Perihan Yiğit1, Oytun Erbaş1

1ERBAS Institute of Experimental Medicine, Illinois, USA & Gebze, Türkiye

Keywords: Adult stem cell, embryonic stem cells, lncRNA, microRNA, non-coding RNA, self-renewal, stem cell.

Abstract

Stem cells are cells that aren't specialized and have the potential to renew themselves and differentiate. Stem cells are critical for tissue regeneration, differentiation, and homeostasis. Recent studies have revealed that long non-coding RNAs and microRNAs, which are non-coding RNAs (ncRNAs) that play an important role in gene expression, development, and course of diseases, have important effects on the functions of stem cells. This chapter aimed to investigate the consequences of ncRNAs on stem cells in normal and diseased states, as well as their significance in differentiation.

Introduction

Stem cells are unspecialized cells that can differentiate into any cell of an organism and have the ability to self-renew. Stem cells are classified as embryonic stem cells (ESCs) and adult stem cells (ASCs) based on their location and totipotent, pluripotent, multipotent, oligopotent, and unipotent stem cells based on their differentiation ability.[1]

Regulatory mechanisms such as various cellular components, signal transduction pathways, epigenetic modifications, and molecules regulating gene expression are involved in the differentiation of stem cells.[2] Non-coding ribonucleic acids (ncRNAs), which are transcripts that do not produce protein, have an important role in stem cell regulation. Long non-coding RNAs (lncRNAs) and microRNAs (miRNAs) are two types of ncRNAs that have an effect on cell proliferation and differentiation.[3]

Long non-coding RNAs are 200 nucleotides long and have critical roles in biological processes such as transcription, translation, splicing, stem cell pluripotency, cell cycle, cellular structural integrity, apoptosis, and protein localization.[4]

Long intergenic ncRNAs (lincRNAs) account for the vast majority of long non-coding RNAs. The remainder is overlapping, antisense, or intronic for protein-coding genes.[5]

The lncRNAs, whose biogenesis varies according to cell type and stage, are transcribed by RNA polymerase II enzyme in different genomic regions such as enhancer, promoter, and intergenic regions and at lower levels in contrast to messenger RNA (mRNA) and comprise intron and exon structures. They lack open reading frames (ORFs), 3'UTRs, and termination regions and therefore have limited coding potential. Long non-coding RNAs are involved in biological processes in both the nucleus and the cytoplasm.[6,7] miRNAs, which are effective in many cellular processes such as stress tolerance, energy metabolism, self-renewal and differentiation of stem cells, cell division and proliferation, and initiation and development of cancer, are short non-coding RNAs of 19-25 nucleotides in length.[5,8]

Mature miRNAs bind to the 3’UTR, 5’UTR, ORF regions, or promoter regions of the target mRNA and cause negative gene expression through translation inhibition or through repressing or binding mRNA,[9,10] as shown in Figure 1.

EFFECT OF MIRNAS ON NORMAL DIFFERENTIATION OF STEM CELLS

Role of miRNAs in Embryonic Stem Cells

Embryonic stem cells have the potential to become all types of cells as they are pluripotent, which is why they can form the three germ layers (endoderm, mesoderm, and ectoderm).

It is a known fact that miRNAs play a critical role in the regulation of stem cell self-renewal and differentiation.[11]

There are specific miRNA transcripts specific to ESCs. These are clusters of miR-290-295, miR-302, miR-17-92, miR-106b-25 and miR-106a-363.[12]

Embryonic stem cells’ miRNAs facilitate G1/S transition by suppressing the expression of RNA binding proteins.[12,13]

Nanog regulates the differentiation of miR-296, miR-470 and miR-134 ESC by affecting the coding regions of Oct4 and Sox2.[14] Co-transfection of miRNAs in lateral and paraxial mesodermal cells transiently increases the differentiation potential of both lateral and paraxial mesodermal cells.[15]

In a study, it was observed that knockout of the Dicer1 enzyme involved in RNA interference increased the level of apoptosis and human embryonic stem cells (hESCs) could not renew themselves. It was also observed that mir-302-367 and mir-371-373 clusters containing the AAGUGC seed sequence can reverse apoptosis.[16]

Wu et al.[17] showed that the mir-290 cluster contributes to the regulation of alternative splicing and gene expression programs for ESC fate determination and that mir-290 enhances the regulation of alternative splicing factors in ESCs by targeting the negative regulator of Mbnl1/2.

Meta-analysis of microRNA-seq, RNA-seq, and metabolomics datasets were taken from humans and mice uncovered 115 miRNAs with a distinctive expression profile as the transition from pure to prime occurred. [18]

Role of miRNAs in Adult Stem Cells

miRNAs are known to have a major impact on ASC proliferation and differentiation. It’s been observed that there are varying miRNA expressions between myeloid and lymphoid lineages, indicating that miRNAs are involved in the differentiation of some hematopoietic lineages.[11,19]

In these stem cells, miRNAs are expressed differently in long-term hematopoietic stem cells (LT-HSCs) compared to short-term HSCs and are characterized by cell surface markers such as c-Kit+/ Sca-1+/Lin- (KSL).[20]

miRNA-320a obstructs the differentiation of adipocytes from bone marrow MSCs (BMSCs), as well as the development of osteoblasts, by targeting RUNX2. miRNA-320a-5p contributes to osteogenesis and adipogenesis homeostasis in bone marrow mesenchymal stem cells (MSCs).[21] microRNA-7-5p promotes osteogenic differentiation in human MSCs (hMSC) by targeting CMKLR1.[22]

In a study, miR-540 was found to be a negative regulator during the differentiation of adipose tissue-derived stromal cells (ADSCs).[23]

EFFECT OF LNCRNAS ON NORMAL DIFFERENTIATION OF STEM CELLS

Effect of lncRNAs in Embryonic Stem Cells

Non-coding RNAs regulate pluripotency and differentiation in ESCs.

There was a study that demonstrated long non-coding RNAs have a crucial role in ploripotency, and that the lncRNA genes of the transcription factor Oct4 and Nanog binding sites, which are necessary for the self-renewal of undifferentiated ESCs, are in close proximity.[24]

In a study investigating lincRNAs in mouse ESCs, they found that lincRNAs affect gene expression in ESCs. They showed that long intergenic ncRNAs are effective in preventing ESC differentiation in the maintenance of pluripotency by affecting the expression levels of Nanog, a transcription factor, and identified lincRNAs involved in the formation of three germ sheets. They also showed that lincRNA transcripts bind with multiple chromatin remodeling proteins.[25]

In a study investigating the effect of lncRNAs on pluripotency and neuronal differentiation in hESCs, hESCs were differentiated into neurons and the expression of thousands of lncRNAs was profiled by microarray. In this study, lncRNAs required for neurogenesis were determined and it was emphasized that lncRNAs have an important role in brain development.[26]

Li YP et al.[27] showed that the knockout of TRIM71-interacting long non-coding RNA 1 (Trincr1) and up-regulation of phosphorylated ERK and ERK pathway target genes caused a decrease in ESC self-renewal.

In another study investigating the role of long non-coding RNAs in the regulation of ESC self-renewal and early embryogenesis, they showed that lncKdm2b, a lncRNA, is conserved among five mammalian species and is highly expressed in ESCs and early embryos. It was also shown that knocked-out LncKdm2b impairs ESC self-renewal and causes early embryonic death, and in addition, LncKdm2b activates the transcription of TF Zbtb3, which in turn contributes to ESC self-renewal.[28]

Impact of lncRNAs in Adult Stem Cells

HOTTIP, a lncRNA, accelerates osteogenic differentiation and accelerates osteogenic differentiation and angiogenesis by interacting with TATA box-binding protein-associated factor 15 (TAF15).[29]

In a study investigating the effects of lncRNA SNHG1 on the osteogenic differentiation of BMSCs, inhibition of lncRNA SNHG1 was found to inhibit the osteogenic differentiation of BMSCs.[30]

The lncRNA NEAT1, which is important in the osteogenic differentiation of human bone marrow-derived MSCs (hBMSCs), regulates miR-29b-3p/BMP1 interaction and affects the osteogenic differentiation of hBMSCs.[31]

In the osteogenic differentiation of periodontal ligament stem cells (PDLSCs) under mechanical force, lncRNAs promoted osteogenic differentiation of PDLSCs by reducing the expression of small nucleolar RNA host gene 8 (SNHG8).[32]

The interaction of lncRNA and miRNA is effective in the osteogenesis of MSCs which are effective in bone development and molecular mechanism.[33,34]

AK141205, a lncRNA induced by osteogenic growth peptide (OGP) in mouse MSCs, is effective in osteogenic differentiation by increasing CXCL13 regulation.[35]

Another lncRNA involved in the osteogenic differentiation of human tooth follicle stem cells is maternally expressed MEG3 (maternally expressed 3). Down-regulation of MEG3, which is effective in reducing gene expression, is involved in the osteogenic differentiation of human dental follicle stem cells.[36]

H19 is another lncRNA that is effective in the development of cancer cell proliferation. Knockdown of H19 suppresses angiogenesis in human amniotic MSCs (HAMSC).[37]

The lncRNA taurine up-regulated gene 1 (TUG1) is involved in the endothelial differentiation of adipose-derived stem cells (ADSCs).[38]

lncRNAs are non-coding RNAs involved in the regulation of hematopoiesis and the development of hematopoietic stem cells. They are effective in the regulation of gene expression during the stages of red blood cell development and differentiation.

There are specific transcripts involved in the expression of transcription factors involved in the differentiation of hematopoietic cell lines.[39]

The Effect of Non-Coding RNAs on Cancer Stem Cells

It is known that non-coding RNAs have an impact on both the normal stem cells and cancer stem cells’ functioning. Cancer stem cells possess the same ability as ordinary stem cells, which is the potential to self-renew and differentiate into various cell types. In contrast to regular stem cells, these are malignant as they advance tumor development and metastasis. Therefore, non-coding RNAs have a crucial role in cancer stem cells.[40]

Cancer cell research demonstrated that transmembrane 4 sub-L family member 1 (TM4SF1), which is usually highly expressed in esophageal cancer cells, is elevated but miR-141 is reduced and TM4SF1 is a direct target of miR-141 and could be a feasible option for the development of new treatments and effective drugs.[41]

It was discovered that miR-34a, which is known to have a negative effect on the growth, movement, and invasion of osteosarcoma cells, can suppress Sox-2 by increasing its expression and has a potential antitumorigenic effect in tumors.[42]

A different study showed that an overabundance of miR-145 in cervical cancer stem cells lessened tumor invasion and colony formation by reducing the expression of important stem cell transcription factors like Sox2, Nanog, and Oct4.[43]

Research revealed that silencing miR-21, which is known to control LATS1 expression in kidney cancer cells, decreased proliferation, invasion, and phenotype of cancer stem cells in an investigation of its effect on kidney cancer cell function and LATS1 expression.[44]

A study of breast cancer highlighted the importance of miR-155, an oncogenic miRNA that is overexpressed in many cancers, as a potential therapeutic target. It was found that when miR-155 was inhibited, breast cancer stem cell formation decreased, and sensitivity to doxorubicin, a chemotherapy drug, increased.[45]

In another study on breast cancer, they showed that miR-29a has a significant effect on EMT (epithelial to mesenchyme transition in cancer) and metastasis of breast cancer cells by targeting SUV420H2 and will lead to new therapeutic approaches.[46]

The researchers found that HAND2-AS1, a lncRNA that is abundant in hepatocellular carcinoma, the most common liver cancer type with high recurrence and heterogeneity, encourages the self-renewal of liver cancer stem cells and may be used as a potential biomarker for liver cancer diagnosis.[47]

In a study conducted to elucidate the molecular mechanisms underlying the self-renewal and chemoresistance of bladder cancer stem cells (BCSCs), it was shown that lncRNA termed low expressed in bladder cancer stem cells (lnc-LBCS) plays an important tumor suppressor role in the selfrenewal and chemoresistance of BCSCs, contributes to differentiated tumor formation and enhanced chemosensitivity, and may represent a therapeutic target for clinical intervention.[48]

H19, a potential oncogenic factor, is upregulated in breast cancer tissues and negatively correlates with miR-138 expression. It contributes to the arrest and apoptosis of the breast cancer cell cycle, according to a study investigating its regulatory function.[49]

In a study investigating the role of H19 in Helicobacter pylori-infected gastric cancer tissues and cells, it was found that overexpression of H19 promoted cell proliferation, migration, and invasion by activating the Nuclear factor kappa B (NF-κB) signaling pathway.[50]

In a study investigating the function of lncRNA KLK8 in colon cancer stem cells, KLK8 was shown to be up-regulated in colon cancer tissues and may be a potential biomarker candidate for colon cancer patients. KLK8’s expression is linked to tumor size and metastasis and is positively correlated with genes associated with cancer stem cells in colon cancer tissues.[51]

An investigation was conducted to explain the mechanism behind the self-renewal of cancer stem cells and it was highlighted that HotairM1, a lncRNA, is only faintly present in human colorectal carcinoma and uveal melanoma and affects the development of cancer stem cells through the HOXA1-nanog signaling cycle.[52]

An examination into the role of lncRNA CASC11, an oncogenic lncRNA in colorectal cancer, in small cell lung cancer, revealed that lncRNA CASC11 and transforming growth factor beta (TGF-β1) was increased in small cell lung cancer and demonstrated that lncRNA CASC11 can upregulate TGF-β1 to augment the stemness of small cell lung cancer cells.[53]

In their research on the function of ADAMTS9-AS1, a lncRNA, when it comes to the stemness of lung adenocarcinoma cancer cells, they showed that it can counter the advancement of the cancer cell stemness by modulating the miR-5009-3p/NPNT axis.[54]

Research into breast cancer revealed that lncRNA SNHG1, a lncRNA, was able to control tumor growth and angiogenesis by stimulating the M2-like polarisation of macrophages.[55]

A study on the part of lncRNA H19 in the development of osteosarcoma and its connection to the NF-κB signaling pathway found that when H19 was silenced, the nuclear factor-κB pathway was also suppressed, leading to a decrease in the migration and invasion of osteosarcoma cells.[56]

In conclusion, this chapter summarises the current data on the effects of lncRNAs in stem cell proliferation and differentiation. The knowledge of how lncRNAs collaborate in stem cells on a molecular level will be beneficial in medical applications down the line.

Cite this article as: Yiğit P, Erbaş O. Non-coding RNAs in Stem Cell. JEB Med Sci 2024;5(1):44-49.

Conflict of Interest

The authors declared no conflicts of interest with respect to the authorship and/or publication of this article.

Financial Disclosure

The authors received no financial support for the research and/or authorship of this article.

Acknowledgments

The Figure (Figure 1) used in this chapter was created with BioRender (BioRender.com).

References

  1. Zakrzewski W, Dobrzyński M, Szymonowicz M, Rybak Z. Stem cells: past, present, and future. Stem Cell Res Ther. 2019 Feb 26;10:68.
  2. Yalçın MB, Bora ES, Erdoğan MA, Çakır A, Erbaş O. The Effect of Adipose-Derived Mesenchymal Stem Cells on Peripheral Nerve Damage in a Rodent Model. J Clin Med. 2023 Oct 9;12:6411.
  3. Schweiger V, Hasimbegovic E, Kastner N, Spannbauer A, Traxler D, Gyöngyösi M, et al. Non-Coding RNAs in Stem Cell Regulation and Cardiac Regeneration: Current Problems and Future Perspectives. Int J Mol Sci. 2021 Aug 25;22:9160.
  4. Yıldız M, Erbaş O. Non-Coding RNAs and Cancer. JEB Med Sci 2021;2:211-7.
  5. Ransohoff JD, Wei Y, Khavari PA. The functions and unique features of long intergenic non-coding RNA. Nat Rev Mol Cell Biol. 2018 Mar;19:143-57.
  6. Dahariya S, Paddibhatla I, Kumar S, Raghuwanshi S, Pallepati A, Gutti RK. Long non-coding RNA: Classification, biogenesis and functions in blood cells. Mol Immunol. 2019 Aug;112:82-92.
  7. Kaptan B, Abusalim M, Erbaş O. Non-Coding RNAs in Brain Function and Disorders. JEB Med Sci 2023;4:28-32.
  8. Yu Z, Li Y, Fan H, Liu Z, Pestell RG. miRNAs regulate stem cell self-renewal and differentiation. Front Genet. 2012 Sep 25;3:191.
  9. Lund E, Güttinger S, Calado A, Dahlberg JE, Kutay U. Nuclear export of microRNA precursors. Science. 2004 Jan 2;303:95-8.
  10. O'Brien J, Hayder H, Zayed Y, Peng C. Overview of MicroRNA Biogenesis, Mechanisms of Actions, and Circulation. Front Endocrinol (Lausanne). 2018 Aug 3;9:402.
  11. Gangaraju VK, Lin H. MicroRNAs: key regulators of stem cells. Nat Rev Mol Cell Biol. 2009 Feb;10:116-25.
  12. Mens MMJ, Ghanbari M. Cell Cycle Regulation of Stem Cells by MicroRNAs. Stem Cell Rev Rep. 2018 Jun;14:309-22.
  13. Hao J, Duan FF, Wang Y. MicroRNAs and RNA binding protein regulators of microRNAs in the control of pluripotency and reprogramming. Curr Opin Genet Dev. 2017 Oct;46:95-103.
  14. Tay Y, Zhang J, Thomson AM, Lim B, Rigoutsos I. MicroRNAs to Nanog, Oct4 and Sox2 coding regions modulate embryonic stem cell differentiation. Nature. 2008 Oct 23;455:1124-8. doi: 10.1038/nature07299. Epub 2008 Sep 17. Erratum in: Nature. 2009 Mar 26;458:538.
  15. Tuysuz EC, Ozbey U, Gulluoglu S, Kuskucu A, Sahin F, Bayrak OF. miRNAs as cell fate determinants of lateral and paraxial mesoderm differentiation from embryonic stem cells. Dev Biol. 2021 Oct;478:212-221.
  16. Teijeiro V, Yang D, Majumdar S, González F, Rickert RW, Xu C, et al. DICER1 Is Essential for Self-Renewal of Human Embryonic Stem Cells. Stem Cell Reports. 2018 Sep 11;11:616-25.
  17. Wu DR, Gu KL, Yu JC, Fu X, Wang XW, Guo WT, et al. Opposing roles of miR-294 and MBNL1/2 in shaping the gene regulatory network of embryonic stem cells. EMBO Rep. 2018 Jun;19:e45657.
  18. Wang Y, Hussein AM, Somasundaram L, Sankar R, Detraux D, Mathieu J, et al. microRNAs Regulating Human and Mouse Naïve Pluripotency. Int J Mol Sci. 2019 Nov 22;20:5864.
  19. Mehta A, Baltimore D. MicroRNAs as regulatory elements in immune system logic. Nat Rev Immunol. 2016 Apr 28;16:279-94.
  20. Crisan M, Dzierzak E. The many faces of hematopoietic stem cell heterogeneity. Development. 2016 Dec 15;143:4571-81. doi: 10.1242/dev.114231. Erratum in: Development. 2017 Nov 15;144:4195.
  21. Zhang Y, Zhang N, Wei Q, Dong Y, Liu Y, Yuan Q, et al. MiRNA-320a-5p contributes to the homeostasis of osteogenesis and adipogenesis in bone marrow mesenchymal stem cell. Regen Ther. 2022 Mar 28;20:32-40.
  22. Chen B, Meng J, Zeng YT, Du YX, Zhang J, Si YM, et al. MicroRNA-7-5p regulates osteogenic differentiation of hMSCs via targeting CMKLR1. Eur Rev Med Pharmacol Sci. 2018 Nov;22:7826-31
  23. Chen L, Chen Y, Zhang S, Ye L, Cui J, Sun Q, et al. MiR-540 as a novel adipogenic inhibitor impairs adipogenesis via suppression of PPARγ. J Cell Biochem. 2015 Jun;116:969-76.
  24. Yildirim N, Simsek D, Kose S, Yildirim AGS, Guven C, Yigitturk G, et al. The protective effect of Gingko biloba in a rat model of ovarian ischemia/reperfusion injury: Improvement in histological and biochemical parameters. Adv Clin Exp Med. 2018 May;27:591-7.
  25. Guttman M, Donaghey J, Carey BW, Garber M, Grenier JK, Munson G, et al. lincRNAs act in the circuitry controlling pluripotency and differentiation. Nature. 2011 Aug 28;477:295-300.
  26. Ng SY, Johnson R, Stanton LW. Human long non-coding RNAs promote pluripotency and neuronal differentiation by association with chromatin modifiers and transcription factors. EMBO J. 2012 Feb 1;31:522-33.
  27. Li YP, Duan FF, Zhao YT, Gu KL, Liao LQ, Su HB, et al. TRIM71 binding long noncoding RNA Trincr1 represses FGF/ERK signaling in embryonic stem cells. Nat Commun. 2019 Mar 25;10:1368.
  28. Ye B, Liu B, Yang L, Zhu X, Zhang D, Wu W, et al. LncKdm2b controls self-renewal of embryonic stem cells via activating expression of transcription factor Zbtb3. EMBO J. 2018 Apr 13;37:e97174.
  29. Zeng X, Dong Q, Liu Q, Tan WJ, Liu XD. LncRNA HOTTIP facilitates osteogenic differentiation in bone marrow mesenchymal stem cells and induces angiogenesis via interacting with TAF15 to stabilize DLX2. Exp Cell Res. 2022 Aug 15;417:113226.
  30. Jiang Y, Wu W, Jiao G, Chen Y, Liu H. LncRNA SNHG1 modulates p38 MAPK pathway through Nedd4 and thus inhibits osteogenic differentiation of bone marrow mesenchymal stem cells. Life Sci. 2019 Jul 1;228:208-14.
  31. Zhang Y, Chen B, Li D, Zhou X, Chen Z. LncRNA NEAT1/miR-29b-3p/BMP1 axis promotes osteogenic differentiation in human bone marrow-derived mesenchymal stem cells. Pathol Res Pract. 2019 Mar;215:525-31.
  32. Zhang Z, He Q, Yang S, Zhao X, Li X, Wei F. Mechanical force-sensitive lncRNA SNHG8 inhibits osteogenic differentiation by regulating EZH2 in hPDLSCs. Cell Signal. 2022 May;93:110285.
  33. Sikora M, Marycz K, Smieszek A. Small and Long Non-coding RNAs as Functional Regulators of Bone Homeostasis, Acting Alone or Cooperatively. Mol Ther Nucleic Acids. 2020 Sep 4;21:792-803.
  34. Lanzillotti C, De Mattei M, Mazziotta C, Taraballi F, Rotondo JC, Tognon M, et al. Long Non-coding RNAs and MicroRNAs Interplay in Osteogenic Differentiation of Mesenchymal Stem Cells. Front Cell Dev Biol. 2021 Apr 9;9:646032.
  35. Başol N, Erbaş O, Çavuşoğlu T, Meral A, Ateş U. Beneficial effects of agomelatine in experimental model of sepsis-related acute kidney injury. Ulus Travma Acil Cerrahi Derg. 2016 Mar;22:121-6.
  36. Deng L, Hong H, Zhang X, Chen D, Chen Z, Ling J, Wu L. Down-regulated lncRNA MEG3 promotes osteogenic differentiation of human dental follicle stem cells by epigenetically regulating Wnt pathway. Biochem Biophys Res Commun. 2018 Sep 10;503:2061-7.
  37. Yuan Z, Bian Y, Ma X, Tang Z, Chen N, Shen M. LncRNA H19 Knockdown in Human Amniotic Giuseppina DivisatoMesenchymal Stem Cells Suppresses Angiogenesis by Associating with EZH2 and Activating Vasohibin-1. Stem Cells Dev. 2019 Jun 15;28:781-90.
  38. Xue YN, Yan Y, Chen ZZ, Chen J, Tang FJ, Xie HQ, Tang SJ, Cao K, Zhou X, Wang AJ, Zhou JD. LncRNA TUG1 regulates FGF1 to enhance endothelial differentiation of adipose-derived stem cells by sponging miR-143. J Cell Biochem. 2019 Nov;120:19087-97.
  39. Ghafouri-Fard S, Niazi V, Taheri M. Role of miRNAs and lncRNAs in hematopoietic stem cell differentiation. Noncoding RNA Res. 2020 Dec 19;6:8-14.
  40. Yang L, Shi P, Zhao G, Xu J, Peng W, Zhang J, et al. Targeting cancer stem cell pathways for cancer therapy. Signal Transduct Target Ther. 2020 Feb 7;5:8.
  41. Xue L, Yu X, Jiang X, Deng X, Mao L, Guo L, et al. TM4SF1 promotes the self-renewal of esophageal cancer stem-like cells and is regulated by miR-141. Oncotarget. 2017 Mar 21;8:19274-84.
  42. Candan Balkan B, Erbaş O. Recent advances in stem cell-based therapeutics in ophthalmology. D J Med Sci 2023;9:175-82.
  43. Zhou X, Yue Y, Wang R, Gong B, Duan Z. MicroRNA-145 inhibits tumorigenesis and invasion of cervical cancer stem cells. Int J Oncol. 2017 Mar;50:853-62.
  44. An F, Liu Y, Hu Y. miR-21 inhibition of LATS1 promotes proliferation and metastasis of renal cancer cells and tumor stem cell phenotype. Oncol Lett. 2017 Oct;14:4684-8.
  45. Zuo J, Yu Y, Zhu M, Jing W, Yu M, Chai H, et al. Inhibition of miR-155, a therapeutic target for breast cancer, prevented in cancer stem cell formation. Cancer Biomark. 2018 Feb 6;21:383-92.
  46. Wu Y, Shi W, Tang T, Wang Y, Yin X, Chen Y, et al. miR-29a contributes to breast cancer cells epithelialmesenchymal transition, migration, and invasion via down-regulating histone H4K20 trimethylation through directly targeting SUV420H2. Cell Death Dis. 2019 Feb 21;10:176.
  47. Wang Y, Zhu P, Luo J, Wang J, Liu Z, Wu W, et al. LncRNA HAND2-AS1 promotes liver cancer stem cell self-renewal via BMP signaling. EMBO J. 2019 Sep 2;38:e101110.
  48. Chen X, Xie R, Gu P, Huang M, Han J, Dong W, et al. Long Noncoding RNA LBCS Inhibits Self-Renewal and Chemoresistance of Bladder Cancer Stem Cells through Epigenetic Silencing of SOX2. Clin Cancer Res. 2019 Feb 15;25:1389-403.
  49. Si H, Chen P, Li H, Wang X. Long non-coding RNA H19 regulates cell growth and metastasis via miR-138 in breast cancer. Am J Transl Res. 2019 May 15;11:3213-25.
  50. Zhang Y, Yan J, Li C, Wang X, Dong Y, Shen X, et al. LncRNA H19 induced by helicobacter pylori infection promotes gastric cancer cell growth via enhancing NF-κB-induced inflammation. J Inflamm (Lond). 2019 Nov 26;16:23.
  51. Wang K, Song W, Shen Y, Wang H, Fan Z. LncRNA KLK8 modulates stem cell characteristics in colon cancer. Pathol Res Pract. 2021 Aug;224:153437.
  52. Li F, Xu Y, Xu X, Ge S, Zhang F, Zhang H, et al. lncRNA HotairM1 Depletion Promotes Self-Renewal of Cancer Stem Cells through HOXA1-Nanog Regulation Loop. Mol Ther Nucleic Acids. 2020 Sep 16;22:456-70.
  53. Fu Y, Zhang P, Nan H, Lu Y, Zhao J, Yang M, et al. LncRNA CASC11 promotes TGF-β1, increases cancer cell stemness and predicts postoperative survival in small cell lung cancer. Gene. 2019 Jul 1;704:91-6.
  54. Wang P, Zhang Y, Lv X, Zhou J, Cang S, Song Y. LncRNA ADAMTS9-AS1 inhibits the stemness of lung adenocarcinoma cells by regulating miR-5009-3p/NPNT axis. Genomics. 2023 Mar 2;115:110596.
  55. Zong S, Dai W, Guo X, Wang K. LncRNA-SNHG1 promotes macrophage M2-like polarization and contributes to breast cancer growth and metastasis. Aging (Albany NY). 2021 Oct 7;13:23169-81.
  56. Zhao J, Ma ST. Downregulation of lncRNA H19 inhibits migration and invasion of human osteosarcoma through the NF-κB pathway. Mol Med Rep. 2018 May;17:7388-94.