Open Access

Coordinated microRNA/mRNA expression profiles reveal a putative mechanism of corneal epithelial cell transdifferentiation from skin epidermal stem cells

  • Authors:
    • Yanjie Guo
    • Xiya Ma
    • Weini Wu
    • Mingyan Shi
    • Junlong Ma
    • Yaping Zhang
    • Erkang Zhao
    • Xueyi Yang
  • View Affiliations

  • Published online on: December 1, 2017     https://doi.org/10.3892/ijmm.2017.3304
  • Pages: 877-887
  • Copyright: © Guo et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Skin epidermal stem cells (SESCs), which share a single origin with corneal epithelial cells (CECs), are considered to be one of the most ideal seed cells for the construction of tissue engineered corneas. However, the mechanism underlying the transdifferentiation of SESCs to CECs has not been fully elucidated. In the present study, to identify critical microRNAs (miRNAs/miRs) and genes that regulate the transdifferentiation of SESCs to CECs, SESCs and CECs were collected from sheep and used for small RNA sequencing and mRNA microarray analyses. Among the differentially expressed miRNAs and genes, 36 miRNAs were downregulated and 123 genes were upregulated in the CECs compared with those in the SESCs. miR‑10b exhibited the largest change in expression between the cell types. Target genes of the 36 downregulated miRNAs were predicted and a computational approach demonstrated that these target genes may be involved in several signaling pathways, including the ‘PI3K signaling pathway’, the ‘Wnt signaling pathway’ and the ‘MAPK signaling pathway’, as well as in ‘focal adhesion’. Comparison of these target genes to the 123 upregulated genes identified 43 intersection genes. A regulatory network of these 43 intersection genes and its correlative miRNAs were constructed, and three genes (dedicator of cytokinesis 9, neuronal differentiation 1 and activated leukocyte cell adhesion molecule) were found to have high interaction frequencies. The expression levels of 7 randomly selected miRNAs and the 3 intersection genes were further validated by reverse transcription-quantitative polymerase chain reaction. It was found that miR‑10b, the Wnt signaling pathway and the 3 intersection genes may act together and serve a critical role in the transdifferentiation process. This study identified miRNAs and genes that were expressed in SESCs and CECs that may assist in uncovering its underlying molecular mechanism, as well as promote corneal tissue engineering using epidermal stem cells for clinical applications.

Introduction

The corneal epithelium (CE) is the exfoliated multilayer epithelium that forms the anterior surface layer of the cornea (1). Corneal epithelial cell (CEC) homeostasis is a requirement for both ocular surface integrity and corneal transparency and visual function (2). Corneal damage, including infection, trauma and denaturation, have severe consequences for the ocular surface, which in turn may affect the integrity of the CE and visual function, ultimately leading to functional blindness (3). There were approximately 4.9–10 million people with corneal blindness worldwide in 2012 (4). One of the most successful strategies for ocular surface reconstruction is the transplantation of epithelial cell sheets engineered from autologous limbal epithelial cells (1,5,6); it is effective in animal models and humans. However, a limbal biopsy is required for the isolation of limbal epithelial cells, which may pose a risk to the contralateral healthy eye and is not useful for treating bilateral limbal stem cell deficiency. Allogeneic limbal epithelium transplantation (7), one optional treatment, has limitations of immunological rejection, donor scarcity and an extremely low long‑term success rate. Thus, it is reasonable to find and evaluate alternative autologous stem cell sources for in vitro culture and transplantation to avoid the risk of immunological rejection and to satisfy the demand from patients with corneal blindness.

Other cell sources, including oral mucosal epithelial cells (810), mesenchymal stem cells (11,12), embryonic stem cells (13) and immature dental pulp stem cells (14), have been established for corneal epithelium replacement, but to date, no long-term results have been reported or achieved. Stem cells residing within the epidermis have the potential of multi-directional differentiation. In addition to differentiating into skin cells and appendages, skin epidermal stem cells (SESCs) can differentiate into other lineages, including nerve cells (15) and germ cells (16). Moreover, SESCs and limbal stem cells are of the same origin, namely, the basal layer of embryonic skin, and are thus keratinocyte stem cells (17,18), which express specific types of cytokeratin protein (K1, K3, K5, K10, K12, K14, K15 and K19) on their surfaces (1921). It is hypothesized that SESCs are ideal seed cells that may be utilized to replace limbal stem cells for tissue engineered cornea construction. There have been certain attempts to use hair follicle stem cells (22) and skin-derived precursor cells (2,23); however, the mechanisms involved, particularly the regulatory role of miRNAs in this process, have not been fully elucidated. In the present study, the expression profile of microRNAs (miRNAs/miRs) was compared in the SESCs and CECs of sheep. The findings of this study may be useful in the elucidation of the mechanisms underlying the molecular regulation of the transdifferentiation process of SESCs into CECs.

Materials and methods

Cells

SESCs and CECs were obtained from 2 male small-tailed Han sheep (obtained from the Experimental Animal Center of Shaanxi Center for Stem Cell Engineering and Technology, Northwest A&F University) following a protocol approved by the Medical Animal Care and Welfare Committee of Luoyang Normal University. The isolation of CECs and SESCs were performed as previously described (24,25).

Small RNA sequencing

Total RNA (3 µg) from the SESCs and CECs was used as input material for the generation of small RNA libraries. Sequencing libraries were generated using the NEBNext® Multiplex Small RNA Library Prep Set for Illumina® (New England Biolabs, Inc., Ipswich, MA, USA) following the manufacturer's instructions, and index codes were added to attribute sequences to each sample. The clustering of the index-coded samples was performed on a cBot Cluster Generation System using TruSeq SR Cluster kit v3-cBot-HS (Illumina, Inc., San Diego, CA, USA), according to the manufacturer's instructions. Following cluster generation, the library preparations were sequenced on an Illumina HiSeq 2500/2000 platform (Illumina, Inc.) and 50-bp single-end reads were generated.

The raw sequence reads were first trimmed from the adapter sequence using a Perl script and adapter-trimmed reads longer than 18 nt were used for further analysis as clean reads. To further parse the clean reads, the Integrative Short Reads Navigator software (freely accessed at http://omicslab.genetics.ac.cn/ISRNA/) was used based on the manufacturer's instructions. Bowtie software (version 1.1.2, open-source software maintained by Johns Hopkins University) was used to map sequences to sheep genomes (Ovis aries). Annotation of the short sequence reads was performed using the BLAST + program (ftp://ftp.ncbi.nlm.nih.gov/blast/executables/blast+/LATEST/) and the Rfam database (http://rfam.xfam.org/). Known miRNA sequences were extracted from miRBase (http://www.mirbase.org/). The frequency of miRNA reads was normalized as transcripts per million, then the normalized expression levels of miRNAs between two samples were conducted to calculate fold-changes. The identification of differentially expressed miRNAs between two different samples was performed based on the log10 ratio of fold-changes. Hierarchical clustering was performed using Heatmap.2 (R language gplots package). The levels of miRNA expression were clustered using average linkage clustering. The average value of the distance of each miRNA was used to measure cluster-to-cluster distance.

mRNA microarray analysis

The transcriptional profiles of the SESCs and CECs were analyzed. Total RNA was extracted and purified using the miRNeasy mini kit (Qiagen GmBH, Hilden, Germany) following the manufacturer's instructions and an RIN number was checked for to determine RNA integrity using an Agilent Bioanalyzer 2100 (Agilent Technologies, Inc., Santa Clara, CA, USA). Total RNA was amplified and labeled using a Low Input Quick Amp Labeling kit, One‑Color (Agilent Technologies, Inc.), following the manufacturer's instructions. Labeled cRNAs were purified using an RNeasy mini kit (Qiagen GmBH). Each slide was hybridized with 600 ng Cy3-labeled cRNA using a gene expression hybridization kit in a hybridization oven (both Agilent Technologies, Inc.), according to the manufacturer's instructions. After 17 h of hybridization, slides were washed in staining dishes (Thermo Fisher Scientific, Inc., Waltham, MA, USA) with a gene expression wash buffer kit (Agilent Technologies, Inc.), following the manufacturer's instructions. Slides were scanned on a Agilent Microarray Scanner (Agilent Technologies) using the following default settings: Dye channel, green; scan resolution, 5 µm; PMT, 100 and 10%, 16-bit. Data were extracted using Feature Extraction 10.7 (Agilent Technologies, Inc.). Raw data were normalized using a Quantile algorithm, Gene Spring Software 11.0 (Agilent Technologies, Inc.).

Target prediction and network construction

As there was no appropriate database or methods for predicting sheep miRNA target genes, the human orthologs of differentially expressed sheep miRNA sequences were used to identify potential target genes by searching the TargetScan database (http://www.targetscan.org/vert_71/). JAVA (version 1.8.0_101; Oracle Software Systems Ltd., Redwood Shore, CA, USA) and Cytoscape 2.6.0 (version 2.6.0, open-source software) software were used to construct regulatory miRNA-target gene networks based on regulatory interactions between differentially expressed miRNAs.

Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses

GO (http://geneon-tology.org/) and KEGG (http://www.kegg.jp/) pathway analyses of differentially expressed genes were performed using the Database for Annotation, Visualization and Integrated Discovery (DAVID; https://david.ncifcrf.gov/); a hypergeometric test with the Benjamini and Hochberg false discovery rate (FDR) was performed using the default parameters to adjust the P-value. A bubble chart was plotted using the OmicShare tools, a free online platform for data analysis (www.omicshare.com/tools).

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

RT-qPCR was performed to validate the expression levels of the analyzed miRNAs by sequencing. Reverse transcription was performed in a 20-µl reaction containing 2 µl total RNA, 1 µl of 5 µM RT primer, 1 µl of 10 mM dNTPs, 4 µl 5X PrimeScript buffer and 200 units of PrimeScript RTase (Takara Bio, Inc., Otsu, Japan). The primers used were as follows: U6 forward, 5′-CTCGCTTCGGCAGCACA-3′ and reverse, 5′-AACGCTTCACGAATTTGCGT-3′; miR-127 forward, 5′-ACACTCCAGCTGGGCTGAAGCTCAGA GGG-3′; miR-376c forward, 5′-ACACTCCAGCTGGGAACATAGAGGAAA-3′; miR-382 forward, 5′-ACACTCCAGCTGGGAATCATTCACGGAC-3′; miR-758 forward, 5′-ACACTCCAGCTGGGTTTGTGACCTGGTC-3′; miR-154 forward, 5′-ACACTCCAGCTGGGTAGGTTATCCGTGTTG-3′; miR-409 forward, 5′-ACACTCCAGCTGGGGAATGTTGCTCGGTG-3′; miR-10b forward, 5′-ACACTCCAGCTGGGTACCCTGTAGAACCGAA-3′ (all miRs had the same reverse primer, 5′-TGGTGTCGTGGAGTCG-3′); DOCK9 forward, 5′-TTAAGGATGCATCTGGAAACC-3′ and reverse, 5′-CTTGAGCATGTCTTCATTGGA-3′; NEUROD1 forward, 5′-CTTCGATAGCCATTCACATCAT-3′ and reverse, 5′-TGAAACTGACGTGCCTCTAATC-3′; ALCAM forward, 5′-CTTCAGCAGCCATCACAGTT-3′ and reverse, 5′-AGAGCATCGCCTATCTGCTT-3′; GAPDH forward, 5′-CTGGCCAAGGTCATCCAT-3′ and reverse, 5′-ACAGTCTTCTGGGTGGCAGT-3′. The reaction was incubated at 42°C for 60 min, and then terminated by heating at 85°C for 5 min. The qPCR amplification was performed using SYBR-Green I® (Roche Diagnostics, Indianapolis, IN, USA). The reactions were performed for 35 cycles at 94°C for 30 sec, 54–60°C for 30 sec, and 72°C for 30 sec. All reactions were run in triplicate. U6 small nuclear RNA was used as an internal reference control, and the data were analyzed using the 2−ΔΔCt method (26).

Statistical analysis

Data were analyzed using SPSS software (version 22.0; SPSS, Inc., Chicago, IL, USA). All measurement data are presented as the mean ± standard deviation. Statistical significance analysis of involved measurement data was performed using paired Student's t-tests. P<0.05 was considered to indicate a statistically significant difference.

Results

Overview of small RNA sequencing data

To identify differentially expressed miRNAs in the SESCs and CECs of sheep, two small RNA libraries were constructed by deep sequencing. The results of 14.36 million (M) and 13.71 M total reads were obtained from the cell libraries of SESCs and CECs, respectively. Subsequent to removing the low-quality and adaptor sequences, a total of 14.15 and 13.48 M clean reads were ultimately obtained. Subsequently, all identical sequence reads were classified as groups, and 0.21 and 0.22 M unique sequences associated with individual sequence reads, respectively, were obtained (Table I). The size distribution of the reads was similar between the two libraries (Fig. 1). The majority of the small RNAs were 21–24 nt in size. Sequences 22 nt in length, the typical size of Dicer-derived products, accounted for 45.47 and 35.85% of the total sequence reads in the SESC and CEC libraries, respectively. The composition of the RNA classes in each library is shown in Fig. 2A. Conserved miRNAs accounted for 95.88 and 95.01% of the total sequence reads (Fig. 2A) in the SESC and CEC small RNA libraries, respectively. The detection of a low proportion of long RNAs, including mRNA (1.02 and 1.03%), rRNA (0.54 and 0.63%) and snoRNA (1.06 and 2.32%) (Fig. 2A), in the respective SESC and CEC small RNA libraries indicated that the samples were not contaminated by degraded RNA. In addition, the highest fraction of unique reads (63 and 65% in the SESC and CEC libraries, respectively) was attributed to unannotated sequences (Fig. 2B). A total of 136 and 94 conserved miRNAs were identified in the SESC and CEC libraries, respectively; 184 and 169 miRNAs were mapped to the sheep genome, with the extended genome sequences having the potential to form hairpins.

Table I

Statistics based on the reads of the sequencing data.

Table I

Statistics based on the reads of the sequencing data.

ReadsCorneal epithelial cell countSkin epidermal stem cell count
Raw reads13,708,25714,358,739
Clean reads13,485,77014,152,453
Annotated reads11,289,22711,969,113
 with miRBase5,774,8596,283,880
 with piRNA127,061155,238
 with Rfam v.104,520,5794,752,923
 with ncRNA866,728777,072
Unannotated reads2,196,5432,183,340
Novel miRNA832,45853,42

[i] miRNA, microRNA.

Differential expression of miRNAs and mRNA in SESCs and CECs

To identify miRNAs that may be involved in the transdifferentiation process of SESCs to CECs, the miRNA and mRNA expression profiles were assessed in these two cells using small RNA deep sequencing and microarray analysis, respectively. The threshold used to screen upregulated or downregulated miRNAs and mRNAs was a fold‑change of ≥2.0. A total of 37 miRNAs were found to be differentially expressed (Fig. 3). Among these miRNAs, 36 were found to be downregulated by <0.5-fold, and 1 (miR-143) was found to be upregulated >2.0-fold in SESCs compared with that in CECs (Fig. 3A). The fold-change of downregulated miRNAs ranged from −425.71 to −3.34, and miR-10b exhibited the greatest decrease in expression in SESCs compared with CECs. Hierarchical clustering was performed with normalized miRNA data (fold-change >2). A total of 37 miRNAs were identified whose expression was significantly altered in the CECs and SESCs (Fig. 3B). Differential expressions of miRNAs and mRNAs were identified as those meeting the criteria of FDR <0.05 and |log2FoldChange| >1.

A total of 241 mRNAs were found to be differentially expressed. Among these mRNAs, 123 were downregulated by <0.5-fold and 118 were upregulated by >2.0-fold in SESCs compared with that in CECs. The fold-change ranged from −62.50 to −2.02 and from 2.00 to 155.76 for the downregulated and upregulated genes in SESCs compared with that in CECs, respectively.

RT-qPCR confirmation of differential miRNA expression

A total of 7 differentially expressed miRNAs (Fig. 4) were randomly chosen for validation by RT-qPCR using the same RNA samples that were applied to the small RNA sequencing. The expression levels of all 7 miRNAs determined by RT-qPCR were concordant with their small RNA sequencing data (Fig. 4), indicating a strong association between miRNA profiling and the RT-qPCR data.

Target prediction and functional analysis of differentially expressed miRNAs

As aforementioned, 36 miRNAs were found to be downregulated in the SESCs compared those in the CECs, and candidate target genes for all downregulated miRNAs were identified using the commonly used prediction algorithm, TargetScan. These targets were sorted by the enrichment of GO categories based on DAVID and mainly clustered into different functional groups (Fig. 5A). Targets were then functionally analyzed using the KEGG pathways database. Pathway analyses showed that these target genes were apparently associated with several pathways, including the 'PI3K-Akt signaling pathway', the 'Wnt signaling pathway', 'focal adhesion', the 'ErbB signaling pathway' and the 'MAPK signaling pathway' (Fig. 5B).

Intersection genes between differentially expressed miRNA target genes and differentially expressed genes

As previously described, a total of 9,660 genes were predicted by TargetScan for 36 downregulated miRNAs, and microarray analysis determined that 123 genes were expressed at higher levels in CECs than in SESCs. These two set of genes were compared and 43 intersection genes were detected. Notably, 24 miRNAs of the 36 downregulated miRNAs were associated with these 43 intersection genes. These findings suggested that these miRNAs likely regulate the same biological process.

Function analysis of differentially expressed miRNA target genes and intersection genes
GO enrichment and KEGG pathway analyses

The intersection genes were sorted by the enrichment of GO categories based on DAVID and mainly clustered into different functional groups (Fig. 6A), and then were functionally analyzed using the KEGG pathways database. Fig. 6B shows 10 of the most significant pathways.

Construction of a regulatory and interaction network

A regulatory network was performed using the differentially expressed miRNAs and its target genes (Fig. 7). As previously described, 24 miRNAs of the 36 downregulated miRNAs were associated with these 43 intersection genes. A regulatory network of these 24 miRNAs and 43 intersection genes was then constructed using Cytoscape (Fig. 8). It was observed that dedicator of cytokinesis 9 (DOCK9), neuronal differentiation 1 (NEUROD1) and activated leukocyte cell adhesion molecule (ALCAM) had more interaction with the miRNAs, and RT-qPCR confirmed that these 3 genes were upregulated in CECs compared with that in SESCs (Fig. 4B). DOCK9, NEUROD1 and ALCAM may thus serve an important role in CEC homeostasis and in SESC to CEC transdifferentiation.

Discussion

The present study elucidated a coordinated miRNA/mRNA expression profile for SESCs and CECs. miRNAs, although non-coding, play a critical role by binding to the 3′-untranslated region (3′UTR) of target genes for post-transcriptional regulation (27). When the expression profile of miRNAs was analyzed in the present study, it was found that miR-10b exhibited the greatest decrease in expression in SESCs compared with that in CECs (Figs. 3 and 4A). miR-10b is considered as a marker of cell pathological transformation, particularly the transformation and migration of malignant tumors (28). A growing number of studies have indicated that miR-10b expression is markedly increased in malignant tumors, including breast cancer (29), pancreatic cancer (30), esophageal cancer (31) and glioblastoma (32). Thus, miR-10b may serve a critical role in cell pathological transformation, and is considered as a target for cancer therapy of, for example, breast cancer and neurofibromatosis (33,34). In pathological conditions, CECs convert into skin-like epithelial cells, which in turn may lead to a loss in corneal transparency (2,3); however, the underlying mechanism remains largely unknown. In this process, the expression of paired box protein Pax-6 (PAX6), a critical transcription factor during eye development, is markedly decreased (3). Analysis of the 3′UTR region of PAX6 indicated that this gene is one of the potential targets of miR-10b (data not shown), and as a tumor suppressor, is suppressed by miR-10b in gliomagenesis (35). Thus, miR-10b may serve an important role in homeostasis and the pathogenesis of CECs through the suppression of PAX6 expression.

KEGG analysis indicated that the Wnt signaling pathway is one of the most significant pathways involved in the differential expression of miRNA target genes. SESCs and limbal stem cells originate from the basal layer of embryonic skin (17,18), which develops into different cell types in response to different micro-environments. The Wnt signal has emerged as the predominant pathway in the dermal condensation message that determines whether the basal layer of embryonic skin should differentiate into skin and its appendages (36), as well as subsequently controlling the function of differentiated skin cells (37). CECs transdifferentiate into skin-like epithelial cells when situated in a pathological condition or induced by dermal developmental signals (2,38). The expression level of β-catenin then sharply increases, indicating that the Wnt signaling pathway has been activated (2,38). Moreover, knockdown of the Wnt signaling inhibitor dickkopf-related protein 2 promotes the differentiation of the corneal epithelia into skin-like epithelial cells and the development of appendages such as hair follicles (39). Thus, the Wnt signaling pathway triggers the pathological transdifferentiation of corneal epithelia into skin-like epithelia and the development, formation and maintenance of the corneal epithelium, which is required to inhibit Wnt signals. It is also possible that the inhibition of a Wnt signal promotes the trans-differentiation of SESCs into CECs.

The present study found 3 intersection genes, DOCK9, NEUROD1 and ALCAM, which had more interactions with miRNAs. DOCK9 (OMIM 607325) encodes a DOCK protein family member that has GTP/GDP exchange factor activity and specifically activates G-protein cell division control protein 42 homolog (40). Furthermore, it has been reported as the most functionally significant gene for keratoconus (41). DOCK3, one of the members of the DOCK protein family, was found as a negative regulator of Wnt signaling, as it inhibited the nuclear expression of β-catenin (42). Therefore, DOCK9 may play a role in corneal epithelium homeostasis by inhibiting Wnt signaling. NEUROD1 is a member of the NeuroD family of basic helix-loop-helix transcription factors and activates the transcription of genes that contain a specific DNA sequence known as the E-box (43,44). There is evidence that PAX6, a critical transcription factor that defines corneal epithelium homeostasis and pathogenesis (2), is activated by NeuroD1 by binding the E-box in the PAX6 promoter (44). The present study found that NeuroD1 was upregulated in CECs, and thus may act as the upstream activator of PAX6 and maintain the characteristics of corneal epithelia. ALCAM, also known as cluster of differentiation (CD)166, serves an important role in mediating adhesion interactions in epithelial cells. In CD166-deficient cells, mRNA expression of the epithelial-mesenchymal transition (EMT) activator zinc finger E-box-binding homeobox 1 is overexpressed (45), which implies that CD166 deficiency may cause EMT in epithelial cells. MET is one of the pathological changes when homeostasis of the corneal epithelia is disturbed by infection, injury, epithelial hypertrophy or stem cell deficiency (46). Thus, ALCAM may maintain the homeostasis of corneal epithelium by regulating adhesion interactions in corneal epithelial cells. In conclusion, DOCK9, NEUROD1 and ALCAM, which are upregulated in CECs, may serve an important role in maintaining the homeostasis of corneal epithelia, and their overexpression induces the transdifferentiation of SESCs into corneal epithelial cells.

In conclusion (Fig. 9), the homeostasis of CECs is maintained by the sustained upregulation of PAX6, which can inhibit the Wnt/β-catenin signaling pathway, and this dermal condensation message causes epidermal changes in the CECs. The inhibitory effect of PAX6 may be enhanced by NEUROD1 and DOCK9. In SESCs, the expression of PAX6 is inhibited by miR-10b and the Wnt/β-catenin signaling pathway. Therefore, the transdifferentiation from SESCs to CECs may be realized by the following approach: i) Forced expression of PAX6 in SESCs; this approach has previously been demonstrated to be feasible (2). ii) Forced expression of DOCK9, NEUROD1, and ALCAM. iii) Inhibition of miR-10b expression in SESCs. iv) Inhibition of the Wnt/β-catenin signaling pathway in SESCs. PAX6, miR-10b and the Wnt/β-catenin signaling pathway are the critical factors that predominate the transdifferentiation process.

The present study investigated the miRNA and mRNA expression profiles of sheep SESCs and CECs. Coordinated miRNA/mRNA expression analysis revealed that miR-10b, DOCK9, NEUROD1 and the Wnt/β-catenin signaling pathway may be critical factors in the construction of a regulatory network to regulate corneal homeostasis and transdifferentiation from SESCs to CECs.

Acknowledgments

This study was supported by grants from the National Natural Science Foundation of China (nos. 31240089 and 31701121) and the Key Project of Higher Education Institutions in Henan Province (nos. 15A180008 and 16A180009).

References

1 

Pellegrini G, Traverso CE, Franzi AT, Zingirian M, Cancedda R and De Luca M: Long-term restoration of damaged corneal surfaces with autologous cultivated corneal epithelium. Lancet. 349:990–993. 1997. View Article : Google Scholar : PubMed/NCBI

2 

Ouyang H, Xue Y, Lin Y, Zhang X, Xi L, Patel S, Cai H, Luo J, Zhang M, Zhang M, et al: WNT7A and PAX6 define corneal epithelium homeostasis and pathogenesis. Nature. 511:358–361. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Li W, Chen YT, Hayashida Y, Blanco G, Kheirkah A, He H, Chen SY, Liu CY and Tseng SC: Down-regulation of Pax6 is associated with abnormal differentiation of corneal epithelial cells in severe ocular surface diseases. J Pathol. 214:114–122. 2008. View Article : Google Scholar

4 

Lamm V, Hara H, Mammen A, Dhaliwal D and Cooper DK: Corneal blindness and xenotransplantation. Xenotransplantation. 21:99–114. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Yang X, Moldovan I, Zhao Q, Mi S, Zhou Z, Chen D, Gao Z, Tong D and Dou Z: Reconstruction of damaged cornea by autologous transplantation of epidermal adult stem cells. Mol Vis. 14:1064–1070. 2008.PubMed/NCBI

6 

Shortt AJ, Secker GA, Notara MD, Limb GA, Khaw PT, Tuft SJ and Daniels JT: Transplantation of ex vivo cultured limbal epithelial stem cells: A review of techniques and clinical results. Surv Ophthalmol. 52:483–502. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Tseng SC, Prabhasawat P, Barton K, Gray T and Meller D: Amniotic membrane transplantation with or without limbal allografts for corneal surface reconstruction in patients with limbal stem cell deficiency. Arch Ophthalmol. 116:431–441. 1998. View Article : Google Scholar : PubMed/NCBI

8 

Nakamura T and Kinoshita S: Ocular surface reconstruction using cultivated mucosal epithelial stem cells. Cornea. 22(Suppl 7): S75–S80. 2003. View Article : Google Scholar

9 

Utheim TP, Utheim OA, Khan QE and Sehic A: Culture of oral mucosal epithelial cells for the purpose of treating limbal stem cell deficiency. J Funct Biomater. 7:52016. View Article : Google Scholar

10 

Nishida K, Yamato M, Hayashida Y, Watanabe K, Yamamoto K, Adachi E, Nagai S, Kikuchi A, Maeda N, Watanabe H, et al: Corneal reconstruction with tissue-engineered cell sheets composed of autologous oral mucosal epithelium. N Engl J Med. 351:1187–1196. 2004. View Article : Google Scholar : PubMed/NCBI

11 

Katikireddy KR, Dana R and Jurkunas UV: Differentiation potential of limbal fibroblasts and bone marrow mesenchymal stem cells to corneal epithelial cells. Stem Cells. 32:717–729. 2014. View Article : Google Scholar

12 

Gu S, Xing C, Han J, Tso MO and Hong J: Differentiation of rabbit bone marrow mesenchymal stem cells into corneal epithelial cells in vivo and ex vivo. Mol Vis. 15:99–107. 2009.PubMed/NCBI

13 

Ahmad S, Stewart R, Yung S, Kolli S, Armstrong L, Stojkovic M, Figueiredo F and Lako M: Differentiation of human embryonic stem cells into corneal epithelial-like cells by in vitro replication of the corneal epithelial stem cell niche. Stem Cells. 25:1145–1155. 2007. View Article : Google Scholar : PubMed/NCBI

14 

Gomes JA, Geraldes Monteiro B, Melo GB, Smith RL, Cavenaghi Pereira da Silva M, Lizier NF, Kerkis A, Cerruti H and Kerkis I: Corneal reconstruction with tissue-engineered cell sheets composed of human immature dental pulp stem cells. Invest Ophthalmol Vis Sci. 51:1408–1414. 2010. View Article : Google Scholar

15 

Toma JG, Akhavan M, Fernandes KJ, Barnabé-Heider F, Sadikot A, Kaplan DR and Miller FD: Isolation of multipotent adult stem cells from the dermis of mammalian skin. Nat Cell Biol. 3:778–784. 2001. View Article : Google Scholar : PubMed/NCBI

16 

Dyce PW, Wen L and Li J: In vitro germline potential of stem cells derived from fetal porcine skin. Nat Cell Biol. 8:384–390. 2006. View Article : Google Scholar : PubMed/NCBI

17 

Chee KY, Kicic A and Wiffen SJ: Limbal stem cells: The search for a marker. Clin Experiment Ophthalmol. 34:64–73. 2006. View Article : Google Scholar : PubMed/NCBI

18 

Lavker RM and Sun TT: Epidermal stem cells: Properties, markers, and location. Proc Natl Acad Sci USA. 97:13473–13475. 2000. View Article : Google Scholar : PubMed/NCBI

19 

Bose A, Teh MT, Mackenzie IC and Waseem A: Keratin k15 as a biomarker of epidermal stem cells. Int J Mol Sci. 14:19385–19398. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Forni MF, Trombetta-Lima M and Sogayar MC: Stem cells in embryonic skin development. Biol Res. 45:215–222. 2012. View Article : Google Scholar

21 

Ghadially R: 25 years of epidermal stem cell research. J Invest Dermatol. 132:797–810. 2012. View Article : Google Scholar

22 

Blazejewska EA, Schlötzer-Schrehardt U, Zenkel M, Bachmann B, Chankiewitz E, Jacobi C and Kruse FE: Corneal limbal microenvironment can induce transdifferentiation of hair follicle stem cells into corneal epithelial-like cells. Stem Cells. 27:642–652. 2009. View Article : Google Scholar :

23 

Saichanma S, Bunyaratvej A and Sila-Asna M: In vitro trans-differentiation of corneal epithelial-like cells from human skin-derived precursor cells. Int J Ophthalmol. 5:158–163. 2012.

24 

Araki K, Ohashi Y, Sasabe T, Kinoshita S, Hayashi K, Yang XZ, Hosaka Y, Aizawa S and Handa H: Immortalization of rabbit corneal epithelial cells by a recombinant SV40-adenovirus vector. Invest Ophthalmol Vis Sci. 34:2665–2671. 1993.PubMed/NCBI

25 

Yang X, Qu L, Wang X, Zhao M, Li W, Hua J, Shi M, Moldovan N, Wang H and Dou Z: Plasticity of epidermal adult stem cells derived from adult goat ear skin. Mol Reprod Dev. 74:386–396. 2007. View Article : Google Scholar

26 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) Method. Methods. 25:402–408. 2001. View Article : Google Scholar

27 

Shukla GC, Singh J and Barik S: MicroRNAs: Processing, maturation, target recognition and regulatory functions. Mol Cell Pharmacol. 3:83–92. 2011.PubMed/NCBI

28 

Baffa R, Fassan M, Volinia S, O'Hara B, Liu CG, Palazzo JP, Gardiman M, Rugge M, Gomella LG, Croce CM, et al: MicroRNA expression profiling of human metastatic cancers identifies cancer gene targets. J Pathol. 219:214–221. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Ma L, Teruya-Feldstein J and Weinberg RA: Tumour invasion and metastasis initiated by microRNA-10b in breast cancer. Nature. 449:682–688. 2007. View Article : Google Scholar : PubMed/NCBI

30 

Bloomston M, Frankel WL, Petrocca F, Volinia S, Alder H, Hagan JP, Liu CG, Bhatt D, Taccioli C and Croce CM: MicroRNA expression patterns to differentiate pancreatic adenocarcinoma from normal pancreas and chronic pancreatitis. JAMA. 297:1901–1908. 2007. View Article : Google Scholar : PubMed/NCBI

31 

Tian Y, Luo A, Cai Y, Su Q, Ding F, Chen H and Liu Z: MicroRNA-10b promotes migration and invasion through KLF4 in human esophageal cancer cell lines. J Biol Chem. 285:7986–7994. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Ciafrè SA, Galardi S, Mangiola A, Ferracin M, Liu CG, Sabatino G, Negrini M, Maira G, Croce CM and Farace MG: Extensive modulation of a set of microRNAs in primary glioblastoma. Biochem Biophys Res Commun. 334:1351–1358. 2005. View Article : Google Scholar : PubMed/NCBI

33 

Ma L, Reinhardt F, Pan E, Soutschek J, Bhat B, Marcusson EG, Teruya-Feldstein J, Bell GW and Weinberg RA: Therapeutic silencing of miR-10b inhibits metastasis in a mouse mammary tumor model. Nat Biotechnol. 28:341–347. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Chai G, Liu N, Ma J, Li H, Oblinger JL, Prahalad AK, Gong M, Chang LS, Wallace M, Muir D, et al: MicroRNA-10b regulates tumorigenesis in neurofibromatosis type 1. Cancer Sci. 101:1997–2004. 2010. View Article : Google Scholar : PubMed/NCBI

35 

Lin J, Teo S, Lam DH, Jeyaseelan K and Wang S: MicroRNA-10b pleiotropically regulates invasion, angiogenicity and apoptosis of tumor cells resembling mesenchymal subtype of glioblastoma multiforme. Cell Death Dis. 3:e3982012. View Article : Google Scholar : PubMed/NCBI

36 

Widelitz RB: Wnt signaling in skin organogenesis. Organogenesis. 4:123–133. 2008. View Article : Google Scholar

37 

Lim X and Nusse R: Wnt signaling in skin development, homeostasis, and disease. Cold Spring Harb Perspect Biol. 5:52013. View Article : Google Scholar

38 

Pearton DJ, Yang Y and Dhouailly D: Transdifferentiation of corneal epithelium into epidermis occurs by means of a multistep process triggered by dermal developmental signals. Proc Natl Acad Sci USA. 102:3714–3719. 2005. View Article : Google Scholar : PubMed/NCBI

39 

Mukhopadhyay M, Gorivodsky M, Shtrom S, Grinberg A, Niehrs C, Morasso MI and Westphal H: Dkk2 plays an essential role in the corneal fate of the ocular surface epithelium. Development. 133:2149–2154. 2006. View Article : Google Scholar : PubMed/NCBI

40 

Kwofie MA and Skowronski J: Specific recognition of Rac2 and Cdc42 by DOCK2 and DOCK9 guanine nucleotide exchange factors. J Biol Chem. 283:3088–3096. 2008. View Article : Google Scholar

41 

Czugala M, Karolak JA, Nowak DM, Polakowski P, Pitarque J, Molinari A, Rydzanicz M, Bejjani BA, Yue BY, Szaflik JP, et al: Novel mutation and three other sequence variants segregating with phenotype at keratoconus 13q32 susceptibility locus. Eur J Hum Genet. 20:389–397. 2012. View Article : Google Scholar :

42 

Caspi E and Rosin-Arbesfeld R: A novel functional screen in human cells identifies MOCA as a negative regulator of Wnt signaling. Mol Biol Cell. 19:4660–4674. 2008. View Article : Google Scholar : PubMed/NCBI

43 

Poulin G, Turgeon B and Drouin J: NeuroD1/beta2 contributes to cell-specific transcription of the proopiomelanocortin gene. Mol Cell Biol. 17:6673–6682. 1997. View Article : Google Scholar : PubMed/NCBI

44 

Marsich E, Vetere A, Di Piazza M, Tell G and Paoletti S: The PAX6 gene is activated by the basic helix-loop-helix transcription factor NeuroD/BETA2. Biochem J. 376:707–715. 2003. View Article : Google Scholar : PubMed/NCBI

45 

Fujiwara K, Ohuchida K, Sada M, Horioka K, Ulrich CD III, Shindo K, Ohtsuka T, Takahata S, Mizumoto K, Oda Y, et al: CD166/ALCAM expression is characteristic of tumorigenicity and invasive and migratory activities of pancreatic cancer cells. PLoS One. 9:e1072472014. View Article : Google Scholar : PubMed/NCBI

46 

Park GB, Kim D, Kim YS, Kim S, Lee HK, Yang JW and Hur DY: The Epstein-Barr virus causes epithelial-mesenchymal transition in human corneal epithelial cells via Syk/src and Akt/Erk signaling pathways. Invest Ophthalmol Vis Sci. 55:1770–1779. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2018
Volume 41 Issue 2

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Guo Y, Ma X, Wu W, Shi M, Ma J, Zhang Y, Zhao E and Yang X: Coordinated microRNA/mRNA expression profiles reveal a putative mechanism of corneal epithelial cell transdifferentiation from skin epidermal stem cells. Int J Mol Med 41: 877-887, 2018
APA
Guo, Y., Ma, X., Wu, W., Shi, M., Ma, J., Zhang, Y. ... Yang, X. (2018). Coordinated microRNA/mRNA expression profiles reveal a putative mechanism of corneal epithelial cell transdifferentiation from skin epidermal stem cells. International Journal of Molecular Medicine, 41, 877-887. https://doi.org/10.3892/ijmm.2017.3304
MLA
Guo, Y., Ma, X., Wu, W., Shi, M., Ma, J., Zhang, Y., Zhao, E., Yang, X."Coordinated microRNA/mRNA expression profiles reveal a putative mechanism of corneal epithelial cell transdifferentiation from skin epidermal stem cells". International Journal of Molecular Medicine 41.2 (2018): 877-887.
Chicago
Guo, Y., Ma, X., Wu, W., Shi, M., Ma, J., Zhang, Y., Zhao, E., Yang, X."Coordinated microRNA/mRNA expression profiles reveal a putative mechanism of corneal epithelial cell transdifferentiation from skin epidermal stem cells". International Journal of Molecular Medicine 41, no. 2 (2018): 877-887. https://doi.org/10.3892/ijmm.2017.3304