Advances in Clinical Medicine
Vol. 13  No. 06 ( 2023 ), Article ID: 67098 , 6 pages
10.12677/ACM.2023.1361308

LncRNA H19在子宫内膜异位症中的研究进展

曾晓艳,万晓慧*

新疆医科大学第一附属医院,新疆 乌鲁木齐

收稿日期:2023年5月13日;录用日期:2023年6月7日;发布日期:2023年6月15日

摘要

子宫内膜异位症(EMs)是一种慢性疾病,其特点是激素反应性是子宫内膜样组织存在子宫体以外的部位。子宫内膜组织的增殖和新生血管形成是子宫内膜异位症发生发展的重要因素。长非编码RNA (Long Non Coding RNAs, LncRNAs)是一组含有200多个核苷酸的非编码单链RNA,参与细胞增殖、分化、染色体重塑、表观遗传调控、转录和转录后修饰等生物学过程。印迹长非编码RNA H19 (LncRNA H19)可能通过调节细胞增殖和凋亡、侵袭、迁移等参与子宫内膜异位症的发病,LncRNA H19表观遗传异常在其发病机制中起着重要作用。本研究旨在探讨LncRNA H19基因的表达及H19-DMR调控区差异甲基化区域(DMR)的表观遗传学改变及LncRNA H19基因与胰岛素样生长因子1 (IGF1)、α-平滑肌肌动蛋白(ACTA2)等在子宫内膜异位症的信号通路调节作用,主要包括遗传表观学、细胞的增殖、凋亡、侵袭、迁移、复发等内容,为临床治疗子宫内膜异位症提供进一步的理论支持。

关键词

LncRNA H19,子宫内膜异位症,发病机制

Research Progress of LncRNA H19 in Endometriosis

Xiaoyan Zeng, Xiaohui Wan*

The First Affiliated Hospital of Xinjiang Medical University, Urumqi Xinjiang

Received: May 13th, 2023; accepted: Jun. 7th, 2023; published: Jun. 15th, 2023

ABSTRACT

Endometriosis, as chronic estrogen-dependent disease, is defined by the presence of endometrial-like tissue outside the uterus. Proliferation of endometrial tissue and neoangiogenesis are critical factors in development of endometriosis. Long non coding RNAs (LncRNAs) are a group of non coding single-stranded RNAs with more than 200 nucleotides that participate in biological processes, including cell proliferation, differentiation, chromosome remodeling, epigenetic regulation, transcription, and posttranscriptional modification. Imprinted long non coding RNA H19 (LncRNA H19) may be involved in the pathogenesis of endometriosis by regulating cell proliferation and apoptosis, invasion, and migration, and epigenetic abnormalities in LncRNA H19 play an important role in its pathogenesis. The aim of this study was to investigate the expression of LncRNA H19 gene and epigenetic alterations in the differentially methylated region (DMR) of the H19-DMR regulatory region and the role of LncRNA H19 gene and insulin-like growth factor 1 (IGF1) and actin alpha 2 (ACTA2) in the regulation of signaling pathways in endometriosis, mainly including genetic epigenetics, cell proliferation, apoptosis, invasion, migration, and recurrence, in order to provide further theoretical support for the clinical treatment of endometriosis.

Keywords:IncRNA H19, Endometriosis, Pathogenesis

Copyright © 2023 by author(s) and Hans Publishers Inc.

This work is licensed under the Creative Commons Attribution International License (CC BY 4.0).

http://creativecommons.org/licenses/by/4.0/

1. 引言

子宫内膜异位症典型症状包括痛经、慢性盆腔疼痛、性交困难、不规则子宫出血和不孕。一些研究表明,25%~50%的不孕妇女同时有子宫内膜异位症,30%~50%的子宫内膜异位症妇女是不孕的。虽然子宫内膜异位症和不孕症之间的联系已被证实,但子宫内膜异位症影响生育的机制尚不清楚。LncRNA有些功能可能与RNA、DNA或蛋白质的基因表达相互作用有关 [1] [2] 。对EMs发病机制包括子宫内膜细胞转移、经血反流和种植、体腔上皮化生和干细胞参与的可能等 [3] [4] ,但是这些机制的具体调控过程尚未完全阐释清楚。

近有研究表明,在位或异位子宫内膜中具有潜在致病可能的LncRNAs中,LncRNA H19在异位子宫内膜中存在明显的差异调控表达。现就LncRNA H19与EMs发病的相关作用机制进行综述。

2. LncRNA H19结构及生物学功能

LncRNA H19因参与细胞功能调节、转录和转录后修饰的能力是最早被发现的长链非编码RNA之一,它主要位于胞质中包括4个内含子和5个外显子,经剪接后产生一个2.3 kb的LncRNA。H19在胚胎发育和生长调控中具有重要作用,在胚胎发育中高度表达,出生后表达显著下降,但在骨骼肌和心脏中持续表达 [5] 。H19可发挥“分子海绵”机制作为内源性竞争性RNA (ceRNA)或靶向结合蛋白等作用调控下游基因转录和翻译 [6] [7] 。H19在细胞核中表达也较高,提示其在细胞核中可能发挥表观遗传调控作用 [8] 。

LncRNA H19是在癌症、血管生成、糖尿病等疾病中研究最广泛的LncRNA之一。先前的研究表明,在子宫内膜异位症患者的异位内膜中,LncRNA H19的下调通过调节miR-124-3p和整合素β3 (ITGB3)的表达来抑制间质细胞的增殖和侵袭能力 [9] 。此外,LncRNA H19的敲除已经被证明在体内可以抑制肿瘤的形成 [10] [11] 。

3. LncRNA H19在EMs中的表观遗传学特征

Nasu K等 [12] 研究表明,子宫内膜异位症可被认为是一种涉及DNA修饰的表观遗传病。表观遗传学由可遗传的表型变化组成,而表型变化不是由DNA序列的改变引起的。证据表明,各种表观遗传修饰,如DNA甲基化,可能在子宫内膜异位症的病因中发挥主要作用。DNA甲基化通常通过将甲基化的DNA结合蛋白如甲基CpG结合蛋白2 (MeCP2)结合到甲基CpG位点来抑制基因转录 [13] 。

Sedigheh K等 [14] 为研究H19基因调节区差异甲基化区(DMR) DNA甲基化状态,采用芯片技术检测了H19基因第二个DMR区域的甲基化水平,与对照组相比,在位内膜组的H19基因第二DMR区出现了显著的低甲基化(P = 0.02),而异位内膜组的低甲基化与对照组相比略有显著性(P = 0.056)。然而,在位和异位子宫内膜样本H19-DMR第二区域的DNA甲基化没有显著差异。H19-DMR区域的DNA甲基化是一种表观遗传改变,可诱导印迹并影响基因表达。H19通过与染色质修饰复合物相互作用调控基因表达的表观遗传机制。也有研究 [15] 表明,不明原因不孕症患者子宫内膜中H19低表达及H19表达产物异常增加继而可能影响子宫内膜细胞的免疫原性及对胚胎的接受性或胚胎的植入,从而考虑LncRNA H19可能在不孕症相关疾病中发挥作用。

4. LncRNA H19在EMs影响在位内膜的作用机制

4.1. LncRNA H19在EMs细胞的增殖和凋亡调控

Liu等 [16] 研究发现EMS患者腹膜液单个核细胞(PFMCs)中LncRNA H19下调,H19过表达通过miR342-3p/IER3降低了IL-17的分泌,降低了Th17细胞/CD4+ T细胞的比例,从而抑制了Th17的分化和子宫内膜基质细胞(ESC)增殖。研究显示,EMS患者较无EMS患者LncRNA H19和即刻早期反应基因3 (IER3)表达减少,miR-342-3p表达增加,Th17细胞/CD4+ T细胞百分比增加。研究证明LncRNA H19过表达通过miR-342-3p/IER3通路抑制Th17细胞分化和ESCs增殖。

子宫LncRNA H19可以调节胰岛素因子1 (IGF1)信号通路,从而调节子宫内膜间质细胞的增殖和凋亡 [17] 。已有研究报道H19与IGF1信号通路关系密切。子宫内膜组织产生IGF1,在子宫内膜细胞的生长和分化中发挥重要作用,结果表明IGF1通过IGF1受体(IGF1R)介导,激活PI3K/AKT和Ras/Raf/MAPK信号转导通路,促进子宫细胞增殖 [18] 。已有研究表明,IGF1可防止子宫内膜细胞的凋亡,并可作为促有丝分裂因子 [19] 。H19的升高导致IGF1R蛋白水平升高。这种增加导致IGF1信号上调,导致子宫内膜基质细胞 [20] 增殖增加。Sedigheh K等研究表明IGF1表达的降低可能是由于H19表达水平的降低,与对照组相比,子宫内膜异位症患者在位内膜和异位内膜IGF1表达降低,降低H19的表达可能会降低IGF1的表达,从而降低IGF1信号,导致子宫内膜异位症中基质细胞增殖率降低。子宫内膜间质细胞增殖的这些改变可能是子宫内膜异位症妇女不孕的一个潜在机制。

4.2. LncRNA H19在EMs基质细胞的侵袭和迁移

Zeitvogel等 [21] 根据植入理论,子宫内膜异位症的形成涉及两个关键过程:迁移和侵袭。Xu等 [22] 研究结果表明雌激素/H19/miR-216a-5p/ACTA2通路的改变调节了子宫内膜异位症子宫内膜基质细胞(euESC)的侵袭和迁移。ACTA2也被称为α-平滑肌肌动蛋白(α-SMA),参与血管收缩和血压稳态 [23] 。持续暴露于活化的血小板会导致子宫内膜间质细胞中ACTA2和分化的平滑肌细胞标志物表达增加,从而导致平滑肌化生。这些发现表明,一个涉及H19、ACTA2和miR-216a-5p的新型调控网络可能是治疗子宫内膜异位症的分子靶点。

4.3. LncRNA H19与EMs复发的关系

子宫内膜异位症有很高的复发率,即使在完全切除病变后;然而,复发机制尚不清楚,一般认为与病变的肿瘤样生物学行为有关,包括增殖、浸润和转移。LncRNA H19过表达与胶质瘤、宫颈癌和子宫内膜癌的不良预后相关,是这些疾病临床预后的潜在预测因子。LncRNA H19已被发现是多种疾病预后不良的预测因子。

Liu等人 [24] 研究分析LncRNA H19表达与子宫内膜异位症临床特征的关系及其作为复发潜在预测因子的价值,随访了104例子宫内膜异位症患者,随访时间中位数为26个月(范围为18~36个月)。复发22例,累计复发率为21.2%,术后1年内复发12例(11.5%),术后2年内复发7例(6.7%),术后2年以上复发3例(2.9%)。分析22例复发患者异位子宫内膜LncRNA H19表达水平及临床特点。结果显示,LncRNA H19过表达且不育且年龄小于40岁的患者复发风险较高。多因素logistic回归分析显示,年龄小于40岁、异位子宫内膜LncRNA H19过表达是子宫内膜异位症复发的独立预后因素,ROC曲线显示当异位子宫内膜LncRNA H19表达水平高于0.0277时,预测复发的敏感性和特异性分别为90.9%和61.0%。

LncRNA H19可能参与了子宫内膜异位症的发展进程特别是复发机制,是子宫内膜异位症复发的一种新的潜在预测因子。确定子宫内膜异位症复发的有效预测因子在临床上非常重要,预测因子不仅可以帮助识别高危患者,以便采取更多措施降低复发风险,还可以保护低危患者避免过度治疗 [25] 。

5. LncRNA H19在疾病中血管形成机制

一项研究表明,LncRNA-H19在体外抑制miR-29a上调血管生成因子VASH2,并调节胶质瘤血管内皮细胞的增殖和血管的形成 [26] 。Sun等 [27] 研究表明LncRNA H19通过与血管平滑肌细胞(VSMCs)中的let-7a直接结合,正向调控细胞周期蛋白D1的表达。证明LncRNA H19/let-7a/cyclin D1是新的信号轴,研究揭示了LncRNA H19在VSMCs增殖和血管重构中的作用机制的新认识,可能以后对子宫内膜异位症新生血管形成诊断和治疗具有新的意义。

6. 总结与展望

LncRNA H19通过调节多种相关因子及调控特定基因的表达而在EMs的发生发展中发挥重要作用。虽然人们对LncRNA H19的生物学特性和功能有了一定的了解,但它对EMs发生发展分子机制及其多个下游靶基因之间复杂的相互作用和协调关系尚未完全阐述清楚,需待进一步深入研究。相信随着研究的深入,LncRNA H19通过非侵入性方法进行测定可提高对内异症识别,从而导致更早的诊断,更快的干预。

文章引用

曾晓艳,万晓慧. LncRNA H19在子宫内膜异位症中的研究进展
Research Progress of LncRNA H19 in Endometriosis[J]. 临床医学进展, 2023, 13(06): 9350-9355. https://doi.org/10.12677/ACM.2023.1361308

参考文献

  1. 1. Xu, J., Bai, J., Zhang, X., et al. (2017) A Comprehensive Overview of LncRNA Annotation Resources. Briefings in Bi-oinformatics, 18, 236-249.

  2. 2. Zhu, J., Fu, H., Wu, Y., et al. (2013) Function of lneRNAs and Approaches to LncRNA-Protein Interactions. Science China Life Sciences, 56, 876-885. https://doi.org/10.1007/s11427-013-4553-6

  3. 3. Taghavipour, M., Sadoughi, F., Mirzaei, H., Yousefi, B., Mo-azzami, B., Chaichian, S., Mansournia, M.A. and Asemi, Z. (2020) Apoptotic Functions of microRNAs in Pathogenesis, Diagnosis, and Treatment of Endometriosis. Cell & Bioscience, 10, Article No. 12. https://doi.org/10.1186/s13578-020-0381-0

  4. 4. Nothnick, W. and Alali, Z. (2016) Recent Advances in the Under-standing of Endometriosis: The Role of Inflammatory Mediators in Disease Pathogenesis and Treatment. F1000Research, 5, F1000 Faculty Rev-186. https://doi.org/10.12688/f1000research.7504.1

  5. 5. Zhang, Y. and Tycko, B. (1992) Monoallelic Expression of the Human H19 Gene. Nature Genetics, 1, 40-44. https://doi.org/10.1038/ng0492-40

  6. 6. Yan, L., Zhou, J., Gao, Y., et al. (2015) Regulation of Tumor Cell Migra-tion and Invasion by the H19/let-7 Axis Is Antagonized by Metformin-Induced DNA Methylation. Oncogene, 34, 3076-3084. https://doi.org/10.1038/onc.2014.236

  7. 7. Gao, Y., Wu, F., Zhou, J., et al. (2014) The H19/let-7 Double-Negative Feedback Loop Contributes to Glucose Metabolism in Muscle Cells. Nucleic Acids Research, 42, 13799-13811. https://doi.org/10.1093/nar/gku1160

  8. 8. Kuchroo, P., Dave, V., Vijayan, A., Viswanathan, C. and Ghosh, D. (2015) Paracrine Factors Secreted by Umbilical Cord-Derived Mesenchymal Stem Cells Induce Angiogenesis in Vitro by a VEGF-Independent Pathway. Stem Cells and Development, 24, 437-450. https://doi.org/10.1089/scd.2014.0184

  9. 9. Liu, S., Qiu, J., Tang, X., et al. (2019) LncRNA-H19 Regulates Cell Proliferation and Invasion of Ectopic Endometrium by Targeting ITGB3 via Modulating miR-124-3p. Experimental Cell Research, 381, 215-222. https://doi.org/10.1016/j.yexcr.2019.05.010

  10. 10. Gao, L.M., Xu, S.F., Zheng, Y., et al. (2019) Long Non-Coding RNA H19 Is Responsible for the Progression of Lung Adenocarcinoma by Mediating Methylation-Dependent Repres-sion of CDH1 Promoter. Journal of Cellular and Molecular Medicine, 23, 6411-6428. https://doi.org/10.1111/jcmm.14533

  11. 11. Peng, F., Li, T.T., Wang, K.L., et al. (2017) H19/let-7/LIN28 Reciprocal Negative Regulatory Circuit Promotes Breast Cancer Stem Cell Maintenance. Cell Death & Disease, 8, e2569. https://doi.org/10.1038/cddis.2016.438

  12. 12. Nasu, K., Kawano, Y., Tsukamoto, Y., et al. (2011) Aberrant DNA Methylation Status of Endometriosis: Epigenetics as the Pathogenesis, Biomarker and Therapeutic Target. Journal of Ob-stetrics and Gynaecology Research, 37, 683-695. https://doi.org/10.1111/j.1447-0756.2011.01663.x

  13. 13. Bogdanović, O. and Veenstra, G.J. (2009) DNA Methyla-tion and methyl-CpG Binding Proteins: Developmental Requirements and Function. Chromosoma, 118, 549-565. https://doi.org/10.1007/s00412-009-0221-9

  14. 14. Sedigheh, K., Elham, A., Firouzeh, G., et al. (2022) Altered Gene Expression of VEGF, IGFs and H19 LncRNA and Epigenetic Profile of H19-DMR Region in Endometrial Tissues of Women with Endometriosis. Reproductive Health, 19, Article No. 100. https://doi.org/10.1186/s12978-022-01406-w

  15. 15. Hurst, B.S., Shimp, K.E., Elliot, M., et al. (2014) Molecular Evaluation of Proliferative-Phase Endometrium May Provide Insight about the Underlying Causes of Infertility in Wom-en with Endometriosis. Archives of Gynecology and Obstetrics, 289, 1119-1124. https://doi.org/10.1007/s00404-013-3103-6

  16. 16. Liu, Z., Liu, L., Zhong, Y., et al. (2019) LncRNA H19 Over-Expression Inhibited Th17 Cell Differentiation to Relieve Endometriosis through miR-342-3p/IER3 Pathway. Cell Bioscience, 9, Article No. 84. https://doi.org/10.1186/s13578-019-0346-3

  17. 17. Lei, Q., Pan, Q., Li, N., et al. (2019) H19 Regulates the Prolifera-tion of Bovine Male Germline Stem Cells via IGF-1 Signaling Pathway. Journal of Cellular Physiology, 234, 915-926. https://doi.org/10.1002/jcp.26920

  18. 18. Ivanga, M., Labrie, Y., Calvo, E., et al. (2007) Temporal Analysis of E2 Transcriptional Induction of PTP and MKP and Downregulation of IGF-I Pathway Key Components in the Mouse Uter-us. Physiological Genomics, 29, 13-23. https://doi.org/10.1152/physiolgenomics.00291.2005

  19. 19. Zhou, Y., Zeng, C., Li, X., et al. (2016) IGF-I Stimu-lates ERβ and Aromatase Expression via IGF1R/PI3K/AKT-Mediated Transcriptional Activation in Endometriosis. Mo-lecular Medicine, 94, 887-897. https://doi.org/10.1007/s00109-016-1396-1

  20. 20. Ghazal, S., McKinnon, B., Zhou, J., et al. (2015) H19 LncRNA Alters Stromal Cell Growth via IGF Signaling in the Endometrium of Women with Endometriosis. EMBO Molecular Medicine, 7, 996-1003. https://doi.org/10.15252/emmm.201505245

  21. 21. Zeitvogel, A., Baumann, R. and Starzinski-Powitz, A. (2001) Identification of an Invasive, N-Cadherin-Expressing Epithelial Cell Type in Endometriosis Using a New Cell Culture Model. The American Journal of Pathology, 159, 1839-1852. https://doi.org/10.1016/S0002-9440(10)63030-1

  22. 22. Xu, Z., Zhang, L.P., Yu, Q., et al. (2019) The Estro-gen-Regulated IncRNA H19/miR-216a-5p Axis Alters Stromal Cell Invasion and Migration via ACTA2 in Endometrio-sis. Molecular Human Reproduction, 25, 550-561. https://doi.org/10.1093/molehr/gaz040

  23. 23. Chen, J., Peters, A., Papke, C.L., et al. (2017) Loss of Smooth Muscle α-Actin Leads to NF-κB-Dependent Increased Sensitivity to Angiotensin II in Smooth Muscle Cells and Aortic Enlarge-ment. Circulation Research, 120, 1903-1915. https://doi.org/10.1161/CIRCRESAHA.117.310563

  24. 24. Liu, S.P., Xin, W.J., Tang, X.Y., et al. (2020) LncRNA H19 Overexpression in Endometriosis and Its Utility as a Novel Bi-omarker for Predicting Recurrence. Reproductive Sciences, 27, 1687-1697.

  25. 25. Selcuk, S., Cam, C., Koc, N., et al. (2016) Evaluation of Risk Factors for the Recurrence of Ovarian Endometriomas. The European Journal of Obstetrics & Gynecology and Reproductive Biology, 203, 56-60. https://doi.org/10.1016/j.ejogrb.2016.05.008

  26. 26. Jia, P., Cai, H., Liu, X., Chen, J., Ma, J., Wang, P., et al. (2016) Long Non-Coding RNA H19 Regulates Glioma Angiogenesis and the Biological Behavior of Glioma Associated Endo-thelial Cells by Inhibiting microRNA-29a. Cancer Letters, 381, 359-369. https://doi.org/10.1016/j.canlet.2016.08.009

  27. 27. Sun, W.F., Lv, J.Y., Duan, L.R., et al. (2018) Long Noncoding RNA H19 Promotes Vascular Remodeling by Sponging let-7a to Upregulate the Expression of Cyclin D1. Biochemical and Biophysical Research Communications, 508, 1038-1042. https://doi.org/10.1016/j.bbrc.2018.11.185

  28. NOTES

    *通讯作者。

期刊菜单