Skip to main content
Erschienen in: BMC Medical Genetics 1/2019

Open Access 01.12.2019 | Research article

Interpreting the MicroRNA-15/107 family: interaction identification by combining network based and experiment supported approach

verfasst von: Si Wang, Wenhua Zhu, Jing Xu, Yuanxu Guo, Jidong Yan, Liesu Meng, Congshan Jiang, Shemin Lu

Erschienen in: BMC Medical Genetics | Ausgabe 1/2019

Abstract

Background

The highly conservative miR-15/107 family (also named as miR-15/107 gene group) including ten miRNA members is currently recognized strongly implicated in multiple human disorders. Some studies focus on the entire family rather than individual miRNA for a bigger picture, while there is also certain signature dysregulation for some of the individual miRNA implicated even in the same disorder.

Methods

Faced with the exponential growth of experimental evidence, our study tries to analyze their function and target interactions using various bioinformatics tools.

Results

Firstly, the evolutionary conservative “AGCAGC” sequence and possible clustered transcriptional pattern were described. Secondly, both the experimentally validated and bioinformatically predicted miRNA-target gene relationship of the entire family was analyzed to understand the mechanism of underlying collective effects for target regulation from the miR-15/107 family. Moreover, pathway analysis among miR-15/107 family was performed and displayed in detail, while its impact on cell proliferation is experimentally validated. Eventually, the dysregulation of miR-15/107 in diseases was discussed.

Conclusions

In summary, our study proposes that the collective functions and implication of miR-15/107 family in various human diseases are achieved relying on the massive overlapping target genes. While the minor differences within target gene interaction among family members could also explain the signature behavior for some of the individual miRNA in aspects such as its disease-specific dysregulation and various participation in pathways.

Background

General background of evolutionarily conservative miR-15/107 family

The miR-15/107 family (also called as miR-15/107 gene group) contains multiple highly conservative miRNA members, including miR-15a-5p, miR-15b-5p, miR-16-5p, miR-103a-3p, miR-107, miR-195-5p, miR-424-5p, miR-497-5p, miR-503-5p and miR-646 [1]. Mature miRNA form of this entire family was found highly expressed in eleven human tissues including the cerebral cortex, frontal cortex, primary visual cortex, thalamus, heart, lung, liver, kidney, spleen, stomach and skeletal muscle [2]. These miRNAs share a common “AGCAGC” sequence within the crucial No.2-No.7 seed binding region for their targets [1], which leads to the potential functional overlap among this family.

Family behavior vs. individual signature

Since this miRNA family was recognized [3], the researchers started to capture the impact of the entire family rather than focus exclusively on a single one of such miRNA, which helped us better understand their biological role [4]. The entire miR-15/107 family was found to suppress the BRCA1 gene at a post-transcriptional level in nine cell lines [5]. The mitogen and growth factor Granulin was dysregulated resulted from the miR-15/107 gene group in multiple human cancers [6]. The miR-15/107 family was found to regulate its target gene CDK5R1/p35 during Alzheimer’s disease (AD) pathogenesis [7].
Besides the important family behavior of miR-15/107, there was also certain signature expression profile for some of individual miRNA implicated even in the same disorder. For example, multiple miR-15/107 family members were developed for therapy of Alzheimer’s disease (AD), among which the miR-16 was selected as the best candidate for simultaneously suppressing multiple AD biomarkers including Aβ and Tau [8]. The investigation focused on this same disorder discovered that all the other members of the miR-15/107 family were down-regulated in gray matter of temporal cortex from the AD patients except for the miR-424, which was up-regulated in white matter in AD [9].

Aim of study

In short, the miR-15/107 family is widely considered with strong influences on human biology in health and disease. Faced with the exponential growth of experimental evidence as well as the mounting bioinformatics tools for miRNA dissection [10], this study aims to analyze the target interactions and its implication in multiple pathways and various diseases.

Methods

miRNA sequence alignment

Analysis of “AGCAGC” sequence within mature miRNA molecules was achieved according to the “mature.fa” dataset downloaded from miRBase website, a well-recognized online repository of miRNA sequences and associated annotation [1115].

Search for genomic position of miRNA host genes

miRNA host genes were found with help of the UCSC Genome Browser [16], a graphical viewing and analyzing tool for genomic information based on the human genome assemblies and annotations.

miRNA-mRNA target relationship analysis

miRNA-mRNA target was analyzed for both experimentally validated interactions using TarBase v8.0 and computationally predicted interactions using Targetscan database (release 7.2, updated in March 2018) [17].

Analysis of multi-miRNA regulatory network

miRTargetLink database calculates the integrated multi-miRNA mode of regulatory network for the understanding of collective effects according to the experimentally validated targets with strong or weak evidence [18].

miRNA pathway analysis

The miRNA regulated KEGG pathway [19] was analyzed using DIANA-miRPath v3.0 [20], a web-server using merging and meta-analysis algorithms according to either predicted or experimentally validated miRNA target interactions. Here in this study, we chose to analyze the validated miRNA interactions using archive in TarBase [21]. Using this algorithm, P < 0.05 in Fisher’s Exact Test (for hypergeometric distribution) was considered as statistically significant.

Cell proliferation assay

SW982 cells were cultured in DMEM high glucose medium (Hyclone, USA) supplemented with 10% FBS (Hyclone, USA) and 0.2% penicillin/streptomycin. Gain of miR-15/107 function was achieved by transfecting 10 nM miRNA mimic (Genepharma company, China) with Lipofectamine 3000 reagent (Invitrogen, USA) according to the manufacturer’s instruction. The scramble miRNA mimic served as negative control (NC). 48 h after cell transfection, CCK8 reagent was applied to the cells and incubated at 37 °C for 1 h. Optical density value was measured at 450 nm. The data was further normalized as percentage against NC group. Results for cell proliferation assay are represented as the mean ± SEM from the three independent cell experiments. Mann-Whitney test was used to analyze the statistical difference between the indicated group and NC group. P value less than 0.05 was considered as statistically significant.

Analysis of dysregulated miR-15/107 in disease

miR2Disease is a manually curated database offering the miRNA-disease relationship, miRNA expression pattern in the disease state, the miRNA expression detection method, experimentally verified miRNA target genes and literature references [22]. With the help of miR2Disease, we could understand the comprehensive potential role in disease state.

Results

The “AGCAGC” sequence for evolutionarily conservative miR-15/107 family

According to the archive in miRBase (Release 22, updated in March 2018), the miR-15/107 family (such as miR-15a-5p, miR-15b-5p, miR-16-5p, miR-103a-3p, miR-107, miR-195-5p, miR-424-5p, miR-497-5p, miR-503-5p and miR-646) all shared an evolutionarily conservative “AGCAGC” sequence at the 5′ end of the miRNAs (Table 1). After investigating all the 2656 mature human miRNAs identified up to date, we found that another miRNA named miR-6838-5p also contains the specific “AGCAGC” sequence at the seed binding region. The systemic concept of miR-15/107 family was characterized in 2010 [1], before the discovery of miR-6838-5p using RNA sequencing in mice and human [23]. Hence, in this study we also included it for further analysis. In addition, there are five other miRNAs (miR-191-5p, miR-4640-5p, miR-6762-5p, miR-6812-5p and miR-6868-5p) harboring an “AGCAGC” sequence within its full length, while for now there is no evidence for any similarity in the potential target binding function with their targets.
Table 1
The “AGCAGC” sequence for evolutionarily conservative miR-15/107 family
miRNAs
miRBase Accession Number
Sequence (from 5′ to 3′)
hsa-miR-15a-5p
MIMAT0000068
UAGCAGCACAUAAUGGUUUGUG
hsa-miR-15b-5p
MIMAT0000417
UAGCAGCACAUCAUGGUUUACA
hsa-miR-16-5p
MIMAT0000069
UAGCAGCACGUAAAUAUUGGCG
hsa-miR-103a-3p
MIMAT0000101
AGCAGCAUUGUACAGGGCUAUGA
hsa-miR-107
MIMAT0000104
AGCAGCAUUGUACAGGGCUAUCA
hsa-miR-195-5p
MIMAT0000461
UAGCAGCACAGAAAUAUUGGC
hsa-miR-424-5p
MIMAT0001341
CAGCAGCAAUUCAUGUUUUGAA
hsa-miR-497-5p
MIMAT0002820
CAGCAGCACACUGUGGUUUGU
hsa-miR-503-5p
MIMAT0002874
UAGCAGCGGGAACAGUUCUGCAG
hsa-miR-646
MIMAT0003316
AAGCAGCUGCCUCUGAGGC
hsa-miR-6838-5p
MIMAT0027578
AAGCAGCAGUGGCAAGACUCCU

Possible transcriptional pattern in clusters

To figure out the possible underlying mechanism of the dysregulated miRNA expression profiles in health and disease at transcriptional level, the precursor transcripts of these miRNAs were searched for gene clusters (host gene and chromosome position of these miRNAs were detailed in Table 2). With the help of UCSC Genome Browser, it was found that there were 4 pairs of neighboring miRNA clusters including miR-15a-5p/miR-16-5p, miR-15b-5p/miR-16-5p, miR-195-5p/ miR-497-5p, miR-424-5p/miR-503-5p within the common host genes. Moreover, the host genes of miR-103a-3p and miR-107 independently but coincidently belong to the pantothenate kinase family, even though located at entirely different chromosomes. All the above-mentioned clues indicated that the miR-15/107 family might be synergistically transcribed in pairs except for the miR-646 and miR-6838-5p.
Table 2
The precursor transcripts of the miR-15/107 family
miRNAs
Host gene
Transcript location
Chromosome position
hsa-miR-15a-5p
deleted in lymphocytic leukemia 2 (DLEU2) non-protein coding gene
Intron
chr13: 50049119–50,049,201 [−]
hsa-miR-15b-5p
structural maintenance of chromosomes 4 (SMC4) protein-coding gene
Intron
chr3: 160404588–160,404,685 [+]
hsa-miR-16-5p
structural maintenance of chromosomes 4 (SMC4) protein-coding gene
Intron
chr3: 160404745–160,404,825 [+]
deleted in lymphocytic leukemia 2 (DLEU2) non-protein coding gene
Intron
chr13: 50048973–50,049,061 [−]
hsa-miR-103a-3p
pantothenate kinase 3 (PANK3) protein-coding gene
Intron
chr5: 168560896–168,560,973 [−]
pantothenate kinase 2 (PANK2) protein-coding gene
Intron
chr20: 3917494–3,917,571 [+]
hsa-miR-107
pantothenate kinase 1 (PANK1) protein-coding gene
Intron
chr10: 89592747–89,592,827 [−]
hsa-miR-195-5p
mir-497-195 cluster host gene (MIR497HG) long non-coding RNA
N/A
chr17: 7017615–7,017,701 [−]
hsa-miR-424-5p
MIR503 host gene (MIR503HG) long non-coding RNA
N/A
chrX: 134546614–134,546,711 [−]
hsa-miR-497-5p
mir-497-195 cluster host gene (MIR497HG) long non-coding RNA
N/A
chr17: 7017911–7,018,022 [−]
hsa-miR-503-5p
MIR503 host gene (MIR503HG) long non-coding RNA
N/A
chrX: 134546328–134,546,398 [−]
hsa-miR-646
MIR646 host gene (MIR646HG) long non-coding RNA
N/A
chr20: 60308474–60,308,567 [+]
hsa-miR-6838-5p
polymerase (DNA) mu (POLM) protein-coding gene
Exon of transcript variant 2, intron of transcript variant 1 and 3
chr7: 44073378–44,073,433 [−]
N/A not applicable

Interaction of miR-15/107 family and their target genes

Mounting evidence and bioinformatics tools for miRNA dissection provided us lots of information for miR-15/107 regulated target genes (Fig. 1a). As above-mentioned, the common “AGCAGC” sequence within the seed binding region is the crucial characteristic of miR-15/107 family. Hence, plenty of shared target genes could be widely expected. There are plenty of attributes shared in common within the miR-15/107 family. However, there are also quite a lot of differences in the target gene profiles among individual members which defined their various function (Fig. 1b and c). The similarity and difference of both the computationally predicted and experimentally validated target gene profiles were analyzed and displayed in Additional files 1 and 2. The results showed that most of these target genes (above 78%) were predicted to be overlapping for more than 8 miR-15/107 members, while the experimentally validated miRNA-target interaction archived in TarBase was relatively less informative for those types of target genes. It was possible that either plenty of predicted targets might be false positive, or the present attention was not paid enough for the part of target genes. So far, the numbers for miR-424-5p, miR-503-5p targets were relatively less than the others, while the miRNA targets for miR-646-5p and miR-6838-5p was still absent in TarBase database.
The overlapping target genes were analyzed and a cluster dendrogram of miR-15/107 family was plotted accordingly (Fig. 2). Concerning the profile of their target genes, it was shown that there are three pairs of miRNAs including miR-15a-5p/miR-15b-5p, miR-103a-3p/miR-107 and miR-424/miR-497 closely resembling each other, while three of them including the miR-6838-5p, miR-503-5p and miR-646 vary independently.

Collective effects of target regulation from the miR-15/107 family

Since there are massive overlapping target genes among the miR-15/107 family, we further calculated and emphasized the collective effects of target regulation from multi members. According to the regulatory network calculated by miRTargetLink database, we can see that dozens of target genes (located in the central part of this network) could be affected by this family collectively, especially for genes such as CCNE1, CCND1, VEGFA and so on which were simultaneously subject to regulation from more than 5 members of this family (Fig. 3).

Pathway analysis of the miR-15/107 family

miR-15/107 regulated pathways were calculated using mirPath v3.0 web-server, and the results showed that 39 KEGG pathways (see Additional file 3) were significantly (P < 0.05 in Fisher’s Exact Test) regulated by the miR-15/107 family (Fig. 4). Pathway calculation was based on merging and meta-analysis algorithms according to experimentally validated miRNA target interactions, hence we could find that cluster dendrogram based on miRNA regulated pathways was consistent with that of miRNA target gene. The pathways regulated by the miR-15a-5p/miR-15b-5p, miR-103a-3p/miR-107 and miR-424/miR-497 closely resembled each other, while the miR-6838-5p, miR-503-5p and miR-646 vary independently. Besides, the cluster dendrogram of these pathways further showed that the most significantly regulated pathways included the fatty acid metabolism/biosynthesis/degradation/elongation, various signaling during carcinogenesis, and some crucial pathways for cell survival such as cell cycle, meiosis, adherent junction et al.

Cell cycle as a representative pathway regulated by the miR-15/107 family

Here, we take the cell cycle pathway as an example, to uncover the implication of miR-15/107 family in determining cell fate. miR-15/107 targeted genes within the cell cycle pathway was highlighted in Fig. 5 and listed in Additional file 4. The results showed that 47 of 65 cell cycle related genes (up to 72%) were regulated by miR-15/107 family. Particularly, some of these target genes play multi-functional roles as molecular contacts within the pathway, such as CDKN2A, RBL1, E2F5, TFDP1, RBX1, SKP2, and MCM3. Besides, cell cycle related complex machineries including ORC (origin recognition complex) and MCM (mini-chromosome maintenance complex) were also regulated by miR-15/107. More importantly, there were up to 15 genes subject to regulation from more than 5 members from the miR-15/107 family simultaneously. These bioinformatics evidence suggested a strong implication of miR-15/107 family in cell cycle regulation, while such prediction was also widely supported by experimental evidence (see Additional file 5). Taken together, miR-15/107 family was strongly implicated in multiple pathways including the cell cycle regulation.

Members of the miR-15/107 gene family have an inhibitory effect on cell proliferation

Based on our previous signaling pathway prediction, this family may play an important role in the cell cycle pathway. We validated their role during cell proliferation in human synovial fibroblast cell line SW982 after gain of miR-15/107 function for 48 h. The results showed that the miRNA mimic for miR-16, miR-497, miR-503, miR-646 and miR-6838 displayed a significant inhibition on cell proliferation (Fig. 6). The results demonstrated that this gene family indeed has impact on cell cycle signaling as we predicted.

Dysregulation of miR-15/107 in diseases and prospect of therapeutics

According to the archives in miR2Disease, dysregulated miR-15/107 family was reported in various human diseases, especially in cardiac hypertrophy [24], chronic lymphocytic leukemia (CLL) [25] and prostate cancer [26] (Table 3). More than 5 of such members were simultaneously found abnormally expressed in those diseases, while there are also some of the members individually participated in various diseases such as miR-103 in cerebellar neurodegeneration [27].
Table 3
Dysregulated miR-15/107 family in human diseases
 
miR-15a
miR-15b
miR-16
miR-103
miR-107
miR-195
miR-424
miR-497
miR-503
sum
Adrenocortical carcinoma
-
-
-
-
-
-
-
-
1
Alzheimer's disease
-
-
-
-
-
-
-
2
Acute lymphoblastic leukemia (ALL)
-
-
-
-
-
-
-
-
1
Acute myeloid leukemia (AML)
-
-
-
-
-
4
Acute promyelocytic leukemia (APL)
-
-
-
-
-
-
-
2
Autism spectrum disorder (ASD)
-
-
-
-
-
-
-
2
B-cell chronic lymphocytic leukemia
-
-
-
-
-
-
-
-
1
Bladder cancer
-
-
-
-
-
-
-
-
1
Breast cancer
-
-
-
-
-
-
-
2
Cardiac hypertrophy
-
-
-
-
5
Cerebellar neurodegeneration
-
-
-
-
-
-
-
-
1
Chronic lymphocytic leukemia (CLL)
-
-
-
-
5
Chronic pancreatitis
-
-
-
-
-
-
-
2
Colorectal cancer
-
-
-
-
-
4
Endometriosis
-
-
-
-
-
-
-
-
1
Epithelial ovarian cancer (EOC)
-
-
-
-
-
-
-
-
1
Esophageal cancer
-
-
-
-
-
-
-
2
Gastric cancer (stomach cancer)
-
-
-
-
-
4
Glioma
-
-
-
-
-
-
3
Heart failure
-
-
-
-
-
-
-
-
1
Head and neck squamous cell carcinoma (HNSCC)
-
-
-
-
-
-
3
Hepatocellular carcinoma (HCC)
-
-
-
-
-
4
Hodgkin's lymphoma
-
-
-
-
-
-
-
-
1
Intrahepatic cholangiocarcinoma (ICC)
-
-
-
-
-
-
-
-
1
Kidney cancer
-
-
-
-
-
-
-
2
Lung cancer
-
-
-
-
-
-
3
Lupus nephritis
-
-
-
-
-
-
-
2
Malignant melanoma
-
-
-
-
-
-
-
2
Non-alcoholic fatty liver disease (NAFLD)
-
-
-
-
-
-
-
2
Non-small cell lung cancer (NSCLC)
-
-
-
-
-
4
Ovarian cancer (OC)
-
-
-
-
-
4
Oral Squamous Cell Carcinoma (OSCC)
-
-
-
-
-
-
-
2
Pancreatic cancer
-
-
-
-
-
4
Papillary thyroid carcinoma (PTC)
-
-
-
-
-
-
-
2
Pituitary adenoma
-
-
-
-
-
-
-
2
Polycystic Kidney Disease
-
-
-
-
-
-
-
-
1
Polycystic liver disease
-
-
-
-
-
-
-
-
1
Prostate cancer
-
-
-
6
Retinoblastoma
-
-
-
-
-
-
-
-
1
Schizophrenia
-
-
-
-
-
4
Serous ovarian cancer
-
-
-
-
-
-
-
-
1
Ulcerative colitis (UC)
-
-
-
-
-
-
-
2
Total (42)
17
12
13
10
13
16
10
5
3
 
Diseases with more than 5 dysregulated miR-15/107 members were set in bold
These dysregulated miRNAs offered a prospect of therapeutics. Some lncRNAs were discovered to serve as miRNA sponge and since might alter disease progression. For example, miR-107 is found upregulated in glioma cell lines and binds to LncRNA nuclear paraspeckle assembly transcript 1 (NEAT1). NEAT1 silencing inhibits glioma progression, and NEAT1 induces glioma progression by regulating miR-107 as its endogenous sponge [28]. Besides, lncRNA RP11-79H23.3 might suppress the pathogenesis and development of bladder cancer by acting as a sponge for miR-107 to increase PTEN expression [29].

Discussion

In this study, function and target interactions of miR-15/107 family was analyzed using various bioinformatics tools. Firstly, the evolutionary conservative “AGCAGC” sequence, possible clustered transcriptional pattern and tissue specific expression profile were described. Secondly, both the experimentally validated and bioinformatically predicted miRNA-target gene relationship of the entire family was fully interpreted to understand the mechanism of underlying collective effects. Moreover, pathway analysis among miR-15/107 family was performed and displayed in detail. Eventually, the dysregulation of miR-15/107 in diseases was discussed.
According to the previous reports, the implication of miR-15/107 family in various diseases has aroused much attention from multiple fields. In chronic lymphocytic lymphoma (CLL), downregulated miR-15a/miR-16-1 was widely studied [3032]. Their CLL related target genes includes BCL2/MCL1/CCND1/WNT3A [30], BAZ2A/RNF41/RASSF5/MKK3/LRIG1 [31] et al. The miR-15/107 was also strongly implicated in Alzheimer’s disease. Alzheimer is firstly found to be associated with miR-107, it’s levels decreased significantly even in patients with the earliest stages of pathology [33]. The role of miR-15/107 family during AD pathogenesis was also found related with the target suppressing on CDK5R1/p35. Our study focuses on explaining how this miR-15/107 family displayed a collective function and similar implication in various human diseases. We agree that the major contributor should be the massive overlapping miRNA: mRNA target interaction shared crossing the entire family. For this part, we have displayed plenty of convincing evidence achieved from both the computationally prediction and the previous experimentally validation.
As above-mentioned, the common “AGCAGC” sequence within the 5′ seed binding region is the crucial characteristic of miR-15/107 family. Hence, plenty of shared target genes could be widely expected. Besides the important 5′ portion similarity, 3’portion also contribute to target binding. For example, 3′ portion of miR-103/107 appears to play a role in causing miRNAs to bind preferentially with CDS of target mRNAs [4].
We believe that there were also some additional clues from the transcriptional level of miRNA themselves. As shown in Table 2, there were several miRNA members within the miR-15/107 family transcribed in the same clusters including miR-15a-5p/miR-16-5p, miR-15b-5p/miR-16-5p, miR-195-5p/miR-497-5p, and miR-424-5p/miR-503-5p. It was considered that miRNAs in the same clusters might evolve to coordinately regulate the functionally related genes [34]. It is known that some evolutionarily conserved miRNAs are significantly enriched in miRNA clusters. According to this theory, the survival of new miRNAs in clusters is related to the function of pre-existing miRNAs in this cluster. For example, clustering miRNAs can synergistically target overlapping genomes, and new miRNAs can share this targeting function with the help of functional co-adaptation patterns, thereby successfully survive in clusters and performing other functions. This functional co-adaptation may be the driving force of clustering and persist in the initial stages of new miRNA clusters formation. Once their coordinated regulation of target genes is established, miRNA clusters will maintained by natural selection. Our existing data support this theory. We displayed that PANK is the host gene for miR-103/107, and this gene is known as a core player in the regulation of intracellular CoA. MiR-103/107 acts on genes in metabolic pathways in a synergistic manner with its host gene [35, 36]. Moreover, we considered that the coordinately regulating network derived from those clustered miRNAs might contribute to the majority of the present complicated regulation pattern to lots of target genes, and eventually lead to the multi-miRNA/target interaction network as we described.

Conclusions

Our study proposes that the collective functions and implication of miR-15/107 family in various human diseases are achieved relying on the massive overlapping target genes. While the minor differences within target gene interaction among family members could also explain the signature behavior for some of the individual miRNA in aspects such as its disease-specific dysregulation and participation in pathways.

Acknowledgements

Not applicable.

Funding

This work was supported by the National Natural Science Foundation of China (No. 81671629, 81401289, 81701619 and 31771262), Postdoctoral Science Foundation of China (2015 M580858), and Shannxi Province Natural Science Foundation (Project No. 2018JM7057). The funding body had no role in the design of the study and collection, analysis, and interpretation of data and in writing the manuscript.

Availability of data and materials

All data generated or analysed during this study are included in the published article [and its supplementary information file] except for the proliferation assay data which is available upon request from the corresponding author.
Not applicable.
Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Finnerty JR, Wang WX, Hebert SS, Wilfred BR, Mao G, Nelson PT. The miR-15/107 group of microRNA genes: evolutionary biology, cellular functions, and roles in human diseases. J Mol Biol. 2010;402(3):491–509.CrossRef Finnerty JR, Wang WX, Hebert SS, Wilfred BR, Mao G, Nelson PT. The miR-15/107 group of microRNA genes: evolutionary biology, cellular functions, and roles in human diseases. J Mol Biol. 2010;402(3):491–509.CrossRef
2.
Zurück zum Zitat Wang WX, Danaher RJ, Miller CS, Berger JR, Nubia VG, Wilfred BS, Neltner JH, Norris CM, Nelson PT. Expression of miR-15/107 family microRNAs in human tissues and cultured rat brain cells. Genomics Proteomics Bioinformatics. 2014;12(1):19–30.CrossRef Wang WX, Danaher RJ, Miller CS, Berger JR, Nubia VG, Wilfred BS, Neltner JH, Norris CM, Nelson PT. Expression of miR-15/107 family microRNAs in human tissues and cultured rat brain cells. Genomics Proteomics Bioinformatics. 2014;12(1):19–30.CrossRef
3.
Zurück zum Zitat Linsley PS, Schelter J, Burchard J, Kibukawa M, Martin MM, Bartz SR, Johnson JM, Cummins JM, Raymond CK, Dai H, et al. Transcripts targeted by the microRNA-16 family cooperatively regulate cell cycle progression. Mol Cell Biol. 2007;27(6):2240–52.CrossRef Linsley PS, Schelter J, Burchard J, Kibukawa M, Martin MM, Bartz SR, Johnson JM, Cummins JM, Raymond CK, Dai H, et al. Transcripts targeted by the microRNA-16 family cooperatively regulate cell cycle progression. Mol Cell Biol. 2007;27(6):2240–52.CrossRef
4.
Zurück zum Zitat Nelson PT, Wang WX, Mao G, Wilfred BR, Xie K, Jennings MH, Gao Z, Wang X. Specific sequence determinants of miR-15/107 microRNA gene group targets. Nucleic Acids Res. 2011;39(18):8163–72.CrossRef Nelson PT, Wang WX, Mao G, Wilfred BR, Xie K, Jennings MH, Gao Z, Wang X. Specific sequence determinants of miR-15/107 microRNA gene group targets. Nucleic Acids Res. 2011;39(18):8163–72.CrossRef
5.
Zurück zum Zitat Quann K, Jing Y, Rigoutsos I. Post-transcriptional regulation of BRCA1 through its coding sequence by the miR-15/107 group of miRNAs. Front Genet. 2015;6:242.CrossRef Quann K, Jing Y, Rigoutsos I. Post-transcriptional regulation of BRCA1 through its coding sequence by the miR-15/107 group of miRNAs. Front Genet. 2015;6:242.CrossRef
6.
Zurück zum Zitat Wang WX, Kyprianou N, Wang X, Nelson PT. Dysregulation of the mitogen granulin in human cancer through the miR-15/107 microRNA gene group. Cancer Res. 2010;70(22):9137–42.CrossRef Wang WX, Kyprianou N, Wang X, Nelson PT. Dysregulation of the mitogen granulin in human cancer through the miR-15/107 microRNA gene group. Cancer Res. 2010;70(22):9137–42.CrossRef
7.
Zurück zum Zitat Moncini S, Lunghi M, Valmadre A, Grasso M, Del Vescovo V, Riva P, Denti MA, Venturin M. The miR-15/107 family of microRNA genes regulates CDK5R1/p35 with implications for Alzheimer's disease pathogenesis. Mol Neurobiol. 2017;54(6):4329–42.CrossRef Moncini S, Lunghi M, Valmadre A, Grasso M, Del Vescovo V, Riva P, Denti MA, Venturin M. The miR-15/107 family of microRNA genes regulates CDK5R1/p35 with implications for Alzheimer's disease pathogenesis. Mol Neurobiol. 2017;54(6):4329–42.CrossRef
8.
Zurück zum Zitat Parsi S, Smith PY, Goupil C, Dorval V, Hebert SS. Preclinical evaluation of miR-15/107 family members as multifactorial drug targets for Alzheimer's disease. Mol Ther. 2015;4:e256. Parsi S, Smith PY, Goupil C, Dorval V, Hebert SS. Preclinical evaluation of miR-15/107 family members as multifactorial drug targets for Alzheimer's disease. Mol Ther. 2015;4:e256.
9.
Zurück zum Zitat Wang WX, Huang Q, Hu Y, Stromberg AJ, Nelson PT. Patterns of microRNA expression in normal and early Alzheimer's disease human temporal cortex: white matter versus gray matter. Acta Neuropathol. 2011;121(2):193–205.CrossRef Wang WX, Huang Q, Hu Y, Stromberg AJ, Nelson PT. Patterns of microRNA expression in normal and early Alzheimer's disease human temporal cortex: white matter versus gray matter. Acta Neuropathol. 2011;121(2):193–205.CrossRef
10.
Zurück zum Zitat Akhtar MM, Micolucci L, Islam MS, Olivieri F, Procopio AD. Bioinformatic tools for microRNA dissection. Nucleic Acids Res. 2016;44(1):24–44.CrossRef Akhtar MM, Micolucci L, Islam MS, Olivieri F, Procopio AD. Bioinformatic tools for microRNA dissection. Nucleic Acids Res. 2016;44(1):24–44.CrossRef
11.
Zurück zum Zitat Kozomara A, Griffiths-Jones S. miRBase: annotating high confidence microRNAs using deep sequencing data. Nucleic Acids Res. 2014;42(Database issue):D68–73.CrossRef Kozomara A, Griffiths-Jones S. miRBase: annotating high confidence microRNAs using deep sequencing data. Nucleic Acids Res. 2014;42(Database issue):D68–73.CrossRef
12.
Zurück zum Zitat Kozomara A, Griffiths-Jones S. miRBase: integrating microRNA annotation and deep-sequencing data. Nucleic Acids Res. 2011;39(Database):D152–7.CrossRef Kozomara A, Griffiths-Jones S. miRBase: integrating microRNA annotation and deep-sequencing data. Nucleic Acids Res. 2011;39(Database):D152–7.CrossRef
13.
Zurück zum Zitat Griffiths-Jones S, Saini HK, van Dongen S, Enright AJ. miRBase: tools for microRNA genomics. Nucleic Acids Res. 2008;36(Database issue):D154–8.PubMed Griffiths-Jones S, Saini HK, van Dongen S, Enright AJ. miRBase: tools for microRNA genomics. Nucleic Acids Res. 2008;36(Database issue):D154–8.PubMed
14.
Zurück zum Zitat Griffiths-Jones S, Grocock RJ, van Dongen S, Bateman A, Enright AJ. miRBase: microRNA sequences, targets and gene nomenclature. Nucleic Acids Res. 2006;34(Database issue):D140–4.CrossRef Griffiths-Jones S, Grocock RJ, van Dongen S, Bateman A, Enright AJ. miRBase: microRNA sequences, targets and gene nomenclature. Nucleic Acids Res. 2006;34(Database issue):D140–4.CrossRef
15.
Zurück zum Zitat Griffiths-Jones S. The microRNA registry. Nucleic Acids Res. 2004;32(Database issue):D109–11.CrossRef Griffiths-Jones S. The microRNA registry. Nucleic Acids Res. 2004;32(Database issue):D109–11.CrossRef
16.
Zurück zum Zitat Kent WJ, Sugnet CW, Furey TS, Roskin KM, Pringle TH, Zahler AM, Haussler D. The human genome browser at UCSC. Genome Res. 2002;12(6):996–1006.CrossRef Kent WJ, Sugnet CW, Furey TS, Roskin KM, Pringle TH, Zahler AM, Haussler D. The human genome browser at UCSC. Genome Res. 2002;12(6):996–1006.CrossRef
17.
Zurück zum Zitat Agarwal V, Bell GW, Nam JW, Bartel DP. Predicting effective microRNA target sites in mammalian mRNAs. eLife. 2015;4:e05005. Agarwal V, Bell GW, Nam JW, Bartel DP. Predicting effective microRNA target sites in mammalian mRNAs. eLife. 2015;4:e05005.
18.
Zurück zum Zitat Hamberg M, Backes C, Fehlmann T, Hart M, Meder B, Meese E, Keller A. MiRTargetLink--miRNAs, genes and interaction networks. Int J Mol Sci. 2016;17(4):564.CrossRef Hamberg M, Backes C, Fehlmann T, Hart M, Meder B, Meese E, Keller A. MiRTargetLink--miRNAs, genes and interaction networks. Int J Mol Sci. 2016;17(4):564.CrossRef
19.
Zurück zum Zitat Kanehisa M, Furumichi M, Tanabe M, Sato Y, Morishima K. KEGG: new perspectives on genomes, pathways, diseases and drugs. Nucleic Acids Res. 2017;45(D1):D353–61.CrossRef Kanehisa M, Furumichi M, Tanabe M, Sato Y, Morishima K. KEGG: new perspectives on genomes, pathways, diseases and drugs. Nucleic Acids Res. 2017;45(D1):D353–61.CrossRef
20.
Zurück zum Zitat Vlachos IS, Zagganas K, Paraskevopoulou MD, Georgakilas G, Karagkouni D, Vergoulis T, Dalamagas T, Hatzigeorgiou AG. DIANA-miRPath v3.0: deciphering microRNA function with experimental support. Nucleic Acids Res. 2015;43(W1):W460–6.CrossRef Vlachos IS, Zagganas K, Paraskevopoulou MD, Georgakilas G, Karagkouni D, Vergoulis T, Dalamagas T, Hatzigeorgiou AG. DIANA-miRPath v3.0: deciphering microRNA function with experimental support. Nucleic Acids Res. 2015;43(W1):W460–6.CrossRef
21.
Zurück zum Zitat Karagkouni D, Paraskevopoulou MD, Chatzopoulos S, Vlachos IS, Tastsoglou S, Kanellos I, Papadimitriou D, Kavakiotis I, Maniou S, Skoufos G, et al. DIANA-TarBase v8: a decade-long collection of experimentally supported miRNA-gene interactions. Nucleic Acids Res. 2018;46(D1):D239–45.CrossRef Karagkouni D, Paraskevopoulou MD, Chatzopoulos S, Vlachos IS, Tastsoglou S, Kanellos I, Papadimitriou D, Kavakiotis I, Maniou S, Skoufos G, et al. DIANA-TarBase v8: a decade-long collection of experimentally supported miRNA-gene interactions. Nucleic Acids Res. 2018;46(D1):D239–45.CrossRef
22.
Zurück zum Zitat Jiang Q, Wang Y, Hao Y, Juan L, Teng M, Zhang X, Li M, Wang G, Liu Y. miR2Disease: a manually curated database for microRNA deregulation in human disease. Nucleic Acids Res. 2009;37(Database):D98–104.CrossRef Jiang Q, Wang Y, Hao Y, Juan L, Teng M, Zhang X, Li M, Wang G, Liu Y. miR2Disease: a manually curated database for microRNA deregulation in human disease. Nucleic Acids Res. 2009;37(Database):D98–104.CrossRef
23.
Zurück zum Zitat Ladewig E, Okamura K, Flynt AS, Westholm JO, Lai EC. Discovery of hundreds of mirtrons in mouse and human small RNA data. Genome Res. 2012;22(9):1634–45.CrossRef Ladewig E, Okamura K, Flynt AS, Westholm JO, Lai EC. Discovery of hundreds of mirtrons in mouse and human small RNA data. Genome Res. 2012;22(9):1634–45.CrossRef
24.
Zurück zum Zitat Sayed D, Hong C, Chen IY, Lypowy J, Abdellatif M. MicroRNAs play an essential role in the development of cardiac hypertrophy. Circ Res. 2007;100(3):416–24.CrossRef Sayed D, Hong C, Chen IY, Lypowy J, Abdellatif M. MicroRNAs play an essential role in the development of cardiac hypertrophy. Circ Res. 2007;100(3):416–24.CrossRef
25.
Zurück zum Zitat Calin GA, Ferracin M, Cimmino A, Di Leva G, Shimizu M, Wojcik SE, Iorio MV, Visone R, Sever NI, Fabbri M, et al. A MicroRNA signature associated with prognosis and progression in chronic lymphocytic leukemia. N Engl J Med. 2005;353(17):1793–801.CrossRef Calin GA, Ferracin M, Cimmino A, Di Leva G, Shimizu M, Wojcik SE, Iorio MV, Visone R, Sever NI, Fabbri M, et al. A MicroRNA signature associated with prognosis and progression in chronic lymphocytic leukemia. N Engl J Med. 2005;353(17):1793–801.CrossRef
26.
Zurück zum Zitat Porkka KP, Pfeiffer MJ, Waltering KK, Vessella RL, Tammela TL, Visakorpi T. MicroRNA expression profiling in prostate cancer. Cancer Res. 2007;67(13):6130–5.CrossRef Porkka KP, Pfeiffer MJ, Waltering KK, Vessella RL, Tammela TL, Visakorpi T. MicroRNA expression profiling in prostate cancer. Cancer Res. 2007;67(13):6130–5.CrossRef
27.
Zurück zum Zitat Schaefer A, O'Carroll D, Tan CL, Hillman D, Sugimori M, Llinas R, Greengard P. Cerebellar neurodegeneration in the absence of microRNAs. J Exp Med. 2007;204(7):1553–8.CrossRef Schaefer A, O'Carroll D, Tan CL, Hillman D, Sugimori M, Llinas R, Greengard P. Cerebellar neurodegeneration in the absence of microRNAs. J Exp Med. 2007;204(7):1553–8.CrossRef
28.
Zurück zum Zitat Zhen YW, Nan Y, Guo SW, Zhang LZ, Li G, Yue SC, Liu XZ. Knockdown of NEAT1 repressed the malignant progression of glioma through sponging miR-107 and inhibiting CDK14. J Cell Physiol. 2019;234(7):10671–9.CrossRef Zhen YW, Nan Y, Guo SW, Zhang LZ, Li G, Yue SC, Liu XZ. Knockdown of NEAT1 repressed the malignant progression of glioma through sponging miR-107 and inhibiting CDK14. J Cell Physiol. 2019;234(7):10671–9.CrossRef
29.
Zurück zum Zitat Chi H, Yang R, Zheng XY, Zhang LY, Jiang R, Chen JX. LncRNA RP11-79H23.3 functions as a competing endogenous RNA to regulate PTEN expression through sponging hsa-miR-107 in the development of bladder Cancer. Int J Mol Sci. 2018;19(9):2531.CrossRef Chi H, Yang R, Zheng XY, Zhang LY, Jiang R, Chen JX. LncRNA RP11-79H23.3 functions as a competing endogenous RNA to regulate PTEN expression through sponging hsa-miR-107 in the development of bladder Cancer. Int J Mol Sci. 2018;19(9):2531.CrossRef
30.
Zurück zum Zitat Aqeilan RI, Calin GA, Croce CM. miR-15a and miR-16-1 in cancer: discovery, function and future perspectives. Cell Death Differ. 2010;17(2):215–20.CrossRef Aqeilan RI, Calin GA, Croce CM. miR-15a and miR-16-1 in cancer: discovery, function and future perspectives. Cell Death Differ. 2010;17(2):215–20.CrossRef
31.
Zurück zum Zitat Hanlon K, Rudin CE, Harries LW. Investigating the targets of MIR-15a and MIR-16-1 in patients with chronic lymphocytic leukemia (CLL). PLoS One. 2009;4(9):e7169.CrossRef Hanlon K, Rudin CE, Harries LW. Investigating the targets of MIR-15a and MIR-16-1 in patients with chronic lymphocytic leukemia (CLL). PLoS One. 2009;4(9):e7169.CrossRef
32.
Zurück zum Zitat Balatti V, Acunzo M, Pekarky Y, Croce CM. Novel mechanisms of regulation of miRNAs in CLL. Trends Cancer. 2016;2(3):134–43.CrossRef Balatti V, Acunzo M, Pekarky Y, Croce CM. Novel mechanisms of regulation of miRNAs in CLL. Trends Cancer. 2016;2(3):134–43.CrossRef
33.
Zurück zum Zitat Wang WX, Rajeev BW, Stromberg AJ, Ren N, Tang GL, Huang QW, Rigoutsos I, Nelson PT. The expression of microRNA miR-107 decreases early in Alzheimer's disease and may accelerate disease progression through regulation of beta-site amyloid precursor protein-cleaving enzyme 1. J Neurosci. 2008;28(5):1213–23.CrossRef Wang WX, Rajeev BW, Stromberg AJ, Ren N, Tang GL, Huang QW, Rigoutsos I, Nelson PT. The expression of microRNA miR-107 decreases early in Alzheimer's disease and may accelerate disease progression through regulation of beta-site amyloid precursor protein-cleaving enzyme 1. J Neurosci. 2008;28(5):1213–23.CrossRef
34.
Zurück zum Zitat Wang Y, Luo J, Zhang H, Lu J. microRNAs in the same clusters evolve to coordinately regulate functionally related genes. Mol Biol Evol. 2016;33(9):2232–47.CrossRef Wang Y, Luo J, Zhang H, Lu J. microRNAs in the same clusters evolve to coordinately regulate functionally related genes. Mol Biol Evol. 2016;33(9):2232–47.CrossRef
35.
Zurück zum Zitat Wilfred BR, Wang WX, Nelson PT. Energizing miRNA research: a review of the role of miRNAs in lipid metabolism, with a prediction that miR-103/107 regulates human metabolic pathways. Mol Genet Metab. 2007;91(3):209–17.CrossRef Wilfred BR, Wang WX, Nelson PT. Energizing miRNA research: a review of the role of miRNAs in lipid metabolism, with a prediction that miR-103/107 regulates human metabolic pathways. Mol Genet Metab. 2007;91(3):209–17.CrossRef
36.
Zurück zum Zitat Polster BJ, Westaway SK, Nguyen TM, Yoon MY, Hayflick SJ. Discordant expression of miR-103/7 and pantothenate kinase host genes in mouse. Mol Genet Metab. 2010;101(2–3):292–5.CrossRef Polster BJ, Westaway SK, Nguyen TM, Yoon MY, Hayflick SJ. Discordant expression of miR-103/7 and pantothenate kinase host genes in mouse. Mol Genet Metab. 2010;101(2–3):292–5.CrossRef
Metadaten
Titel
Interpreting the MicroRNA-15/107 family: interaction identification by combining network based and experiment supported approach
verfasst von
Si Wang
Wenhua Zhu
Jing Xu
Yuanxu Guo
Jidong Yan
Liesu Meng
Congshan Jiang
Shemin Lu
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
BMC Medical Genetics / Ausgabe 1/2019
Elektronische ISSN: 1471-2350
DOI
https://doi.org/10.1186/s12881-019-0824-9

Weitere Artikel der Ausgabe 1/2019

BMC Medical Genetics 1/2019 Zur Ausgabe