Liu
- Department of Neurology, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China; and
| | - Ling-Juan Zhu
- Department of Neurology, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China; and
| | - Chun-Yi Li
- Department of Neurology, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China; and
| | - Wen-Li Chen
- Department of Pharmacology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Lei Zhang
- Department of Neurology, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China; and
| |
Collapse
|
222
|
Systematic analysis of genes and diseases using PheWAS-Associated networks. Comput Biol Med 2019; 109:311-321. [PMID: 31128465 DOI: 10.1016/j.compbiomed.2019.04.037] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/28/2019] [Accepted: 04/28/2019] [Indexed: 02/08/2023]
|
223
|
Xia P, Gu R, Zhang W, Shao L, Li F, Wu C, Sun Y. MicroRNA-377 exerts a potent suppressive role in osteosarcoma through the involvement of the histone acetyltransferase 1-mediated Wnt axis. J Cell Physiol 2019; 234:22787-22798. [PMID: 31152456 DOI: 10.1002/jcp.28843] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 04/30/2019] [Accepted: 04/30/2019] [Indexed: 12/18/2022]
Abstract
It has been demonstrated that microRNAs (miRNAs) may contribute to tumorigenesis and tumor growth in osteosarcoma (OS), which is a primary malignant tumor of bone frequently diagnosed in adolescents and young people. The purpose of our investigation was to evaluate the functional relevance of miR-377 in OS and to investigate whether the mechanism was related to the histone acetyltransferase 1 (HAT1)-mediated Wnt signaling pathway. By screening differentially expressed genes in microarray GSE47572, HAT1 was found to be a candidate gene of interest. Besides, the regulatory miRNA (miR-377) of HAT1 was also selected. The interaction among miR-377, HAT1, and the Wnt signaling pathway was evaluated. In addition, the miR-377 expression was altered in OS cells (U-2OS and SOSP-9607) to assess the in vitro cell apoptosis and the in vivo tumor growth. OS tissues presented elevated HAT1 expression and decreased miR-377 expression. A putative miR-377 binding site in HAT1 3'-UTR HAT1 was verified. Cells with miR-377 overexpression or HAT1 silencing were observed to exhibit reduced HAT1 expression and promoted apoptosis, accompanied by blockade of Wnt signaling. Moreover, the in vivo experiment revealed that miR-377 overexpression or HAT1 silencing inhibited tumor growth and reduced tumor size in nude mice. Taken together, our results conclude that miR-377 may promote OS cell apoptosis through inactivation of the HAT1-mediated Wnt signaling pathway, highlighting the potential therapeutic effect of miR-377 on OS treatment.
Collapse
Affiliation(s)
- Peng Xia
- Department of Orthopeadics, The Second Hospital of Jilin University, Changchun, Jilin, P.R. China
| | - Rui Gu
- Department of Orthopeadics, China-Japan Union Hospital of Jilin University, Changchun, Jilin, P.R. China
| | - Wei Zhang
- Department of Orthopeadics, The Second Hospital of Jilin University, Changchun, Jilin, P.R. China
| | - Liwei Shao
- Department of Orthopeadics, China-Japan Union Hospital of Jilin University, Changchun, Jilin, P.R. China
| | - Fang Li
- Department of Orthopeadics, China-Japan Union Hospital of Jilin University, Changchun, Jilin, P.R. China
| | - Changyan Wu
- Department of Orthopeadics, China-Japan Union Hospital of Jilin University, Changchun, Jilin, P.R. China
| | - Yifu Sun
- Department of Orthopeadics, China-Japan Union Hospital of Jilin University, Changchun, Jilin, P.R. China
| |
Collapse
|
224
|
Duan Q, Si E. MicroRNA-25 aggravates Aβ1-42-induced hippocampal neuron injury in Alzheimer's disease by downregulating KLF2 via the Nrf2 signaling pathway in a mouse model. J Cell Biochem 2019; 120:15891-15905. [PMID: 31144355 DOI: 10.1002/jcb.28861] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 02/12/2019] [Accepted: 02/14/2019] [Indexed: 12/21/2022]
Abstract
Recently, numerous microRNAs (miRNAs) have been considered as key players in the regulation of neuronal processes. The purpose of the present study is to explore the effect of miR-25 on hippocampal neuron injury in Alzheimer's disease (AD) induced by amyloid β (Aβ) peptide fragment 1 to 42 (Aβ1-42) via Kruppel-like factor 2 (KLF2) through the nuclear factor-E2-related factor 2 (Nrf2) signaling pathway. A mouse model of AD was established through Aβ1-42 induction. The underlying regulatory mechanisms of miR-25 were analyzed through treatment of miR-25 mimics, miR-25 inhibitors, or small interfering RNA (siRNA) against KLF2 in hippocampal tissues and cells isolated from AD mice. The targeting relationship between miR-25 and KLF2 was predicted using a target prediction program and verified by luciferase activity determination. MTT assay was used to evaluate the proliferative ability and flow cytometry to detect cell cycle distribution and apoptosis. KLF2 was confirmed as a target gene of miR-25. When the mice were induced by Aβ1-42, proliferation was suppressed while apoptosis was promoted in hippocampal neurons as evidenced by lower levels of KLF2, Nrf2, haem oxygenase, glutathione S transferase α1, glutathione, thioredoxin, and B-cell lymphoma-2 along with higher bax level. However, such alternations could be reversed by treatment of miR-25 inhibitors. These findings indicate that miR-25 may inhibit hippocampal neuron proliferation while promoting apoptosis, thereby aggravating hippocampal neuron injury through downregulation of KLF2 via the Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Qiang Duan
- Department of Neurology, Heze Third People's Hospital, Heze, People's Republic of China
| | - Erwang Si
- Department of Neurology, Heze Third People's Hospital, Heze, People's Republic of China
| |
Collapse
|
225
|
Zhao X, Wang Y, Yang J, Liu H, Wang L. MicroRNA‐326 suppresses iNOS expression and promotes autophagy of dopaminergic neurons through the JNK signaling by targeting XBP1 in a mouse model of Parkinson's disease. J Cell Biochem 2019; 120:14995-15006. [PMID: 31135066 DOI: 10.1002/jcb.28761] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 12/26/2018] [Accepted: 01/10/2019] [Indexed: 02/03/2023]
Affiliation(s)
- Xiao‐Hui Zhao
- Department of Neurology Shanghai University of Medicine & Health Sciences Affiliated Pudong New District People's Hospital Shanghai P. R. China
| | - Yong‐Bing Wang
- Department of General Surgery Shanghai University of Medicine & Health Sciences Affiliated Pudong New District People's Hospital Shanghai P. R. China
| | - Juan Yang
- Department of Neurology Shanghai University of Medicine & Health Sciences Affiliated Pudong New District People's Hospital Shanghai P. R. China
| | - Hui‐Qin Liu
- Department of Neurology Shanghai University of Medicine & Health Sciences Affiliated Pudong New District People's Hospital Shanghai P. R. China
| | - Ling‐Ling Wang
- Department of Neurology Shanghai University of Medicine & Health Sciences Affiliated Pudong New District People's Hospital Shanghai P. R. China
| |
Collapse
|
226
|
Wen Y, Liu G, Zhang Y, Li H. MicroRNA-205 is associated with diabetes mellitus-induced erectile dysfunction via down-regulating the androgen receptor. J Cell Mol Med 2019; 23:3257-3270. [PMID: 30729682 PMCID: PMC6484320 DOI: 10.1111/jcmm.14212] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 12/12/2018] [Accepted: 01/17/2019] [Indexed: 12/15/2022] Open
Abstract
As a major class of regulatory genes in majority metazoans, microRNAs (miRs) play an important role in various diseases including diabetes mellitus (DM). Lack of androgens has previously been associated with DM-induced erectile dysfunction (DMED). In addition, the biological functioning of androgen is mediated by androgen receptor (AR). Herein, we sought to investigate whether miRs participate in AR-associated DMED. Sprague-Dawlay rats were employed to establish DMED models. After modelling, levels of miR-205 and AR in their cavernous bodies were measured. The relationship between miR-205 and AR was verified using a dual-luciferase reporter gene assay. The underlying regulatory mechanisms of miR-205 were investigated in concert with the treatment of mimics or inhibitors of miR-205, or AR overexpression in the cavernous smooth muscle cells (CSMCs) isolated from rats with DMED. Meanwhile, the effects of miR-205 and AR on cell proliferation and apoptosis were evaluated using MTT assay and flow cytometry respectively. Rats with DMED presented with increased miR-205 and decreased AR levels in the cavernous bodies. AR was identified as a target gene of miR-205. Down-regulation of miR-205 or up-regulation of AR could increase proliferation and inhibits apoptosis of CSMCs in addition to improvements in the erectile functioning of rats with DMED. In summary, miR-205 may contribute to the pathogenesis of DMED via down-regulation of AR expressions.
Collapse
Affiliation(s)
- Yan Wen
- Department of EndocrinologyChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Guohui Liu
- Department of CardiologyChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Yun Zhang
- Department of UrologyChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Hai Li
- Department of UrologyChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| |
Collapse
|
227
|
Yu HM, Yao XD, Zhang RM, Zhuang HF, Wang PW, Li YZ. Repression of let-7b-5p prevents the development of multifidus muscle dysfunction by promoting vitamin D accumulation via upregulation of electron transfer flavoprotein alpha subunit in a rat model of multifidus muscle injury. J Cell Biochem 2019; 120:7458-7473. [PMID: 30387180 DOI: 10.1002/jcb.28020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 10/15/2018] [Indexed: 01/24/2023]
Abstract
Multifidus muscle dysfunction is associated with the multifidus muscle injury (MMI), which ultimately result in the low-back pain. Increasing evidence shows that microRNAs (miRs) may be involved in multifidus muscle dysfunction. In this study, we tested the hypothesis that downregulation of let-7b-5p may inhibit the multifidus muscle dysfunction development and progression. The target prediction program and luciferase activity determination confirmed electron transfer flavoprotein alpha subunit (ETFA) as a direct target gene of let-7b-5p. To study the mechanisms and functions of let-7b-5p in relation to ETFA in MMI progression, we prepared rats with experimental MMI, and a lentivirus-based packaging system was designed to upregulate expressions of let-7b-5p, and downregulate the expression of ETFA. ETFA was identified as a target gene of let-7b-5p. Older age, a longer duration of pain, and higher visual analog scale and Oswestry disability index scores for the patients with chronic low-back pain were linked to a more severe degree of degenerative muscle atrophy and fatty infiltration. Increased expression of let-7b-5p and decreased expression of ETFA and vitamin D receptor (VDR) were positively correlated with multifidus muscle dysfunction. Downregulated let-7b-5p could inhibit infiltration of collagen fibers, reverse the ultrastructural changes of multifidus muscle, and induce the VDR expression, thereby repair the MMI. The results provided a potential basis for let-7b-5p that could support targeted intervention in multifidus muscle dysfunction. Collectively, this study confirmed that downregulation of let-7b-5p has a potential inhibitory effect on the development of the function of the musculus myocytes by upregulating ETFA.
Collapse
Affiliation(s)
- Hai-Ming Yu
- Department of Orthopaedics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Xue-Dong Yao
- Department of Orthopaedics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Rong-Mou Zhang
- Department of Orthopaedics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Hua-Feng Zhuang
- Department of Orthopaedics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Pei-Wen Wang
- Department of Orthopaedics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Yi-Zhong Li
- Department of Orthopaedics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| |
Collapse
|
228
|
Wu S, Han M, Zhang C. Overexpression of microRNA-186 inhibits angiogenesis in retinoblastoma via the Hedgehog signaling pathway by targeting ATAD2. J Cell Physiol 2019; 234:19059-19072. [PMID: 30993715 DOI: 10.1002/jcp.28545] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 02/27/2019] [Accepted: 03/06/2019] [Indexed: 12/13/2022]
Abstract
Retinoblastoma (RB) represents an aggressive malignancy in the eye during the period of infancy and childhood. We delineated the ability of microRNA-186 (miR-186) to influence viability, invasion, migration, angiogenesis, and apoptosis of RB via the Hedgehog signaling pathway by targeting AAA domain-containing protein 2 (ATAD2). The microarray-based analysis was adopted to identify differentially expressed genes (DEGs) related to RB. Subsequently, RB cells were treated with miR-186 mimic, miR-186 inhibitor, or si-ATAD2. The expression of miR-186, ATAD2, Hedgehog signaling pathway-related genes were evaluated, and the target relationship between miR-186 and ATAD2 was verified. Finally, cell proliferation, invasion, migration, apoptosis, and angiogenesis were assessed. ATAD2 was identified as a DEG and modulated by miR-186. Moreover, we revealed that ATAD2 was highly expressed, whereas miR-186 was lowly expressed, and the Hedgehog signaling pathway was activated in RB. Then, ATAD2 as a putative target of miR-186 was validated using a luciferase assay. miR-186 mimic or siRNA-ATAD2 in RB cells reduced cell viability, invasion, and migration coordinating with elevated apoptosis via impairing the Hedgehog signaling pathway, where repressed angiogenesis was observed. Overexpression of miR-186 attenuates RB via the inactivation of the Hedgehog signaling pathway by downregulating ATAD2.
Collapse
Affiliation(s)
- Shuai Wu
- Department of Orbital Disease & Ocular Plastic Surgery, The Second Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Mei Han
- Department of Strabismus & Pediatric Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Chao Zhang
- Department of Strabismus & Pediatric Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| |
Collapse
|
229
|
Sun G, Liu M, Han H. Overexpression of microRNA‐190 inhibits migration, invasion, epithelial‐mesenchymal transition, and angiogenesis through suppression of protein kinase B‐extracellular signal‐regulated kinase signaling pathway via binding to stanniocalicin 2 in breast cancer. J Cell Physiol 2019; 234:17824-17838. [PMID: 30993707 DOI: 10.1002/jcp.28409] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 01/30/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Guiming Sun
- Department of Oncology Liaocheng People's Hospital Liaocheng P.R. China
| | - Meirong Liu
- Department of Oncology Liaocheng People's Hospital Liaocheng P.R. China
| | - Hui Han
- Department of Oncology Liaocheng People's Hospital Liaocheng P.R. China
| |
Collapse
|
230
|
MicroRNA-143-5p targeting eEF2 gene mediates intervertebral disc degeneration through the AMPK signaling pathway. Arthritis Res Ther 2019; 21:97. [PMID: 30987676 PMCID: PMC6466769 DOI: 10.1186/s13075-019-1863-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 03/11/2019] [Indexed: 11/10/2022] Open
Abstract
Background Intervertebral disc degeneration (IDD) is a major contributor to back, neck, and radicular pain, and the treatment of IDD is costly and relatively ineffective. Dysregulation of microRNAs (miRNAs) has been reported to be involved in IDD. The purpose of our study is to illustrate the potential that miR-143-5p targeting eEF2 gene mediates IDD. Methods Following the establishment of the IDD rat models, expression of miR-143-5p, eEF2, Bcl-2, Bax, AMPK, mTOR, cyclinD, COL2, ACAN, and DCN was detected. The NP cells isolated from degenerative intervertebral disc (IVD) were introduced with a series of mimic, inhibitor, or AICAR to explore the functional role of miR-143-5p in IDD and to characterize the relationship between miR-143-5p and eEF2. Cell viability, cell cycle, apoptosis, and senescence were also evaluated. Results A reduction in eEF2, an increase in miR-143-5p, and activation of the AMPK signaling pathway were observed in degenerative IVD. Moreover, increased senescent NP cells were observed in degenerative IVD. eEF2 was confirmed as a target gene of miR-143-5p. miR-143-5p was found to activate the AMPK signaling pathway. The restoration of miR-143-5p or the activation of AMPK signaling pathway decreased COL2, ACAN, and DCN expression, coupled with the inhibition of NP cell proliferation and differentiation, and promotion of NP apoptosis and senescence. On the contrary, the inhibition of miR-143-5p led to the reversed results. Conclusion The results demonstrated that the inhibition of miR-143-5p may act as a suppressor for the progression of IDD.
Collapse
|
231
|
Yang LX, Wu J, Guo ML, Zhang Y, Ma SG. Suppression of long non-coding RNA TNRC6C-AS1 protects against thyroid carcinoma through DNA demethylation of STK4 via the Hippo signalling pathway. Cell Prolif 2019; 52:e12564. [PMID: 30938030 PMCID: PMC6536409 DOI: 10.1111/cpr.12564] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 09/28/2018] [Accepted: 11/02/2018] [Indexed: 01/09/2023] Open
Abstract
Objectives Thyroid carcinoma (TC) represents a malignant neoplasm affecting the thyroid. Current treatment strategies include the removal of part of the thyroid; however, this approach is associated with a significant risk of developing hypothyroidism. In order to adequately understand the expression profiles of TNRC6C‐AS1 and STK4 and their potential functions in TC, an investigation into their involvement with Hippo signalling pathway and the mechanism by which they influence TC apoptosis and autophagy were conducted. Methods A microarray analysis was performed to screen differentially expressed lncRNAs associated with TC. TC cells were employed to evaluate the role of TNRC6C‐AS1 by over‐expression or silencing means. The interaction of TNRC6C‐AS1 with methylation of STK4 promoter was evaluated to elucidate its ability to elicit autophagy, proliferation and apoptosis. Results TNRC6C‐AS1 was up‐regulated while STK4 was down‐regulated, where methylation level was elevated. STK4 was verified as a target gene of TNRC6C‐AS1, which was enriched by methyltransferase. Methyltransferase’s binding to STK4 increased expression of its promoter. Over‐expressed TNRC6C‐AS1 inhibited STK4 by promoting STK4 methylation and reducing the total protein levels of MST1 and LATS1/2. The phosphorylation of YAP1 phosphorylation was decreased, which resulted in the promotion of SW579 cell proliferation and tumorigenicity. Conclusion Based on our observations, we subsequently confirmed the anti‐proliferative, pro‐apoptotic and pro‐autophagy capabilities of TNRC6C‐AS1 through STK4 methylation via the Hippo signalling pathway in TC.
Collapse
Affiliation(s)
- Liu-Xue Yang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Ji Wu
- Department of Thyroid and Breast Surgery, Suqian Hospital Affiliated to Xuzhou Medical University, Suqian, China.,Department of Thyroid and Breast Surgery, Nanjing Drum Tower Hospital, Suqian, China
| | - Man-Li Guo
- Department of Endocrinology and Metabolism, Suqian People's Hospital, Nanjing Drum Tower Hospital, Suqian, China
| | - Yong Zhang
- Department of Endocrinology and Metabolism, Huai'an Hospital Affiliated to Xuzhou Medical College and Huai'an Second People's Hospital, Huai'an, China.,Department of Endocrinology and Metabolism, Suqian First Hospital, Suqian, China
| | - Shao-Gang Ma
- Department of Endocrinology and Metabolism, Suqian First Hospital, Suqian, China.,Department of Endocrinology and Metabolism, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
| |
Collapse
|
232
|
Chen YN, Ren CC, Yang L, Nai MM, Xu YM, Zhang F, Liu Y. MicroRNA let‑7d‑5p rescues ovarian cancer cell apoptosis and restores chemosensitivity by regulating the p53 signaling pathway via HMGA1. Int J Oncol 2019; 54:1771-1784. [PMID: 30816441 DOI: 10.3892/ijo.2019.4731] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 10/04/2018] [Indexed: 11/06/2022] Open
Abstract
Ovarian cancer (OC) is the gynecological malignancy type with the highest mortality rate in females. The regulatory effect of microRNAs (miRs) on their target genes serves a key role in tumor development. Therefore, in the present study, whether miR let‑7d‑5p targeting high mobility group A1 (HMGA1) regulated biological characteristics and chemosensitivity of OC cells by mediating the p53 signaling pathway was investigated. The let‑7d‑5p level was detected in OC tissues and adjacent normal tissues, followed by detection in OC cell lines SKOV3, A2780, OVCAR‑3 and CaOV3, and human normal ovarian epithelial cell line (IOSE‑80), in order to select the OC cell line for the following experiments. Subsequently, OC cells were treated with the let‑7d‑5p mimic, siHMGA1 and Tenovin‑1. The targeting association between let‑7d‑5p and HMGA1 was then examined, and the OC cell viability, migration, cycle and apoptosis were evaluated. Subsequently, the chemosensitivity of OC cells to cisplatin was verified. Finally, expression levels of let‑7d‑5p, HMGA1, p21, B‑cell lymphoma‑2 (Bcl‑2)‑associated X (Bax), p27, p53 wild‑type (p53wt), p53 mutated (p53mut), proliferating cell nuclear antigen (PCNA), cyclin‑dependent kinase 2 (CDK2), matrix metallopeptidase (MMP)2, MMP9 and Bcl‑2 were determined. As demonstrated in the results, let‑7d‑5p expression was low in OC tissues and had an increased reduction in the OVCAR‑3 cell line. HMGA1 was confirmed as a target of let‑7d‑5p, and its expression was also silenced by let‑7d‑5p. let‑7d‑5p repressed OC cell viability, migration, cell cycle progression and apoptosis, while it promoted the chemosensitivity of OC cells to cisplatin by targeting HMGA1. The expression of let‑7d‑5p, p21, Bax, p27 and p53wt was increased, while that of HMGA1, p53mut, PCNA, CDK2, MMP2, MMP9 and Bcl‑2 was reduced following cell transfection. The results in the present study provided evidence that let‑7d‑5p may suppress proliferation, and facilitate apoptosis and cisplatin chemosensitivity of OC cells by silencing HMGA1 via the p53 signaling pathway.
Collapse
Affiliation(s)
- Yan-Nan Chen
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Chen-Chen Ren
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Li Yang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Man-Man Nai
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yi-Ming Xu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Feng Zhang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yan Liu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
233
|
Qu Y, Zhang Y, Wu J, Jie L, Deng J, Zhao D, Yu Q. Retracted
: Downregulated microRNA‐135a ameliorates rheumatoid arthritis by inactivation of the phosphatidylinositol 3‐kinase/AKT signaling pathway via phosphatidylinositol 3‐kinase regulatory subunit 2. J Cell Physiol 2019; 234:17663-17676. [DOI: 10.1002/jcp.28390] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 01/10/2019] [Accepted: 01/16/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Yuan Qu
- Department of Rheumatology and Clinical Immunology Zhujiang Hospital of Southern Medical University Guangzhou China
| | - Yu‐Ping Zhang
- Department of Rheumatology and Clinical Immunology Zhujiang Hospital of Southern Medical University Guangzhou China
| | - Jing Wu
- Department of Rheumatology and Clinical Immunology Zhujiang Hospital of Southern Medical University Guangzhou China
| | - Li‐Gang Jie
- Department of Rheumatology and Clinical Immunology Zhujiang Hospital of Southern Medical University Guangzhou China
| | - Jia‐Xin Deng
- Department of Rheumatology and Clinical Immunology Zhujiang Hospital of Southern Medical University Guangzhou China
| | - Dong‐Bao Zhao
- Department of Rheumatology and Immunology Changhai Hospital, Second Military Medical University Shanghai China
| | - Qing‐Hong Yu
- Department of Rheumatology and Clinical Immunology Zhujiang Hospital of Southern Medical University Guangzhou China
| |
Collapse
|
234
|
Song S, Lin S, Liu J, Zhang M, Du Y, Zhang D, Xu W, Wang H. Retracted
: Targeting of SPP1 by microRNA‐340 inhibits gastric cancer cell epithelial–mesenchymal transition through inhibition of the PI3K/AKT signaling pathway. J Cell Physiol 2019; 234:18587-18601. [DOI: 10.1002/jcp.28497] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 02/19/2019] [Accepted: 02/20/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Su‐Zhen Song
- Department of Internal Medicine Shandong University of Traditional Chinese Medicine Jinan Shandong People's Republic of China
| | - Sen Lin
- Department of Digestive Disease The Second Hospital of Shandong University Jinan Shandong People's Republic of China
| | - Jia‐Ning Liu
- Department of Thyroid and Pancreatic Disease The Second Hospital of Shandong University Jinan Shandong People's Republic of China
| | - Ming‐Bao Zhang
- Department of Digestive Disease The Second Hospital of Shandong University Jinan Shandong People's Republic of China
| | - Ya‐Ting Du
- Department of Digestive Disease The Second Hospital of Shandong University Jinan Shandong People's Republic of China
| | - Dong‐Dong Zhang
- Department of Digestive Disease The Second Hospital of Shandong University Jinan Shandong People's Republic of China
| | - Wei‐Hua Xu
- Department of Digestive Disease The Second Hospital of Shandong University Jinan Shandong People's Republic of China
| | - Hong‐Bo Wang
- Department of Digestive Disease The Second Hospital of Shandong University Jinan Shandong People's Republic of China
| |
Collapse
|
235
|
Dong Y, Wang J, Du KX, Jia TM, Zhu CL, Zhang Y, Xu FL. MicroRNA-135a participates in the development of astrocytes derived from bacterial meningitis by downregulating HIF-1α. Am J Physiol Cell Physiol 2019; 316:C711-C721. [PMID: 30726113 DOI: 10.1152/ajpcell.00440.2018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Accumulating evidence has highlighted the potential of microRNAs (miRs) as biomarkers in various human diseases. However, the roles of miRs in bacterial meningitis (BM), a severe infectious condition, still remain unclear. Thus, the present study aimed to investigate the effects of miR-135a on proliferation and apoptosis of astrocytes in BM. Neonatal rats were injected with Streptococcus pneumoniae to establish the BM model. The expression of miR-135a and hypoxia-inducible factor 1α (HIF-1α) in the BM rat models were characterized, followed by determination of their interaction. Using gain- and loss-of-function approaches, the effects of miR-135a on proliferation, apoptosis, and expression of glial fibrillary acidic protein (GFAP), in addition to apoptosis-related factors in astrocytes were examined accordingly. The regulatory effect of HIF-1α was also determined along with the overexpression or knockdown of HIF-1α. The results obtained indicated that miR-135a was poorly expressed, whereas HIF-1α was highly expressed in the BM rat models. In addition, restored expression levels of miR-135a were determined to promote proliferation while inhibiting the apoptosis of astrocytes, along with downregulated Bax and Bad, as well as upregulated Bcl-2, Bcl-XL, and GFAP. As a target gene of miR-135a, HIF-1α expression was determined to be diminished by miR-135a. The upregulation of HIF-1α reversed the miR-135a-induced proliferation of astrocytes. Taken together, the key findings of the current study present evidence suggesting that miR-135a can downregulate HIF-1α and play a contributory role in the development of astrocytes derived from BM, providing a novel theoretical perspective for BM treatment approaches.
Collapse
Affiliation(s)
- Yan Dong
- Department of Pediatrics, Third Affiliated Hospital of Zhengzhou University , Zhengzhou , China.,Henan Provincial Key Laboratory of Child Brain Injury , Zhengzhou , China
| | - Jun Wang
- Department of Children Rehabilitation, Third Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Kai-Xian Du
- Department of Pediatrics, Third Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Tian-Ming Jia
- Department of Pediatrics, Third Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Chang-Lian Zhu
- Department of Pediatrics, Third Affiliated Hospital of Zhengzhou University , Zhengzhou , China.,Henan Provincial Key Laboratory of Child Brain Injury , Zhengzhou , China.,Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | - Yan Zhang
- Clinical Laboratory, Henan Red Cross Blood Center , Zhengzhou , China
| | - Fa-Lin Xu
- Department of Pediatrics, Third Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| |
Collapse
|
236
|
Zhou ZQ, Chen Y, Chai M, Tao R, Lei YH, Jia YQ, Shu J, Ren J, Li G, Wei WX, Han YD, Han Y. Adipose extracellular matrix promotes skin wound healing by inducing the differentiation of adipose‑derived stem cells into fibroblasts. Int J Mol Med 2019; 43:890-900. [PMID: 30535488 PMCID: PMC6317660 DOI: 10.3892/ijmm.2018.4006] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 11/20/2018] [Indexed: 12/25/2022] Open
Abstract
Fibroblasts are the major effector cells of skin wound healing. Adipose‑derived stem cells can differentiate into fibroblasts under certain conditions. In the present study, it was hypothesized that adipose‑derived stem cells (ADSCs) could be induced by the adipose extracellular matrix (ECM) to differentiate into fibroblasts in order to promote skin wound healing. First, flow cytometry was used to detect the ratio of fibroblasts and relative expression of the fibroblast markers cytokeratin 19 (CK19) and vimentin in ADSCs. Then, the effect of the adipose ECM during the differentiation of ADSCs into fibroblasts was investigated by detecting the total amount of collagen fibers and degree of fibrosis, and the proliferation and cell cycle of differentiated fibroblasts, using the MTT assay and flow cytometry analysis respectively. Finally, a mouse skin wound model was established and treated with PBS, ADSC suspension or ECM + ADSCs to compare wound healing rate and expression of collagen I and collagen III by immunohistochemistry. Following induction of ADSCs with the adipose ECM, more fibroblasts were found, expression of CK19 and vimentin increased, and a greater degree of fibrosis occurred, which revealed the positive effect of the adipose ECM on the differentiation of ADSCs into fibroblasts. In addition, the induced fibroblasts had enhanced proliferation activity, with more cells in the S phase and fewer in the G2/M phase. The in vivo experiment indicated that the ECM produced by the ADSCs had a faster wound healing rate and increased expression of collagen I and collagen III compared with mice injected with PBS or ADSCs alone, which verified that ADSCs induced by the adipose ECM had a positive effect on skin wound healing. The present study demonstrated that the adipose ECM in combination with ADSCs may be a novel therapeutic target for the repair of skin injury, due to the ability of the adipose ECM to induce the differentiation of ADSCs into fibroblasts and to facilitate the wound healing process.
Collapse
Affiliation(s)
- Zhi-Qiang Zhou
- Department of Plastic and Reconstructive Surgery, General Hospital of Chinese PLA, Beijing 100853
| | - Yi Chen
- Institute of Bioengineering, Academy of Military Medical Research, Academy of Military Science of Chinese PLA, Beijing 100071, P.R. China
| | - Mi Chai
- Department of Plastic and Reconstructive Surgery, General Hospital of Chinese PLA, Beijing 100853
| | - Ran Tao
- Department of Plastic and Reconstructive Surgery, General Hospital of Chinese PLA, Beijing 100853
| | - Yong-Hong Lei
- Department of Plastic and Reconstructive Surgery, General Hospital of Chinese PLA, Beijing 100853
| | - Yi-Qing Jia
- Department of Plastic and Reconstructive Surgery, General Hospital of Chinese PLA, Beijing 100853
| | - Jun Shu
- Department of Plastic and Reconstructive Surgery, General Hospital of Chinese PLA, Beijing 100853
| | - Jing Ren
- Department of Plastic and Reconstructive Surgery, General Hospital of Chinese PLA, Beijing 100853
| | - Guo Li
- Department of Plastic and Reconstructive Surgery, General Hospital of Chinese PLA, Beijing 100853
| | - Wen-Xin Wei
- Department of Plastic and Reconstructive Surgery, General Hospital of Chinese PLA, Beijing 100853
| | - Yu-Di Han
- Department of Plastic and Reconstructive Surgery, General Hospital of Chinese PLA, Beijing 100853
| | - Yan Han
- Department of Plastic and Reconstructive Surgery, General Hospital of Chinese PLA, Beijing 100853
| |
Collapse
|
237
|
Efficacy of Biophysical Energies on Healing of Diabetic Skin Wounds in Cell Studies and Animal Experimental Models: A Systematic Review. Int J Mol Sci 2019; 20:ijms20020368. [PMID: 30654555 PMCID: PMC6359711 DOI: 10.3390/ijms20020368] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/04/2019] [Accepted: 01/14/2019] [Indexed: 12/22/2022] Open
Abstract
We have systematically assessed published cell studies and animal experimental reports on the efficacy of selected biophysical energies (BPEs) in the treatment of diabetic foot ulcers. These BPEs include electrical stimulation (ES), pulsed electromagnetic field (PEMF), extracorporeal shockwave (ECSW), photo energies and ultrasound (US). Databases searched included CINAHL, MEDLINE and PubMed from 1966 to 2018. Studies reviewed include animal and cell studies on treatment with BPEs compared with sham, control or other BPEs. Information regarding the objective measures of tissue healing and data was extracted. Eighty-two studies were eventually selected for the critical appraisal: five on PEMF, four each on ES and ECSW, sixty-six for photo energies, and three about US. Based on the percentage of original wound size affected by the BPEs, both PEMF and low-level laser therapy (LLL) demonstrated a significant clinical benefit compared to the control or sham treatment, whereas the effect of US did not reveal a significance. Our results indicate potential benefits of selected BPEs in diabetic wound management. However, due to the heterogeneity of the current clinical trials, comprehensive studies using well-designed trials are warranted to confirm the results.
Collapse
|
238
|
Liu L, Yang L, Chang H, Chen YN, Zhang F, Feng S, Peng J, Ren CC, Zhang XA. CP‑31398 attenuates endometrial cancer cell invasion, metastasis and resistance to apoptosis by downregulating MDM2 expression. Int J Oncol 2019; 54:942-954. [PMID: 30628640 PMCID: PMC6365028 DOI: 10.3892/ijo.2019.4681] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 10/26/2018] [Indexed: 12/21/2022] Open
Abstract
Endometrial cancer (EC) is one of the most common malignancies of the female reproductive system, and metastasis is a major cause of mortality. In this study, we aimed to explore the role of CP-31398 in the migration, invasion and apoptosis of EC cells by its regulation of the expression of the murine double minute 2 (MDM2) gene. For this purpose, EC tissues and adjacent normal tissues were collected, and the positive expression rate of MDM2 in these tissues was assessed. Subsequently, the cellular 50% inhibitory concentration (IC50) of CP-31398 was measured. The EC RL95-2 and KLE cell lines had a higher MDM2 expression and were thus selected for use in subsequent experiments. The EC cells were then treated with CP-31398 (2 µg/ml), and were transfected with siRNA against MDM2 or an MDM2 overexpression plasmid in order to examine the effects of CP-31398 and MDM2 on EC cell activities. The expression of p53, p21, Bad, Bax, B-cell lymphoma-2 (Bcl-2), cytochrome c (Cyt-c), caspase-3, Cox-2, matrix metalloproteinase (MMP)-2 and MMP-9 was measured to further confirm the effects of CP-31398 on cell migration, invasion and apoptosis. Our results indicated that MDM2 was highly expressed in EC tissues. Notably, EC cell viability decreased with the increasing concentrations of CP-31398. The EC cells treated with CP-31398 or siRNA against MDM2 exhibited an increased apoptosis and a suppressed migration and invasion, corresponding to an increased expression of p53, p21, Bad, Bax, Cyt-c and caspase-3, as well as to a decreased expression of Bcl-2, Cox-2, MMP-2 and MMP-9. Moreover, treatment with CP-31398 and siRNA against MDM2 further enhanced these effects. Taken together, the findings of this study indicate that the CP-31398-mediated downregulation of MDM2 may suppress EC progression via its inhibitory role in EC cell migration, invasion and resistance to apoptosis. Therefore, treatment with CP-31398 may prove to be possible therapeutic strategy for EC.
Collapse
Affiliation(s)
- Ling Liu
- Department of Gynecologic Oncology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Li Yang
- Department of Gynecologic Oncology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Hui Chang
- Laboratory of Tumor Precision Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yan-Nan Chen
- Department of Gynecologic Oncology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Feng Zhang
- Department of Gynecologic Oncology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Shuo Feng
- Department of Gynecologic Oncology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Juan Peng
- Department of Gynecologic Oncology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Chen-Chen Ren
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Xiao-An Zhang
- Department of Imaging, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
239
|
Gao LM, Zheng Y, Wang P, Zheng L, Zhang WL, Di Y, Chen LL, Yin XB, Tian Q, Shi SS, Xu SF. Tumor-suppressive effects of microRNA-181d-5p on non-small-cell lung cancer through the CDKN3-mediated Akt signaling pathway in vivo and in vitro. Am J Physiol Lung Cell Mol Physiol 2019; 316:L918-L933. [PMID: 30628487 DOI: 10.1152/ajplung.00334.2018] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The involvement of several microRNAs (miRs) in the initiation and development of tumors through the suppression of the target gene expression has been highlighted. The aberrant expression of miR-181d-5p and cyclin-dependent kinase inhibitor 3 (CDKN3) in non-small-cell lung cancer (NSCLC) was then screened by microarray analysis. In the present study, we performed a series of in vivo and in vitro experiments for the purpose of investigating their roles in NSCLC and the underlying mechanism. There was a high expression of CDKN3, whereas miR-181d-5p was downregulated in NSCLC. Quantitative RT-PCR, Western blot analysis, and dual-luciferase reporter gene assay further identified that CDKN3 could be negatively regulated by miR-181d-5p. Moreover, the upregulation of miR-181d-5p or silencing of CDKN3 could inactivate the Akt signaling pathway. A549 with the lowest miR-181d-5p and H1975 with the highest CDKN3 among the five NSCLC cell lines (H1299, A549, H1975, NCI-H157, and GLC-82) were adopted for in vitro experiments, in which expression of miR-181d-5p and CDKN3 was altered by transfection of miR-181d-5p mimic/inhibitor or siRNA-targeting CDKN3. Afterwards, cell proliferation, apoptosis, invasion, migration, and angiogenesis, as well as epithelial-mesenchymal transition (EMT), were evaluated, and tumorigenicity was assessed. In addition, an elevation in miR-181d-5p or depletion in CDKN3 led to significant reductions in proliferation, invasion, migration, angiogenesis, EMT, and tumorigenicity of NSCLC cells, coupling with increased cell apoptosis. In conclusion, this study highlights the tumor-suppressive effects of miR-181d-5p on NSCLC via Akt signaling pathway inactivation by suppressing CDKN3, thus providing a promising therapeutic strategy for the treatment of NSCLC.
Collapse
Affiliation(s)
- Li-Ming Gao
- Department of Oncology, the First Hospital of Qinhuangdao , Qinhuangdao , People's Republic of China
| | - Yue Zheng
- Department of Gastroenterology, the First Hospital of Qinhuangdao , Qinhuangdao , People's Republic of China
| | - Ping Wang
- Department of Respiratory, Chinese PLA General Hospital , Beijing , People's Republic of China
| | - Lei Zheng
- Department of Oncology, the First Hospital of Qinhuangdao , Qinhuangdao , People's Republic of China
| | - Wen-Li Zhang
- Department of Imaging, the First Hospital of Qinhuangdao , Qinhuangdao , People's Republic of China
| | - Ya Di
- Department of Oncology, the First Hospital of Qinhuangdao , Qinhuangdao , People's Republic of China
| | - Lan-Lan Chen
- Department of Oncology, the First Hospital of Qinhuangdao , Qinhuangdao , People's Republic of China
| | - Xiao-Bo Yin
- Department of Respiratory, the First Hospital of Qinhuangdao , Qinhuangdao , People's Republic of China
| | - Qi Tian
- Department of Respiratory, the First Hospital of Qinhuangdao , Qinhuangdao , People's Republic of China
| | - Shan-Shan Shi
- Department of Respiratory, the First Hospital of Qinhuangdao , Qinhuangdao , People's Republic of China
| | - Shu-Feng Xu
- Department of Respiratory, the First Hospital of Qinhuangdao , Qinhuangdao , People's Republic of China
| |
Collapse
|
240
|
Wang HB, Wei H, Wang JS, Li L, Chen AY, Li ZG. Down-regulated expression of LINC00518 prevents epithelial cell growth and metastasis in breast cancer through the inhibition of CDX2 methylation and the Wnt signaling pathway. Biochim Biophys Acta Mol Basis Dis 2019; 1865:708-723. [PMID: 30611858 DOI: 10.1016/j.bbadis.2019.01.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 12/07/2018] [Accepted: 01/02/2019] [Indexed: 01/02/2023]
Abstract
Breast cancer (BC)-related mortality is associated with the potential metastatic properties of the primary breast tumors. The following study was conducted with the main focus on the effect of LINC00518 on the growth and metastasis of BC epithelial cells via the Wnt signaling pathway through regulation of the methylation of CDX2 gene. Initially, differentially expressed long intergenic non-protein coding RNAs (lincRNAs) related to BC were screened out in the Cancer Genome Atlas (TCGA) database, after which we detected the LINC00518 expression and localization in BC tissues and cells. Then the CDX2 positive expression and methylation level were identified. The targeting relationship of LINC00518 and CDX2, and binding methyltransferase in the promoter region were examined. BC epithelial cell proliferation, colony formation ability, invasion, migration and apoptosis were further evaluated. The lincRNA expression data related to BC downloaded from the TCGA database revealed that there was a high expression of LINC00518 in BC, and a negative correlation between LINC00518 and CDX2. In addition, LINC00518 promotes CDX2 methylation by recruiting DNA methyltransferase through activating the Wnt signaling pathway. The down-regulation of LINC00518 inhibited proliferation, invasion, migration, and EMT of BC epithelial cells while enhancing apoptosis. The inhibitory effects of LINC00518 down-regulation was reversed by CDX2 down-regulation. In conclusion, our findings revealed that down-regulation of LINC00518 might have the ability to suppress BC progression by up-regulating CDX2 expression through the reduction of methylation and blockade of the Wnt signaling pathway, resulting in the inhibition of proliferation and promotion of apoptosis of BC epithelial cells.
Collapse
Affiliation(s)
- Hong-Bin Wang
- Department of Breast Surgery (No. 2 Sickroom), Harbin Medical University Cancer Hospital, Harbin 150081, PR China
| | - Hong Wei
- Department of In-Patient Ultrasound, The 2nd Affiliated Hospital of Harbin Medical University, Harbin 150081, PR China
| | - Jin-Song Wang
- Department of Breast Surgery (No. 2 Sickroom), Harbin Medical University Cancer Hospital, Harbin 150081, PR China
| | - Lin Li
- Department of Breast Surgery (No. 2 Sickroom), Harbin Medical University Cancer Hospital, Harbin 150081, PR China
| | - An-Yue Chen
- Department of Breast Surgery (No. 2 Sickroom), Harbin Medical University Cancer Hospital, Harbin 150081, PR China
| | - Zhi-Gao Li
- Department of Breast Surgery (No. 2 Sickroom), Harbin Medical University Cancer Hospital, Harbin 150081, PR China.
| |
Collapse
|
241
|
Zhang S, Jiang H, Xu Z, Jiang Y, She Y, Huang X, Feng S, Chen W, Chen S, Chen Y, Qiu G, Zhong S. The resistance of esophageal cancer cells to paclitaxel can be reduced by the knockdown of long noncoding RNA DDX11-AS1 through TAF1/TOP2A inhibition. Am J Cancer Res 2019; 9:2233-2248. [PMID: 31720085 PMCID: PMC6834486 DOI: pmid/31720085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 08/18/2019] [Indexed: 02/05/2023] Open
Abstract
Esophageal cancer (EC) is one of the most common malignancies in the world. The currently used chemotherapeutic drug for the treatment of EC is paclitaxel (PTX), the efficacy of which is affected by the development of drug resistance. The present study aims to define the role of the long noncoding RNA (lncRNA) DDX11-AS1 in the progression of EC with the involvement of PTX-resistant EC cells. First, EC and adjacent normal tissue samples were collected from 82 patients with EC, after which the expression levels of DDX11-AS1, TOP2A and TAF1 were determined. The results showed that DDX11-AS1, TOP2A and TAF1 were highly expressed in EC tissues, and there was a positive correlation between the expression levels of DDX11-AS1 and TOP2A. A PTX-resistant EC cell line was constructed. Next, we evaluated the effects of DDX11-AS1 and TOP2A on the resistance of EC cells to PTX, and the regulatory relationships between DDX11-AS1, TOP2A and TAF1 were investigated. DDX11-AS1 could promote TOP2A transcription via TAF1, and the knockdown of TOP2A or DDX11-AS1 could increase the sensitivity of EC cells to PTX. The effect of DDX11-AS1 on the growth of PTX-inhibited tumors was confirmed using a tumor formation assay in nude mice. It was verified that knocking down DDX11-AS1 reduced the expression level of TOP2A and inhibited tumor growth. In conclusion, our findings suggest that DDX11-AS1 knockdown results in reduced resistance of EC cells to PTX by inhibiting TOP2A transcription via TAF1. Therefore, DDX11-AS1 knockdown could be a promising therapeutic strategy for EC.
Collapse
Affiliation(s)
- Shuyao Zhang
- Department of Pharmacy, Guangzhou Red Cross Hospital Affiliated of Ji-Nan University Medical CollegeGuangzhou 510220, Guangdong Province, P. R. China
- Clinical Pharmacy Research Center, Shantou University Medical CollegeShantou 515031, Guangdong Province, P. R. China
| | - Hong Jiang
- Department of Nursing, Guangzhou Red Cross Hospital Affiliated of Ji-Nan University Medical CollegeGuangzhou 510220, Guangdong Province, P. R. China
| | - Zhe Xu
- Department of Urology, Cancer Hospital of Shantou University Medical CollegeShantou 515031, Guangdong Province, P. R. China
| | - Yi Jiang
- Department of Digestive Oncology, Cancer Hospital of Shantou University Medical CollegeShantou 515031, Guangdong Province, P. R. China
| | - Yuqi She
- Clinical Pharmacy Research Center, Shantou University Medical CollegeShantou 515031, Guangdong Province, P. R. China
| | - Xiaoting Huang
- Clinical Pharmacy Research Center, Shantou University Medical CollegeShantou 515031, Guangdong Province, P. R. China
| | - Shanna Feng
- Clinical Pharmacy Research Center, Shantou University Medical CollegeShantou 515031, Guangdong Province, P. R. China
| | - Wanying Chen
- Clinical Pharmacy Research Center, Shantou University Medical CollegeShantou 515031, Guangdong Province, P. R. China
| | - Shuang Chen
- Clinical Pharmacy Research Center, Shantou University Medical CollegeShantou 515031, Guangdong Province, P. R. China
| | - Yun Chen
- Clinical Pharmacy Research Center, Shantou University Medical CollegeShantou 515031, Guangdong Province, P. R. China
| | - Guodong Qiu
- Clinical Pharmacy Research Center, Shantou University Medical CollegeShantou 515031, Guangdong Province, P. R. China
| | - Shilong Zhong
- Clinical Pharmacy Research Center, Shantou University Medical CollegeShantou 515031, Guangdong Province, P. R. China
- Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical SciencesGuangzhou 510080, Guangdong Province, P. R. China
| |
Collapse
|
242
|
Lu S, Xu Q. MicroRNA-23a inhibits melanoma cell proliferation, migration, and invasion in mice through a negative feedback regulation of sdcbp and the MAPK/ERK signaling pathway. IUBMB Life 2018; 71:587-600. [PMID: 30589231 DOI: 10.1002/iub.1979] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 10/23/2018] [Accepted: 11/10/2018] [Indexed: 12/15/2022]
Abstract
Melanoma is the main cause of death associated with skin cancer. Surgical resection and adjuvant therapy are currently effective treatments, but the recurrence rate is very high. The understanding of microRNA (miR) dynamics after surgical resection of melanoma is essential to accurately explain the changes in the recurrence of melanoma. In this study, we hypothesized that microRNA-23a (miR-23a) affects the cell proliferation, migration, and invasion of melanoma with a mechanism related to SDCBP and the MAPK/ERK signaling pathway. To validate this, we performed a series of experiments in cells of melanoma modeled. Initially, positive expression of SDCBP and morphology of normal and melanoma tissues and cells were observed. Expression of miR-23a, SDCBP, and MAPK/ERK signaling pathway-related genes was identified in melanoma tissues. Melanoma cells transfected with mimic or inhibitor of miR-23a or si-SDCBP were detected to validate effect of miR-23a on SDCBP and the MAPK/ERK signaling pathway. MTT assay, scratch test, transwell assay, and flow cytometry were performed to evaluate cell viability, invasion, metastasis, and apoptosis in vitro, respectively. Tumorigenicity assay in nude mice was conducted to test the tumorigenesis of the transfected cells in vivo. High positive expression of SDCBP and abnormal morphology were observed in melanoma tissues and cells. Reduced expression of miR-23a and increased expression of SDCBP and MAPK/ERK signaling pathway-related genes were identified in the melanoma tissues of melanoma mice. Overexpressed miR-23a dampened SDCBP and the MAPK/ERK signaling pathway. The melanoma cells with overexpressed miR-23a presented ascended cell apoptosis and descended cell proliferation, migration, invasion as well as tumor size. Taken together, our study demonstrated that miR-23a could inhibit the development of melanoma in mice through a negative feedback regulation of SDCBP and the MAPK/ERK signaling pathway. © 2018 IUBMB Life, 71(5):587-600, 2019.
Collapse
Affiliation(s)
- Shelian Lu
- Department of Neurobiology, Beijing Institute for Brain Disorders, Beijing Center of Neural Regeneration and Repair, Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Capital Medical University, Beijing, China
| | - Qunyuan Xu
- Department of Neurobiology, Beijing Institute for Brain Disorders, Beijing Center of Neural Regeneration and Repair, Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Capital Medical University, Beijing, China
| |
Collapse
|
243
|
Lan Y, Li YJ, Li DJ, Li P, Wang JY, Diao YP, Ye GD, Li YF. Long noncoding RNA MEG3 prevents vascular endothelial cell senescence by impairing miR-128-dependent Girdin downregulation. Am J Physiol Cell Physiol 2018; 316:C830-C843. [PMID: 30576236 DOI: 10.1152/ajpcell.00262.2018] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Long noncoding RNAs (lncRNAs) are commonly associated with various biological functions, in which the function of lncRNA maternally expressed gene 3 (MEG3) has been identified in various cancers. Strikingly, an association between MEG3 with microRNAs (miRNAs), mRNAs, and proteins has been reported. This study investigates the role of MEG3 in vascular endothelial cell (VEC) senescence. Expression of Girdin and miR-128 was monitored in the blood vessel samples of young and old mice/healthy volunteers, along with the measurement of human umbilical vein endothelial cells (HUVECs). The relationship between MEG3/Girdin and miR-128 was determined and verified. Loss- and gain-of-function approaches were applied to analyze the regulatory effects of MEG3 on platelet phagocytosis and lipoprotein oxidation of HUVEC membrane. In addition, the effect of MEG3 on HUVEC senescence was evaluated by detection of the reactive oxygen species, telomerase activity, and telomere length. To further analyze the MEG3-mediated regulatory mechanism, miR-128 upregulation and inhibition were introduced into the HUVECs. Downregulated Girdin and upregulated miR-128 were found in the blood vessels of old individuals and old mice, as well as in senescent HUVECs. MEG3 downregulation was found to be capable of inhibiting Girdin but enhancing miR-128 expression. It was also indicated to inhibit platelet phagocytosis and reduce telomerase activity and telomere length, while enhancing lipoprotein oxidation and reactive oxygen species production, which ultimately contributed in preventing and protecting HUEVCs from senescence. These findings provide evidence supporting that MEG3 leads to miR-128 downregulation and Girdin upregulation, which promotes platelet phagocytosis, thus protecting VECs from senescence.
Collapse
Affiliation(s)
- Yong Lan
- National Center of Gerontology, Department of Vascular Surgery, Beijing Hospital , Beijing , People's Republic of China
| | - Yong-Jun Li
- National Center of Gerontology, Department of Vascular Surgery, Beijing Hospital , Beijing , People's Republic of China
| | - Da-Jun Li
- National Center of Gerontology, Department of Vascular Surgery, Beijing Hospital , Beijing , People's Republic of China
| | - Peng Li
- National Center of Gerontology, Department of Vascular Surgery, Beijing Hospital , Beijing , People's Republic of China
| | - Ji-Yang Wang
- National Center of Gerontology, Department of Vascular Surgery, Beijing Hospital , Beijing , People's Republic of China
| | - Yong-Peng Diao
- National Center of Gerontology, Department of Vascular Surgery, Beijing Hospital , Beijing , People's Republic of China
| | - Guo-Dong Ye
- National Center of Gerontology, Department of Vascular Surgery, Beijing Hospital , Beijing , People's Republic of China
| | - Yang-Fang Li
- Beijing Neurosurgical Institute, Capital Medical University , Beijing , People's Republic of China
| |
Collapse
|
244
|
Zhu ZD, Ye JM, Fu XM, Wang XC, Ye JY, Wu XR, Hua P, Liao YQ, Xuan W, Duan JL, Li WY, Fu H, Xia ZH, Zhang X. DDAH2 alleviates myocardial fibrosis in diabetic cardiomyopathy through activation of the DDAH/ADMA/NOS/NO pathway in rats. Int J Mol Med 2018; 43:749-760. [PMID: 30569164 PMCID: PMC6317674 DOI: 10.3892/ijmm.2018.4034] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 11/27/2018] [Indexed: 01/02/2023] Open
Abstract
Diabetic cardiomyopathy (DCM) is a form of idiopathic heart disease, with signs including hypertrophy of myocardial cells, hypertension‑independent fibrosis and coronary artery disease. Considering the involvement of dimethylarginine dimethylaminohydrolase 2 (DDAH2) in diabetes, it was hypothesized that DDAH2 may be beneficial to cardiac function and myocardial fibrosis during the progression of DCM with involvement of the DDAH/asymmetric NG, NGdimethyl‑L‑arginine (ADMA)/nitric oxide synthase (NOS)/nitric oxide (NO) signaling pathway. Following establishment of diabetic rat models, diabetes‑related blood biochemical indices and cardiac function were measured in diabetic rats treated with lentivirus expressing DDAH2, short hairpin RNA against DDAH2, or L‑NNA (inhibitor of NOS) to identify the roles of DDAH2 in DCM. The functional roles of DDAH2 in DCM were further determined through detection of the levels of collagen I, matrix metalloproteinase 2 (MMP2) and tissue inhibitor of metalloproteinase 2 (TIMP2). The H9C2 myocardial cell line was selected for in vitro experiments. The effects of DDAH2 on the migration of myocardial cells under high glucose conditions were also examined. To further investigate the underlying regulatory mechanism of DDAH2 in DCM, the contents of ADMA and NO, and the activities of DDAH and NOS were observed. The DCM model rats treated with DDAH2 exhibited reduced left ventricular end‑diastolic pressure, and decreased blood glucose, total cholesterol, triglyceride, fasting blood glucose, and fasting insulin levels, but exhibited increased left ventricular systolic pressure and maximum rate of left ventricular pressure rise/fall levels in myocardial tissues. Myocardial cells under high glucose conditions treated with DDAH2 showed reductions in collagen I, MMP2 and TIMP2, indicating that DDAH2 reduced cell migration. Decreased levels of ADMA and NO but increased levels of DDAH and NOS were observed following treatment with DDAH2, indicating that the DDAH/ADMA/NOS/NO pathway was activated. These results reveal that the overexpression of DDAH2 attenuates myocardial fibrosis and protects against DCM through activation of the DDAH/ADMA/NOS/NO pathway in DCM rats. These results indicate that DDAH2 is a potential therapeutic candidate for the treatment of DCM.
Collapse
Affiliation(s)
- Zhen-Dong Zhu
- Yunnan Research Center for Geriatric Diseases, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming Science and Technology University, Kunming, Yunnan 650032, P.R. China
| | - Ji-Ming Ye
- Department of Pharmacy, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming Science and Technology University, Kunming, Yunnan 650032, P.R. China
| | - Xue-Mei Fu
- Department of Geriatrics, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming Science and Technology University, Kunming, Yunnan 650032, P.R. China
| | - Xue-Chang Wang
- Department of Pharmacy, the Third People's Hospital of Yunnan Province, The Second Affiliated Hospital of Dali University, Kunming, Yunnan 650011, P.R. China
| | - Ji-Yun Ye
- Pathogenic Organisms Department of Experimental Center, School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Xin-Ran Wu
- Center Laboratory, The Third People's Hospital of Yunnan Province, The Second Affiliated Hospital of Dali University, Kunming, Yunnan 650011, P.R. China
| | - Peng Hua
- Department of Pharmacy, the Third People's Hospital of Yunnan Province, The Second Affiliated Hospital of Dali University, Kunming, Yunnan 650011, P.R. China
| | - Yu-Qiong Liao
- Department of Geriatrics, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming Science and Technology University, Kunming, Yunnan 650032, P.R. China
| | - Wei Xuan
- Department of Geriatrics, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming Science and Technology University, Kunming, Yunnan 650032, P.R. China
| | - Jin-Lan Duan
- Department of Geriatrics, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming Science and Technology University, Kunming, Yunnan 650032, P.R. China
| | - Wei-Yuan Li
- Department of Geriatrics, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming Science and Technology University, Kunming, Yunnan 650032, P.R. China
| | - Hui Fu
- Clinic Laboratory, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming Science and Technology University, Kunming, Yunnan 650032, P.R. China
| | - Zhong-Hua Xia
- Clinical Medical College of Dali University, Dali, Yunnan 671003, P.R. China
| | - Xuan Zhang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
245
|
Ye CY, Zheng CP, Ying WW, Weng SS. Up-regulation of microRNA-497 inhibits the proliferation, migration and invasion but increases the apoptosis of multiple myeloma cells through the MAPK/ERK signaling pathway by targeting Raf-1. Cell Cycle 2018; 17:2666-2683. [PMID: 30382763 PMCID: PMC6343711 DOI: 10.1080/15384101.2018.1542895] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Multiple myeloma (MM) is a cancer that occurs in plasma cells, which fall under the category of white blood cells that are in charge of antibody production. According to previous studies, microRNA-497 (miR-497) functions as a tumor suppressor in several types of cancer, including gastric cancer and colorectal cancer. Therefore, the present study aims to investigate the effects of miR-497 on cellular function of human MM cells through the mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) signaling pathway by targeting Raf-1. The differentially expressed genes and miRs in MM, and the relationship between the miR and gene were verified. It was found that Raf-1 was a target gene of miR-497. The data obtained from MM tissues showed increased Raf-1 level and decreased miR-497 level. MM cells were treated with mimic, inhibitor and siRNA in order to evaluate the role of miR-497, Raf-1 and MAPK/ERK in MM. The expression pattern of miR-497, Raf-1, ERK1/2, survivin, B-cell lymphoma-2 (Bcl-2) and BCL2-Associated X (Bax) as well as the extent of ERK1/2 phosphorylation were determined. Retored miR-497 and si-Raf-1 resulted in increases in the Bax expression and cell apoptosis and decreases in the expressions of Raf-1, MEK-2, survivin, Bcl-2, along with the extent of ERK1/2 phosphorylation. In addition, the biological function evaluations of MM cells revealed that miR-497 mimic or si-Raf-1 led to suppression in cell proliferation, invasion and migration. In conclusion, our results have demonstrated that miR-497 targets Raf-1 in order to inhibit the progression of MM by blocking the MAPK/ERK signaling pathway.
Collapse
Affiliation(s)
- Cheng-Yu Ye
- a Department of Hematologic Oncology , Wenzhou Central Hospital, Dingli Clinical Medical School of Wenzhou Medical University , Wenzhou , P.R. China
| | - Cui-Ping Zheng
- a Department of Hematologic Oncology , Wenzhou Central Hospital, Dingli Clinical Medical School of Wenzhou Medical University , Wenzhou , P.R. China
| | - Wei-Wei Ying
- b Wenzhou Medical University , Wenzhou , P.R. China
| | - Shan-Shan Weng
- a Department of Hematologic Oncology , Wenzhou Central Hospital, Dingli Clinical Medical School of Wenzhou Medical University , Wenzhou , P.R. China
| |
Collapse
|
246
|
Dong Y, Han LL, Xu ZX. Suppressed microRNA-96 inhibits iNOS expression and dopaminergic neuron apoptosis through inactivating the MAPK signaling pathway by targeting CACNG5 in mice with Parkinson's disease. Mol Med 2018; 24:61. [PMID: 30486773 PMCID: PMC6263543 DOI: 10.1186/s10020-018-0059-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 11/04/2018] [Indexed: 12/15/2022] Open
Abstract
Background There have been a number of reports implicating the association of microRNAs (miRs) and the MAPK signaling pathway with the dopaminergic neuron, which is involved in the development of Parkinson’s disease (PD). The present study was conducted with aims of exploring the role of miR-96 in the activation of iNOS and apoptosis of dopaminergic neuron through the MAPK signaling pathway in mice with PD. Methods The miR and the differentially expressed gene in PD were screened out and the relationship between them was verified. A mouse model of PD induced by MPTP and was then constructed and treated with miR-96 mimic/inhibitor and CACNG5 overexpression plasmid to extract nigral dopaminergic neuron for the purpose of detecting the effect of miR-96 on PD. The TH and iNOS positive neuronal cells, the apoptotic neuronal cells by TUNEL staining, and expression of miR-96, CACNG5, iNOS, p38MAPK, p-p38MAPK, c-Fos, Bax, and Bcl-2 in substantia nigra dopaminergic neuronal tissues were evaluated. Results The results obtained from the aforementioned procedure were then verified by cell culture of the SH-SY5Y cells, followed by treatment with miR-96 mimic/inhibitor, CACNG5 overexpression plasmid and the inhibitor of the MAPK signaling pathway. CACNG5 was confirmed as a target gene of miR-96. The inhibition of miR-96 resulted in a substantial increase in nigral cells, TH positive cells and expression of CACNG5 and Bcl-2 in nigral dopaminergic neuronal tissues, and a decrease in iNOS positive cells, apoptotic neuronal cells, and expression of iNOS, p38MAPK, p-p38MAPK, c-Fos, and Bax. Conclusion The above results implicated that the downregulation of miR-96 inhibits the activation of iNOS and apoptosis of dopaminergic neuron through the blockade of the MAPK signaling pathway by promoting CACNG5 in mice with PD.
Collapse
Affiliation(s)
- Yue Dong
- Department of Neurology , China-Japan Union Hospital, Jilin University, No. 126, Xiantai Street, Erdao District, Changchun, 130012, Jilin Province, People's Republic of China
| | - Li-Li Han
- Department of Neurology, Cangzhou Central Hospital, Cangzhou, 061000, People's Republic of China
| | - Zhong-Xin Xu
- Department of Neurology , China-Japan Union Hospital, Jilin University, No. 126, Xiantai Street, Erdao District, Changchun, 130012, Jilin Province, People's Republic of China.
| |
Collapse
|
247
|
Liu HC, Zeng J, Zhang B, Liu XQ, Dai M. Inhibitory effect of MSH6 gene silencing in combination with cisplatin on cell proliferation of human osteosarcoma cell line MG63. J Cell Physiol 2018; 234:9358-9369. [PMID: 30456894 DOI: 10.1002/jcp.27620] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 09/25/2018] [Indexed: 12/23/2022]
Abstract
Osteosarcoma (OS) is one of the most common primary bone malignancies, with the survival rate of patients with OS remaining low. Therefore, we conducted this study to identify the potential role combination of both MSH6 gene silencing and cisplatin (DDP) plays in OS cell proliferation and apoptosis. Microarray-based gene expression profiling was used to identify the differentially expressed genes (DEGs) in patients with OS, as well as microRNAs (miRNAs) that regulate the candidate gene. OS tissues from 67 patients with OS along with normal tissues from 24 amputee patients were collected for detection of the positive expression of mutS homolog 6 (MSH6) protein, mRNA, and protein expressions of c-myc, cyclin D1, l-2, B-cell lymphoma 2 (Bcl-2), Stathmin, proliferating cell nuclear antigen (PCNA), and Bcl-2-associated X (Bax). Moreover, after MSH6 silencing and DDP were treated on the selected human OS cell line MG63 with the highest expression of MSH6, cell viability, cell cycle distribution, and apoptosis were detected. The microarray analysis showed that MSH6 was upregulated in OS chip data. Furthermore, silencing MSH6 combined with DDP reduced expressions of c-myc, cyclin D1, Bcl-2, Stathmin, and PCNA, and elevated Bax expression, whereas inhibiting OS cell viability, impeding cell cycle distribution, and inducing apoptosis. In conclusion, our preliminary results indicated that the combination of MSH6 gene silencing coupled with DDP may have a better effect on the inhibition of OS cell proliferation and promote apoptosis, potentially providing targets for the OS treatment.
Collapse
Affiliation(s)
- Hu-Cheng Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jin Zeng
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Bin Zhang
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xu-Qiang Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Min Dai
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
248
|
Zhou WM, Wu GL, Huang J, Li JG, Hao C, He QM, Chen XD, Wang GX, Tu XH. Low expression of PDK1 inhibits renal cell carcinoma cell proliferation, migration, invasion and epithelial mesenchymal transition through inhibition of the PI3K-PDK1-Akt pathway. Cell Signal 2018; 56:1-14. [PMID: 30465826 DOI: 10.1016/j.cellsig.2018.11.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 11/16/2018] [Accepted: 11/19/2018] [Indexed: 11/18/2022]
Abstract
As the most commonly occurring form of primary renal tumor, renal cell carcinoma (RCC) is a malignancy accompanied by a high mortality rate. 3-phosphoinositide-dependent protein kinase 1 (PDK1) has been established as a protein target and generated considerable interest in both the pharmaceutical and academia industry. The aim of the current study was to investigate the effect of si-PDK1 on the RCC cell apoptosis, proliferation, migration, invasion and epithelial mesenchymal transition (EMT) in connection with the PI3K-PDK1-Akt pathway. Microarray analysis from the GEO database was adopted to identify differentially expressed genes (DEGs) related to RCC, after which the positive expression of the PDK1 protein in tissue was determined accordingly. The optimal silencing si-RNA was subsequently selected and RCC cell lines 786-O and A498 were selected and transfected with either a si-PDK1 or activator of the PI3K-PDK1-Akt pathway for grouping purposes. The mRNA and protein expressions of PDK1, the PI3K-PDK1-Akt pathway-, EMT- and apoptosis-related genes were then evaluated. The effect of si-PDK1 on cell proliferation, apoptosis, invasion and migration was then analyzed. Through microarray analysis of GSE6344, GSE53757, GSE14762 and GSE781, PDK1 was examined. PDK1 was determined to be highly expressed in RCC tissues. Si-PDK1 exhibited marked reductions in relation to the mRNA and protein expression of PDK1, PI3K, AKT as well as Vimentin while elevated mRNA and protein expressions of E-cadherin were detected, which ultimately suggested that cell migration, proliferation and invasion had been inhibited coupled with enhanced levels of cell apoptosis. While a notable observation was made highlighting that the PI3K-PDK1-Akt pathway antagonized the effect of PDK1 silencing. Taken together, the key observations of this study provide evidence suggesting that high expressions of PDK1 are found in RCC, while highlighting that silencing PDK1 could inhibit RCC cell proliferation, migration, invasion and EMT by repressing the PI3K-PDK1-Akt pathway.
Collapse
Affiliation(s)
- Wei-Min Zhou
- Jiangxi Medical College, Nanchang University, Nanchang 330006, PR China; Department of Urology, Jiangxi Cancer Hospital, Nanchang 330029, PR China
| | - Gao-Liang Wu
- Department of Urology, Jiangxi Cancer Hospital, Nanchang 330029, PR China
| | - Ji Huang
- Department of Urology, Jiangxi Cancer Hospital, Nanchang 330029, PR China
| | - Jin-Gao Li
- Department of Radiotherapy, Jiangxi Cancer Hospital, Nanchang 330029, PR China
| | - Chao Hao
- Department of Urology, Jiangxi Cancer Hospital, Nanchang 330029, PR China
| | - Qiu-Ming He
- Department of Urology, Jiangxi Cancer Hospital, Nanchang 330029, PR China
| | - Xiao-Dan Chen
- Department of Science and Education, Jiangxi Cancer Hospital, Nanchang 330029, PR China
| | - Gong-Xian Wang
- Jiangxi Medical College, Nanchang University, Nanchang 330006, PR China; Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, PR China.
| | - Xin-Hua Tu
- Department of Urology, Jiangxi Cancer Hospital, Nanchang 330029, PR China.
| |
Collapse
|
249
|
Gao JX, Li Y, Wang SN, Chen XC, Lin LL, Zhang H. Overexpression of microRNA-183 promotes apoptosis of substantia nigra neurons via the inhibition of OSMR in a mouse model of Parkinson's disease. Int J Mol Med 2018; 43:209-220. [PMID: 30431059 PMCID: PMC6257840 DOI: 10.3892/ijmm.2018.3982] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 10/24/2018] [Indexed: 12/13/2022] Open
Abstract
The present study aimed to investigate the effect of microRNA-183 (miR-183) on substantia nigra neurons by targeting oncostatin M receptor (OSMR) in a mouse model of Parkinson’s disease (PD). The positive expression rates of OSMR and the apoptosis of substantia nigra neurons were detected by immunohistochemistry and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end-labeling, respectively. Substantia nigra neurons in normal and PD mice were cultured in vitro. The association between miR-183 and OSMR was verified using a dual luciferase reporter gene assay. The expression of miR-183 and the phosphoinositide 3-kinase-Akt signaling pathway-associated genes were detected by reverse transcription-quantitative polymerase chain reaction and western blot analysis, respectively. Cell apoptosis was detected by flow cytometry. OSMR is the target gene of miR-183. The number of OSMR-positive cells and the apoptotic rate of substantia nigra neurons were increased in the PD group. Neurons transfected with miR-183 mimic exhibited elevated expression levels of miR-183, B-cell lymphoma 2 (Bcl-2)-associated X protein (Bax) and caspase-9 and increased apoptotic rate, and reduced expression levels of OSMR, Akt, phosphorylated (p-)Akt, glycogen synthase kinase-3 (GSK-3β), p-GSK-3β, Bcl-2, insulin-like growth factor 1 (IGF-1), mammalian target of rapamycin (mTOR) and p-mTOR. The miR-183 inhibitor decreased the expression levels of miR-183, Bax and caspase-9 and the apoptotic rate; however, increased the expression of OSMR, Akt, p-Akt, GSK-3β, p-GSK-3β, Bcl-2, IGF-1, mTOR and p-mTOR. The results of the present study provide evidence that the overexpression of miR-183 promotes the apoptosis of substantia nigra neurons by inhibiting the expression of OSMR.
Collapse
Affiliation(s)
- Jin-Xia Gao
- Department of Anesthesiology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116027, P.R. China
| | - Yu Li
- Department of Neurology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116027, P.R. China
| | - Sai-Nan Wang
- Department of Neurology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116027, P.R. China
| | - Xing-Chi Chen
- Department of Neurology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116027, P.R. China
| | - Lu-Lu Lin
- Department of Neurology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116027, P.R. China
| | - Hui Zhang
- Department of Neurology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116027, P.R. China
| |
Collapse
|
250
|
Huang YX, Nie XG, Li GD, Fan DS, Song LL, Zhang XL. Downregulation of microRNA‑182 inhibits cell viability, invasion and angiogenesis in retinoblastoma through inhibition of the PI3K/AKT pathway and CADM2 upregulation. Int J Oncol 2018; 53:2615-2626. [PMID: 30320366 DOI: 10.3892/ijo.2018.4587] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 07/02/2018] [Indexed: 11/06/2022] Open
Abstract
Retinoblastoma (RB) is a well‑vascularized tumor dependent on angiogenesis. The present study aimed to explore whether microRNA (miR)‑182 regulates cell viability, invasion and angiogenesis in RB via the phosphatidylinositol‑3‑OH kinase (PI3K)/protein kinase B (AKT) signaling pathway and by targeting cell adhesion molecule 2 (CADM2). The expression levels of miR‑182 and CADM2 were initially detected in RB tissues from patients with RB who underwent ophthalmectomy, and normal retinal tissues collected from other trauma patients who underwent eye enucleation. To determine whether CADM2 was targeted by miR‑182, a dual luciferase reporter assay was conducted. Subsequently, Y79 and WERI‑Rb‑1 RB cells were transfected with a miR‑182 mimic or miR‑182 inhibitor, or small interfering RNA against CADM2, in order to investigate the effects of miR‑182 on viability and invasion, which were detected using MTT and Transwell assays, respectively. In addition, to determine whether the regulatory mechanism underlying the effects of miR‑182 was associated with the PI3K/AKT signaling pathway, the expression levels of associated genes were detected by reverse transcription‑quantitative polymerase chain reaction and western blot analysis. A xenograft tumor model in nude mice was also established, in order to evaluate the effects of miR‑182 on tumor growth and angiogenesis. The results indicated that miR‑182 expression was increased and CADM2 expression was reduced in RB tissues; CADM2 was confirmed to be targeted and negatively regulated by miR‑182. When the expression of miR‑182 was downregulated, cell viability, invasion, tumor volume and angiogenesis were significantly decreased. Furthermore, the expression levels of PI3K/AKT signaling pathway‑associated genes were increased in response to miR‑182 overexpression or CADM2 silencing. Taken together, these results suggested that inhibition of miR‑182 may suppress cell viability, invasion and angiogenesis in RB through inactivation of the PI3K/AKT pathway and CADM2 upregulation. This mechanism may reveal a novel potential therapeutic target.
Collapse
Affiliation(s)
- Yan-Xia Huang
- Department of Ophthalmology, Luoyang Central Hospital, Luoyang, Henan 471009, P.R. China
| | - Xin-Gang Nie
- Department of Ophthalmology, Luoyang Central Hospital, Luoyang, Henan 471009, P.R. China
| | - Guang-Da Li
- Department of Ophthalmology, Linyi People's Hospital, Linyi, Shandong 276003, P.R. China
| | - Dong-Sheng Fan
- Department of Ophthalmology, Luoyang Central Hospital, Luoyang, Henan 471009, P.R. China
| | - Li-Li Song
- Department of Ophthalmology, Luoyang Central Hospital, Luoyang, Henan 471009, P.R. China
| | - Xin-Lin Zhang
- Department of Ophthalmology, Luoyang Central Hospital, Luoyang, Henan 471009, P.R. China
| |
Collapse
|