1
|
Wang J, Ke N, Wu X, Zhen H, Hu J, Liu X, Li S, Zhao F, Li M, Shi B, Zhao Z, Ren C, Hao Z. MicroRNA-148a Targets DNMT1 and PPARGC1A to Regulate the Viability, Proliferation, and Milk Fat Synthesis of Ovine Mammary Epithelial Cells. Int J Mol Sci 2024; 25:8558. [PMID: 39201245 PMCID: PMC11354201 DOI: 10.3390/ijms25168558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 09/02/2024] Open
Abstract
In this study, the expression profiles of miR-148a were constructed in eight different ovine tissues, including mammary gland tissue, during six different developmental periods. The effect of miR-148a on the viability, proliferation, and milk fat synthesis of ovine mammary epithelial cells (OMECs) was investigated, and the target relationship of miR-148a with two predicted target genes was verified. The expression of miR-148a exhibited obvious tissue-specific and temporal-specific patterns. miR-148a was expressed in all eight ovine tissues investigated, with the highest expression level in mammary gland tissue (p < 0.05). Additionally, miR-148a was expressed in ovine mammary gland tissue during each of the six developmental periods studied, with its highest level at peak lactation (p < 0.05). The overexpression of miR-148a increased the viability of OMECs, the number and percentage of Edu-labeled positive OMECs, and the expression levels of two cell-proliferation marker genes. miR-148a also increased the percentage of OMECs in the S phase. In contrast, transfection with an miR-148a inhibitor produced the opposite effect compared to the miR-148a mimic. These results indicate that miR-148a promotes the viability and proliferation of OMECs in Small-tailed Han sheep. The miR-148a mimic increased the triglyceride content by 37.78% (p < 0.01) and the expression levels of three milk fat synthesis marker genes in OMECs. However, the miR-148a inhibitor reduced the triglyceride level by 87.11% (p < 0.01). These results suggest that miR-148a promotes milk fat synthesis in OMECs. The dual-luciferase reporter assay showed that miR-148a reduced the luciferase activities of DNA methyltransferase 1 (DNMT1) and peroxisome proliferator-activated receptor gamma coactivator 1-A (PPARGC1A) in wild-type vectors, suggesting that they are target genes of miR-148a. The expression of miR-148a was highly negatively correlated with PPARGC1A (r = -0.789, p < 0.001) in ovine mammary gland tissue, while it had a moderate negative correlation with DNMT1 (r = -0.515, p = 0.029). This is the first study to reveal the molecular mechanisms of miR-148a underlying the viability, proliferation, and milk fat synthesis of OMECs in sheep.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Zhiyun Hao
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (J.W.); (N.K.); (X.W.); (H.Z.); (J.H.); (X.L.); (S.L.); (F.Z.); (M.L.); (B.S.); (Z.Z.); (C.R.)
| |
Collapse
|
2
|
Marzoog BA. Transcription Factors - the Essence of Heart Regeneration: A Potential Novel Therapeutic Strategy. Curr Mol Med 2023; 23:232-238. [PMID: 35170408 DOI: 10.2174/1566524022666220216123650] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 12/02/2021] [Accepted: 12/07/2021] [Indexed: 02/08/2023]
Abstract
Myocardial cell injury and following sequelae are the primary reasons for death globally. Unfortunately, myocardiocytes in adults have limited regeneration capacity. Therefore, the generation of neo myocardiocytes from non-myocardial cells is a surrogate strategy. Transcription factors (TFs) can be recruited to achieve this tremendous goal. Transcriptomic analyses have suggested that GATA, Mef2c, and Tbx5 (GMT cocktail) are master TFs to transdifferentiate/reprogram cell linage of fibroblasts, somatic cells, mesodermal cells into myocardiocytes. However, adding MESP1, MYOCD, ESRRG, and ZFPM2 TFs induces the generation of more efficient and physiomorphological features for induced myocardiocytes. Moreover, the same cocktail of transcription factors can induce the proliferation and differentiation of induced/pluripotent stem cells into myocardial cells. Amelioration of impaired myocardial cells involves the activation of healing transcription factors, which are induced by inflammation mediators; IL6, tumor growth factor β, and IL22. Transcription factors regulate the cellular and subcellular physiology of myocardiocytes to include mitotic cell cycling regulation, karyokinesis and cytokinesis, hypertrophic growth, adult sarcomeric contractile protein gene expression, fatty acid metabolism, and mitochondrial biogenesis and maturation. Cell therapy by transcription factors can be applied to cardiogenesis and ameliorating impaired cardiocytes. Transcription factors are the cornerstone in cell differentiation.
Collapse
Affiliation(s)
- Basheer Abdullah Marzoog
- Department of Normal and Pathological Physiology, National Research Mordovia State University, Bolshevitskaya Street, 68, Saransk, Rep. Mordovia, 430005, Russia
| |
Collapse
|
3
|
Smurf2-induced degradation of SMAD2 causes inhibition of hair follicle stem cell differentiation. Cell Death Dis 2022; 8:160. [PMID: 35379779 PMCID: PMC8980066 DOI: 10.1038/s41420-022-00920-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 02/15/2022] [Accepted: 02/24/2022] [Indexed: 11/08/2022]
Abstract
Hair follicle stem cells (HFSCs) are implicated in the formation of hair follicles and epidermis. This study aims to clarify the role of SMAD2 in regulating the differentiation of HFSCs, which is involved with Smurf2. Functional assays were carried out in human HFSCs to assess the effect of SMAD2 and Smurf2 with altered expression on growth dynamics of HFSCs. Ubiquitination of SMAD2 and its protein stability were assessed. The binding relationship between NANOG and DNMT1 was assessed. A mouse skin wound model was induced to verify the effects of Smurf2/SMAD2/NANOG/DNMT1 on wound healing. SMAD2 overexpression was observed in HFSCs during differentiation and its ectopic expression contributed to promotion of differentiation and apoptosis of HFSCs while arresting cell proliferation. Mechanistic investigations indicated that Smurf2 promoted the ubiquitination and degradation of SMAD2, thus causing downregulation of SMAD2 expression. By this mechanism, NANOG expression was reduced and the subsequent DNMT1 transcriptional expression was also diminished, leading to suppression of differentiation and apoptosis of HFSCs while stimulating cell proliferation. Moreover, in vivo data showed that Smurf2 upregulation limited epidermal wound healing in mice by inhibiting the SMAD2/NANOG/DNMT1 axis. Our work proposed a potential target regarding SMAD2 restoration in promoting HFSC differentiation and skin wound healing.
Collapse
|
4
|
Silvestro S, Gugliandolo A, Chiricosta L, Diomede F, Trubiani O, Bramanti P, Pizzicannella J, Mazzon E. MicroRNA Profiling of HL-1 Cardiac Cells-Derived Extracellular Vesicles. Cells 2021; 10:cells10020273. [PMID: 33573156 PMCID: PMC7912193 DOI: 10.3390/cells10020273] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 01/19/2021] [Accepted: 01/27/2021] [Indexed: 11/20/2022] Open
Abstract
HL-1 is a cell line that shows a phenotype similar to adult cardiomyocytes. All major cardiac cell types release extracellular vesicles (EVs) that emerge as key mediators of intercellular communication. EVs can mediate intercellular cross-talk through the transfer of specific microRNAs (miRNAs). MiRNAs are known to play important regulatory roles during tissue differentiation and regeneration processes. Furthermore, miRNAs have recently been shown to be involved in the proliferation of adult cardiomyocytes. In this context, the purpose of this study was to analyze the transcriptomic profile of miRNAs expressed from HL-1 cardiac muscle cell-derived EVs, using next generation sequencing (NGS). Specifically, our transcriptomic analysis showed that the EVs derived from our HL-1 cells contained miRNAs that induce blood vessel formation and increase cell proliferation. Indeed, our bioinformatics analysis revealed 26 miRNAs expressed in EVs derived from our HL-1 that target genes related to cardiovascular development. In particular, their targets are enriched for the following biological processes related to cardiovascular development: heart morphogenesis, positive regulation of angiogenesis, artery development, ventricular septum development, cardiac atrium development, and myoblast differentiation. Consequently, EVs could become important in the field of regenerative medicine.
Collapse
Affiliation(s)
- Serena Silvestro
- IRCCS Centro Neurolesi “Bonino-Pulejo”, 98124 Messina, Italy; (S.S.); (A.G.); (L.C.); (P.B.)
| | - Agnese Gugliandolo
- IRCCS Centro Neurolesi “Bonino-Pulejo”, 98124 Messina, Italy; (S.S.); (A.G.); (L.C.); (P.B.)
| | - Luigi Chiricosta
- IRCCS Centro Neurolesi “Bonino-Pulejo”, 98124 Messina, Italy; (S.S.); (A.G.); (L.C.); (P.B.)
| | - Francesca Diomede
- Department of Innovative Technologies in Medicine & Dentistry, University “G. d’Annunzio”, Chieti-Pescara, Via dei Vestini, 31, 66100 Chieti, Italy; (F.D.); (O.T.)
| | - Oriana Trubiani
- Department of Innovative Technologies in Medicine & Dentistry, University “G. d’Annunzio”, Chieti-Pescara, Via dei Vestini, 31, 66100 Chieti, Italy; (F.D.); (O.T.)
| | - Placido Bramanti
- IRCCS Centro Neurolesi “Bonino-Pulejo”, 98124 Messina, Italy; (S.S.); (A.G.); (L.C.); (P.B.)
| | | | - Emanuela Mazzon
- IRCCS Centro Neurolesi “Bonino-Pulejo”, 98124 Messina, Italy; (S.S.); (A.G.); (L.C.); (P.B.)
- Correspondence: ; Tel.: +39-090-6012-8172
| |
Collapse
|
5
|
Myocyte-specific enhancer factor 2c triggers transdifferentiation of adipose tissue-derived stromal cells into spontaneously beating cardiomyocyte-like cells. Sci Rep 2021; 11:1520. [PMID: 33452355 PMCID: PMC7810870 DOI: 10.1038/s41598-020-80848-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 12/29/2020] [Indexed: 01/10/2023] Open
Abstract
Cardiomyocyte regeneration is limited in adults. The adipose tissue-derived stromal vascular fraction (Ad-SVF) contains pluripotent stem cells that rarely transdifferentiate into spontaneously beating cardiomyocyte-like cells (beating CMs). However, the characteristics of beating CMs and the factors that regulate the differentiation of Ad-SVF toward the cardiac lineage are unknown. We developed a simple culture protocol under which the adult murine inguinal Ad-SVF reproducibly transdifferentiates into beating CMs without induction. The beating CMs showed the striated ventricular phenotype of cardiomyocytes and synchronised oscillation of the intracellular calcium concentration among cells on day 28 of Ad-SVF primary culture. We also identified beating CM-fated progenitors (CFPs) and performed single-cell transcriptome analysis of these CFPs. Among 491 transcription factors that were differentially expressed (≥ 1.75-fold) in CFPs and the beating CMs, myocyte-specific enhancer 2c (Mef2c) was key. Transduction of Ad-SVF cells with Mef2c using a lentiviral vector yielded CFPs and beating CMs with ~ tenfold higher cardiac troponin T expression, which was abolished by silencing of Mef2c. Thus, we identified the master gene required for transdifferentiation of Ad-SVF into beating CMs. These findings will facilitate the development of novel cardiac regeneration therapies based on gene-modified, cardiac lineage-directed Ad-SVF cells.
Collapse
|
6
|
Wang F, Zhang H, Wang C. MiR-590-3p regulates cardiomyocyte P19CL6 proliferation, apoptosis and differentiation in vitro by targeting PTPN1 via JNK/STAT/NF-kB pathway. Int J Exp Pathol 2020; 101:196-202. [PMID: 33058302 DOI: 10.1111/iep.12377] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/26/2020] [Accepted: 08/27/2020] [Indexed: 12/17/2022] Open
Abstract
Cardiomyocyte differentiation is a multi-step process which involves a number of signalling pathways. microRNAs exhibit regulatory functions in various diseases and are involved in the signalling pathways in multiple physiological processes, but the specific functions of particular mRNAs is often not fully understood. of an example of this is that the role of miR-590-3p in the differentiation of cardiomyocytes remains unclear. In the current study, RT-qPCR was used to determine the expression of miR-590-3p in cardiomyocytes differentiated from the embryonic carcinoma cell line P19CL6. MTT, EdU, caspase-3 activity and flow cytometry assays were performed to examine the influence of miR-590-3p on cell behaviour. A luciferase assay was used to confirm binding between miR-590-3p and PTPN1. Western blotting was used to determine the relationship between the JNK/STAT/NF-kB pathway and PTPN1. The results inferred that miR-590-3p became heavily expressed in differentiated P19CL6. Knockdown miR-590-3p suppressed the cell proliferation while at the same time, accelerated apoptosis. Moreover, PTPN1 was identified as the target of miR-590-3p. More importantly, PTPN1 overexpression activated the JNK/STAT/NF-kB pathway and limited the differentiation of P19CL6. Thus the conclusions from this study are that miR-590-3p has the potential to regulate the proliferation, apoptosis and differentiation of cardiomyocyte P19CL6 in vitro by targeting PTPN1 via the JNK/STAT/NF-kB pathway.
Collapse
Affiliation(s)
- Fanshun Wang
- Department of Cardiac Surgery, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Hongqiang Zhang
- Department of Cardiac Surgery, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Chunsheng Wang
- Department of Cardiac Surgery, Zhongshan Hospital of Fudan University, Shanghai, China
| |
Collapse
|
7
|
Liu J, Wu M, Feng G, Li R, Wang Y, Jiao J. Downregulation of LINC00707 promotes osteogenic differentiation of human bone marrow‑derived mesenchymal stem cells by regulating DKK1 via targeting miR‑103a‑3p. Int J Mol Med 2020; 46:1029-1038. [PMID: 32705245 PMCID: PMC7387089 DOI: 10.3892/ijmm.2020.4672] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 05/21/2020] [Indexed: 12/17/2022] Open
Abstract
Human bone marrow-derived mesenchymal stem cells (HBMSCs) have the potential of multidirectional differentiation and self-renewal, which is important for the formation of human bone. It has been reported that long non-coding RNAs (lncRNAs) serve important roles in HBMSC osteogenic differentiation. The current study aimed to investigate the roles of long intergenic non-protein coding RNA 00707 (LINC00707) and microRNA (miR)-103a-3p in the osteogenic differentiation of HBMSCs. Reverse transcription-quantitative PCR (RT-qPCR) was performed to detect the expression levels of LINC00707, miR-103a-3p and osteogenesis-related genes (Alkaline phosphatase, osteocalcin, osteopontin and RUNX family transcription factor 2) in HBMSCs cultured in proliferation medium (PM) and osteogenic medium (OM). Mineralized matrix deposition was measured using Alizarin Red S staining. The protein expression levels of osteogenesis-related genes were detected by western blotting. The relationships between LINC00707, miR-103a-3p and dickkopf WNT signaling pathway inhibitor 1 (DKK1) were predicted using Starbase and TargetScan7.2, and were further assessed with a dual-luciferase reporter assay. After 21 days of cell culture, the results indicated that expression of LINC00707 was downregulated, and those of miR-103a-3p and osteogenesis-related genes were upregulated in OM-cultured HBMSCs. However, there was no significant difference in the aforementioned gene expression levels in PM-cultured HBMSCs. Small interfering (si)LINC00707 increased the deposition of mineralized matrix and promoted the expression levels of osteogenesis-related proteins. Furthermore, miR-103a-3p was predicted to be a target gene of LINC00707, its expression was significantly upregulated by siLINC00707, while overexpression of miR-103a-3p increased the expression levels of osteogenesis-related proteins. DKK1 was also predicted to be a target gene of miR-103a-3p and could inhibit the expression levels of osteogenesis-related proteins, but such effect of DKK1 could be reversed by the miR-103a-3p mimic. In conclusion, the present results suggested that LINC00707 regulated DKK1 expression by targeting miR-103a-3p to regulate osteogenic differentiation.
Collapse
Affiliation(s)
- Jun Liu
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Minfei Wu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Guang Feng
- The Fourth Medical Center of PLA General Hospital, Beijing 100048, P.R. China
| | - Rui Li
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Yang Wang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Jianhang Jiao
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| |
Collapse
|
8
|
Dai Y, Wang S, Chang S, Ren D, Shali S, Li C, Yang H, Huang Z, Ge J. M2 macrophage-derived exosomes carry microRNA-148a to alleviate myocardial ischemia/reperfusion injury via inhibiting TXNIP and the TLR4/NF-κB/NLRP3 inflammasome signaling pathway. J Mol Cell Cardiol 2020; 142:65-79. [PMID: 32087217 DOI: 10.1016/j.yjmcc.2020.02.007] [Citation(s) in RCA: 175] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 02/15/2020] [Accepted: 02/18/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND Reperfusion may cause injuries to the myocardium in ischemia situation. Emerging studies suggest that exosomes may serve as key mediators in myocardial ischemia/reperfusion (MI/R) injury. OBJECTIVE The study was conducted to figure out the mechanism of M2 macrophage-derived exosomes (M2-exos) in MI/R injury with the involvement of microRNA-148a (miR-148a). METHODS AND RESULTS M2 macrophages were prepared and M2-exos were collected and identified. Neonatal rat cardiomyocytes (NCMs) were extracted for in vitro hypoxia/reoxygenation (H/R) model establishment, while rat cardiac tissues were separated for in vivo MI/R model establishment. Differentially expressed miRNAs in NCMs and H/R-treated NCMs after M2-exos treatment were evaluated using microarray analysis. The target relation between miR-148a and thioredoxin-interacting protein (TXNIP) was identified using dual luciferase reporter gene assay. Gain- and loss- of function studies of miR-148a and TXNIP were performed to figure out their roles in MI/R injury. Meanwhile, the activation of the TLR4/NF-κB/NLRP3 inflammasome signaling pathway and pyroptosis of NCMs were evaluated. M2 macrophages carried miR-148a into NCMs. Over-expression of miR-148a enhanced viability of H/R-treated NCMs, reduced infarct size in vivo, and alleviated dysregulation of cardiac enzymes and Ca2+ overload in both models. miR-148a directly bound to the 3'-untranslated region (3'UTR) of TXNIP. Over-expressed TXNIP triggered the TLR4/NF-κB/NLRP3 signaling pathway activation and induced cell pyroptosis of NCMs, and the results were reproduced in in vivo studies. CONCLUSION This study demonstrated that M2-exos could carry miR-148a to mitigate MI/R injury via down-regulating TXNIP and inactivating the TLR4/NF-κB/NLRP3 inflammasome signaling pathway. This study may offer new insights into MI/R injury treatment.
Collapse
Affiliation(s)
- Yuxiang Dai
- Department of Cardiology, Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Shanghai 200032, China
| | - Shen Wang
- Department of Cardiology, the Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310005, China
| | - Shufu Chang
- Department of Cardiology, Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Shanghai 200032, China
| | - Daoyuan Ren
- Department of Cardiology, Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Shanghai 200032, China
| | - Shalaimaiti Shali
- Department of Cardiology, Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Shanghai 200032, China
| | - Chenguang Li
- Department of Cardiology, Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Shanghai 200032, China
| | - Hongbo Yang
- Department of Cardiology, Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Shanghai 200032, China
| | - Zheyong Huang
- Department of Cardiology, Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Shanghai 200032, China
| | - Junbo Ge
- Department of Cardiology, Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Shanghai 200032, China.
| |
Collapse
|
9
|
Shi H, Chen X, Jiang H, Wang X, Yu H, Sun P, Sui X. miR-148a suppresses cell invasion and migration in gastric cancer by targeting DNA methyltransferase 1. Oncol Lett 2018. [PMID: 29541249 PMCID: PMC5835867 DOI: 10.3892/ol.2018.7907] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Gastric cancer (GC) is the fourth most common malignant tumor globally. The highest incidence of GC is found in Eastern Asia, particularly in China. It is therefore imperative to further elucidate the molecular pathogenesis of GC in order to identify new biomarkers and targets for effective therapy. In the present study, we determined whether miR-148a was aberrantly downregulated in gastric cancer tissues and significantly correlated with aggressive clinicopathological characteristics in the MGC-803, HGC-27 and GES-1 cell lines using reverse transcription-quantitative PCR and western blot analysis. The cell lines were obtained from 60 patients who presented at our hospital between September 2010 and July 2015. The results showed that, miR-148a was aberrantly downregulated in GC tissues and its expression was relatively lower in the MGC-803 and HGC-27 GC cell lines than in the normal gastric epithelial cell line, GES-1. The clinicopathological analysis revealed that a decrease of miR-148a was significantly correlated with lymph-node metastasis (P<0.01) and tumor node metastasis (TNM) stage (P<0.05). The transwell assay showed that the re-expression of miR-148a significantly reduced cell migratory and invasive abilities in vitro (P<0.01). The luciferase assay confirmed that, DNA methyltransferase 1 (DNMT1) was a direct and functional target of miR-148a. The miR-148a inhibitor increased the expression of DNMT1 in HGC-27 cells and the re-expression of miR-148a reduced the expression of DNMT1 in MGC-803 cells as confirmed by western blot analysis. Furthermore, we found that the re-expression of DNMT1 reversed the inhibition of cell migration and invasion induced by miR-148a. Taken together, we demonstrated that miR-148a suppresses cell invasion and migration in gastric cancer by regulating DNMT1 expression. The miR-148a/DNMT1 axis may therefore be a new potential target for GC therapy.
Collapse
Affiliation(s)
- Huaijie Shi
- Department of Gastrointestinal Surgery, Weihai Central Hospital, Weihai, Shandong 264400, P.R. China
| | - Xiaojing Chen
- First Department of Radiotherapy, Qingdao Center Hospital, Qingdao, Shandong 266000, P.R. China
| | - Hao Jiang
- Department of Gastrointestinal Surgery, Weihai Central Hospital, Weihai, Shandong 264400, P.R. China
| | - Xujie Wang
- Department of Gastrointestinal Surgery, Weihai Central Hospital, Weihai, Shandong 264400, P.R. China
| | - Hao Yu
- Department of Gastrointestinal Surgery, Weihai Central Hospital, Weihai, Shandong 264400, P.R. China
| | - Pijiang Sun
- Department of Gastrointestinal Surgery, Weihai Central Hospital, Weihai, Shandong 264400, P.R. China
| | - Xin Sui
- Department of Gastrointestinal Surgery, Weihai Central Hospital, Weihai, Shandong 264400, P.R. China
| |
Collapse
|