Skip to main content
American Journal of Stem Cells logoLink to American Journal of Stem Cells
. 2020 Apr 25;9(2):16–24.

Role of microRNA in inner ear stem cells and related research progress

Xia Wu 1, Shengyu Zou 1, Fan Wu 2, Zuhong He 1, Weijia Kong 1
PMCID: PMC7218733  PMID: 32419976

Abstract

Deafness is one of the major global health problems that seriously affects the quality of human life. At present, there are no successful treatments for deafness caused by cochlear hair cell (HC) damage. The irreversibility of mammalian hearing impairment is that the inner ear’s sensory epithelium cannot repair lost hair cells and neurons through spontaneous regeneration. The goal of stem cell therapy for sensorineural hearing loss is to reconstruct the damaged inner ear structure and achieve functional repair. microRNA (miRNA), as a class of highly conserved endogenous non-coding small RNAs, plays an important role in the development of cochlea and HCs. miRNA also participates in the regulation of stem cell proliferation and differentiation, and plays an important role in the process of regeneration of inner ear HCs, miRNA has a broad application prospect of clinical treatment of hearing loss, which is conducive to solving the medical problem of inner ear HC regeneration.

Keywords: Hair cell, stem cell, miRNA, inner ear, hearing loss

Introduction

According to WHO analysis data in 2018, 5% of the world’s population suffers from hearing loss, of which 432 million are adults and 34 million are children. In addition, it is estimated that more than 900 million people will experience disabling hearing loss in 2050 (www.who.int). Therefore, how to intervene or treat hearing loss has become a global issue. The mammalian inner ear is a highly differentiated and complex structured organ. The mature inner ear hair cells (HCs) lack the ability to regenerate. Therefore, with the accumulation of hair cell damage, hearing loss will occur [1]. With the advancement of diagnosis and treatment technology, there have been many new developments in the treatment and intervention methods for deafness. Including cochlear implant therapy, gene therapy, drug-induced stem cell differentiation, molecular therapy, etc [2,3]. In addition, with the deepening of research on inner ear stem cells in recent years, it has been discovered that by regulating signaling pathways such as Wnt, Notch, Atoh1, and β-catenin, the potential of supporter cells to differentiate into HCs can be activated in the inner ear of newborn mice [4-7]. Therefore, finding the most effective way to induce inner ear stem cells to differentiate into HCs will provide a very valuable reference for the intervention of hearing loss due to hair cell damage.

microRNAs (miRNAs) are a class of evolutionarily conserved single-stranded small molecule RNA sequences that contain approximately 19-23 nucleotide sequences [8]. miRNA is a non-protein-encoding RNA and usually plays a role in post-transcriptional regulation of gene expression in the body [9]. With the increase of research on miRNA, it has been found that it has a wide range of biological functions, such as regulating cell differentiation, proliferation, development, apoptosis and immune response [10-12]. Previous studies have reported that miRNAs are involved in the regulation of neural stem cell differentiation. In addition, miRNAs also play an important role in the regulation of bone marrow mesenchymal stem cell differentiation [13]. With the increase of research on the regulation mechanism of miRNA and its relationship with stem cells, it becomes possible to use miRNA to regulate the proliferation and differentiation of inner ear stem cells to protect or restore hearing.

miRNA formation and regulation

miRNA is a type of non-coding small RNA that is endogenously expressed in cells consisting of 19 to 25 nucleotides. miRNA is widely found in vertebrates, drosophila, nematodes, plants, and even viruses. miRNA can form an RNA-induced silencing complex (RISC) by binding with Argonaute (AGO) protein, completely or incompletely complementary pairing with the target mRNA, leading to mRNA degradation (complete pairing) or inhibiting its translation (incomplete pairing) to achieve post-transcriptional levels regulation of gene expression [8]. In the nucleus, miRNAs are encoded by nucleotides located in the intergenic region and transcribed by RNA polymerase II to form a longer primitive miRNA (Pri-miRNA) with a stem-loop structure. Then the pri-miRNA is processed into a precursor miRNA (pre-miRNA) with a hairpin structure with a length of 70~100 nt in the nucleus under the action of a complex composed of the RNase III family Drosha enzyme and the double-stranded RNA-specific binding protein DGCR8. Pre-miRNA is then transported to the cytoplasm by a nuclear transporter (Exportin-5), and is cleaved into shorter double-stranded strands miRNA by the RNase III-Dicer and transactivating response RNA-binding protein (TRBP) complex. After the double-stranded miRNA is melted, one of the strands will be degraded, and the other strand forms a single-stranded miRNA, which is the mature miRNA (mature miRNA) [14,15]. In animals, mature miRNAs can be combined with specific ribonucleoprotein AGO protein to form a silencing complex (RNA-induced silencing complex, RISC). RISC can recognize target genes and mediate mRNA degradation or inhibit translation through complementary binding with mRNA 3’UTR or Open Reading Frame (ORF) regions, thereby achieving the goal of regulating target gene expression [16,17]. The way in which the target mRNA is treated by the RSC depends on the characteristics of the mRNA. If mRNA and miRNA are highly complementary, miRNA will guide the target mRNA to break at specific sites and activate endonucleases to degrade mRNA. If they are partially complementary, miRNA will specifically bind to the target mRNA through 3’UTR, which will inhibit translation after transcription of mRNA, but will not affect the stability of mRNA. Therefore, the correct recognition of the binding region between miRNA and target mRNA will directly affect the regulatory function of miRNA. In addition, some studies have found that some miRNAs can accelerate mRNA deadenylation, reduce the effective abundance of mRNA in the cell, and thus downregulate gene expression [18,19]. In some plants, miRNAs can mediate methylation of their loci or target genes, thereby regulating gene expression at the epigenetic level [20]. One miRNA can regulate multiple target genes, and multiple miRNAs can also regulate one gene expression at the same time, so the miRNA regulatory network is both complex and sophisticated.

The role of miRNA in other organs and tissues

It has been found that miRNA expression in animals, plants, and fungi is significantly tissue-specific and plays a variety of regulatory roles in cell growth and development. At present, there are more than 2,000 miRNAs found in the human body and regulating 30% of gene expression [21], not only participating in the regulation of normal physiological processes in the body, such as cell proliferation, differentiation, development, apoptosis, etc. It is also closely related to the occurrence and development of tumors [22], heart disease [23], and neurological diseases [24]. miRNAs play an important role in tumorigenesis and development, and abnormalities in the mutual regulation between miRNAs and genes can lead to tumorigenesis. The biological function of miRNAs in tumors mainly depends on the diversity of their regulation of target genes. Tumors related miRNAs can be generally divided into onco miRNA and suppressor miRNA. Suppressive miRNA negatively regulates tumorigenesis, and its down-regulation or inactivation will directly lead to the occurrence and development of tumors, such as Let-7, miR-9, miR-34, and miR-145, miR-451, etc [25-29]. Onco-miRNAs are positively correlated with tumorigenesis and can form a complex regulatory network with target genes and upstream transcription factors to regulate tumorigenesis, such as miR-130b, miR-182, miR-222, miR-137, miR-708, miR-96, miRNA-21, etc [30-33]. In addition, miRNAs also play important roles in cognition and memory. Alzheimer’s disease-related research found that miR-188-5p expression dysregulation may be the pathophysiological cause of synaptic dysfunction and cognitive-related diseases [34]. miR-148a has been shown to play an important regulatory role in T cell and B cell mediated chronic inflammatory diseases and autoimmune diseases, and its abnormal expression may be one of the pathogenesis of autoimmune diseases [35-37]. miR-155 can regulate the recruitment and retention of inflammatory cells in the synovium of rheumatoid arthritis patients by affecting the production of chemokines and the expression of pro-inflammatory chemokine receptors [38,39]. miR-23b can inhibit the activation of NF-κB induced by pro-inflammatory cytokines and the expression of inflammatory cytokines by targeting TAB2, TAB3 and IKK-α, thereby inhibiting autoimmune inflammation [40]. Cardiovascular related research has found that miR-1 is directly involved in the regulation of the pathological process of myocardial infarction, including myocardial cell apoptosis, inflammation, angiogenesis, fibrosis, and affecting the survival and proliferation activity of myocardial cells [41,42]. In aging-related research, it was found that miRNA-335 and miRNA-34a can inhibit the production of two antioxidant enzymes, such as Superoxide Dismutase 2 (SOD2) and Thioredoxin 2 (TRX2), in the mitochondria, thereby increasing the production of intracellular ROS and promoting cell aging [43]. Therefore, micRNA plays a very important role in the pathological process of many diseases. However, due to the diversity and complexity of miRNA regulation, it is difficult to locate a specific disease in a particular miRNA. Future research on miRNA should not only be limited to miRNA itself, but also focus on the identification and functional analysis of miRNA target genes in order to explore the related molecular mechanisms more thoroughly.

The role of miRNA in auditory system related diseases

Auditory system consists of peripheral auditory system and central auditory system. The peripheral auditory system includes the outer ear, the middle ear, and the inner ear. The inner ear has mechanosensory HCs that convert acoustic energy into neural signalling [44]. Mammal inner ear HCs cannot be regenerated, so once damaged, they can cause permanent hearing loss. The loss of cochlear HCs will also cause the spiral ganglion degeneration, which will affect the effect of cochlear implantation. Hearing loss can usually be divided into two types: conductive hearing loss and sensorineural hearing loss [45]. Conductive hearing loss is caused by structural abnormalities in the outer and middle ears. Sensorineural hearing loss is caused by damage or dysfunction of inner ear HCs, auditory neurons synapses, or stria vascularis. Currently known causes of these tissue or cell damage include aging, genetic mutations, noise trauma, ototoxic drugs and other metabolic diseases [2,44].

miRNA plays an important role in the formation of the inner ear during embryonic development and the maintenance of inner ear function after birth. Over 100 miRNAs have been detected in various types of cochlear cells, such as miR-183, miR-96, miR-182, miR-124, miR-34a, miR-376, and miR-135b [46,47]. miR-183, miR-96, and miR-182 in the cochlea affect the maturation of cochlear function through the temporal and spatial specificity of their expression [48]. Researchers have reported that abnormal expression of miRNAs can directly lead to loss of inner ear structure and hearing dysfunction. miRNA-124 regulates inner ear structure development and cell type differentiation [49]. miR-96 is closely related to the maturation of the ciliated bundles of inner ear hair cells and the development of cochlear nerves, and its mutation will cause non-syndromic progressive hearing loss [50,51]. miRNA-34a can affect hair cell apoptosis through SIRT1/p53 signaling pathway [52]. miR-183 is not only important for the development and function of animal sensory organs, but also plays an important role in the occurrence and development of noise-induced hearing loss [53,54]. With the development of experimental technology and the deepening of scientific understanding, the important role of miRNAs in hearing development and deafness is being continuously discovered.

The role of stem cells in the inner ear

In many mammalian organs, stem cells can sustain continued tissue formation by generating tissue progeny while renewing themselves through division. Renewal and replenishment of cells in blood, bones, epithelium and many other tissues. In the past few decades, stem cell treatment has made great progress in clinical application [55]. For example, bone marrow transplantation is used to treat lymphoma, leukemia and various autoimmune diseases [56]. Pluripotent stem cells derived from retinal pigment epithelium is used to treat blindness (US National Library of Medicine. ClinicalTrials.gov). Gene-edited stem cells are used to treat bullous epidermolysis due to genetic defects [57].

In many non-mammalian animals, researchers have observed HC regeneration, such as fish and birds [58,59]. In addition, some studies have reported that after induction of inner ear HCs death in newborn mice (P3-P4), some support cells in the cochlea can be observed to differentiate into hair cells. These indicate that the cochlea support cells of newborn mice have the potential to differentiate into HCs [60]. We believe that these supporter cells that differentiate into HCs are due to their stem cell properties during this period. There are also reports that the capacity to generate HCs was limited to a subset of supporting cells: inner pillar cells and third-row Deiters cells [61]. Notch and Wnt signaling pathways have important regulatory effects on stem cell self-renewal and differentiation in various tissues [62-64]. Therefore, some researchers have used specific inhibitor to inhibit the Notch pathway in the in-vitro cultured neonatal mouse basilar membrane. After 72 hours, it was observed that new HCs appeared in abnormal areas, and the new cells could express myosin VIIa and phalloidin which are HC specific marker. Notch inhibitor treatment not only causes an increase in the number of HCs but also reduces hearing loss caused by noise [65,66]. In addition, studies have reported that activation of the Wnt/β-catenin pathway in the cochlea of newborn mice can also induce supporter cells to differentiate into HCs [4,6]. For example, Atoh1, a downstream factor of Wnt/β-catenin pathway overexpresses in the cochlea of embryos and newborn mice, can promote the transformation of support cells into HCs [67]. Recent studies have found that in adult mouse cochlea, there are two main stages in inducing support cells to differentiate into HCs. The activation of Myc/NICD induces support cells into reprogramming and proliferation stage, and then inactivates Myc/NICD and overexpresses Atoh1, thereby inducing reprogrammed support cells to differentiate into HC-like cells. These newly generated HC-like cells can form connections with neurons [68]. With the deepening of research on inner ear stem cells, new discoveries provide more ideas and methods for the treatment of hearing loss due to HC loss.

The role of miRNAs in inner ear stem cells

In recent years, many researchers have found that miRNAs can affect the differentiation and proliferation of stem cells by participating in the post-transcriptional regulation of stem cells, and the regulation of specific miRNA expression levels can induce stem cells to differentiate into specific tissue cells [69,70]. Therefore, miRNA is closely related to the fate decision of stem cells, which provides new ideas with important value for the treatment of diseases. In the embryonic stem cell related research, it was found that miRNA-1 and miRNA-133 can promote the mesoderm formation of embryonic stem cells. Among them, miRNA-1 can promote the differentiation of mouse and human embryonic stem cells into heart cells, while miRNA-133 can prevents differentiation of sarcoplasmic progenitor cells [71]. In mesenchymal stem cells (MSCs) related researches, it was found that miR-145, miR-495, miR-29a directly regulate the differentiation of MSCs into chondrocytes [72-74]. miR128 negatively regulates the expression of Wnt3a, and inhibition of miR128 can induce the differentiation of MSCs into neuron-like cells [75]. miR-124 can regulates MSCs differentiation into cardiomyocytes by inhibiting STAT3 gene expression [76,77]. miR-9 promotes stem cell differentiation into neural cells by regulating Notch pathway and zinc finger protein 521 expression [78,79]. Therefore, miRNAs play an important role in the differentiation of pancreas, heart muscle, nerves, osteoblasts, and chondroblasts. In addition, miRNAs also have the ability to regulate cell reprogramming, such as the mir-200c, mir-302 family and mir-369 family can directly induce somatic cell reprogramming [80,81], and the miR-34 family has an inhibitory effect on somatic cell reprogramming [82].

In inner ear HC regeneration related research, overexpression of miR-96 and miR-182 leads to abnormal inner ear HC generation [83]. Due to vestibular HCs have a certain degree of regenerative ability, researchers have compared the expression differences of miRNA between cochlea and vestibule of newborn mice, and found that let-7a, let-7b, miR-24, miR-220, miR-423, miR-190, miR-204, miR-195 and miR-125b have significant differences [84]. Previous research reported that miR-182 can affect the differentiation of cochlear stem cells to HCs by regulating Sox2 and Tbx1 expression [85]. The low expression of miR-182 in cochlear support cells can inhibit trans-differentiation of the support cells into HCs, and its overexpression promotes the differentiation of cochlear stem cells into hair cells, resulting in an increase in the number of cochlear hair cells [85]. The miR-183 family can regulate the Wnt/β-catenin signaling pathway by inhibiting the expression of LRP6 [86], and Wnt signaling can promote cochlear sensory precursors proliferation [4], so we believe that miR-183 is closely related to the proliferation and differentiation of inner ear stem cells. In addition, miR-183 can also inhibits the Notch signaling pathway by inhibiting the expression of NICD3 and NICD4, thereby affecting HC differentiation and regeneration [87]. These findings corresponds to previous research that inhibiting the Notch signaling pathway and activating the Wnt signaling pathway can effectively promote the proliferation and differentiation of Lgr5+ cochlear precursor cells into hair cells [88]. In addition, previous research have reported that miR-384-5p can negatively regulate Notch1 expression [89]. These findings indicate that miRNAs can affect the development of the inner ear through its epigenetic function, especially the regulation of inner ear stem cell proliferation and differentiation. Therefore, miRNA provides a potential tool for gene editing to achieve the regulation of the cell fate and differentiation of inner ear stem cells.

Conclusions

It is known that the loss of HCs in mammals is non-renewable, and stem cells play an important role in the development and regeneration of many other tissues due to their stem maintenance and differentiation capabilities. Therefore, the use of stem cell-related technologies to make auditory system damage repair, especially the regeneration and repair of damaged HCs, has become a hot spot in deafness research. Researches have shown that inner ear stem cells can self-renew and have multiple differentiation potential that can differentiate into HCs under appropriate induction conditions. However, there are still many problems in the application of inner ear stem cells. For example, the distribution and duration of inner ear stem cells in the inner ear are still limited and the molecular mechanism of in vitro differentiation of inner ear stem cells into hair cells is not clear. Although newborn HCs can express HC-related marker proteins, they still do not meet the standard of typical HC mature structure and functions. Therefore, new methods and technologies are needed to intervene in the process of HC regeneration. Since hundreds of transcripts are simultaneously regulated by miRNAs, miRNAs can be considered potential therapeutic tools for the manipulation of the processes of cell differentiation and the fate of hearing cells and the repair or the regeneration of HCs.

Acknowledgements

This work was supported by grants from the National Natural Science Foundation of China (Nos. 81873700, 81800915).

Disclosure of conflict of interest

None.

References

  • 1.Géléoc GS, Holt JR. Sound strategies for hearing restoration. Science. 2014;344:1241062. doi: 10.1126/science.1241062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Ma Y, Wise AK, Shepherd RK, Richardson RT. New molecular therapies for the treatment of hearing loss. Pharmacol Ther. 2019;200:190–209. doi: 10.1016/j.pharmthera.2019.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Roccio M, Senn P, Heller S. Novel insights into inner ear development and regeneration for refined hearing loss therapies. Hear Res. 2019:107859. doi: 10.1016/j.heares.2019.107859. [DOI] [PubMed] [Google Scholar]
  • 4.Chai R, Kuo B, Wang T, Liaw EJ, Xia A, Jan TA, Liu Z, Taketo MM, Oghalai JS, Nusse R, Zuo J, Cheng AG. Wnt signaling induces proliferation of sensory precursors in the postnatal mouse cochlea. Proc Natl Acad Sci U S A. 2012;109:8167–8172. doi: 10.1073/pnas.1202774109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Kelly MC, Chang Q, Pan A, Lin X, Chen P. Atoh1 directs the formation of sensory mosaics and induces cell proliferation in the postnatal mammalian cochlea in vivo. J Neurosci. 2012;32:6699–6710. doi: 10.1523/JNEUROSCI.5420-11.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Shi F, Hu L, Edge AS. Generation of hair cells in neonatal mice by β-catenin overexpression in Lgr5-positive cochlear progenitors. Proc Natl Acad Sci U S A. 2013;110:13851–13856. doi: 10.1073/pnas.1219952110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Doetzlhofer A, Basch ML, Ohyama T, Gessler M, Groves AK, Segil N. Hey2 regulation by FGF provides a Notch-independent mechanism for maintaining pillar cell fate in the organ of Corti. Dev Cell. 2009;16:58–69. doi: 10.1016/j.devcel.2008.11.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Dong H, Lei J, Ding L, Wen Y, Ju H, Zhang X. MicroRNA: function, detection, and bioanalysis. Chem Rev. 2013;113:6207–6233. doi: 10.1021/cr300362f. [DOI] [PubMed] [Google Scholar]
  • 9.He L, Hannon GJ. MicroRNAs: small RNAs with a big role in gene regulation. Nat Rev Genet. 2004;5:522. doi: 10.1038/nrg1379. [DOI] [PubMed] [Google Scholar]
  • 10.Wang Y, Keys DN, Au-Young JK, Chen C. MicroRNAs in embryonic stem cells. J Cell Physiol. 2009;218:251–255. doi: 10.1002/jcp.21607. [DOI] [PubMed] [Google Scholar]
  • 11.Brennecke J, Hipfner DR, Stark A, Russell RB, Cohen SM. Bantam encodes a developmentally regulated microRNA that controls cell proliferation and regulates the proapoptotic gene hid in Drosophila. Cell. 2003;113:25–36. doi: 10.1016/s0092-8674(03)00231-9. [DOI] [PubMed] [Google Scholar]
  • 12.O’Connell RM, Rao DS, Chaudhuri AA, Baltimore D. Physiological and pathological roles for microRNAs in the immune system. Nat Rev Immunol. 2010;10:111. doi: 10.1038/nri2708. [DOI] [PubMed] [Google Scholar]
  • 13.Martin EC, Qureshi AT, Dasa V, Freitas MA, Gimble JM, Davis TA. MicroRNA regulation of stem cell differentiation and diseases of the bone and adipose tissue: perspectives on miRNA biogenesis and cellular transcriptome. Biochimie. 2016;124:98–111. doi: 10.1016/j.biochi.2015.02.012. [DOI] [PubMed] [Google Scholar]
  • 14.Son YH, Ka S, Kim AY, Kim JB. Regulation of adipocyte differentiation via microRNAs. Endocrinol Metab (Seoul) 2014;29:122–135. doi: 10.3803/EnM.2014.29.2.122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Ha M, Kim VN. Regulation of microRNA biogenesis. Nat Rev Mol Cell Biol. 2014;15:509. doi: 10.1038/nrm3838. [DOI] [PubMed] [Google Scholar]
  • 16.Shenoy A, Blelloch RH. Regulation of microRNA function in somatic stem cell proliferation and differentiation. Nat Rev Mol Cell Biol. 2014;15:565. doi: 10.1038/nrm3854. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116:281–297. doi: 10.1016/s0092-8674(04)00045-5. [DOI] [PubMed] [Google Scholar]
  • 18.Wu L, Fan J, Belasco JG. MicroRNAs direct rapid deadenylation of mRNA. Proc Natl Acad Sci U S A. 2006;103:4034–4039. doi: 10.1073/pnas.0510928103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Djuranovic S, Nahvi A, Green R. miRNA-mediated gene silencing by translational repression followed by mRNA deadenylation and decay. Science. 2012;336:237–240. doi: 10.1126/science.1215691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Matzke MA, Mosher RA. RNA-directed DNA methylation: an epigenetic pathway of increasing complexity. Nat Rev Genet. 2014;15:394. doi: 10.1038/nrg3683. [DOI] [PubMed] [Google Scholar]
  • 21.Lewis BP, Burge CB, Bartel DP. Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell. 2005;120:15–20. doi: 10.1016/j.cell.2004.12.035. [DOI] [PubMed] [Google Scholar]
  • 22.Lee YS, Dutta A. MicroRNAs in cancer. Annu Rev Pathol. 2009;4:199–227. doi: 10.1146/annurev.pathol.4.110807.092222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Chen JF, Murchison EP, Tang R, Callis TE, Tatsuguchi M, Deng Z, Rojas M, Hammond SM, Schneider MD, Selzman CH, Meissner G, Patterson C, Hannon GJ, Wang DZ. Targeted deletion of dicer in the heart leads to dilated cardiomyopathy and heart failure. Proc Natl Acad Sci U S A. 2008;105:2111–2116. doi: 10.1073/pnas.0710228105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Lukiw WJ, Zhao Y, Cui JG. An NF-κB-sensitive micro RNA-146a-mediated inflammatory circuit in Alzheimer disease and in stressed human brain cells. J Biol Chem. 2008;283:31315–31322. doi: 10.1074/jbc.M805371200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Barh D, Malhotra R, Ravi B, Sindhurani P. MicroRNA let-7: an emerging next-generation cancer therapeutic. Curr Oncol. 2010;17:70. doi: 10.3747/co.v17i1.356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Wang LQ, Kwong YL, Kho CSB, Wong KF, Wong KY, Ferracin M, Calin GA, Chim CS. Epigenetic inactivation of miR-9 family microRNAs in chronic lymphocytic leukemia-implications on constitutive activation of NFκB pathway. Mol Cancer. 2013;12:173. doi: 10.1186/1476-4598-12-173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Wong KY, Yu L, Chim CS. DNA methylation of tumor suppressor miRNA genes: a lesson from the miR-34 family. Epigenomics. 2011;3:83–92. doi: 10.2217/epi.10.74. [DOI] [PubMed] [Google Scholar]
  • 28.Yang XW, Zhang LJ, Huang XH, Chen LZ, Su Q, Zeng WT, Li W, Wang Q. miR-145 suppresses cell invasion in hepatocellular carcinoma cells: miR-145 targets ADAM 17. Hepatol Res. 2014;44:551–559. doi: 10.1111/hepr.12152. [DOI] [PubMed] [Google Scholar]
  • 29.Andrew AS, Marsit CJ, Schned AR, Seigne JD, Kelsey KT, Moore JH, Perreard L, Karagas MR, Sempere LFJ. Expression of tumor suppressive micro RNA-34a is associated with a reduced risk of bladder cancer recurrence. Int J Cancer. 2015;137:1158–1166. doi: 10.1002/ijc.29413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Colangelo T, Fucci A, Votino C, Sabatino L, Pancione M, Laudanna C, Binaschi M, Bigioni M, Maggi CA, Parente D, Forte N, Colantuoni V. MicroRNA-130b promotes tumor development and is associated with poor prognosis in colorectal cancer. Neoplasia. 2013;15:1086. doi: 10.1593/neo.13998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Tang T, Wong HK, Gu W, Yu MY, To KF, Wang CC, Wong YF, Cheung TH, Chung TK, Choy KW. MicroRNA-182 plays an onco-miRNA role in cervical cancer. Gynecol Oncol. 2013;129:199–208. doi: 10.1016/j.ygyno.2012.12.043. [DOI] [PubMed] [Google Scholar]
  • 32.Wu Z, Liu K, Wang Y, Xu Z, Meng J, Gu S. Upregulation of microRNA-96 and its oncogenic functions by targeting CDKN1A in bladder cancer. Cancer Cell Int. 2015;15:107. doi: 10.1186/s12935-015-0235-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Škrha P, Hořínek A, Anděl M, Škrha J. miRNA-192, miRNA-21 and miRNA-200: new pancreatic cancer markers in diabetic patients? Vnitr Lek. 2015;61:351–354. [PubMed] [Google Scholar]
  • 34.Lee K, Kim H, An K, Kwon OB, Park S, Cha JH, Kim MH, Lee Y, Kim JH, Cho K, Kim HS. Replenishment of microRNA-188-5p restores the synaptic and cognitive deficits in 5XFAD mouse model of Alzheimer’s disease. Sci Rep. 2016;6:34433. doi: 10.1038/srep34433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Haftmann C, Stittrich AB, Zimmermann J, Fang Z, Hradilkova K, Bardua M, Westendorf K, Heinz GA, Riedel R, Siede J, Lehmann K, Weinberger EE, Zimmel D, Lauer U, Häupl T, Sieper J, Backhaus M, Neumann C, Hoffmann U, Porstner M, Chen W, Grün JR, Baumgrass R, Matz M, Löhning M, Scheffold A, Wittmann J, Chang HD, Rajewsky N, Jäck HM, Radbruch A, Mashreghi MF. miR-148a is upregulated by Twist1 and T-bet and promotes Th1-cell survival by regulating the proapoptotic gene Bim. Eur J Immunol. 2015;45:1192–1205. doi: 10.1002/eji.201444633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Porstner M, Winkelmann R, Daum P, Schmid J, Pracht K, Côrte-Real J, Schreiber S, Haftmann C, Brandl A, Mashreghi MF, Gelse K, Hauke M, Wirries I, Zwick M, Roth E, Radbruch A, Wittmann J, Jäck HM. miR-148a promotes plasma cell differentiation and targets the germinal center transcription factors Mitf and Bach2. Eur J Immunol. 2015;45:1206–1215. doi: 10.1002/eji.201444637. [DOI] [PubMed] [Google Scholar]
  • 37.Gonzalez-Martin A, Adams BD, Lai M, Shepherd J, Salvador-Bernaldez M, Salvador JM, Lu J, Nemazee D, Xiao C. The microRNA miR-148a functions as a critical regulator of B cell tolerance and autoimmunity. Nat Immunol. 2016;17:433. doi: 10.1038/ni.3385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.O’Connell RM, Kahn D, Gibson WS, Round JL, Scholz RL, Chaudhuri AA, Kahn ME, Rao DS, Baltimore D. MicroRNA-155 promotes autoimmune inflammation by enhancing inflammatory T cell development. Immunity. 2010;33:607–619. doi: 10.1016/j.immuni.2010.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Elmesmari A, Fraser AR, Wood C, Gilchrist D, Vaughan D, Stewart L, McSharry C, McInnes IB, Kurowska-Stolarska M. MicroRNA-155 regulates monocyte chemokine and chemokine receptor expression in rheumatoid arthritis. Rheumatology (Oxford) 2016;55:2056–2065. doi: 10.1093/rheumatology/kew272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Zhu S, Pan W, Song X, Liu Y, Shao X, Tang Y, Liang D, He D, Wang H, Liu W, Shi Y, Harley JB, Shen N, Qian Y. The microRNA miR-23b suppresses IL-17-associated autoimmune inflammation by targeting TAB2, TAB3 and IKK-α. Nat Med. 2012;18:1077. doi: 10.1038/nm.2815. [DOI] [PubMed] [Google Scholar]
  • 41.Huang F, Li ML, Fang ZF, Hu XQ, Liu QM, Liu ZJ, Tang L, Zhao YS, Zhou SH. Overexpression of MicroRNA-1 improves the efficacy of mesenchymal stem cell transplantation after myocardial infarction. Cardiology. 2013;125:18–30. doi: 10.1159/000347081. [DOI] [PubMed] [Google Scholar]
  • 42.van Mil A, Vrijsen KR, Goumans MJ, Metz CH, Doevendans PA, Sluijter JP. MicroRNA-1 enhances the angiogenic differentiation of human cardiomyocyte progenitor cells. J Mol Med (Berl) 2013;91:1001–1012. doi: 10.1007/s00109-013-1017-1. [DOI] [PubMed] [Google Scholar]
  • 43.Bai XY, Ma Y, Ding R, Fu B, Shi S, Chen XM. miR-335 and miR-34a promote renal senescence by suppressing mitochondrial antioxidative enzymes. J Am Soc Nephrol. 2011;22:1252–1261. doi: 10.1681/ASN.2010040367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Cunningham LL, Tucci DL. Hearing loss in adults. N Engl J Med. 2017;377:2465–2473. doi: 10.1056/NEJMra1616601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Yueh B, Shapiro N, MacLean CH, Shekelle PG. Screening and management of adult hearing loss in primary care: scientific review. JAMA. 2003;289:1976–1985. doi: 10.1001/jama.289.15.1976. [DOI] [PubMed] [Google Scholar]
  • 46.Matsunami T, Suzuki T, Hisa Y, Takata K, Takamatsu T, Oyamada M. Gap junctions mediate glucose transport between GLUT1-positive and-negative cells in the spiral limbus of the rat cochlea. Cell Commun Adhes. 2006;13:93–102. doi: 10.1080/15419060600631805. [DOI] [PubMed] [Google Scholar]
  • 47.Lee S, Ju H, Choi J, Ahn Y, Lee S, Seo YJ. Circulating serum miRNA-205 as a diagnostic biomarker for ototoxicity in mice treated with aminoglycoside antibiotics. Int J Mol Sci. 2018;19:2836. doi: 10.3390/ijms19092836. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Berrettini S, De Vito A, Bruschini L, Fortunato S, Forli F. Idiopathic sensorineural hearing loss in the only hearing ear. Acta Otorhinolaryngol Ital. 2016;36:119. doi: 10.14639/0392-100X-587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Friedman LM, Avraham KB. MicroRNAs and epigenetic regulation in the mammalian inner ear: implications for deafness. Mamm Genome. 2009;20:581–603. doi: 10.1007/s00335-009-9230-5. [DOI] [PubMed] [Google Scholar]
  • 50.Mencía A, Modamio-Høybjør S, Redshaw N, Morín M, Mayo-Merino F, Olavarrieta L, Aguirre LA, del Castillo I, Steel KP, Dalmay T, Moreno F, Moreno-Pelayo MA. Mutations in the seed region of human miR-96 are responsible for nonsyndromic progressive hearing loss. Nat Genet. 2009;41:609. doi: 10.1038/ng.355. [DOI] [PubMed] [Google Scholar]
  • 51.Patel M, Cai Q, Ding D, Salvi R, Hu Z, Hu BH. The miR-183/Taok1 target pair is implicated in cochlear responses to acoustic trauma. PLoS One. 2013;8:e58471. doi: 10.1371/journal.pone.0058471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Xiong H, Pang J, Yang H, Dai M, Liu Y, Ou Y, Huang Q, Chen S, Zhang Z, Xu Y, Lai L, Zheng Y. Activation of miR-34a/SIRT1/p53 signaling contributes to cochlear hair cell apoptosis: implications for age-related hearing loss. Neurobiol Aging. 2015;36:1692–1701. doi: 10.1016/j.neurobiolaging.2014.12.034. [DOI] [PubMed] [Google Scholar]
  • 53.Zhang Z, Liu K, Chen Y, Li Z, Yan N, Zhang J. The expression of miR-183 family in the pathogenesis and development of noise-induced deafness. Lin Chung Er Bi Yan Hou Tou Jing Wai Ke Za Zhi. 2014;28:468–472. [PubMed] [Google Scholar]
  • 54.Li H, Fekete DM. MicroRNAs in hair cell development and deafness. Curr Opin Otolaryngol Head Neck Surg. 2010;18:459. doi: 10.1097/MOO.0b013e32833e0601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Tewary M, Shakiba N, Zandstra PW. Stem cell bioengineering: building from stem cell biology. Nat Rev Genet. 2018;19:595–614. doi: 10.1038/s41576-018-0040-z. [DOI] [PubMed] [Google Scholar]
  • 56.Henig I, Zuckerman T. Hematopoietic stem cell transplantation-50 years of evolution and future perspectives. Rambam Maimonides Med J. 2014;5:e0028. doi: 10.5041/RMMJ.10162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Hirsch T, Rothoeft T, Teig N, Bauer JW, Pellegrini G, De Rosa L, Scaglione D, Reichelt J, Klausegger A, Kneisz D, Romano O, Secone Seconetti A, Contin R, Enzo E, Jurman I, Carulli S, Jacobsen F, Luecke T, Lehnhardt M, Fischer M, Kueckelhaus M, Quaglino D, Morgante M, Bicciato S, Bondanza S, De Luca M. Regeneration of the entire human epidermis using transgenic stem cells. Nature. 2017;551:327. doi: 10.1038/nature24487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Jiang L, Romero-Carvajal A, Haug JS, Seidel CW, Piotrowski T. Gene-expression analysis of hair cell regeneration in the zebrafish lateral line. Proc Natl Acad Sci U S A. 2014;111:E1383–E1392. doi: 10.1073/pnas.1402898111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Oesterle EC, Tsue TT, Reh TA, Rubel EW. Hair-cell regeneration in organ cultures of the postnatal chicken inner ear. Hear Res. 1993;70:85–108. doi: 10.1016/0378-5955(93)90054-5. [DOI] [PubMed] [Google Scholar]
  • 60.Cox BC, Chai R, Lenoir A, Liu Z, Zhang L, Nguyen DH, Chalasani K, Steigelman KA, Fang J, Rubel EW, Cheng AG, Zuo J. Spontaneous hair cell regeneration in the neonatal mouse cochlea in vivo. Development. 2014;141:816–829. doi: 10.1242/dev.103036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Bramhall NF, Shi F, Arnold K, Hochedlinger K, Edge AS. Lgr5-positive supporting cells generate new hair cells in the postnatal cochlea. Stem Cell Reports. 2014;2:311–322. doi: 10.1016/j.stemcr.2014.01.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Pinto D, Clevers H. Wnt, stem cells and cancer in the intestine. Biol Cell. 2005;97:185–196. doi: 10.1042/BC20040094. [DOI] [PubMed] [Google Scholar]
  • 63.Kuwabara T, Hsieh J, Muotri A, Yeo G, Warashina M, Lie DC, Moore L, Nakashima K, Asashima M, Gage FH. Wnt-mediated activation of NeuroD1 and retro-elements during adult neurogenesis. Nat Neurosci. 2009;12:1097. doi: 10.1038/nn.2360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Demitrack ES, Gifford GB, Keeley TM, Carulli AJ, VanDussen KL, Thomas D, Giordano TJ, Liu Z, Kopan R, Samuelson LC. Notch signaling regulates gastric antral LGR5 stem cell function. EMBO J. 2015;34:2522–2536. doi: 10.15252/embj.201490583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Jeon SJ, Fujioka M, Kim SC, Edge AS. Notch signaling alters sensory or neuronal cell fate specification of inner ear stem cells. J Neurosci. 2011;31:8351–8358. doi: 10.1523/JNEUROSCI.6366-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Mizutari K, Fujioka M, Hosoya M, Bramhall N, Okano HJ, Okano H, Edge AS. Notch inhibition induces cochlear hair cell regeneration and recovery of hearing after acoustic trauma. Neuron. 2013;77:58–69. doi: 10.1016/j.neuron.2012.10.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Zheng JL, Gao WQ. Overexpression of Math1 induces robust production of extra hair cells in postnatal rat inner ears. Nat Neurosci. 2000;3:580. doi: 10.1038/75753. [DOI] [PubMed] [Google Scholar]
  • 68.Shu Y, Li W, Huang M, Quan YZ, Scheffer D, Tian C, Tao Y, Liu X, Hochedlinger K, Indzhykulian AA, Wang Z, Li H, Chen ZY. Renewed proliferation in adult mouse cochlea and regeneration of hair cells. Nat Commun. 2019;10:1–15. doi: 10.1038/s41467-019-13157-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Ren J, Jin P, Wang E, Marincola FM, Stroncek DF. MicroRNA and gene expression patterns in the differentiation of human embryonic stem cells. J Transl Med. 2009;7:20. doi: 10.1186/1479-5876-7-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Makeyev EV, Maniatis T. Multilevel regulation of gene expression by microRNAs. Science. 2008;319:1789–1790. doi: 10.1126/science.1152326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Ivey KN, Muth A, Arnold J, King FW, Yeh RF, Fish JE, Hsiao EC, Schwartz RJ, Conklin BR, Bernstein HS, Srivastava D. MicroRNA regulation of cell lineages in mouse and human embryonic stem cells. Cell Stem Cell. 2008;2:219–229. doi: 10.1016/j.stem.2008.01.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Yang B, Guo H, Zhang Y, Chen L, Ying D, Dong S. MicroRNA-145 regulates chondrogenic differentiation of mesenchymal stem cells by targeting Sox9. PLoS One. 2011;6:e21679. doi: 10.1371/journal.pone.0021679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Lee S, Yoon DS, Paik S, Lee KM, Jang Y, Lee JW. microRNA-495 inhibits chondrogenic differentiation in human mesenchymal stem cells by targeting Sox9. Stem Cells Dev. 2014;23:1798–1808. doi: 10.1089/scd.2013.0609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Guérit D, Brondello JM, Chuchana P, Philipot D, Toupet K, Bony C, Jorgensen C, Noël D. FOXO3A regulation by miRNA-29a controls chondrogenic differentiation of mesenchymal stem cells and cartilage formation. Stem Cells Dev. 2014;23:1195–1205. doi: 10.1089/scd.2013.0463. [DOI] [PubMed] [Google Scholar]
  • 75.Wu R, Tang Y, Zang W, Wang Y, Li M, Du Y, Zhao G, Xu Y. MicroRNA-128 regulates the differentiation of rat bone mesenchymal stem cells into neuron-like cells by Wnt signaling. Mol Cell Biochem. 2014;387:151–158. doi: 10.1007/s11010-013-1880-7. [DOI] [PubMed] [Google Scholar]
  • 76.Cai B, Li J, Wang J, Luo X, Ai J, Liu Y, Wang N, Liang H, Zhang M, Chen N, Wang G, Xing S, Zhou X, Yang B, Wang X, Lu Y. microRNA-124 regulates cardiomyocyte differentiation of bone marrow-derived mesenchymal stem cells via targeting STAT3 signaling. Stem Cells. 2012;30:1746–1755. doi: 10.1002/stem.1154. [DOI] [PubMed] [Google Scholar]
  • 77.Xu H, Yang YJ, Qian HY, Tang YD, Wang H, Zhang QJ. Rosuvastatin treatment activates JAK-STAT pathway and increases efficacy of allogeneic mesenchymal stem cell transplantation in infarcted hearts. Circ J. 2011;75:1476–1485. doi: 10.1253/circj.cj-10-1275. [DOI] [PubMed] [Google Scholar]
  • 78.Jing L, Jia Y, Lu J, Han R, Li J, Wang S, Peng T, Jia Y. MicroRNA-9 promotes differentiation of mouse bone mesenchymal stem cells into neurons by notch signaling. Neuroreport. 2011;22:206–211. doi: 10.1097/WNR.0b013e328344a666. [DOI] [PubMed] [Google Scholar]
  • 79.Han R, Kan Q, Sun Y, Wang S, Zhang G, Peng T, Jia Y. MiR-9 promotes the neural differentiation of mouse bone marrow mesenchymal stem cells via targeting zinc finger protein 521. Neurosci Lett. 2012;515:147–152. doi: 10.1016/j.neulet.2012.03.032. [DOI] [PubMed] [Google Scholar]
  • 80.Miyoshi N, Ishii H, Nagano H, Haraguchi N, Dewi DL, Kano Y, Nishikawa S, Tanemura M, Mimori K, Tanaka F, Saito T, Nishimura J, Takemasa I, Mizushima T, Ikeda M, Yamamoto H, Sekimoto M, Doki Y, Mori M. Reprogramming of mouse and human cells to pluripotency using mature microRNAs. Cell Stem Cell. 2011;8:633–638. doi: 10.1016/j.stem.2011.05.001. [DOI] [PubMed] [Google Scholar]
  • 81.Anokye-Danso F, Trivedi CM, Juhr D, Gupta M, Cui Z, Tian Y, Zhang Y, Yang W, Gruber PJ, Epstein JA, Morrisey EE. Highly efficient miRNA-mediated reprogramming of mouse and human somatic cells to pluripotency. Cell Stem Cell. 2011;8:376–388. doi: 10.1016/j.stem.2011.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Choi YJ, Lin CP, Ho JJ, He X, Okada N, Bu P, Zhong Y, Kim SY, Bennett MJ, Chen C, Ozturk A, Hicks GG, Hannon GJ, He L. miR-34 miRNAs provide a barrier for somatic cell reprogramming. Nat Cell Biol. 2011;13:1353. doi: 10.1038/ncb2366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Li H, Kloosterman W, Fekete DM. MicroRNA-183 family members regulate sensorineural fates in the inner ear. J Neurosci. 2010;30:3254–3263. doi: 10.1523/JNEUROSCI.4948-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Friedland DR, Eernisse R, Erbe C, Gupta N, Cioffi JA. Cholesteatoma growth and proliferation: post-transcriptional regulation by microRNA-21. Otol Neurotol. 2009;30:998. doi: 10.1097/MAO.0b013e3181b4e91f. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Wang XR, Zhang XM, Du J, Jiang H. MicroRNA-182 regulates otocyst-derived cell differentiation and targets T-box1 gene. Hear Res. 2012;286:55–63. doi: 10.1016/j.heares.2012.02.005. [DOI] [PubMed] [Google Scholar]
  • 86.Chen C, Xiang H, Peng Yl, Peng J, Jiang SW. Mature miR-183, negatively regulated by transcription factor GATA3, promotes 3T3-L1 adipogenesis through inhibition of the canonical Wnt/β-catenin signaling pathway by targeting LRP6. Cell Signal. 2014;26:1155–1165. doi: 10.1016/j.cellsig.2014.02.003. [DOI] [PubMed] [Google Scholar]
  • 87.Zhou W, Du J, Jiang D, Wang X, Chen K, Tang H, Zhang X, Cao H, Zong L, Dong C, Jiang H. microRNA-183 is involved in the differentiation and regeneration of notch signaling-prohibited hair cells from mouse cochlea. Mol Med Rep. 2018;18:1253–1262. doi: 10.3892/mmr.2018.9127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Li W, Wu J, Yang J, Sun S, Chai R, Chen ZY, Li H. Notch inhibition induces mitotically generated hair cells in mammalian cochleae via activating the Wnt pathway. Proc Natl Acad Sci U S A. 2015;112:166–171. doi: 10.1073/pnas.1415901112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Chen Z, Pu M, Yao J, Cao X, Cheng L. Screening of microRNAs targeting Notch signaling pathway implicated in inner ear development and the role of microRNA-384-5p. Zhonghua Er Bi Yan Hou Tou Jing Wai Ke Za Zhi. 2018;53:830–837. doi: 10.3760/cma.j.issn.1673-0860.2018.11.007. [DOI] [PubMed] [Google Scholar]

Articles from American Journal of Stem Cells are provided here courtesy of e-Century Publishing Corporation

RESOURCES