Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Characterization of MicroRNA Expression Profiles and the Discovery of Novel MicroRNAs Involved in Cancer during Human Embryonic Development

  • Yi Lin,

    Affiliation State Key Laboratory of Virology & College of Life Sciences, Wuhan University, Wuhan, China

  • Yan Zeng,

    Affiliation State Key Laboratory of Virology & College of Life Sciences, Wuhan University, Wuhan, China

  • Fan Zhang,

    Affiliation State Key Laboratory of Virology & College of Life Sciences, Wuhan University, Wuhan, China

  • Lu Xue,

    Affiliation State Key Laboratory of Virology & College of Life Sciences, Wuhan University, Wuhan, China

  • Zan Huang,

    Affiliation Department of Biochemistry and Molecular Biology, College of Life Sciences, Wuhan University, Wuhan, China

  • Wenxin Li ,

    guomx@whu.edu.cn (MG); liwxlab@whu.edu.cn(WL)

    Affiliation State Key Laboratory of Virology & College of Life Sciences, Wuhan University, Wuhan, China

  • Mingxiong Guo

    guomx@whu.edu.cn (MG); liwxlab@whu.edu.cn(WL)

    Affiliation State Key Laboratory of Virology & College of Life Sciences, Wuhan University, Wuhan, China

Abstract

MicroRNAs (miRNAs), approximately 22-nucleotide non-coding RNA molecules, regulate a variety of pivotal physiological or pathological processes, including embryonic development and tumorigenesis. To obtain comprehensive expression profiles of miRNAs in human embryos, we characterized miRNA expression in weeks 4-6 of human embryonic development using miRNA microarrays and identified 50 human-embryo-specific miRNAs (HES-miRNAs). Furthermore, we selected three non-conserved or primate-specific miRNAs, hsa-miR-638, -720, and -1280, and examined their expression levels in various normal and tumor tissues. The results show that expression of most miRNAs is extremely low during early human embryonic development. In addition, the expression of some non-conserved or primate-specific miRNAs is significantly different between tumor and the corresponding normal tissue samples, suggesting that the miRNAs are closely related to the pathological processes of various tumors. This study presents the first comprehensive overview of miRNA expression during human embryonic development and offers immediate evidence of the relationship between human early embryonic development and tumorigenesis.

Introduction

MicroRNAs (miRNAs) are ~22-nucleotide (nt) non-coding RNA molecules that regulate gene expression at the level of messenger RNA degradation or translation, exert essential functions in disparate biological processes ranging from cell cycle regulation, differentiation, and metabolism, to normal tissue and embryo development, and even tumorigenesis [14]. Alterations in miRNA expression are involved in the initiation, progression, and metastasis of human tumors [5]. Gradually increasing evidence suggests a direct link between miRNAs and many diseases, particularly cancer.

Using various experimental methods, previous studies in animal embryogenesis found that certain miRNAs were continuously reported, and most play crucial roles in differentiation or the maintenance of tissue identity [610]. Human development in the first 8 weeks of embryogenesis is potentially one of the most exciting areas of biological research. Specifically, some important tissues and organs, including neurogenesis and the development of the liver, began to form during weeks 4-6 (Carnegie Stages 10-17) of human embryogenesis [11]. Recently, our research group and others have characterized the transcriptional profiles of genome-wide mRNA expression during human embryogenesis using microarray [12,13]. However, human miRNA expression in this period remains to be elucidated.

Many studies have demonstrated that miRNAs are implicated in various aspects of tumorigenesis and animal embryogenesis (Reviewed in 4,5,1421). In 1892, the French biologists Lobstein and Recamier speculated the concept of the embryonic origin of tumors for the first time. In the 1970s, Dr. Pierce proposed the theory ‘cancer, a developmental biology’ and pointed out that tumorigenesis was intimately involved with developmental biology to a large extent [2227]. Currently, however, relevant evidence of the relationship between early human embryo development and tumorigenesis is still limited [2832]. To fill this important gap in knowledge, therefore, more studies are necessary.

In this study, the miRNA expression profiles during human embryonic development were identified and characterized using miRNAs microarrays. The results show that nearly all miRNAs of known function that are highly expressed or exhibit expression changes during human embryonic development have been implicated in several malignant diseases. Furthermore, tissue expression investigations of several miRNAs of unknown function that are highly expressed or undergo expression changes during human embryo development indicate that the expression of these miRNAs was significantly different between cancer samples and corresponding normal tissues, thereby suggesting that these miRNAs may also play important roles in tumorigenesis. These results provide evidence regarding the close relationship between human embryogenesis and carcinogenesis, and further reveal that miRNAs related to embryonic development with unknown functions may be involved in the development and progression of cancer.

Materials and Methods

Embryo collection and cell line

Human embryo collection was performed as previously described [13]. Appropriate written consent was obtained from the patients and approval gained from the Medical Ethics Committee of Zhongnan Hospital at Wuhan University by following national guidelines.

Microarray and Bioinformatics Analyses

The microRNAs microarray assay was performed using a service provider (LC Sciences). The assay started with 2 to 5 µg total RNA sample, which was size fractionated using an YM-100 Microcon centrifugal filter (Millipore). The isolated small RNAs (<300 nt) were 3’-extended with a poly(A) tail using poly(A) polymerase. An oligonucleotide tag was ligated to the poly(A) tail for later fluorescent dye staining; two different tags were used for the two RNA samples in dual-sample experiments. Hybridization was performed overnight on a μParaflo™ microfluidic chip using a microcirculation pump (Atactic Technologies) [33,34]. On the microfluidic chip, each detection probe consisted of a chemically modified nucleotide-coding segment complementary to target microRNA (from miRBase, http://www.mirbase.org/) or other RNA (control or customer defined sequences) as well as a spacer segment of polyethylene glycol to extend the coding segment away from the substrate. The detection probes were generated from in situ synthesis using PGR (photogenerated reagent) chemistry. The hybridization melting temperatures were balanced by chemical modifications of the detection probes. Hybridization used 100 µL 6xSSPE buffer (0.90 M NaCl, 60 mM Na2HPO4, 6 mM EDTA, pH 6.8) containing 25% formamide at 34° C. After hybridization, detection used fluorescence labeling with tag-specific Cy3 and Cy5 dyes. Hybridization images were collected using a laser scanner (GenePix 4000B, Molecular Device) and digitized using Array-Pro image analysis software (Media Cybernetics). Data were analyzed by first subtracting the background and normalizing the signals using a LOWESS filter (Locally-weighted Regression) [35]. For two-color experiments, the ratio of the two sets of detected signals (log2 transformed, balanced) and p-values of the t-tests were calculated; differentially detected signals were those with p-values less than 0.01. The normalized signal intensity from 8 samples (Week 4 of human embryonic development: ZN18, ZN46/47, ZN75; Week 5: ZN38, ZN43, ZN63-1; Week 6: ZN61, ZN70) was used the clustering analysis using a one-way analysis of variance (ANOVA) by a service provider (LC Sciences). Normalized data for all arrays have been deposited in the Gene Expression Omnibus (GEO) at the National Center for Biotechnology Information (NCBI), accessible through GEO Series accession number GSE46795 (http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE46795).

High-density multiple organ tumor and normal tissue microarrays (catalog MC5003), containing 18 tumor types (20 cases per type) and normal corresponding controls (5 cases per type), were purchased from US Biomax. The miRNA and scrambled oligonucleotides for in situ hybridization were purchased as digoxigenin-labeled locked nucleic acid (LNA) probes from Exiqon (Denmark). The sequence of the LNA detection probes were: LNA-638: 5’-DIG-AGGCCGCCACCCGCCCGCGATCCT-3’; LNA-720: 5’-DIG-TGGAGGCCCCAGCGAGA-3’; and LNA-1280: 5’-DIG-GGGTGGCAGCGGTGGGA-3’. LNA-U6: 5’-DIG-CACGAATTTGCGTGTCATCCTT-3’ and LNA-scramble: 5’-DIG-GTGTAACACGTCTATACGCCCA-3’ were used as positive and negative control probes, respectively. The in situ hybridization of tissue array for miRNAs experssion was performed using a service provider (Shaanxi Chaoying Biotechnology CO, LTD, Shaanxi China), and slides were read by two independent researchers. The intensity of the staining was scored as negative (-/0), weak (+/1), moderate (++/2), or strong (+++/3) as previously described [36,37].

miRNA real-time polymerase chain reaction (PCR)

miRNA RT-PCR assay was performed using a service provider (LC Sciences). Briefly, miRNA quantifications were examined using TaqMan® MicroRNA Assays and TaqMan® Universal PCR Master Mix and analyzed by ABI PRISM 7000 Sequence Detection System. RNU24 was used as an internal control to determine relative miRNA expression. Each sample was performed in triplicate.

Statistical analyses

One-way analysis of variance (ANOVA) was performed using the normalized data for week 4–6 to identify miRNAs whose expression changed. To compare miRNAs expression in human normal and cancer tissues, two-tailed Student’s t tests were performed using the above scores of samples.

Results and Discussion

Characterization of miRNA expression profiles during human embryonic development

To identify the miRNA expression profiles of human embryos, miRNA microarrays assays were performed in human embryo samples (weeks 4, 5, and 6 after fertilization) using µParaflo® technology, and the microarrays raw data were analyzed. The array covers all miRNA transcripts available in the Sanger miRBase database (release 10.1). For details, see the Experimental Procedures and Supplemental Data (Table S1 available online). Accordingly, miRNAs exhibiting signal strengths greater than 32 were considered as detected expression. After normalization, a strong signal threshold of miRNA detection was defined as 500 (Figure 1). We designated these 50 miRNAs (~6%), in which the expression signal was greater than 500 of any stage at weeks 4, 5, or 6 during human embryonic development, as human embryo-specific miRNAs (HES-miRNAs) (Table 1).

thumbnail
Figure 1. Expression overview of the 835 miRNAs during human embryonic development

MiRNA expression levels during human embryonic development (weeks 4, 5, and 6 after fertilization) with signal strength greater 32 were considered as detected expression. A strong signal threshold of miRNA detection was defined as 500 after normalization. MiRNAs with a fluorescence signal greater than 500 at any stage at weeks 4, 5 or 6 were defined as HES-miRNAs.

https://doi.org/10.1371/journal.pone.0069230.g001

miRNAWeek 4Week 5Week 6
hsa-miR-1031020.97589.63775.30
hsa-miR-106a3717.471283.56988.08
hsa-miR-106b705.36336.58217.08
hsa-miR-107837.20446.95520.07
hsa-miR-10b1679.971511.97688.29
hsa-miR-1224805.009396.8510671.26
hsa-miR-1246356.13816.55100.26
hsa-miR-125a-5p387.96827.091146.67
hsa-miR-125b558.09937.533407.28
hsa-miR-126513.43257.52195.18
hsa-miR-126867.10212.18725.03
hsa-miR-1275238.04928.53413.71
hsa-miR-1280189.87292.811238.08
hsa-miR-130a522.76130.33161.38
hsa-miR-130b827.85593.00936.95
hsa-miR-151-5p713.59579.19423.27
hsa-miR-15b1041.17788.21368.30
hsa-miR-161080.15395.87286.75
hsa-miR-174108.911437.681210.55
hsa-miR-181a312.21269.73741.07
hsa-miR-181b192.98187.58873.77
hsa-miR-182341.78579.71453.59
hsa-miR-191482.51483.55631.69
hsa-miR-199a-3p1284.651201.391165.51
hsa-miR-19b758.72559.73223.40
hsa-miR-206791.121067.481658.21
hsa-miR-20a3614.131105.53804.23
hsa-miR-20b1471.15540.45323.50
hsa-miR-2142654.733566.049605.29
hsa-miR-23b745.25594.28767.96
hsa-miR-251871.251553.321021.11
hsa-miR-26a4645.233390.432536.21
hsa-miR-320a1849.633185.424495.39
hsa-miR-320b1358.111904.943209.74
hsa-miR-320c1666.092358.804068.96
hsa-miR-320d790.281072.821897.73
hsa-miR-361-5p751.92719.17643.41
hsa-miR-423-5p553.06888.43400.99
hsa-miR-432499.71807.49534.52
hsa-miR-4511355.12622.2018.66
hsa-miR-483-5p480.512580.411537.07
hsa-miR-574-5p163.53375.89709.17
hsa-miR-6384622.856092.422891.58
hsa-miR-663290.15525.9244.41
hsa-miR-720101.92181.661255.10
hsa-miR-92a11338.0811838.016909.45
hsa-miR-92b4171.404315.192556.64
hsa-miR-93872.20552.87445.45
hsa-miR-936323.52512.7235.61
hsa-miR-99b510.57635.731232.78

Table 1. List of human-embryo-specific-miRNAs.

After normalization, a strong signal threshold of miRNA detection was defined as 500 (Figure 1), and 50 miRNAs (~6%, 50/835), in which the expression signal was greater than 500 of any stage at weeks 4, 5, or 6 during human embryonic development, were designated as human-embryo-specific miRNAs (HES-miRNAs).
CSV
Download CSV

The expression profiles show that the expression of most miRNAs is low during early human embryonic development, in which approximately 80% of miRNAs (666 of 835 miRNAs) exhibiting signal strength were considered as undetected expression (Figure 1 and Table S1). These data are similar to the findings that most miRNAs were not detected during early zebrafish development [10]. Hierarchical cluster assays of HES-miRNA expression (Figure 2A) and 169 miRNAs exhibiting signal strength over 32 (Figure S1) during human embryonic development are shown. In Figure 2A, the following three clusters of HES-miRNAs expressions were identified: Cluster a, upregulated at week 6; Cluster b, downregulated at week 6; and Cluster c, upregulated at week 4. In Figure S1, four clusters of 169 miRNAs were identified: clusters 1, 2, 3, and 4. Cluster a miRNAs in Figure 2A increased during the three human embryonic stages (week 6 > week 5 > week 4), and included some miRNA species reported to be enriched in various developmental and disease processes (e.g., hsa-miR-122, -206, -214, -181 family, and -125 family) [38]. Cluster c miRNAs that decreased during the three human embryonic stages (week 4 > week 5 > week 6) included some miRNAs that were also enriched in various developmental and disease processes (e.g., hsa-miR-451, -106, -16, and -17) (Figure 2A) [38]. However, the expression of many cluster b members that increased from week 4 to week 5, but decreased from week 5 to week 6, have unknown functions in development and disease processes.

thumbnail
Figure 2. Hierarchical clustering analyses of the expression of 50 HES-miRNAs

(A) Fifty HES-miRNAs were divided into 3 clusters: clusters a, b, and c. The arrow shows the miRNAs that were selected to further validate the microarray data using microRNA qRT-PCR (Figure 3). The asterisk shows the non-conserved or primate-specific HES-miRNAs (Figure S4). The hollow triangle indicates miRNAs harborring the same seed region sequence among cluster c. (B) Figure 2B shows the mature sequence of six miRNAs (miR-17, -106a, -106b, -20a, -20b, -93) with the identical seed region tagged with light red shadow. (C) Pie chart shows functional pattern analysis (Gene Ontology) of the conserved targets (1245 transcripts), predicted by TargetScan (http://www.targetscan.org/vert_60/), of the six miRNAs.

https://doi.org/10.1371/journal.pone.0069230.g002

Among the HES-miRNAs (Table 1 and Figure 2A), some have been experimentally confirmed to play pivotal roles in differentiation and disease. For instance, the mammalian liver-specific miRNA, miR-122, which had the highest expression level in week 6 of human embryonic development, is frequently suppressed in primary hepatocellular carcinomas [39,40], and is also essential for hepatitis C virus RNA accumulation in cultured liver cells [10,4147]. Hsa-miR-214 is related to muscle development and bone formation, and is involved in gastric and ovarian cancer [4853]. Hsa-miR-92a/b, as members of the miR-17~92 family, is involved in many solid tumors and leukemia [5457]. Hsa-miR-106a is involved in regulation of induced pluripotent stem cell generation, posttranscriptional regulation of interleukin-10 expression, gastric cancer, and astrocytoma [5861]. Therefore, our results strongly support the notion that there is striking similarity between early embryonic development and tumorigenesis [22].

Moreover, these highly expressed miRNAs, as well as those that have low expression, may play different regulatory roles in different stages of human embryonic development. Alternatively, these miRNAs with high expression levels have strong tissue specificity, such as hsa-miR-122.

Among cluster c, it’s amazing that we noticed 6 miRNAs (hsa-miR-17, -106a, -106b, -20a, -20b and -93), which contain the same seed region sequcence AAAGUG (Figure 2B), were downregulated at week 6 of human embryonic development, and might play the similar biological functions in human embryogenesis. The Hannon and Hammond laboratories provided direct experimental results that these microRNAs, encoded by the miR-17-92 cluster and its paralogs (the miR-106a-363 cluster and miR-106b-25 clusters), have oncogenic activity in solid tumor [62]. Ventura and colleagues documented strong evidences that deletion of the miR-17-92 cluster resulted in severely hypoplastic lungs and reduction in pre-B cells numbers in mice, finally leading to smaller embryos even immediate postnatal death [63]. Considering the vital role of these miRNAs in development and tumorigenesis, we predicted all targets of these 6 miRNAs by TargetScan, and then the conserved targets (1245 transcripts) were further analyzed. During early stage of human embryonic development, one important event associated with the developmental transition from the early cell proliferation phase to organogenesis and histogenesis is that the number of stem cells or undifferentiated cells is reduced because more cells begin to differentiate into different organ/tissue-specific cell types. During early stage of human embryonic development, one important event associated with the developmental transition from the early cell proliferation phase to organogenesis and histogenesis is presented. Functional pattern analysis (Gene Ontology) from the conserved target (1245 transcripts) of 6 miRNAs shows that the vast majority of these genes in the GO categories related to biological regulation, regulation of biological process, metabilic process, cellular process, multicellular organismal process and developmental process (Figure 2C). Recently reported some targets of miR-17~92 family, such as CDKN1A (p21) [64], BIM [65], RUNX1 [66], both of them are included in these GO categories, which suggest that these genes are irreplaceable position in controlling of cell cycle, cell apopotosis, and differentiation of hematopoietic progenitor cells. And these results could help us better understand the role of HES-miRNAs in human embryogenesis and tumorigenesis.

In addition, to validate the quality of the miRNA microarray data, four miRNAs (hsa-miR-125b, -720, -1280, and -20b, Figure 3 and Figure S2) whose expression levels changed significantly during development were validated using TaqMan® real-time RT-PCR miRNA assays. The trends of miRNA expression were consistent with that observed in the miRNA microarray data (Figure 3B, C, D, and E).

thumbnail
Figure 3. miRNA qRT-PCR analyses validated the miRNA microarray results

Four miRNAs [hsa-miR-125b (B), hsa-miR-720 (C), hsa-miR-1280 (D), and hsa-miR-20b (E)], which were differentially expressed (p<0.05, Figure 3A) and (p<0.10, Figure S2) during weeks 4, 5, and 6 of human embryonic development, were chosen. qRT-PCR was performed and U6 snRNA was used as an internal control to determine the relative miRNA expression.

https://doi.org/10.1371/journal.pone.0069230.g003

Human-Mouse Comparative Analyses of miRNA Expression During Embryonic Development

To compare the miRNA expression profiles between human and mouse embryonic development, we conducted a comparative analysis relative to previously published microRNA expression profiles in mouse embryos covering prenatal development (E9.5, E10.5, and E11.5) [8], which correspond with weeks 4, 5, and 6 of human embryonic development [67,68]. Given the identical miRNAs in human and mouse, we constructed an intersection analysis between 835 human miRNAs in this study and 390 mouse miRNAs, and identified 195 homologous miRNAs (Table S2). The hierarchical clustering assays of human-mouse homology miRNAs are shown in Figure S3A and B. Venn diagrams (Figures S3C and S3D) showed that 38 or 32 human-mouse homology miRNAs were upregulated or downregulated, respectively, during human and mouse embryonic development. Among these miRNAs, let-7 family members, miR-34a, miR-221/222, miR-145, miR-125b, miR-15a, and miR-223 have been proven to play multifarious roles during development and pathogenesis in various mammalian organs [38]. These results further suggest that some homologous human-mouse miRNAs are important for development and pathogenesis, whereas those that are not homologous are likely involved in different development processes that reflect the developmental divergence between human and mouse.

Conservative properties of HES-miRNAs

A previous study showed that many known miRNA genes have a typical conservation pattern, persistent throughout the clusters, suggesting evolutionary and functional implications [69]. However, a substantial portion of microRNAs are confirmed as non-conserved or primate-specific miRNAs because many new human miRNAs have been continuously identified [70]. To define the conservation of HES-miRNAs, we used the miRviewer [71], which shows conservation of miRNA genes grouped by name. As shown in Figure S4, most HES-miRNAs are highly conserved, and their functions are well known. Remarkably, among HES-miRNAs, eight miRNAs, hsa-miR-638, -663, -720, -936, -1246, -1268, -1275, and -1280, are extremely non-conserved, even though most are primate-specific (Figure S4) [70,72]. Noticeably, the peak expression values of 5 of these miRNAs, miR-638, -663, -936, -1246, and -1275, are at 5 weeks of human embryonic development. In other words, these miRNAs may function only at a specific human embryonic developmental stage. Furthermore, we know very little about the biological functions of these miRNAs in mammalian organogenesis and pathogenesis. However, because the expression of these non-conserved or primate-specific miRNAs is high during human embryonic development, these miRNAs likely play pivotal roles in mammalian organogenesis and/or pathogenesis. In fact, a few studies indicate that these non-conserved or primate-specific HES-miRNAs may be associated with all kinds of disease [7382]. The function of these non-conserved or primate-specific HES-miRNAs remains to be determined.

Comparative analyses of the expression of hsa-miR-638, -720, and -1280 in normal and malignant tissues

Many studies have indicated that miRNAs are involved in diverse aspects of animal developmental processes and disease [4,1621,38,8389]. To further validate the relationship between the HES-miRNAs and various solid malignant tumors, we focus on three non-conserved or primate-specific HES-miRNAs, hsa-miR-638, -720, and -1280 (Table 1), of highly expressed or exhibit expression changes during week 4-6 of human embryonic development for further functional investigation. We examined whether the expression of the three HES-miRNAs differs significantly between various human tumors and the corresponding normal tissue.

Using the tissue microarray MC5003 and highly specific and sensitive LNA-modified oligonucleotide probes, the expression profiles of the 3 miRNAs in normal and cancerous tissues were detected using in situ hybridization. U6 snoRNA and scramble-miR probes were used as positive and negative control probes, respectively. The statistical results show that the expression of hsa-miR-638 is significantly upregulated in hepatocellular liver cancer tissues (n=20) versus normal liver tissues (n=5) (p=0.0092), and in cervix uteri squamous cell carcinoma tissues (n=18) versus normal cervix uteri tissues (n=7) (p=0.0003). In contrast, the expression of hsa-miR-638 was significantly downregulated in stomach adenocarcinoma tissues (n=19) versus normal stomach tissues (n=6) (p=0.0095) (Figure 4A). These data agree with the results from Tsukamoto et al. showing downregulation of this miRNA in gastric cancer [90]. hsa-miR-720 expression is significantly upregulated in cervix uteri squamous cell carcinoma tissues (n=18) versus normal cervix uteri tissues (n=6) (p=0.0086), in lung squamous cell carcinoma/adenocarcinoma tissues (n=17) versus normal lung tissues (n=6) (p=0.0386), in ovary cystadenocarcinoma/adenocarcinoma tissues (n=18) versus normal ovary tissue (n=6) (p=0.04045), and in urothelial carcinoma tissues (n=17) versus normal vesica urinaria tissues (n=5) (p=0.03504) (Figure 4B). In addition, hsa-miR-720 expression is significantly downregulated in intestinal mucosa malignant tissues (n=19) versus normal skin tissues (n=9) (p=0.0017) (Figure 4B). The expression of hsa-miR-1280 is significantly downregulated in squamous cell carcinoma tissues of head and neck skin (n=20) versus normal skin tissues (n=9) (p<0.0001), in intestinal mucosa malignant tissues (n=20) versus normal skin tissues (n=9) (p=0.0009), and in pancreatic adenocarcinoma tissues (n=17) versus normal pancreatic tissues (n=6) (p=0.0270) (Figure 4C). In contrast, hsa-miR-1280 expression is significantly upregulated in cervix uteri squamous cell carcinoma tissues (n=17) versus normal cervix uteri tissues (n=7) (p=0.01076) (Figure 4C). Representative examples of three miRNAs that were differentially expressed in tumor tissues versus normal tissues are shown in Figures 5, S5, S6, and S7. The results suggest that these HES-miRNAs are closely related to the pathological processes of the various tumors.

thumbnail
Figure 4. Statistical analyses of three HES-miRNAs (hsa-miR-638, -720, and -1280) expression using in situ hybridization

High-density multiple organ tumor and normal tissue microarrays containing 500 tissue-dots with 18 tumor types and normal corresponding control tissues. Digoxigenin-labeled locked nucleic acid (LNA) probes (LNA-638, LNA-720, and LNA-1280) were used to specifically detect the miRNA expression on the tissue chip. (A) Aberrant expression of hsa-miR-638 on tissue microarray. Significant up-regulation of miR-638 can be observed in hepatocellular liver cancer and cervix uteri squamous cell carcinoma. miR-638 down-regulation is found in stomach adenocarcinoma versus the corresponding normal tissues (p=0.0092, p=0.0003, and p=0.0095, respectively) (B) Aberrant expression of hsa-miR-720 on tissue microarray. Hsa-miR-720 is significantly upregulated in cervix uteri squamous cell carcinoma, lung squamous cell adenocarcinoma, ovary adenocarcinoma, and urothelial carcinoma versus the corresponding normal tissues (p=0.0086, p=0.0386, p=0.0404, and p=0.035, respectively). Hsa-miR-720 is significantly downregulated in intestinal mucosa malignant tissues versus normal skin tissues (p=0.0017). (C) Aberrant expression of hsa-miR-1280 on tissue microarray. Hsa-miR-1280 is downregulated in squamous cell carcinoma, intestinal mucosa malignant tissue, and pancreatic adenocarcinoma versus the corresponding normal tissues (p<0.0001, p=0.0009, and p=0.0270, respectively). Hsa-miR-1280 is upregulated significantly in cervix uteri squamous cell carcinoma tissues versus normal cervix uteri tissues (p=0.01076). MiRNA expression levels (y-axis) with a score=0, 1, 2, or 3, indicate negative, weak, medium, or strong staining intensity, respectively. n indicates the numbers of the specimens studied. NT indicates normal tissue samples; CT indicates carcinoma tissue samples. Two-tailed student t tests were performed to compare miRNA expression in normal and cancerous tissues.

https://doi.org/10.1371/journal.pone.0069230.g004

thumbnail
Figure 5. hsa-miR-638 expression in stomach adenocarcinoma and corresponding normal tissues.

Panels a through t show stomach adenocarcinoma samples and panels u through z show normal stomach tissue. The blue staining signal indicates expression of hsa-miR-638. The red staining shows the cell nucleus (scale 200μm as Figure 5a). j2 shows higher magnification (5x) of the indicated area in figure 5j, and z2 shows higher magnification (5x) of the indicated area in figure 5z.

https://doi.org/10.1371/journal.pone.0069230.g005

In summary, our studies herein reveal that the expression levels of most miRNAs during human embryogenesis are very low. We designated 50 (~6%) of the 835 miRNAs regulated in weeks 4 through 6 of human embryonic development as HES-miRNAs, and demonstrated that some non-conserved or primate-specific HES-miRNAs are involved in tumorigenesis, thus supporting the hypothesis that early embryonic development shares many similarities with cancer development in biological behavior as well as molecularly [22]. However, the functions of the non-conserved or primate-specific HES-miRNAs remain to be experimentally established, which will help us further understand the complicated molecular modulation network that occurs during human embryonic development.

Supporting Information

Figure S1.

Hierarchical clustering analyses of the expression of 169 miRNAs exhibiting signal strengths greater than 32 miRNAs (n=169) were divided into 4 clusters: clusters 1, 2, 3, and 4. The arrow shows the miRNAs that were selected be validated by microRNA qRT-PCR (Figure 3). The asterisk shows the non-conserved or primate-specific HES-miRNAs (Figure S4).

https://doi.org/10.1371/journal.pone.0069230.s001

(TIF)

Figure S2.

Clustering analyses of miRNA expression (p<0.10) during human embryonic development Red and green indicate high and low expression levels, respectively.

https://doi.org/10.1371/journal.pone.0069230.s002

(TIF)

Figure S3.

Conserved miRNA expression patterns and biological characteristics of human-mouse homologs during embryonic development. Unsupervised hierarchical clustering analyses of miRNA expression profiles in humans (weeks 4, 5, and 6, Figure S3A) and mouse embryo samples (E9.5, E10.5, and E11.5, Figure S3B). The mouse embryo microarray data were published in 2006 by Mineno and colleagues. The color in each lattice reflects the expression level of the miRNA in the corresponding sample. The increasing intensities of red indicate that a specific miRNA has a higher expression in the given sample. The increasing intensities of green indicate that this miRNA has lower expression. Venn diagrams depict the intersection miRNAs between humans (weeks 4, 5, and 6) and mouse embryo samples (E9.5, E10.5, and E11.5) and show that 38 and 32 human-mouse homology miRNAs were upregulated (C) and downregulated (D), respectively, during human and mouse embryonic development.

https://doi.org/10.1371/journal.pone.0069230.s003

(TIF)

Figure S4.

Conservation analyses of the HES-miRNAs. The miRviewer shows conservation of the HES-miRNA genes, grouped by name. The miRNAs that only can be discovered in human or other primates, including miR-638, -663, -720, -936, -1246, -1268, -1275, and -1280, were separated below. The increasing intensities of green indicate that a specific miRNA (or miRNA family) has higher conservation in the given species. Numbers in brackets indicate the numbers of the miRNAs in a given miRNA family. Most share similar conservation results.

https://doi.org/10.1371/journal.pone.0069230.s004

(TIF)

Figure S5.

hsa-miR-638 expression in cervix uteri adenocarcinoma and corresponding normal tissues. Panels a through t show cervix uteri adenocarcinoma samples and panels u through z show normal cervix uteri tissues. The blue staining signal indicates expression of hsa-miR-638. The red staining shows the cell nucleus (scale 200µm as Figure S5a).

https://doi.org/10.1371/journal.pone.0069230.s005

(TIF)

Figure S6.

hsa-miR-720 expression in cervix uteri adenocarcinoma and corresponding normal tissues. Panels a through t show cervix uteri adenocarcinoma samples and panels u through z show normal cervix uteri tissues. The blue staining signal indicates expression of hsa-miR-720. The red staining shows the cell nucleus (scale 200µm as Figure S6a).

https://doi.org/10.1371/journal.pone.0069230.s006

(TIF)

Figure S7.

hsa-miR-1280 expression in skin tumors and corresponding normal tissues. Panels a through t show skin tumor samples and panels u through z show normal skin tissues. The blue staining signal indicates expression of hsa-miR-1280. The red staining shows the cell nucleus (scale 200µm as Figure S7a).

https://doi.org/10.1371/journal.pone.0069230.s007

(TIF)

Table S1.

The expressions of 835 miRNAs during week 4-6 of human embryonic development.

https://doi.org/10.1371/journal.pone.0069230.s008

(XLS)

Table S2.

List of 195 homologous human and mouse miRNAs.

https://doi.org/10.1371/journal.pone.0069230.s009

(XLS)

Author Contributions

Conceived and designed the experiments: MG WL. Performed the experiments: MG YL YZ FZ LX. Analyzed the data: MG WL YL ZH. Contributed reagents/materials/analysis tools: YZ YL FZ. Wrote the manuscript: MG YL WL.

References

  1. 1. Rigoutsos I, Furnari F (2010) Gene-expression forum: Decoy for microRNAs. Nature 465: 1016-1017. doi:https://doi.org/10.1038/4651016a. PubMed: 20577197.
  2. 2. Kosik KS (2010) MicroRNAs and cellular phenotypy. Cell 143: 21-26. doi:https://doi.org/10.1016/j.cell.2010.09.008. PubMed: 20887887.
  3. 3. Chitwood DH, Timmermans MC (2010) Small RNAs are on the move. Nature 467: 415-419. doi:https://doi.org/10.1038/nature09351. PubMed: 20864994.
  4. 4. Ambros V (2004) The functions of animal microRNAs. Nature 431: 350-355. doi:https://doi.org/10.1038/nature02871. PubMed: 15372042.
  5. 5. Spizzo R, Nicoloso MS, Croce CM, Calin GA (2009) SnapShot: MicroRNAs in Cancer. Cell 137: 586-586 e581 doi:https://doi.org/10.1016/j.cell.2009.04.040. PubMed: 19410551.
  6. 6. Li M, Xia Y, Gu Y, Zhang K, Lang Q et al. (2010) MicroRNAome of porcine pre- and postnatal development. PLOS ONE 5: e11541. doi:https://doi.org/10.1371/journal.pone.0011541. PubMed: 20634961.
  7. 7. Castro FO, Sharbati S, Rodríguez-Alvarez LL, Cox JF, Hultschig C et al. (2010) MicroRNA expression profiling of elongated cloned and in vitro-fertilized bovine embryos. Theriogenology 73: 71-85. doi:https://doi.org/10.1016/j.theriogenology.2009.08.003. PubMed: 19836069.
  8. 8. Mineno J, Okamoto S, Ando T, Sato M, Chono H et al. (2006) The expression profile of microRNAs in mouse embryos. Nucleic Acids Res 34: 1765-1771. doi:https://doi.org/10.1093/nar/gkl096. PubMed: 16582102.
  9. 9. Darnell DK, Kaur S, Stanislaw S, Konieczka JH, Yatskievych TA et al. (2006) MicroRNA expression during chick embryo development. Dev Dyn 235: 3156-3165. doi:https://doi.org/10.1002/dvdy.20956. PubMed: 17013880.
  10. 10. Wienholds E, Kloosterman WP, Miska E, Alvarez-Saavedra E, Berezikov E et al. (2005) MicroRNA expression in zebrafish embryonic development. Science 309: 310-311. doi:https://doi.org/10.1126/science.1114519. PubMed: 15919954.
  11. 11. Hill MA (2007) Early human development. Clin Obstet Gynecol 50: 2-9. doi:https://doi.org/10.1097/GRF.0b013e31802f119d. PubMed: 17304021.
  12. 12. Fang H, Yang Y, Li C, Fu S, Yang Z et al. (2010) Transcriptome analysis of early organogenesis in human embryos. Dev Cell 19: 174-184. doi:https://doi.org/10.1016/j.devcel.2010.06.014. PubMed: 20643359.
  13. 13. Yi H, Xue L, Guo MX, Ma J, Zeng Y et al. (2010) Gene expression atlas for human embryogenesis. FASEB J 24: 3341-3350. doi:https://doi.org/10.1096/fj.10-158782. PubMed: 20430792.
  14. 14. Esquela-Kerscher A, Slack FJ (2006) Oncomirs - microRNAs with a role in cancer. Nat Rev Cancer 6: 259-269. doi:https://doi.org/10.1038/nrc1840. PubMed: 16557279.
  15. 15. Bartel DP (2004) MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 116: 281-297. doi:https://doi.org/10.1016/S0092-8674(04)00045-5. PubMed: 14744438.
  16. 16. Alvarez-Garcia I, Miska EA (2005) MicroRNA functions in animal development and human disease. Development 132: 4653-4662. doi:https://doi.org/10.1242/dev.02073. PubMed: 16224045.
  17. 17. Kloosterman WP, Plasterk RH (2006) The diverse functions of microRNAs in animal development and disease. Dev Cell 11: 441-450. doi:https://doi.org/10.1016/j.devcel.2006.09.009. PubMed: 17011485.
  18. 18. Plasterk RH (2006) Micro RNAs in animal development. Cell 124: 877-881. doi:https://doi.org/10.1016/j.cell.2006.02.030. PubMed: 16530032.
  19. 19. Bushati N, Cohen SM (2007) microRNA functions. Annu Rev Cell Dev Biol 23: 175-205. doi:https://doi.org/10.1146/annurev.cellbio.23.090506.123406. PubMed: 17506695.
  20. 20. Croce CM (2009) Causes and consequences of microRNA dysregulation in cancer. Nat Rev Genet 10: 704-714. doi:https://doi.org/10.1038/nrg2634. PubMed: 19763153.
  21. 21. Pauli A, Rinn JL, Schier AF (2011) Non-coding RNAs as regulators of embryogenesis. Nat Rev Genet 12: 136-149. doi:https://doi.org/10.1038/nrm3070. PubMed: 21245830.
  22. 22. Ma Y, Zhang P, Wang F, Yang J, Yang Z et al. (2010) The relationship between early embryo development and tumourigenesis. J Cell Mol Med 14: 2697-2701. doi:https://doi.org/10.1111/j.1582-4934.2010.01191.x. PubMed: 21029369.
  23. 23. Bignold LP, Coghlan BL, Jersmann HP (2006) Hansemann, Boveri, chromosomes and the gametogenesis-related theories of tumours. Cell Biol Int 30: 640-644. doi:https://doi.org/10.1016/j.cellbi.2006.04.002. PubMed: 16753311.
  24. 24. Reya T, Morrison SJ, Clarke MF, Weissman IL (2001) Stem cells, cancer, and cancer stem cells. Nature 414: 105-111. doi:https://doi.org/10.1038/35102167. PubMed: 11689955.
  25. 25. Hanahan D, Weinberg RA (2000) The hallmarks of cancer. Cell 100: 57-70. doi:https://doi.org/10.1016/S0092-8674(00)81683-9. PubMed: 10647931.
  26. 26. Papaioannou VE (1993) Ontogeny, pathology, oncology. Int J Dev Biol 37: 33-37. PubMed: 8507567.
  27. 27. Pierce GB (1983) The cancer cell and its control by the embryo. Rous-Whipple Award lecture. Am J Pathol 113: 117-124. PubMed: 6312802.
  28. 28. Tzukerman M, Rosenberg T, Reiter I, Ben-Eliezer S, Denkberg G et al. (2006) The influence of a human embryonic stem cell-derived microenvironment on targeting of human solid tumor xenografts. Cancer Res 66: 3792-3801. doi:https://doi.org/10.1158/0008-5472.CAN-05-3467. PubMed: 16585206.
  29. 29. Hochedlinger K, Blelloch R, Brennan C, Yamada Y, Kim M et al. (2004) Reprogramming of a melanoma genome by nuclear transplantation. Genes Dev 18: 1875-1885. doi:https://doi.org/10.1101/gad.1213504. PubMed: 15289459.
  30. 30. Cheng TC, Huang CC, Chen CI, Liu CH, Hsieh YS et al. (2004) Leukemia inhibitory factor antisense oligonucleotide inhibits the development of murine embryos at preimplantation stages. Biol Reprod 70: 1270-1276. doi:https://doi.org/10.1095/biolreprod.103.023283. PubMed: 14695905.
  31. 31. Dürr M, Harder F, Merkel A, Bug G, Henschler R et al. (2003) Chimaerism and erythroid marker expression after microinjection of human acute myeloid leukaemia cells into murine blastocysts. Oncogene 22: 9185-9191. doi:https://doi.org/10.1038/sj.onc.1207134. PubMed: 14668800.
  32. 32. Papaioannou VE, Waters BK, Rossant J (1984) Interactions between diploid embryonal carcinoma cells and early embryonic cells. Cell Differ 15: 175-179. doi:https://doi.org/10.1016/0045-6039(84)90071-X. PubMed: 6535642.
  33. 33. Gao X, Gulari E, Zhou X (2004) In situ synthesis of oligonucleotide microarrays. Biopolymers 73: 579-596. doi:https://doi.org/10.1002/bip.20005. PubMed: 15048782.
  34. 34. Zhu Q, Hong A, Sheng N, Zhang X, Matejko A et al. (2007) microParaflo biochip for nucleic acid and protein analysis. Methods Mol Biol 382: 287-312. doi:https://doi.org/10.1007/978-1-59745-304-2_19. PubMed: 18220239.
  35. 35. Bolstad BM, Irizarry RA, Astrand M, Speed TP (2003) A comparison of normalization methods for high density oligonucleotide array data based on variance and bias. Bioinformatics 19: 185-193. doi:https://doi.org/10.1093/bioinformatics/19.2.185. PubMed: 12538238.
  36. 36. Chiyomaru T, Enokida H, Tatarano S, Kawahara K, Uchida Y et al. (2010) miR-145 and miR-133a function as tumour suppressors and directly regulate FSCN1 expression in bladder cancer. Br J Cancer 102: 883-891. doi:https://doi.org/10.1038/sj.bjc.6605570. PubMed: 20160723.
  37. 37. Ropponen KM, Eskelinen MJ, Lipponen PK, Alhava EM, Kosma VM (1999) Reduced expression of alpha catenin is associated with poor prognosis in colorectal carcinoma. J Clin Pathol 52: 10-16. doi:https://doi.org/10.1136/jcp.52.1.10. PubMed: 10343606.
  38. 38. Sayed D, Abdellatif M (2011) MicroRNAs in development and disease. Physiol Rev 91: 827-887. doi:https://doi.org/10.1152/physrev.00006.2010. PubMed: 21742789.
  39. 39. Coulouarn C, Factor VM, Andersen JB, Durkin ME, Thorgeirsson SS (2009) Loss of miR-122 expression in liver cancer correlates with suppression of the hepatic phenotype and gain of metastatic properties. Oncogene 28: 3526-3536. doi:https://doi.org/10.1038/onc.2009.211. PubMed: 19617899.
  40. 40. Bai S, Nasser MW, Wang B, Hsu SH, Datta J et al. (2009) MicroRNA-122 inhibits tumorigenic properties of hepatocellular carcinoma cells and sensitizes these cells to sorafenib. J Biol Chem 284: 32015-32027. doi:https://doi.org/10.1074/jbc.M109.016774. PubMed: 19726678.
  41. 41. Lanford RE, Hildebrandt-Eriksen ES, Petri A, Persson R, Lindow M et al. (2010) Therapeutic silencing of microRNA-122 in primates with chronic hepatitis C virus infection. Science 327: 198-201. doi:https://doi.org/10.1126/science.1178178. PubMed: 19965718.
  42. 42. Elmén J, Lindow M, Schütz S, Lawrence M, Petri A et al. (2008) LNA-mediated microRNA silencing in non-human primates. Nature 452: 896-899. doi:https://doi.org/10.1038/nature06783. PubMed: 18368051.
  43. 43. Esau C, Davis S, Murray SF, Yu XX, Pandey SK et al. (2006) miR-122 regulation of lipid metabolism revealed by in vivo antisense targeting. Cell Metab 3: 87-98. doi:https://doi.org/10.1016/j.cmet.2006.01.005. PubMed: 16459310.
  44. 44. Krützfeldt J, Rajewsky N, Braich R, Rajeev KG, Tuschl T et al. (2005) Silencing of microRNAs in vivo with 'antagomirs'. Nature 438: 685-689. doi:https://doi.org/10.1038/nature04303. PubMed: 16258535.
  45. 45. Jopling CL, Yi M, Lancaster AM, Lemon SM, Sarnow P (2005) Modulation of hepatitis C virus RNA abundance by a liver-specific MicroRNA. Science 309: 1577-1581. doi:https://doi.org/10.1126/science.1113329. PubMed: 16141076.
  46. 46. Chang J, Nicolas E, Marks D, Sander C, Lerro A et al. (2004) miR-122, a mammalian liver-specific microRNA, is processed from hcr mRNA and may downregulate the high affinity cationic amino acid transporter CAT-1. RNA Biol 1: 106-113. doi:https://doi.org/10.4161/rna.1.2.1066. PubMed: 17179747.
  47. 47. Li Y, Masaki T, Yamane D, McGivern DR, Lemon SM (2013) Competing and noncompeting activities of miR-122 and the 5' exonuclease Xrn1 in regulation of hepatitis C virus replication. Proc Natl Acad Sci U S A 110: 1881-1886. doi:https://doi.org/10.1073/pnas.1213515110. PubMed: 23248316.
  48. 48. Yin G, Chen R, Alvero AB, Fu HH, Holmberg J et al. (2010) TWISTing stemness, inflammation and proliferation of epithelial ovarian cancer cells through MIR199A2/214. Oncogene 29: 3545-3553. doi:https://doi.org/10.1038/onc.2010.111. PubMed: 20400975.
  49. 49. Ueda T, Volinia S, Okumura H, Shimizu M, Taccioli C et al. (2010) Relation between microRNA expression and progression and prognosis of gastric cancer: a microRNA expression analysis. Lancet Oncol 11: 136-146. doi:https://doi.org/10.1016/S1470-2045(09)70343-2. PubMed: 20022810.
  50. 50. Marchini S, Cavalieri D, Fruscio R, Calura E, Garavaglia D et al. (2010) Association between miR-200c and the survival of patients with stage I epithelial ovarian cancer: a retrospective study of two independent tumour tissue collections. Lancet Oncol 12: 273-285.
  51. 51. Juan AH, Kumar RM, Marx JG, Young RA, Sartorelli V (2009) Mir-214-dependent regulation of the polycomb protein Ezh2 in skeletal muscle and embryonic stem cells. Mol Cell 36: 61-74. doi:https://doi.org/10.1016/j.molcel.2009.08.008. PubMed: 19818710.
  52. 52. Flynt AS, Li N, Thatcher EJ, Solnica-Krezel L, Patton JG (2007) Zebrafish miR-214 modulates Hedgehog signaling to specify muscle cell fate. Nat Genet 39: 259-263. doi:https://doi.org/10.1038/ng1953. PubMed: 17220889.
  53. 53. Wang X, Guo B, Li Q, Peng J, Yang Z et al. (2013) miR-214 targets ATF4 to inhibit bone formation. Nat Med 19: 93-100. PubMed: 23223004.
  54. 54. Al-Nakhle H, Burns PA, Cummings M, Hanby AM, Hughes TA et al. (2010) Estrogen receptor {beta}1 expression is regulated by miR-92 in breast cancer. Cancer Res 70: 4778-4784. doi:https://doi.org/10.1158/0008-5472.CAN-09-4104. PubMed: 20484043.
  55. 55. Uziel T, Karginov FV, Xie S, Parker JS, Wang YD et al. (2009) The miR-17~92 cluster collaborates with the Sonic Hedgehog pathway in medulloblastoma. Proc Natl Acad Sci U S A 106: 2812-2817. doi:https://doi.org/10.1073/pnas.0809579106. PubMed: 19196975.
  56. 56. Tanaka M, Oikawa K, Takanashi M, Kudo M, Ohyashiki J et al. (2009) Down-regulation of miR-92 in human plasma is a novel marker for acute leukemia patients. PLOS ONE 4: e5532. doi:https://doi.org/10.1371/journal.pone.0005532. PubMed: 19440243.
  57. 57. Volinia S, Calin GA, Liu CG, Ambs S, Cimmino A et al. (2006) A microRNA expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci U S A 103: 2257-2261. doi:https://doi.org/10.1073/pnas.0510565103. PubMed: 16461460.
  58. 58. Li Z, Yang CS, Nakashima K, Rana TM (2011) Small RNA-mediated regulation of iPS cell generation. EMBO J 30: 823-834. doi:https://doi.org/10.1038/emboj.2011.2. PubMed: 21285944.
  59. 59. Zhi F, Chen X, Wang S, Xia X, Shi Y et al. (2010) The use of hsa-miR-21, hsa-miR-181b and hsa-miR-106a as prognostic indicators of astrocytoma. Eur J Cancer 46: 1640-1649. doi:https://doi.org/10.1016/j.ejca.2010.02.003. PubMed: 20219352.
  60. 60. Tsujiura M, Ichikawa D, Komatsu S, Shiozaki A, Takeshita H et al. (2010) Circulating microRNAs in plasma of patients with gastric cancers. Br J Cancer 102: 1174-1179. doi:https://doi.org/10.1038/sj.bjc.6605608. PubMed: 20234369.
  61. 61. Sharma A, Kumar M, Aich J, Hariharan M, Brahmachari SK et al. (2009) Posttranscriptional regulation of interleukin-10 expression by hsa-miR-106a. Proc Natl Acad Sci U S A 106: 5761-5766. doi:https://doi.org/10.1073/pnas.0808743106. PubMed: 19307576.
  62. 62. He L, Thomson JM, Hemann MT, Hernando-Monge E, Mu D et al. (2005) A microRNA polycistron as a potential human oncogene. Nature 435: 828-833. doi:https://doi.org/10.1038/nature03552. PubMed: 15944707.
  63. 63. Ventura A, Young AG, Winslow MM, Lintault L, Meissner A et al. (2008) Targeted deletion reveals essential and overlapping functions of the miR-17 through 92 family of miRNA clusters. Cell 132: 875-886. doi:https://doi.org/10.1016/j.cell.2008.02.019. PubMed: 18329372.
  64. 64. Ivanovska I, Ball AS, Diaz RL, Magnus JF, Kibukawa M et al. (2008) MicroRNAs in the miR-106b family regulate p21/CDKN1A and promote cell cycle progression. Mol Cell Biol 28: 2167-2174. doi:https://doi.org/10.1128/MCB.01977-07. PubMed: 18212054.
  65. 65. Koralov SB, Muljo SA, Galler GR, Krek A, Chakraborty T et al. (2008) Dicer ablation affects antibody diversity and cell survival in the B lymphocyte lineage. Cell 132: 860-874. doi:https://doi.org/10.1016/j.cell.2008.02.020. PubMed: 18329371.
  66. 66. Fontana L, Pelosi E, Greco P, Racanicchi S, Testa U et al. (2007) MicroRNAs 17-5p-20a-106a control monocytopoiesis through AML1 targeting and M-CSF receptor upregulation. Nat Cell Biol 9: 775-787. doi:https://doi.org/10.1038/ncb1613. PubMed: 17589498.
  67. 67. Theiler K (1972) The house mouse: atlas of embryonic development. New York.
  68. 68. O’Rahilly R, Müller F (1987) Developmental stages in human embryos : including a revision of Streeter’s "Horizons" and a survey of the Carnegie collection. Washington, D.C.: Carnegie Institution of Washington.
  69. 69. Altuvia Y, Landgraf P, Lithwick G, Elefant N, Pfeffer S et al. (2005) Clustering and conservation patterns of human microRNAs. Nucleic Acids Res 33: 2697-2706. doi:https://doi.org/10.1093/nar/gki567. PubMed: 15891114.
  70. 70. Bentwich I, Avniel A, Karov Y, Aharonov R, Gilad S et al. (2005) Identification of hundreds of conserved and nonconserved human microRNAs. Nat Genet 37: 766-770. doi:https://doi.org/10.1038/ng1590. PubMed: 15965474.
  71. 71. Kiezun A, Artzi S, Modai S, Volk N, Isakov O et al. (2012) miRviewer: a multispecies microRNA homologous viewer. BMC Res Notes 5: 92. doi:https://doi.org/10.1186/1756-0500-5-92. PubMed: 22330228.
  72. 72. Lin S, Cheung WK, Chen S, Lu G, Wang Z et al. (2010) Computational identification and characterization of primate-specific microRNAs in human genome. Comput Biol Chem 34: 232-241. doi:https://doi.org/10.1016/j.compbiolchem.2010.08.001. PubMed: 20863765.
  73. 73. Li D, Wang Q, Liu C, Duan H, Zeng X et al. (2012) Aberrant expression of miR-638 contributes to benzo(a)pyrene-induced human cell transformation. Toxicol Sci 125: 382-391. doi:https://doi.org/10.1093/toxsci/kfr299. PubMed: 22048643.
  74. 74. Yi C, Wang Q, Wang L, Huang Y, Li L et al. (2012) MiR-663, a microRNA targeting p21(WAF1/CIP1), promotes the proliferation and tumorigenesis of nasopharyngeal carcinoma. Oncogene 31: 4421-4433. doi:https://doi.org/10.1038/onc.2011.629. PubMed: 22249270.
  75. 75. Jian P, Li ZW, Fang TY, Jian W, Zhuan Z et al. (2011) Retinoic acid induces HL-60 cell differentiation via the upregulation of miR-663. Hematol Oncol 4: 20. doi:https://doi.org/10.1186/1756-8722-4-20. PubMed: 21518431.
  76. 76. Jones CI, Zabolotskaya MV, King AJ, Stewart HJ, Horne GA et al. (2012) Identification of circulating microRNAs as diagnostic biomarkers for use in multiple myeloma. Br J Cancer 107: 1987-1996. doi:https://doi.org/10.1038/bjc.2012.525. PubMed: 23169280.
  77. 77. Shinozuka E, Miyashita M, Mizuguchi Y, Akagi I, Kikuchi K et al. (2013) SnoN/SKIL modulates proliferation through control of hsa-miR-720 transcription in esophageal cancer cells. Biochem Biophys Res Commun 430: 101-106. doi:https://doi.org/10.1016/j.bbrc.2012.11.005. PubMed: 23154181.
  78. 78. Liao JM, Zhou X, Zhang Y, Lu H (2012) MiR-1246: a new link of the p53 family with cancer and Down syndrome. Cell Cycle 11: 2624-2630. doi:https://doi.org/10.4161/cc.20809. PubMed: 22751441.
  79. 79. Zhang Y, Liao JM, Zeng SX, Lu H (2011) p53 downregulates Down syndrome-associated DYRK1A through miR-1246. EMBO Rep 12: 811-817. doi:https://doi.org/10.1038/embor.2011.98. PubMed: 21637297.
  80. 80. Katsushima K, Shinjo K, Natsume A, Ohka F, Fujii M et al. (2012) Contribution of microRNA-1275 to Claudin11 protein suppression via a polycomb-mediated silencing mechanism in human glioma stem-like cells. J Biol Chem 287: 27396-27406. doi:https://doi.org/10.1074/jbc.M112.359109. PubMed: 22736761.
  81. 81. Majid S, Dar AA, Saini S, Shahryari V, Arora S et al. (2012) MicroRNA-1280 inhibits invasion and metastasis by targeting ROCK1 in bladder cancer. PLOS ONE 7: e46743. doi:https://doi.org/10.1371/journal.pone.0046743. PubMed: 23056431.
  82. 82. Piepoli A, Tavano F, Copetti M, Mazza T, Palumbo O et al. (2012) Mirna expression profiles identify drivers in colorectal and pancreatic cancers. PLOS ONE 7: e33663. doi:https://doi.org/10.1371/journal.pone.0033663. PubMed: 22479426.
  83. 83. Iorio MV, Croce CM (2009) MicroRNAs in cancer: small molecules with a huge impact. J Clin Oncol 27: 5848-5856. doi:https://doi.org/10.1200/JCO.2009.24.0317. PubMed: 19884536.
  84. 84. Friedman JM, Jones PA (2009) MicroRNAs: critical mediators of differentiation, development and disease. Swiss Med Wkly 139: 466-472. PubMed: 19705306.
  85. 85. Stefani G, Slack FJ (2008) Small non-coding RNAs in animal development. Nat Rev Mol Cell Biol 9: 219-230. doi:https://doi.org/10.1038/nrm2347. PubMed: 18270516.
  86. 86. Kato M, Slack FJ (2008) microRNAs: small molecules with big roles - C. elegans to human cancer. Biol Cell 100: 71-81. doi:https://doi.org/10.1042/BC20070078. PubMed: 18199046.
  87. 87. Garzon R, Croce CM (2008) MicroRNAs in normal and malignant hematopoiesis. Curr Opin Hematol 15: 352-358. doi:https://doi.org/10.1097/MOH.0b013e328303e15d. PubMed: 18536574.
  88. 88. Kusenda B, Mraz M, Mayer J, Pospisilova S (2006) MicroRNA biogenesis, functionality and cancer relevance. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 150: 205-215. doi:https://doi.org/10.5507/bp.2006.029. PubMed: 17426780.
  89. 89. Kluiver J, Kroesen BJ, Poppema S, van den Berg A (2006) The role of microRNAs in normal hematopoiesis and hematopoietic malignancies. Leukemia 20: 1931-1936. doi:https://doi.org/10.1038/sj.leu.2404387. PubMed: 16990772.
  90. 90. Tsukamoto Y, Nakada C, Noguchi T, Tanigawa M, Nguyen LT et al. (2010) MicroRNA-375 is downregulated in gastric carcinomas and regulates cell survival by targeting PDK1 and 14-3-3zeta. Cancer Res 70: 2339-2349. doi:https://doi.org/10.1158/1538-7445.AM10-2339. PubMed: 20215506.