留言板

尊敬的读者、作者、审稿人, 关于本刊的投稿、审稿、编辑和出版的任何问题, 您可以本页添加留言。我们将尽快给您答复。谢谢您的支持!

姓名
邮箱
手机号码
标题
留言内容
验证码

Role of microRNAs in the process of metformin treating multiple diseases

Ningning Ma Jing Chen Jin Ren

Role of microRNAs in the process of metformin treating multiple diseases[J]. 寒地医学, 2021, 1(2): 69-78. doi: 10.2478/fzm-2021-0009
引用本文: Role of microRNAs in the process of metformin treating multiple diseases[J]. 寒地医学, 2021, 1(2): 69-78. doi: 10.2478/fzm-2021-0009
Ningning Ma, Jing Chen, Jin Ren. Role of microRNAs in the process of metformin treating multiple diseases[J]. Frigid Zone Medicine, 2021, 1(2): 69-78. doi: 10.2478/fzm-2021-0009
Citation: Ningning Ma, Jing Chen, Jin Ren. Role of microRNAs in the process of metformin treating multiple diseases[J]. Frigid Zone Medicine, 2021, 1(2): 69-78. doi: 10.2478/fzm-2021-0009

Role of microRNAs in the process of metformin treating multiple diseases

doi: 10.2478/fzm-2021-0009
More Information
  • Table  1.   List of miRNAs regulated by metformin with various cancers

    Cancer Related miRNAs Metformin effects Target genes Cell line
    Lung cancer miR-381 Upregulate YAP A549, H1299, Calu6, H520
    miR-7 Upregulate AKT/mTOR A549
    Erk, NF-κB
    miR-222 Downregulate p27, p57 A549
    PTEN NCI-H358
    Pancreatic cancer miR-26a Upregulate HMGA1 Sw1990
    Panc1
    miR-200b Upregulate AsPC-1
    AsPC-1-GTR
    MiaPaCa-2
    miR-34a Upregulate Notch, Slug Panc02
    miR-150 Upregulate
    Breast cancer miR-21-5p Downregulate SESN1 SUM159PT
    CAB39L MCF-7, BT-474
    BT-549
    miR-27a Downregulate AMPKα2 MCF-7
    miR-26a Upregulate PTEN MDA-MB-231
    MCL-1 MDA-MB-468
    MTDH MCF-7
    EZH2
    miR-155 Downregulate
    miR-200c Upregulate c-Myc MDA-MB-231
    AKT/Bcl-2 MCF-7
    Let-7a Upregulate
    miR-96
    miR-181a Downregulate
    miR-193b Upregulate FASN MDA-MB-231
    MDA-MB-468
    BT-549
    Renal cell miR-26a Upregulate Bcl-2 786-O
    carcinoma Cyclin D1
    miR-21 Downregulate PTEN CAKI-1
    CAKI-2
    Oral Cancer miR-26a Upregulate Mcl-1 KB cell
    Skin cancer miR-21 Downregulate PTEN/AKT HaCaT
    Colon cancer miR-21 Downregulate PTEN/AKT HuTu80
    miR-1915 Upregulate Bcl-2 HuTu80
    Cervical cancer miR-142-3p Upregulate HMGA2 SiHa
    HeLa
    Esophageal miR-497 Upregulate PELP1 ESCC
    squamous cell
    carcinoma
    Osteosarcom a miR-570-3p Upregulate LCMR1 143B
    ATG12 U2OS
    下载: 导出CSV

    Table  2.   List of miRNAs regulated by metformin for treating glycolipid metabolic disease

    Metabolic Disease Related miRNA Metformin effects Target genes Cell line
    T2DM miR-140-5p Downregulate
    miR-222 Downregulate
    miR-142-3p Upregulate
    miR-192 Upregulate
    miR-33b Downregulate CPT1 HepG2
    CROT
    miR-291-3b Downregulate AMPKα1 NCTC1469
    下载: 导出CSV

    Table  3.   Lists of miRNAs involved in metformin treating other diseases

    Disease Related miRNA Metformin effects Target genes Cell line
    Anti-angioge nesis miR-21 Downregulate PTEN, HUVEC
    SMAD7
    miR-221/222 Upregulate p27Kip1
    miR-34a Downregulate Sirtuin1 MMEC
    Atheroscleros is Inflammation miR-124 Downregulate P4Hα1 VSMC
    miR-34a-5p Upregulate RAW264.7
    miR-125b-5p
    miR-146a Upregulate
    miR-99b Upregulate AKT
    miR-155 Downregulate AKT
    miR-206 Downregulate HDAC4/ cyclin D1
    下载: 导出CSV
  • [1] Sanchez-Rangel E, Inzucchi SE. Metformin: clinical use in type 2 diabetes. Diabetologia, 2017; 60(9): 1586-1593. doi: 10.1007/s00125-017-4336-x
    [2] Anisimov V N. Metformin for aging and cancer prevention. Aging, 2010; 2(11): 760-774. doi: 10.18632/aging.100230
    [3] An H, He L. Current understanding of metformin effect on the control of hyperglycemia in diabetes. The Journal of Endocrinology, 2016; 228(3): R97-R106. doi: 10.1530/JOE-15-0447
    [4] Hardie D G, Ross F A, Hawley S A. AMPK: a nutrient and energy sensor that maintains energy homeostasis. Nature Reviews. Molecular cell biology, 2012; 13(4): 251-262. doi: 10.1038/nrm3311
    [5] Castellano L, Stebbing J. Deep sequencing of small RNAs identifies canonical and non-canonical miRNA and endogenous siRNAs in mammalian somatic tissues. Nucleic Acids Research, 2013; 41(5): 3339-3351. doi: 10.1093/nar/gks1474
    [6] Pu M, Chen J, Tao Z, et al. Regulatory network of miRNA on its target: coordination between transcriptional and post-transcriptional regulation of gene expression. Cellular and Molecular Life Sciences: CMLS, 2019; 76(3): 441-451. doi: 10.1007/s00018-018-2940-7
    [7] Catalanotto C, Cogoni C, Zardo G. MicroRNA in Control of Gene Expression: An Overview of Nuclear Functions. International Journal of Molecular Sciences, 2016; 17(10): 1712. doi: 10.3390/ijms17101712
    [8] Kim Y K, Kim B, Kim V N. Re-evaluation of the roles of DROSHA, Export in 5, and DICER in microRNA biogenesis. Proceedings of the National Academy of Sciences of the United States of America, 2016; 113(13): e1881-e1889. doi: 10.1073/pnas.1602532113
    [9] Blandino G, Valerio M, Cioce M, et al. Metformin elicits anticancer effects through the sequential modulation of DICER and c-MYC. Nature Communications, 2012; 3(1): 865. doi: 10.1038/ncomms1859
    [10] Abdelmohsen K, Tominaga-Yamanaka K, Srikantan S, et al. RNA-binding protein AUF1 represses Dicer expression. Nucleic Acids Research, 2012; 40(22): 11531-11544. doi: 10.1093/nar/gks930
    [11] Noren Hooten N, Martin-Montalvo A, Dluzen D F, et al. Metformin-mediated increase in DICER1 regulates microRNA expression and cellular senescence. Aging Cell, 2016; 15: 572-581. doi: 10.1111/acel.12469
    [12] Bridgeman S C, Ellison G C, Melton P E, et al. Epigenetic effects of metformin: From molecular mechanisms to clinical implications. Diabetes, Obesity & Metabolism 2018; 20(7): 1553-1562. http://www.ncbi.nlm.nih.gov/pubmed/30148122
    [13] Zhong T, Men Y, Lu L, et al. Metformin alters DNA methylation genome-wide via the H19/SAHH axis. Oncogene, 2017; 36(17): 2345-2354. doi: 10.1038/onc.2016.391
    [14] Yan L, Zhou J, Gao Y, et al. Regulation of tumor cell migration and invasion by the H19/let-7 axis is antagonized by metformin-induced DNA methylation. Oncogene, 2015; 34(23): 3076-3084. doi: 10.1038/onc.2014.236
    [15] Whyte F. Metastasis: the deadly part of cancer. British Journal of Nursing (Mark Allen Publishing), 1996; 5(9): 535-538. doi: 10.12968/bjon.1996.5.9.535
    [16] Park S M, Park S H, Ryu K J, et al. Downregulation of CHIP promotes ovarian cancer metastasis by inducing Snail-mediated epithelial-mesenchymal transition. Molecular Oncology, 2019; 13(5): 1280-1295. doi: 10.1002/1878-0261.12485
    [17] Kang H, Kim H, Lee S, et al. Role of metabolic reprogramming in epithelial(-)mesenchymal transition (EMT). Int J Mol Sci, 2019; 20(8): 2042. doi: 10.3390/ijms20082042
    [18] Muti P, Berrino F, Krogh V, et al. Metformin, diet and breast cancer: an avenue for chemoprevention. Cell Cycle (Georgetown, Tex. ), 2009; 8(16): 2661. doi: 10.4161/cc.8.16.9226
    [19] Vona-Davis L, Rose D P. Type 2 diabetes and obesity metabolic interactions: common factors for breast cancer risk and novel approaches to prevention and therapy. Current Diabetes Reviews, 2012; 8(2): 116-130. doi: 10.2174/157339912799424519
    [20] Zhou J Y, Xu B, Li L. A new role for an old drug: metformin targets MicroRNAs in treating diabetes and cancer. Drug Development Research, 2015; 76(6): 263-269. doi: 10.1002/ddr.21265
    [21] Xue Y, Xu W, Zhao W, et al. miR-381 inhibited breast cancer cells proliferation, epithelial-to-mesenchymal transition and metastasis by targeting CXCR4. Biomedicine & Pharmacotherapy, 2017; 86: 426-433. doi: 10.1016/j.biopha.2016.12.051
    [22] Qiao G, Li J, Wang J, et al. miR-381 functions as a tumor suppressor by targeting ETS1 in pancreatic cancer. International Journal of Molecular medicine, 2019; 44(2): 593-607. http://d.wanfangdata.com.cn/periodical/2403624fc6721680ececec7f4cc9bff3
    [23] Liu C, Tian X, Zhang J, et al. Long non-coding RNA DLEU1 promotes proliferation and invasion by interacting with miR-381 and enhancing HOXA13 expression in cervical cancer. Front Genet, 2018; 9: 629. doi: 10.3389/fgene.2018.00629
    [24] Cao Q, Liu F, Ji K, et al. MicroRNA-381 inhibits the metastasis of gastric cancer by targeting TMEM16A expression. J Exp Clin Cancer Res, 2017; 36: 29. doi: 10.1186/s13046-017-0499-z
    [25] Jin D, Guo J, Wu Y, et al. Metformin-repressed miR-381-YAP-snail axis activity disrupts NSCLC growth and metastasis. J Exp Clin Cancer Res, 2020; 39(1): 6. doi: 10.1186/s13046-019-1503-6
    [26] Li M, Pan M, You C, et al. The therapeutic potential of miR-7 in cancers. Mini Reviews in Medicinal Chemistry, 2019; 19(20): 1707-1716. doi: 10.2174/1389557519666190904141922
    [27] Dong J, Peng H, Yang X, et al. Metformin mediated microRNA-7 upregulation inhibits growth, migration, and invasion of non-small cell lung cancer A549 cells. Anticancer Drugs, 2020; 31: 345-352. doi: 10.1097/CAD.0000000000000875
    [28] Chen W X, Hu Q, Qiu M T, et al. miR-221/222: promising biomarkers for breast cancer. Tumour Biology: The Journal of the International Society for Oncodevelopmental Biology and Medicine, 2013; 34(3): 1361-1370. doi: 10.1007/s13277-013-0750-y
    [29] Gong L, Zhang W, Yuan Y, et al. miR-222 promotes invasion and migration of ovarian carcinoma by targeting PTEN. Oncology Letters, 2018; 16(1): 984-990. http://www.onacademic.com/detail/journal_1000040552652110_78db.html
    [30] Yang Y F, Wang F, Xiao J J, et al. MiR-222 overexpression promotes proliferation of human hepatocellular carcinoma HepG2 cells by downregulating p27. International Journal of Clinical and Experimental Medicine, 2014; 7(4): 893-902. http://europepmc.org/articles/PMC4057838/pdf/ijcem0007-0893.pdf
    [31] Wang Y, Dai W, Chu X, et al. Metformin inhibits lung cancer cells proliferation through repressing microRNA-222. Biotechnol Lett, 2013; 35(12): 2013-2019. doi: 10.1007/s10529-013-1309-0
    [32] Deng J, He M, Chen L, et al. The loss of miR-26a-mediated post-transcriptional regulation of cyclin E2 in pancreatic cancer cell proliferation and decreased patient survival. PloS One, 2013; 8: e76450. doi: 10.1371/journal.pone.0076450
    [33] Ren G, Li H, He X, et al. Downregulation of serum miR-26a predicts poor clinical outcome of papillary thyroid carcinoma. International Journal of Clinical And Experimental Pathology, 2017; 10(8): 9042-9047. http://www.researchgate.net/publication/338802062_Downregulation_of_serum_miR-26a_predicts_poor_clinical_outcome_of_papillary_thyroid_carcinoma
    [34] Hwang J, Min B H, Jang J, et al. MicroRNA Expression Profiles in Gastric Carcinogenesis. Scientific Reports, 2018; 8(1): 14393. doi: 10.1038/s41598-018-32782-8
    [35] Taheriazam A, Bahador R, Karbasy S H, et al. Down-regulation of microRNA-26a and up-regulation of microRNA-27a contributes to aggressive progression of human osteosarcoma. Diagnostic Pathology, 2015; 10(1): 166. doi: 10.1186/s13000-015-0400-3
    [36] Liang L, Zeng J H, Wang J Y, et al. Down-regulation of miR-26a-5p in hepatocellular carcinoma: a qRT-PCR and bioinformatics study. Pathology, Research and Practice, 2017; 213(12): 1494-1509. doi: 10.1016/j.prp.2017.10.001
    [37] Yang Y, Zhang P, Zhao Y, et al. Decreased MicroRNA-26a expression causes cisplatin resistance in human non-small cell lung cancer. Cancer Biology & Therapy, 2016; 17(5): 515-525. http://www.onacademic.com/detail/journal_1000038331651910_039a.html
    [38] Wan D, Hua M, Liu T, et al. Evaluation and application of highway route scheme based on projection method. Journal of Chongqing Jiaotong University(Natural Science), 2012. http://en.cnki.com.cn/Article_en/CJFDTOTAL-CQJT201206009.htm
    [39] Bao B, Wang Z, Ali S, et al. Metformin inhibits cell proliferation, migration and invasion by attenuating CSC function mediated by deregulating miRNAs in pancreatic cancer cells. Cancer Prev Res (Phila), 2012; 5(3): 355-364. doi: 10.1158/1940-6207.CAPR-11-0299
    [40] Bracken C P, Khew-Goodall Y, Goodall G J. Network-based approaches to understand the roles of miR-200 and other microRNAs in cancer. Cancer Res, 2015; 75(13): 2594-2599. doi: 10.1158/0008-5472.CAN-15-0287
    [41] Ji Q, Hao X, Zhang M, et al. MicroRNA miR-34 inhibits human pancreatic cancer tumor-initiating cells. PLoS One, 2009; 4(8): e6816. doi: 10.1371/journal.pone.0006816
    [42] Cifarelli V, Lashinger L M, Devlin K L, et al. Metformin and rapamycin reduce pancreatic cancer growth in obese prediabetic mice by distinct microrna-regulated mechanisms. Diabetes, 2015; 64(5): 1632-1642. doi: 10.2337/db14-1132
    [43] Kato K, Iwama H, Yamashita T, et al. The anti-diabetic drug metformin inhibits pancreatic cancer cell proliferation in vitro and in vivo: Study of the microRNAs associated with the antitumor effect of metformin. Oncol Rep, 2016; 35(3): 1582-1592. doi: 10.3892/or.2015.4496
    [44] Ganci F, Sacconi A, Bossel Ben-Moshe N, et al. Expression of TP53 mutation-associated microRNAs predicts clinical outcome in head and neck squamous cell carcinoma patients. Ann Oncol, 2013; 24: 3082-3088. doi: 10.1093/annonc/mdt380
    [45] Sekar D, Krishnan R, Thirugnanasambantham K, et al. Significance of microRNA 21 in gastric cancer. Clinics and Research in Hepatology and Gastroenterology, 2016; 40(5): 538-545. doi: 10.1016/j.clinre.2016.02.010
    [46] Ghorbanmehr N, Gharbi S, Korsching E, et al. miR-21-5p, miR-141-3p, and miR-205-5p levels in urine-promising biomarkers for the identification of prostate and bladder cancer. The Prostate, 2019; 79: 88-95. doi: 10.1002/pros.23714
    [47] Qu K, Zhang X, Lin T, et al. Circulating miRNA-21-5p as a diagnostic biomarker for pancreatic cancer: evidence from comprehensive miRNA expression profiling analysis and clinical validation. Scientific Reports, 2017; 7(1): 1692. doi: 10.1038/s41598-017-01904-z
    [48] Pulito C, Mori F, Sacconi A, et al. Metformin-induced ablation of microRNA 21-5p releases Sestrin-1 and CAB39L antitumoral activities. Cell Discov, 2017; 3: 17022. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC5501975/pdf/celldisc201722.pdf
    [49] Li X, Xu M, Ding L, et al. MiR-27a: a novel biomarker and potential therapeutic target in tumors. Journal of Cancer, 2019; 10: 2836-2848. doi: 10.7150/jca.31361
    [50] Zhao W, Zhang X, Liu J, et al. miR-27a-mediated antiproliferative effects of metformin on the breast cancer cell line MCF-7. Oncol Rep, 2016; 36(6): 3691-3699. doi: 10.3892/or.2016.5199
    [51] Cabello P, Pineda B, Tormo E, et al. The antitumor effect of metformin is mediated by mir-26a in breast cancer. International Journal of Molecular Sciences, 2016; 17: 1298. doi: 10.3390/ijms17081298
    [52] Zhang J, Li G, Chen Y, et al. Metformin Inhibits Tumorigenesis and Tumor Growth of Breast Cancer Cells by Upregulating miR-200c but Downregulating AKT2 Expression. Journal of Cancer, 2017; 8(10): 1849-1864. doi: 10.7150/jca.19858
    [53] Sharma P, Kumar S. Metformin inhibits human breast cancer cell growth by promoting apoptosis via a ROS-independent pathway involving mitochondrial dysfunction: pivotal role of superoxide dismutase (SOD). Cell Oncol (Dordr), 2018; 41(6): 637-650. http://www.ncbi.nlm.nih.gov/pubmed/30088260
    [54] Oliveras-Ferraros C, Cufi S, Vazquez-Martin A, et al. Micro(mi)RNA expression profile of breast cancer epithelial cells treated with the anti-diabetic drug metformin: induction of the tumor suppressor miRNA let-7a and suppression of the TGFbeta-induced oncomiR miRNA-181a. Cell Cycle (Georgetown, Tex. ) 2011; 10: 1144-1151.
    [55] Wahdan-Alaswad R S, Cochrane D R, Spoelstra N S, et al. Metformin-induced killing of triple-negative breast cancer cells is mediated by reduction in fatty acid synthase via miRNA-193b. Hormones & Cancer, 2014; 5(6): 374-389. http://www.pubmedcentral.nih.gov/picrender.fcgi?artid=PMC4570735&blobtype=pdf
    [56] Yang F Q, Wang J J, Yan J S, et al. Metformin inhibits cell growth by upregulating microRNA-26a in renal cancer cells. International Journal of Clinical and Experimental Medicine, 2014; 7(10): 3289-3296. http://www.pubmedcentral.nih.gov/picrender.fcgi?artid=PMC4238495&blobtype=pdf
    [57] Wang F, Xu J, Liu H, et al. Metformin induces apoptosis by microRNA-26a-mediated downregulation of myeloid cell leukaemia-1 in human oral cancer cells. Mol Med Rep, 2016; 13(6): 4671-4676. doi: 10.3892/mmr.2016.5143
    [58] Kalogirou C, Schafer D, Krebs M, et al. Metformin-Derived Growth Inhibition in Renal Cell Carcinoma Depends on miR-21-Mediated PTEN Expression. Urol Int, 2016; 96(1): 106-115. doi: 10.1159/000441011
    [59] Deng Y, Ma W. Metformin inhibits HaCaT cell viability via the miR-21/ PTEN/Akt signaling pathway. Mol Med Rep, 2018; 17(3): 4062-4066. http://www.onacademic.com/detail/journal_1000041698470199_847b.html
    [60] Roy S, Yu Y, Padhye S B, et al. Difluorinated-curcumin (CDF) restores PTEN expression in colon cancer cells by down-regulating miR-21. PLoS One, 2013; 8(7): e68543. doi: 10.1371/journal.pone.0068543
    [61] Fujihara S, Kato K, Morishita A, et al. Antidiabetic drug metformin inhibits esophageal adenocarcinoma cell proliferation in vitro and in vivo. Int J Oncol, 2015; 46(5): 2172-2180. doi: 10.3892/ijo.2015.2903
    [62] Chiyo T, Kato K, Iwama H, et al. Therapeutic potential of the antidiabetic drug metformin in small bowel adenocarcinoma. Int J Oncol, 2017; 50(6): 2145-2153. doi: 10.3892/ijo.2017.3971
    [63] Xia C, Liang S, He Z, et al. Metformin, a first-line drug for type 2 diabetes mellitus, disrupts the MALAT1/miR-142-3p sponge to decrease invasion and migration in cervical cancer cells. Eur J Pharmacol, 2018; 830: 59-67. doi: 10.1016/j.ejphar.2018.04.027
    [64] Wang L, Li K, Lin X, et al. Metformin induces human esophageal carcinoma cell pyroptosis by targeting the miR-497/PELP1 axis. Cancer Lett, 2019; 450: 22-31. doi: 10.1016/j.canlet.2019.02.014
    [65] Bao X, Zhao L, Guan H, et al. Inhibition of LCMR1 and ATG12 by demethylation-activated miR-570-3p is involved in the anti-metastasis effects of metformin on human osteosarcoma. Cell Death Dis, 2018; 9(6): 611. doi: 10.1038/s41419-018-0620-z
    [66] Miyoshi H, Kato K, Iwama H, et al. Effect of the anti-diabetic drug metformin in hepatocellular carcinoma in vitro and in vivo. Int J Oncol, 2014; 45(1): 322-332. doi: 10.3892/ijo.2014.2419
    [67] Ye D W, Rong X L, Xu A M, et al. Liver-adipose tissue crosstalk: a key player in the pathogenesis of glucolipid metabolic disease. Chinese Journal of Integrative Medicine, 2017; 23(6): 410-414. doi: 10.1007/s11655-017-2810-4
    [68] Guo J. Research progress on prevention and treatment of glucolipid metabolic disease with integrated traditional Chinese and Western medicine. Chinese Journal of Integrative Medicine, 2017; 23(6): 403-409. doi: 10.1007/s11655-017-2811-3
    [69] Vienberg S, Geiger J, Madsen S, et al. MicroRNAs in metabolism. Acta Physiologica (Oxford, England), 2017; 219: 346-361. doi: 10.1111/apha.12681
    [70] Herrera B M, Lockstone H E, Taylor J M, et al. Global microRNA expression profiles in insulin target tissues in a spontaneous rat model of type 2 diabetes. Diabetologia, 2010; 53(6): 1099-1109. doi: 10.1007/s00125-010-1667-2
    [71] Ortega F J, Mercader J M, Moreno-Navarrete J M, et al. Profiling of circulating microRNAs reveals common microRNAs linked to type 2 diabetes that change with insulin sensitization. Diabetes Care, 2014; 37(5): 1375-1383. doi: 10.2337/dc13-1847
    [72] Ghai V, Kim T K, Etheridge A, et al. Extracellular vesicle encapsulated microRNAs in patients with type 2 diabetes are affected by metformin treatment. J Clin Med, 2019; 8(5): 617. doi: 10.3390/jcm8050617
    [73] Li D, Song H, Shuo L, et al. Gonadal white adipose tissue-derived exosomal MiR-222 promotes obesity-associated insulin resistance. Aging, 2020; 12(22): 22719-22743. http://www.researchgate.net/publication/346983258_Gonadal_white_adipose_tissue-derived_exosomal_MiR-222_promotes_obesity-associated_insulin_resistance
    [74] Prattichizzo F, Giuliani A, De Nigris V, et al. Extracellular microRNAs and endothelial hyperglycaemic memory: a therapeutic opportunity? Diabetes, Obesity & Metabolism, 2016; 18(9): 855-867. http://europepmc.org/articles/PMC5094499?pdf=render
    [75] Buzzetti E, Pinzani M, Tsochatzis E A. The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD). Metabolism: Clinical and Experimental, 2016; 65(8): 1038-1048. doi: 10.1016/j.metabol.2015.12.012
    [76] Katsura A, Morishita A, Iwama H, et al. MicroRNA profiles following metformin treatment in a mouse model of non-alcoholic steatohepatitis. International Journal of Molecular Medicine, 2015; 35(4): 877-884. doi: 10.3892/ijmm.2015.2092
    [77] Guo J, Zhou Y, Cheng Y, et al. Metformin-induced changes of the coding transcriptome and non-coding RNAs in the livers of non-alcoholic fatty liver disease mice. Cell Physiol Biochem, 2018; 45(4): 1487-1505. doi: 10.1159/000487575
    [78] Zare M, Panahi G, Koushki M, et al. Metformin reduces lipid accumulation in HepG2 cells via downregulation of miR-33b. Arch Physiol Biochem, 2019; 5: 1-8. doi: 10.1080/13813455.2019.1680700
    [79] Latorre J, Ortega F J, Liñares-Pose L, et al. Compounds that modulate AMPK activity and hepatic steatosis impact the biosynthesis of microRNAs required to maintain lipid homeostasis in hepatocytes. EBioMedicine, 2020; 53: 102697. doi: 10.1016/j.ebiom.2020.102697
    [80] Meng X, Guo J, Fang W, et al. Liver MicroRNA-291b-3p promotes hepatic lipogenesis through negative regulation of adenosine 5'-monophosphate (AMP)-activated protein kinase alpha1. J Biol Chem, 2016; 291(20): 10625-10634. doi: 10.1074/jbc.M116.713768
    [81] Fujita K, Iwama H, Oura K, et al. Metformin-suppressed differentiation of human visceral preadipocytes: Involvement of microRNAs. International Journal of Molecular Medicine, 2016; 38(4): 1135-1140. doi: 10.3892/ijmm.2016.2729
    [82] Flowers E, Froelicher E S, Aouizerat B E. MicroRNA regulation of lipid metabolism. Metabolism, 2013; 62(1): 12-20. doi: 10.1016/j.metabol.2012.04.009
    [83] Lynn F C. Meta-regulation: microRNA regulation of glucose and lipid metabolism. Trends Endocrinol Metab, 2009; 20(9): 452-459. doi: 10.1016/j.tem.2009.05.007
    [84] Bakhashab S, Ahmed F W, Schulten H J, et al. Metformin improves the angiogenic potential of human CD34(+) cells co-incident with downregulating CXCL10 and TIMP1 gene expression and increasing VEGFA under hyperglycemia and hypoxia within a therapeutic window for myocardial infarction. Cardiovasc Diabetol, 2016; 15(1): 27. doi: 10.1186/s12933-016-0344-2
    [85] Ahmed F W, Bakhashab S, Bastaman I T, et al. Anti-Angiogenic miR-222, miR-195, and miR-21a Plasma Levels in T1DM are improved by metformin therapy, thus elucidating its cardioprotective effect: the MERIT study. Int J Mol Sci, 2018; 19. http://www.ncbi.nlm.nih.gov/pubmed/30347712
    [86] Luo M, Tan X, Mu L, et al. MiRNA-21 mediates the antiangiogenic activity of metformin through targeting PTEN and SMAD7 expression and PI3K/AKT pathway. Sci Rep, 2017; 7: 43427. doi: 10.1038/srep43427
    [87] Coleman C B, Lightell D J, Moss S C, et al. Elevation of miR-221 and -222 in the internal mammary arteries of diabetic subjects and normalization with metformin. Mol Cell Endocrinol, 2013; 374(1-2): 125-129. doi: 10.1016/j.mce.2013.04.019
    [88] Arunachalam G, Lakshmanan A P, Samuel S M, et al. Molecular interplay between microRNA-34a and sirtuin1 in hyperglycemia-mediated impaired angiogenesis in endothelial cells: effects of metformin. J Pharmacol Exp Ther, 2016; 356(2): 314-323. doi: 10.1124/jpet.115.226894
    [89] Liang W J, Zhou S N, Shan M R, et al. AMPKalpha inactivation destabilizes atherosclerotic plaque in streptozotocin-induced diabetic mice through AP-2alpha/miRNA-124 axis. J Mol Med (Berl), 2018; 96(5): 403-412. doi: 10.1007/s00109-018-1627-8
    [90] Luo X, Hu R, Zheng Y, et al. Metformin shows anti-inflammatory effects in murine macrophages through Dicer/microribonucleic acid-34a-5p and microribonucleic acid-125b-5p. J Diabetes Investig, 2020; 11(1): 101-109. doi: 10.1111/jdi.13074
    [91] Rahman F, McEvoy J W. Dangers of overly aggressive blood pressure control. Curr Cardiol Rep, 2018; 20(11): 108. doi: 10.1007/s11886-018-1063-y
    [92] Amara V R, Surapaneni S K, Tikoo K. Metformin attenuates cardiovascular and renal injury in uninephrectomized rats on DOCA-salt: Involvement of AMPK and miRNAs in cardioprotection. Toxicol Appl Pharmacol, 2019; 362: 95-104. doi: 10.1016/j.taap.2018.10.004
    [93] Manuyakorn W, Howarth P H, Holgate S T. Airway remodelling in asthma and novel therapy. Asian Pacific Journal of Allergy and Immunology, 2013; 31(1): 3-10. http://www.siaip.it/upload/1530_holgate-2013.pdf
    [94] Cong X, Xu X, Zhang Y, et al. Temperature drop and the risk of asthma: a systematic review and meta-analysis. Environmental Science and Pollution Research International, 2017; 24(28): 22535-22546. doi: 10.1007/s11356-017-9914-4
    [95] Pan Y, Liu L, Li S, et al. Activation of AMPK inhibits TGF-beta1-induced airway smooth muscle cells proliferation and its potential mechanisms. Sci Rep, 2018; 8(1): 3624. doi: 10.1038/s41598-018-21812-0
    [96] Banerjee P, Dutta S, Pal R. Dysregulation of wnt-signaling and a candidate set of miRNAs underlie the effect of metformin on neural crest cell development. Stem Cells, 2016; 34(2): 334-345. doi: 10.1002/stem.2245
    [97] Fast I, Hewel C, Wester L, et al. Temperature-responsive miRNAs in Drosophila orchestrate adaptation to different ambient temperatures. RNA (New York, N.Y. ), 2017; 23(9): 1352-1364. doi: 10.1261/rna.061119.117
    [98] Lin Y, Zhang L, Zhao Y, et al. Comparative analysis and functional identification of temperature-sensitive miRNA in Arabidopsis anthers. Biochemical and Biophysical Research Communications, 2020; 532(1): 1-10. doi: 10.1016/j.bbrc.2020.05.033
  • 加载中
表(3)
计量
  • 文章访问数:  399
  • HTML全文浏览量:  228
  • PDF下载量:  11
  • 被引次数: 0
出版历程
  • 收稿日期:  2021-01-28
  • 录用日期:  2021-04-24
  • 网络出版日期:  2021-12-22

目录

    /

    返回文章
    返回