Abstract
Haploinsufficiency of tumor suppressor genes, wherein the reduced production and activity of proteins results in the inability of the cell to maintain normal cellular function, is one among the various causes of cancer. However the precise molecular mechanisms underlying this condition remain unclear. Here we hypothesize that single nucleotide polymorphisms (SNPs) in the 3′untranslated region (UTR) of mRNAs and microRNA seed sequence (miR-SNPs) may cause haploinsufficiency at the level of proteins through altered binding specificity of microRNAs (miRNAs). Bioinformatics analysis of haploinsufficient genes for variations in their 3′UTR showed that the occurrence of SNPs result in the creation of new binding sites for miRNAs, thereby bringing the respective mRNA variant under the control of more miRNAs. In addition, 19 miR-SNPs were found to result in non-specific binding of microRNAs to tumor suppressors. Networking analysis suggests that the haploinsufficient tumor suppressor genes strongly interact with one another, and any subtle alterations in this network will contribute to tumorigenesis.
Keywords: haploinsufficiency, microRNA, single nucleotide polymorphism, miR-SNPs, tumor suppressor genes, cancer
Introduction
Cancer is a complex genetic disease involving structural and expression abnormalities of both coding and non-coding genes. The search for cancer causing genes has identified two major classes namely the oncogenes and tumor suppressor genes (TSGs). Activation or over expression of a proto-oncogene, such as cMyc,1 or inactivation of a tumor suppressor, such as TP53,2,3 causes tumorigenicity. Oncogene activation in tumors is straightforward, while the inactivation of TSGs is a complex process attained by different means including mutations, deletions, down regulation by microRNAs, epigenetic silencing etc. Although complete loss of TSG is common in tumors, recent studies indicate that a partial compromise in TSG function termed ‘haploinsufficiency’ contributes to the development and progression of many cancers.4 The condition where one functional allele of a gene is lost by mutation or deletion, and the remaining normal allele is insufficient to execute its original physiological function, is called haploinsufficiency. This phenomenon is extensively applicable to TSGs5 as they offer a benefit for cancer cells in regards to proliferation,6 survival,7 and metastasis.8
In principle, haploinsufficient TSGs are impaired by a 50% reduction in expression or activity. However, in vivo studies demonstrate that even a subtle 20% reduction of PTEN protein level—termed as ‘quasi-insufficiency’— could contribute to the development of cancer.9 Another example is that TP53, when targeted by short hairpin RNAs (shRNAs), is shown to elicit distinct phenotypes ranging from hyperplasia to malignancy in mouse models depending on the reduction in its protein level.10 This proves that some TSGs, like PTEN, are exquisitely sensitive to dose, while some, like TP53, are intermediately sensitive. Based on such observations, Berger et al has proposed a continuum model that accounts for subtle dosage effects of tumor suppressors including their regulation by microRNAs.11 The dosage and function of haploinsufficient genes are critical, and understanding the impact of haploinsufficiency is important for assessment of interindividual genetic variation, as well as the molecular basis of haploinsufficiency disorders.
Haploinsufficiency of multiple genes cooperate to promote tumorigenesis, a phenomenon called ‘compound haploinsufficiency’. The 5q deletion syndrome (5q-) is a paradigm of compound haploinsufficiency and demonstrates the importance of combinatorial interactions to elicit specific phenotypes.12 Experimental evidence has shown that co-suppression of linked 8p TSGs promotes tumor formation more potently than any individual gene.13 All the available evidence indicates that the functioning of a cell depends on the appropriate expression levels of proteins and, more importantly, that cell signalling pathways involve complex interactions between many proteins. Not only genes, but also microRNAs (miRNAs), a class of small noncoding RNAs, are shown to be haploinsufficient and to cause developmental abnormalities in humans.14 In addition, genes involved in miRNA biosynthesis pathway, such as DICER1, TARBP2 and XPO5, are identified as haploinsufficient tumor suppressors.15–17
miRNAs act at the post-transcriptional level of gene regulation through RNA-induced silencing complex (RISC) mediated translational inhibition or mRNA cleavage. The miRNA target recognition is mediated through the sequence complementarity between the 2–8 nt at the 5′ end of miRNA (seed sequence) and the 3′-UTR of target mRNA.18 Based on the degree of complementarity, the mRNA is either guided for inhibition of translation or degradation resulting in the decrease of protein encoded by the target messenger.19 This has extended the dimensions of haploinsufficiency as miRNAs can lead to the production of insufficient amount of proteins. Bioinformatics prediction and experimental analysis suggested that miRNAs can regulate approximately half of the mammalian genes, with a significant number of important oncogenes and tumor suppressor genes involved.20,21 A recent genome-wide association study has suggested that a gene with more than two miRNA target sites will have higher variability of expression than a gene which is not regulated by a miRNA. The variability is further increased by SNPs in the miRNA target sites.22 Polymorphisms in the miRNA regulatory pathway are a novel class of functional polymorphisms present in the human genome. The initial demonstration that miRNA binding site variations can result in a phenotype was provided by Abelson et al who identified a mutation in the miR-189 binding site of SLITRK1 and its association with Tourette’s syndrome.23 A pioneering study conducted by Carlo Croce’s group showed that a germline mutation in pri-miR-16-1 resulted in low levels of miR-16-1 expression in familial chronic lymphocytic leukemia,24,25 providing evidence that sequence variation in miRNA genes may affect function and result in cancer susceptibility. Pre- and pri-miRNA SNPs in miR-124-1, miR-146a, miR-196-a2, miR-218, miR-219-1, miR-26a-1, miR-27a, miR-423, miR-492, and miR-499 have already been shown to increase/decrease cancer risk in various populations.26
Single nucleotide polymorphisms (SNPs) are single base pair changes in DNA that occur with a frequency of about 1 in 12,500 bp in the genome.27 Several studies have shown that SNPs in microRNA networks moderately increase the risk of cancer incidence.28 In general, sequence variations in pri-miRNAs, premiRNAs, mature miRNAs, and microRNA binding sites potentially affects the processing and/or target selection of miRNAs. As a consequence, aberrant expression of hundreds of genes and pathways greatly affecting miRNA function may occur.29 SNPs in mature miRNAs and miRNA binding sites function analogously to modulate the miRNA-mRNA interaction and create or destroy miRNA binding sites. Supporting this idea, SNPs within the miRNA binding sites of genes have been implicated in susceptibility to various types of cancer.30–33 Additionally, functional support for individual miR-SNPs implicated in cancer do exist.
In this study we analyzed the role of SNPs that occur at miRNA binding sites and miR-SNPs and their contribution towards haploinsufficiency of tumor suppressor genes.
Materials and Methods
Datasets
A total of 110 haploinsufficient genes known to have a role in tumor initiation and progression were considered for the study. Among the total list, 63 tumorigenic haploinsufficient genes were obtained from Dang et al, 200834 while the remaining 47 genes were compiled using keyword search in PubMed and Online Mendelian Inheritance in Man (OMIM) database. For a comprehensive list of the haploinsufficient tumorigenic genes included in this study with their corresponding reference, see Supplement 1.
Analysis of 3′UTR polymorphisms in mRNA targets and miR seed
The haploinsufficient genes were analyzed for polymorphisms in their 3′UTR by submitting the respective gene ID in ‘Polymorphism in microRNA Target Site’ (PolymiRTS) database,35 http://compbio.uthsc.edu/miRSNP/, designed to identify SNPs that disrupt the regulation of gene expression by miRNAs in human and mouse (last accessed on June 7th, 2012). The database is organized to provide links between SNPs in miRNA target sites, cis-acting expression quantitative trait loci (eQTLs), and the results of genome-wide association studies (GWAS) of human diseases. We applied filters to identify only the SNPs in the 3′UTR that were known to create binding sites for miRNAs and SNPs occurring in the seed region of miRNA themselves.
Classification and validation of SNPs obtained from polymiRTS
The SNPs predicted by the polymiRTS database were classified based on their effect into the following categories: (i) SNPs that lead to the creation of miRNA binding sites in the mRNA, (ii) SNPs that disrupted the originally existing miRNA binding sites and also resulted in creation of new miRNA binding sites in the mRNA, and (iii) miR-SNPs that altered the binding specificity of miRNAs. After classification, the SNPs were verified by submitting their rs ID in the NCBI’s dbSNP Short genetic variation database http://www.ncbi.nlm.nih.gov/projects/SNP/using the batch query option. The output was downloaded in BED format and analyzed. Among the total 158 SNPs submitted, 156 SNPs were validated by dbSNP while 2 miR-SNPs (rs116596918 and rs116838571) were found to be new entries and validated by microRNA-related Single Nucleotide Polymorphism database (http://www.bioguo.org/miRNASNP/miRNA_details.php).36
Functional network prediction using GeneMANIA Cytoscape plug-in
To identify the functional significance, GeneMANIA Cytoscape plug-in which employs the GeneMANIA algorithm37,38 was utilized with default settings to plot the interactions among the haploinsufficient TSGs. The complete list of tumorigenic haploinsufficient genes were uploaded to Cytoscape39 using GeneMANIA plug-in (http://www.genemania.org/plugin/). Further, only the genes that were found to have SNPs in their miRNA binding sites and those that are non-specifically targeted by miRNAs harboring miR-SNPs were uploaded individually to study the interactions among this subgroup. The GeneMANIA Cytoscape plug-in integrates association networks from multiple sources into a single composite network using a conjugate gradient optimization algorithm. This algorithm produces networks from the data either directly (as in the case of protein or genetic interactions) or by using an in-house analysis pipeline to convert profiles to functional association networks.
Results and Discussion
Polymorphisms in 3′UTR of mRNA
Our analysis of tumorigenic haploinsufficient genes for genetic variations in the 3′UTR showed that 26% (29 out of 110) of them had at least one 3′UTR SNP resulting in creation of new binding sites for miRNAs thereby bringing the respective mRNA variant under the control of more new miRNAs. In total, 129 SNPs were found to create binding sites for 234 miRNAs in 29 genes (Table 1). Altogether, these SNPs contribute to haploinsufficiency by bringing the polymorphic mRNA under the control of more new microRNAs thereby leading to translational repression or mRNA degradation. PPARA, a nuclear transcription factor and KIF1B, a kinesin implicated in neuronal and non-neuronal tumors was found to harbor 17 SNPs creating putative binding sites for many miRNAs. On the contrary, the SNP rs121912664 in TP53 creates putative miRNA binding site for miRNA-302 family, miR-520 family, miR-372, and miR-373-3p. The cell adhesion molecule CDH1, an important gene in tumor invasion with frequent allelic loss in metastatic tumors, was found to have 5 different SNPs that create putative binding sites for new miRNAs thereby altering the repertoire of miRNAs controlling this gene. Even RB1, the classical TSG, was predicted to have 3 SNPs that may down regulate its expression. If one allele is already lost in tumors, the remaining allele having at least one of these SNPs may be targeted by miRNAs and thereby lead to complete loss of cellular function, as postulated by Knudson’s “two-hit hypothesis.”
Table 1.
Gene | Location | SNP ID | Allele change | microRNA binding site created for |
---|---|---|---|---|
ATM | 108236469 | rs3092834 | C to T | hsa-miR-4252 |
108237460 | rs187308124 | C to T | hsa-miR-4428 | |
108237837 | rs227091 | C to T | 4 microRNAs* | |
108238032 | rs114847811 | C to T | hsa-miR-600 | |
108239075 | rs186644530 | A to G | hsa-miR-2117 | |
108239099 | rs191399133 | T to C | hsa-miR-4448 | |
108239463 | rs3092852 | C to T | hsa-miR-1265 | |
BAG1 | 33253621 | rs191119067 | G to C | hsa-miR-4452, hsa-miR-4680-3p |
CAMTA1 | 7827003 | rs183056496 | G to A | hsa-miR-582-3p |
7828815 | rs112733335 | G to A | hsa-miR-342-3p | |
CDH1 | 68867609 | rs35942505 | C to T | 6 microRNAs* |
68868146 | rs140240766 | C to A | hsa-miR-427 | |
68868576 | rs9282654 | T to G | hsa-miR-129-1-3p, hsa-miR-129-2-3p | |
68869062 | rs33967108 | G to C | hsa-miR-1273a | |
68869394 | rs146556488 | C to A | hsa-miR-369-3p | |
CDKN1B | 12874056 | rs114975362 | G to T | hsa-miR-3919 |
CHEK1 | 125525308 | rs148807246 | A to G | hsa-miR-1184, hsa-miR-1205, hsa-miR-3158-5p |
125545904 | rs192805943 | G to A | hsa-miR-2964a-3p | |
CLU | 27454684 | rs9331949 | A to G | hsa-miR-1283 |
CTCF | 67672530 | rs187258944 | G to T | hsa-miR-2276 |
DFFB | 3800550 | rs146888429 | G to A | hsa-miR-1227 |
3800570 | rs140704651 | C to T | 4 microRNAs* | |
DIRC2 | 122598847 | rs150233566 | G to C | hsa-miR-4797-5p |
122599452 | rs180703954 | A to C | hsa-miR-3927, hsa-miR-4451, hsa-miR-4732-5p | |
ETV6 | 12043995 | rs183579990 | C to G | hsa-miR-3160-3p, hsa-miR-558 |
12044403 | rs79663578 | T to A | hsa-miR-5011-5p | |
12045614 | rs183305984 | G to A | hsa-miR-3690 | |
12045861 | rs72550771 | T to C | hsa-miR-4774-5p | |
FBXW7 | 153243322 | rs4399968 | G to C | hsa-miR-338-3p, hsa-miR-634 |
FEN1 | 61564571 | rs141406755 | T to C | hsa-miR-3654 |
KIF1B | 10365509 | rs180831292 | C to G | hsa-miR-4768-5p |
10365953 | rs72640311 | T to G | hsa-miR-1972 | |
10366284 | rs115354754 | A to C | hsa-miR-3934, hsa-miR-764 | |
10366763 | rs56164683 | A to G | hsa-miR-27a-3p, hsa-miR-27b-3p | |
10366766 | rs187140427 | A to T | hsa-miR-376c | |
10367056 | rs41274464 | G to A | hsa-miR-148b-5p, hsa-miR-3168 | |
10367348 | rs142468272 | C to A | 4 microRNAs* | |
10367422 | rs2004034 | G to A | 6 microRNAs* | |
10367577 | rs143823495 | G to T | hsa-miR-4494, hsa-miR-4499 | |
10367816 | rs145128126 | A to T | hsa-miR-3941, hsa-miR-466, hsa-miR-4672 | |
10437247 | rs2155760 | C to T | 8 microRNAs* | |
10437802 | rs4240913 | G to A | hsa-miR-645 | |
10438336 | rs189264558 | A to G | hsa-miR-516a-3p, hsa-miR-516b-3p | |
10439362 | rs186201500 | A to C | hsa-miR-199a-5p, hsa-miR-199b-5p, hsa-miR-644a | |
10439538 | rs184232081 | G to A | hsa-miR-4425 | |
10440756 | rs192312673 | C to T | hsa-miR-4797-5p | |
10440757 | rs148005450 | G to A | hsa-miR-4534 | |
KLF5 | 73650341 | rs117651109 | C to T | hsa-miR-5579-5p |
MDM2 | 69235650 | rs144972734 | A to G | hsa-miR-671-5p |
69236143 | rs190072365 | C to T | hsa-miR-934 | |
69236269 | rs142063414 | G to A | hsa-miR-3198, hsa-miR-4309 | |
69236548 | rs1690917 | G to T | 4 microRNAs* | |
69237299 | rs183077346 | T to A | hsa-miR-889 | |
69237446 | rs150137388 | A to G | hsa-miR-9-3p | |
69238249 | rs141938174 | C to T | hsa-miR-4705 | |
69238568 | rs148584832 | G to A | hsa-miR-4522 | |
69238940 | rs184278637 | G to A | 6 microRNAs | |
PAX5 | 36839029 | rs184505592 | G to C | hsa-miR-4520a-3p |
PPARA | 46631658 | rs4253801 | A to G | hsa-miR-4709-5p |
46631718 | rs183307497 | A to G | hsa-miR-5095, hsa-miR-600 | |
46631838 | rs188241700 | T to G | hsa-miR-1470, hsa-miR-4287, hsa-miR-4685-3p | |
46632587 | rs9615264 | G to A | hsa-miR-1231 | |
46632932 | rs181140325 | T to A | hsa-miR-4277 | |
46633120 | rs78922364 | G to C | hsa-miR-499b-5p | |
46633549 | rs115356860 | C to T | hsa-miR-892b | |
46633564 | rs148478205 | C to T | hsa-miR-4252 | |
46634352 | rs184920139 | G to A | hsa-miR-301a-5p | |
46634514 | rs41418950 | C to T | hsa-miR-936 | |
46635061 | rs41403648 | G to A | hsa-miR-3944-5p, hsa-miR-5094 | |
46636623 | rs56180992 | G to A | hsa-miR-515-5p, hsa-miR-519e-5p | |
46637760 | rs181781384 | A to G | hsa-miR-4733-3p | |
46638155 | rs1055658 | G to C | hsa-miR-744-5p | |
46638209 | rs79700435 | G to A | hsa-miR-3191-5p, hsa-miR-518c-5p | |
46639161 | rs187605878 | A to G | hsa-miR-3157-3p | |
46639611 | rs117317496 | G to C | hsa-miR-5584-5p | |
PTCH1 | 98205709 | rs28413010 | A to T | hsa-miR-126-5p, hsa-miR-4795-3p |
98206995 | rs41316950 | C to G | hsa-miR-4690-3p, hsa-miR-5685 | |
PTEN | 89725452 | rs3895069 | T to G | hsa-miR-5003-5p |
89726622 | rs79395513 | C to A | hsa-miR-651 | |
89726726 | rs180953647 | C to T | hsa-miR-510 | |
RAD50 | 131980172 | rs146790944 | A to G | hsa-miR-300, hsa-miR-381 |
131980267 | rs139468574 | A to G | hsa-miR-4524a-5p, hsa-miR-4524b-5p | |
131980270 | rs75939007 | A to G | 4 microRNAs* | |
RB1 | 49054521 | rs187845274 | T to A | hsa-miR-4638-3p |
49054779 | rs150194261 | T to C | hsa-miR-145-5p, hsa-miR-5195-3p | |
49055241 | rs192758219 | G to T | hsa-miR-1323, hsa-miR-548o-3p | |
RXRA | 137328831 | rs1805346 | G to A | hsa-miR-3688-3p |
137328936 | rs189590058 | C to T | hsa-miR-4738-3p | |
137329304 | rs4240711 | G to A | hsa-miR-3184-3p | |
137329411 | rs34449651 | G to C | hsa-miR-3919 | |
137329652 | rs35283205 | G to C | hsa-miR-4685-5p | |
137330306 | rs180869938 | T to C | hsa-miR-1908, hsa-miR-663a | |
137330727 | rs17847916 | C to T | hsa-miR-4721 | |
137330802 | rs10119893 | G to A | 5 microRNAs* | |
137330980 | rs139882385 | T to C | hsa-miR-518a-5p, hsa-miR-527 | |
137331443 | rs139749844 | G to A | hsa-miR-3170, hsa-miR-5572 | |
137331999 | rs11103561 | G to A | hsa-miR-4722-3p, hsa-miR-4769-3p | |
137332362 | rs187745805 | G to A | hsa-miR-5010-3p, hsa-miR-599 | |
137332378 | rs137912651 | G to C | hsa-miR-301a-5p | |
SERPINB5 | 61171674 | rs112795011 | A to C | hsa-miR-4535, hsa-miR-4776-5p |
SMAD4 | 48605247 | rs28403611 | A to G | hsa-miR-552 |
48605297 | rs191822473 | G to A | hsa-miR-656 | |
48605643 | rs183984903 | A to G | hsa-miR-5095 | |
48607107 | rs189547031 | G to C | hsa-miR-5584-5p | |
48607497 | rs191637734 | A to T | hsa-miR-421, hsa-miR-4272 | |
48608123 | rs191317135 | C to T | hsa-miR-3163 | |
48609056 | rs150556996 | G to T | hsa-miR-4496 | |
48609588 | rs112739300 | C to T | hsa-miR-297, hsa-miR-3149, hsa-miR-675-3p | |
48609716 | rs184516281 | T to C | hsa-miR-584-3p | |
48609918 | rs145596898 | G to A | hsa-miR-4694-3p | |
48610254 | rs146551171 | T to C | 23 microRNAs* | |
48610822 | rs12954419 | T to A | hsa-miR-5696 | |
48610844 | rs181250637 | G to C | hsa-miR-636 | |
48611357 | rs183472455 | C to T | hsa-miR-371b-5p, hsa-miR-373-5p, hsa-miR-616-5p | |
SOX9 | 70121452 | rs74999341 | T to C | hsa-miR-4759 |
70121525 | rs185467299 | C to T | hsa-miR-4514, hsa-miR-4692, hsa-miR-4742-5p | |
TNFAIP3 | 138202555 | rs140042278 | G to T | hsa-miR-208a, hsa-miR-208b, hsa-miR-499a-5p |
138202864 | rs76710415 | A to C | hsa-miR-4533 | |
138203096 | rs73778206 | T to C | hsa-miR-223-3p | |
138203386 | rs191520762 | T to G | hsa-miR-4772-3p | |
138203884 | rs73778207 | G to A | hsa-miR-96-3p | |
TP53 | 7572039 | rs35659787 | G to A | hsa-miR-516b-5p, hsa-miR-518c-5p |
7574010 | rs17882252 | G to A | hsa-miR-127-5p | |
7574015 | rs121912664 | G to A | 12 microRNAs* | |
VHL | 10191771 | rs34362094 | C to T | hsa-miR-5579-5p |
10192172 | rs190102073 | A to G | hsa-miR-4448 | |
10193434 | rs115303528 | G to A | hsa-miR-592, hsa-miR-599 | |
10193848 | rs145666034 | G to A | hsa-miR-3545-3p | |
10194195 | rs187719061 | A to G | hsa-miR-371a-5p |
Notes: This table enlists the 129 SNPs that create new miRNA binding sites in the 3′UTR of their respective haploinsufficient tumor suppressor gene. Table compiled from the data provided by PolymiRTS database.
For convenience these microRNAs are not indicated in the table, and for details the reader is referred to Supplement 2.
Another 10 SNPs were found to bring 8 different genes under the control of additional new miRNAs due to the presence of the respective SNP (Fig. 1). A single SNP can create miR binding site for several miRNAs as well as delete a miR binding site. For example, the SNP rs186304832 in the 3′UTR of KIF1B results in the loss of binding of 2 different microRNAs to the variant mRNA while 7 new microRNAs will be targeting this particular variant. This alters the miRNA mediated gene expression control resulting in aberrant regulation of expression. A gene that was previously under the control of one miRNA comes under the control of 3 different new miRNAs which leads to extensive translational repression resulting in protein haploinsufficiency as represented by the polymorphism rs1138533 in CDH1. This mechanism is totally new, since one allele is not lost due to mutation or deletion, as in the case of classical haploinsufficiency. Rather there is a decrease in the overall protein product. In homozygous state, this SNP may result in complete or partial reduction in the expression based on the ability of the repressing miRNA. However, in heterozygous state, the same SNP can lead to 50% or even subtler reduction in expression, which can explain ‘quasi-insufficiency’.
Ours is the first study to identify the SNP and miRNA mediated control of tumor suppressor dose as well as highlighting the importance of genetic variations in cancer. This mechanism is in agreement with the continuum model for tumor suppression that is related to the level of expression or activity of the TSG rather than to the discrete step-by-step changes in gene copy number.11
Polymorphisms in microRNA seed sequence
In addition, SNPs that occur in the seed sequence of miRNAs can alter the binding specificity of these miRNAs resulting in the binding to new non-target mRNAs. We analyzed 19 such miR-SNPs that cause miRNAs to bind new haploinsufficient tumor suppressor genes (Table 2). The SNP rs4636784 occurring in the seed of hsa-miR-4305 alters the binding specificity, thereby enabling it to target 6 new haploinsufficient genes (CDH1, KIF1B, SMAD4, DFFB, FOXP1, and PTEN). This can contribute to compound haploinsufficiency, wherein multiple genes cooperate to cause a particular phenotype.7,13 Alteration in one miRNA can alter the entire set of genes under its control thereby providing an advantage for cancer initiation and progression. In addition, the acquisition of miR-SNPs in tumors is simple enough to deregulate many genes than the need for individual mutations in all of them. This mechanism of miR-SNP mediated down regulation can also be extended to the unexplained allelic loss of KIF1B in neuroblastomas, where the tumor suppressor was shown to have no mutations or promoter methylation.40 Tumors not showing genetic alterations in the TSG, but with aberrant TSG levels due to miRNA misregulation, could behave like tumors with deletion or mutation of the gene. Importantly, mapping the interactions between miRNAs and TSGs could be useful for defining and predicting cancer susceptibility and therapeutic response.
Table 2.
miR ID | SNP ID | miR seed | Allele | Haploinsufficient gene targets |
---|---|---|---|---|
hsa-mir-125a | rs12975333 | CCCUGA[G/U] | G/T | ATM, MSH2 |
hsa-miR-1268 | rs28599926 | GGGC[G/A]UG | G/A | ATM, PAX5, PPARA, PTEN, NBN |
hsa-miR-1469 | rs116596918 | UCGGC[G/C]C | C/G | DDB2, FOXP1 |
hsa-miR-3610 | rs112072631 | AAUCGG[A/C] | C/A | SERPINB5, SMAD4 |
hsa-miR-3615 | rs112977728 | CU[C/U]UCGG | C/T | RAD50, NBN, TP53BP2 |
hsa-miR-3618 | rs12159555 | GU[C/G]UACA | C/G | DFFB, PPARA, SMAD4, FOXP1 |
hsa-miR-3689b | rs116838571 | GUGAU[A/U]U | T/A | CDH1, CDKN1B, FBXW7, RB1, VHL, BRCA2, ROR2, TP53BP2, YWHAE |
hsa-miR-379 | rs61991156 | GGUAG[A/G]C | A/G | CAMTA1, DFFB, TNFAIP3, VHL |
hsa-miR-4257 | rs74743733 | CAGA[G/A]GU | A/G | CHEK1, DIRC2, ETV6, PAX5, SOX9 |
hsa-miR-4284 | rs11973069 | GGCUCA[C/U] | C/T | BAG1, ETV6, TP53BP2 |
hsa-miR-4305 | rs4636784 | CUA[G/C]ACA | G/C | CDH1, DFFB, FBXW7, KIF1B, PTEN, SMAD4, FOXP1, NBN, MDM2 |
hsa-miR-4322 | rs114399468 | UGUGG[G/A]C | A/G | CHEK1, KIF1B, PTEN, RAD50, SERPINB5 |
hsa-miR-557 | rs78825966 | UUUGCA[C/U] | C/T | BAG1, CHEK1, BRCA2, RPL37A, KLF5 |
hsa-miR-585 | rs62376935 | GG[G/A]CGUA | G/A | BAG1, KIF1B, NBN, TP53BP2 |
hsa-miR-605 | rs113212828 | [A/G]AAUCCC | A/G | RAD50, RB1, VHL, MSH2 |
hsa-miR-642a | rs78902025 | UCCC[U/G]CU | G/T | ATM, PPARA, SERPINB5 |
hsa-miR-642b | rs111664333 | GA[C/U]ACAU | T/C | ATM, RAD50, SMAD4, NBN, TP53BP2 |
hsa-miR-936 | rs79924817 | C[A/G]GUAGA | G/A | ATM, BAG1, PPARA, RAD50, XPO5 |
hsa-miR-938 | rs12416605 | [G/A]CCCUUA | G/A | BAG1, PPARA, PTEN, SMAD4, SOX9, TNFAIP3, XPO5, MDM2 |
Notes: This table represents the miR-SNPs that render microRNAs to bind with non-specificity to haploinsufficient tumor suppressor genes. Table compiled from the data extracted from PolymiRTS database.
Networks
Although reports on haploinsufficient genes associated with cancer is limited, analysis of the existing data shows that at least one third of total haploinsufficient genes to be tumorigenic. Tumor suppressors often act as components of complex networks, the overall function of which can be impaired by genetic and epigenetic alterations.41,42 For this reason we analyzed whether the haploinsufficient tumor suppressor genes share a common pathway or fall in a common interacting or functional network. The GeneMANIA algorithm was utilized for this purpose, and results indicate that almost 99% of haploinsufficient genes have a strong interaction, either directly or indirectly. SMAD5 was excluded from the list as it was not recognized by the algorithm. Co-expression, co-localization, physical and genetic interactions, pathway interactions, and other interactions that arise through shared structural domains were shown to exist between the queried genes (Fig. 2). SPRED1 was the only gene to fall out of this network however co-expression links it to the group. Apart from the queried list of genes, the algorithm identified several other genes to be a part of this network including DROSHA, DFFA, MRE11A, TRAPPC2L, TERF2, CDK6, BUB1, WRN, CCNA2, and RAD51. Interestingly, the haploinsufficient status of some of these genes has already been described in developmental abnormalities and other diseases,43–45 while some others are shown to have a role in tumor formation and their haploinsufficient status is yet to be established.46
Next we tested whether the 39 genes with SNPs in miRNA binding sites or targeted by miRNAs with altered specificity due to miR-SNPs fall in to a particular network or pathway. Results of GeneMANIA networking analysis suggested strong interactions between these genes (Fig. 3). DNA repair genes and cell cycle checkpoint genes were enriched in this network and any aberrations in their expression offers selective advantage for the tumors to acquire additional genomic changes or mutations. DFFB was linked by co-expression, while DIRC2 was shown to be completely out of the group. The following genes were found to be a part of this network and to mediate interactions between the queried genes: MRE11A, DFFA, CCNB1, POLQ, AP1B1, RAD51, CCNG1, NPM1, DDB1, and WRN. The identification of the mRNA targets that mediate the actions of miRNAs in disease pathways can reveal previously unrecognized components that may serve as targets for more traditional drug development.
Conclusion
With the expanding understanding of the roles of miRNAs in various cellular processes and diseases, this is the first report to discuss the role of miRNAs and its contribution towards tumor suppressor gene haploinsufficiency at the protein level via 3′UTR SNPs and miR-SNPs. Further, this could be an alternate means for attaining compound heterozygosity observed in several tumors and the mechanism is in complete agreement with the continuum model of tumor suppression.11 Our data suggests that 26% of the tumorigenic haploinsufficient genes were brought under the control of new miRNAs due to 3′UTR SNPs. The identification of 10 SNPs that drive haploinsufficiency by bringing the polymorphic mRNA under the control of miRNAs, other than the miRNAs which are deleted, needs experimental validation in tumors. This alteration in the miRNA mediated gene regulation may cause predisposition to cancer initiation and progression.
Evidence for co-operative contribution of oncogenic mutations with tumor suppressor haploinsufficiency also exist.47 The realization that miR-SNPs play central roles in the aberrant regulation of tumor suppressor genes has provided a new perspective on our understanding of pathophysiologic mechanisms. In addition, networking analysis reveals strong interactions between the haploinsufficient tumor suppressor genes. Any subtle alteration in this network of genes due to SNPs at the 3′UTR and miR-SNPs may contribute to pathogenesis. We suggest that at least a few among these SNPs and their effect on miRNA binding will aid in the diagnosis and/or prognosis of such type of cancers, if experimentally validated. While challenges remain in this regard, the pace of development in this field suggests that new discoveries are forthcoming.
Our approach has currently focused only on analyzing 3′UTRs, although a small subset of miRNAs can target 5′UTRs, coding regions, and gene promoters. miR-SNPs leading to non-specific binding of miRNAs to tumor suppressors may also result in the loss of binding to their original targets. If the target is an oncogene, it will be over expressed and will result in tumorigenesis. This has not been focused on in this study. Since the rules for miRNA binding to its target changes constantly,48 the databases need constant updating. The 110 tumor suppressor genes chosen for our study were already proven to be haploinsufficient in several cancers through experimental evidence. This list may likely grow due to continuous identification of haploinsufficient tumor suppressors.
Supplementary Data
Supplement I.
Haploinsufficient genes | References |
---|---|
ANX7, APC, ARF, ATM, BAG1, BECN1, BRCA1, BRCA2, BUBR1, CAMTA1, CAV1, CDKN1B, CDKN1C, CDKN2B, CSN5, CTCF, DFFB, DIRC2, EEF1E1, ETV6, FBXW7, FOXP1, H2 AX, INI1, INK4C, KLF6, LIG4, LIS1, LKB1, MEL-18, MYH, NHERF1, NKX3–1, NPM1, P53, PAX5, PLK4, PPARA, PPARG, PRKAR1 A, PTCH1, PTEN, RAD50, RASSF1 A, ROR2, RPRM, SAM68, SERCA2, SMAD5, SOCS1, SOX9, SPRED1, ST7, TGFB1, TP53BP2, TP73, TRAPPC2, TSC1, TSC2, VHL, WT1, WWOX, YWHAE | 11 |
ATR | 18 |
BIM (BCL2 L11) | 17 |
BLM | 21 |
BUB3 | 30 |
CDH1 | 44 |
CDKN1 A | 28 |
CHK1 (CHEK1) | 33 |
CLU | 9 |
CREBBP (CBP) | 49, 47, 5 |
DDB2 | 26 |
DGCR8 | 42 |
DICER1 | 32, 2, 41 |
DLL4 | 19, 23 |
DMP1 | 25, 51, 34 |
DOK2 | 4 |
FBXO11 | 14 |
FEN1 | 31 |
IKZF1 (IKAROS) | 39, 29, 52 |
KIF1Bβ | 40 |
KLF5 | 10 |
MAD2 L1 | 38 |
MDM2 and MDM4 | 50 |
MLH1 | 48 |
MSH2 | 12, 13, 6 |
MUS81 | 36 |
NBN | 15 |
NF1 | 27, 59 |
NF2 | 35 |
PINX1 | 58 |
RAD17 | 8 |
RB1 | 53, 57 |
Ribosomal proteins (RPL35, RPL37 A, RPS19 and RPS8) | 1 |
RPS14 | 16, 3 |
RUNX1 | 45 |
RXRA | 24 |
SERPINB5 (Maspin) | 43 |
SMAD4/DPC4 | 56 |
TARBP2 | 37 |
TIP60 | 20 |
TNFAIP3 | 55, 7 |
TP53BP1 | 54 |
TP63 (TAp63) | 46 |
XPO5 | 22 |
Supplement 2.
Gene | Location | SNP ID | Allele change | miR ID |
---|---|---|---|---|
ATM | 108237837 | rs227091 | C to T | hsa-miR-3664-3p, hsa-miR-4433-3p, hsa-miR-4768-3p, hsa-miR-512-5p |
CDH1 | 68867609 | rs35942505 | C to T | hsa-miR-548ae, hsa-miR-548ah-3p, hsa-miR-548aj-3p, hsa-miR-548am-3p, hsa-miR-548aq-3p, hsa-miR-548x-3p |
DFFB | 3800570 | rs140704651 | C to T | hsa-miR-3664-3p, hsa-miR-4433-3p, hsa-miR-4768-3p, hsa-miR-512-5p |
KIF1B | 10367348 | rs142468272 | C to A | hsa-miR-4435, hsa-miR-4701-5p, hsa-miR-548s, hsa-miR-588 |
10367422 | rs2004034 | G to A | hsa-miR-25-3p, hsa-miR-32-5p, hsa-miR-363-3p, hsa-miR-367-3p, hsa-miR-92a-3p, hsa-miR-92b-3p | |
10437247 | rs2155760 | C to T | hsa-miR-125a-5p, hsa-miR-125b-5p, hsa-miR-4319, hsa-miR-4446-5p, hsa-miR-4732-3p, hsa-miR-4755-5p, hsa-miR-5006-3p, hsa-miR-670 | |
MDM2 | 69236548 | rs1690917 | G to T | hsa-miR-548ac, hsa-miR-548d-3p, hsa-miR-548h-3p, hsa-miR-548z |
69238940 | rs184278637 | G to A | hsa-miR-3609, hsa-miR-4796-3p, hsa-miR-519a-3p, hsa-miR-519b-3p, hsa-miR-519c-3p, hsa-miR-548ah-5p | |
RAD50 | 131980270 | rs75939007 | A to G | hsa-miR-199a-3p, hsa-miR-199b-3p, hsa-miR-3129-5p, hsa-miR-936 |
RXRA | 137330802 | rs10119893 | G to A | hsa-miR-1254, hsa-miR-1271-3p, hsa-miR-3116, hsa-miR-550a-3-5p, hsa-miR-550a-5p |
SMAD4 | 48610254 | rs146551171 | T to C | hsa-miR-142-5p, hsa-miR-548a-5p, hsa-miR-548ab, hsa-miR-548ak, hsa-miR-548am-5p, hsa-miR-548ap-5p, hsa-miR-548aq-5p, hsamiR-548ar-5p, hsa-miR-548as-5p, hsa-miR-548au-5p, hsa-miR-548av-5p, hsa-miR-548b-5p, hsa-miR-548c-5p, hsa-miR-548d-5p, hsa-miR-548h-5p, hsa-miR-548i, hsa-miR-548j, hsa-miR-548k, hsamiR-548o-5p, hsa-miR-548w, hsa-miR-548y, hsa-miR-559, hsa-miR- 5590-3p |
TP53 | 7574015 | rs121912664 | G to A | hsa-miR-302a-3p, hsa-miR-302b-3p, hsa-miR-302c-3p, hsa-miR-302d-3p, hsa-miR-302e, hsa-miR-372, hsa-miR-373-3p, hsa-miR-520a-3p, hsa-miR-520b, hsa-miR-520c-3p, hsa-miR- 520d-3p, hsa-miR-520e |
References
- 1.Amsterdam A, Sadler KC, et al. Many ribosomal protein genes are cancer genes in zebrafish. PLoS Biol. 2004;2(5):E139. doi: 10.1371/journal.pbio.0020139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Bahubeshi A, Bal N, et al. Germline DICER1 mutations and familial cystic nephroma. J Med Genet. 2010;47(12):863–6. doi: 10.1136/jmg.2010.081216. [DOI] [PubMed] [Google Scholar]
- 3.Barlow JL, Drynan LF, et al. A p53-dependent mechanism underlies macrocytic anemia in a mouse model of human 5q- syndrome. Nat Med. 2010;16(1):59–66. doi: 10.1038/nm.2063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Berger AH, Niki M, et al. Identification of DOK genes as lung tumor suppressors. Nat Genet. 2010;42(3):216–23. doi: 10.1038/ng.527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Blough RI, Petrij F, et al. Variation in microdeletions of the cyclic AMP-responsive element-binding protein gene at chromosome band 16p13.3 in the Rubinstein-Taybi syndrome. Am J Med Genet. 2000;90(1):29–34. doi: 10.1002/(sici)1096-8628(20000103)90:1<29::aid-ajmg6>3.0.co;2-z. [DOI] [PubMed] [Google Scholar]
- 6.Bouffler SD, Hofland N, et al. Evidence for Msh2 haploinsufficiency in mice revealed by MNU-induced sister-chromatid exchange analysis. Br J Cancer. 2000;83(10):1291–4. doi: 10.1054/bjoc.2000.1422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Braggio E, Keats JJ, et al. Identification of copy number abnormalities and inactivating mutations in two negative regulators of nuclear factor-kappaB signaling pathways in Waldenstrom’s macroglobulinemia. Cancer Res. 2009;69(8):3579–88. doi: 10.1158/0008-5472.CAN-08-3701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Bric A, Miething C, et al. Functional identification of tumor-suppressor genes through an in vivo RNA interference screen in a mouse lymphoma model. Cancer Cell. 2009;16(4):324–35. doi: 10.1016/j.ccr.2009.08.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Chayka O, Corvetta D, et al. Clusterin, a haploinsufficient tumor suppressor gene in neuroblastomas. J Natl Cancer Inst. 2009;101(9):663–77. doi: 10.1093/jnci/djp063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Chen C, Bhalala HV, et al. A possible tumor suppressor role of the KLF5 transcription factor in human breast cancer. Oncogene. 2002;21(43):6567–72. doi: 10.1038/sj.onc.1205817. [DOI] [PubMed] [Google Scholar]
- 11.Dang VT, Kassahn KS, et al. Identification of human haploinsufficient genes and their genomic proximity to segmental duplications. Eur J Hum Genet. 2008;16(11):1350–7. doi: 10.1038/ejhg.2008.111. [DOI] [PubMed] [Google Scholar]
- 12.de Wind N, Dekker M, et al. Mouse models for hereditary nonpolyposis colorectal cancer. Cancer Res. 1998;58(2):248–55. [PubMed] [Google Scholar]
- 13.DeWeese TL, Shipman JM, et al. Mouse embryonic stem cells carrying one or two defective Msh2 alleles respond abnormally to oxidative stress inflicted by low-level radiation. Proc Natl Acad Sci U S A. 1998;95(20):11915–20. doi: 10.1073/pnas.95.20.11915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Duan S, Cermak L, et al. FBXO11 targets BCL6 for degradation and is inactivated in diffuse large B-cell lymphomas. Nature. 2012;481(7379):90–3. doi: 10.1038/nature10688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Dumon-Jones V, Frappart PO, et al. Nbn heterozygosity renders mice susceptible to tumor formation and ionizing radiation-induced tumorigenesis. Cancer Res. 2003;63(21):7263–9. [PubMed] [Google Scholar]
- 16.Ebert BL, Pretz J, et al. Identification of RPS14 as a 5q- syndrome gene by RNA interference screen. Nature. 2008;451(7176):335–9. doi: 10.1038/nature06494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Egle A, Harris AW, et al. Bim is a suppressor of Myc-induced mouse B cell leukemia. Proc Natl Acad Sci U S A. 2004;101(16):6164–9. doi: 10.1073/pnas.0401471101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Fang Y, Tsao CC, et al. ATR functions as a gene dosage-dependent tumor suppressor on a mismatch repair-deficient background. EMBO J. 2004;23(15):3164–74. doi: 10.1038/sj.emboj.7600315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Gale NW, Dominguez MG, et al. Haploinsufficiency of delta-like 4 ligand results in embryonic lethality due to major defects in arterial and vascular development. Proc Natl Acad Sci U S A. 2004;101(45):15949–54. doi: 10.1073/pnas.0407290101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Gorrini C, Squatrito M, et al. Tip60 is a haplo-insufficient tumour suppressor required for an oncogene-induced DNA damage response. Nature. 2007;448(7157):1063–7. doi: 10.1038/nature06055. [DOI] [PubMed] [Google Scholar]
- 21.Goss KH, Risinger MA, et al. Enhanced tumor formation in mice heterozygous for Blm mutation. Science. 2002;297(5589):2051–3. doi: 10.1126/science.1074340. [DOI] [PubMed] [Google Scholar]
- 22.Grosshans H, Bussing I. MicroRNA biogenesis takes another single hit from microsatellite instability. Cancer Cell. 2010;18(4):295–7. doi: 10.1016/j.ccr.2010.10.005. [DOI] [PubMed] [Google Scholar]
- 23.Hellstrom M, Phng LK, et al. Dll4 signalling through Notch1 regulates formation of tip cells during angiogenesis. Nature. 2007;445(7129):776–80. doi: 10.1038/nature05571. [DOI] [PubMed] [Google Scholar]
- 24.Huang J, Powell WC, et al. Prostatic intraepithelial neoplasia in mice with conditional disruption of the retinoid X receptor alpha allele in the prostate epithelium. Cancer Res. 2002;62(16):4812–9. [PubMed] [Google Scholar]
- 25.Inoue K, Zindy F, et al. Dmp1 is haplo-insufficient for tumor suppression and modifies the frequencies of Arf and p53 mutations in Myc-induced lymphomas. Genes Dev. 2001;15(22):2934–9. doi: 10.1101/gad.929901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Itoh T, Iwashita S, et al. Ddb2 is a haploinsufficient tumor suppressor and controls spontaneous germ cell apoptosis. Hum Mol Genet. 2007;16(13):1578–86. doi: 10.1093/hmg/ddm107. [DOI] [PubMed] [Google Scholar]
- 27.Jacks T, Shih TS, et al. Tumour predisposition in mice heterozygous for a targeted mutation in Nf1. Nat Genet. 1994;7(3):353–61. doi: 10.1038/ng0794-353. [DOI] [PubMed] [Google Scholar]
- 28.Jackson RJ, Engelman RW, et al. p21Cip1 nullizygosity increases tumor metastasis in irradiated mice. Cancer Res. 2003;63(12):3021–5. [PubMed] [Google Scholar]
- 29.Jager R, Gisslinger H, et al. Deletions of the transcription factor Ikaros in myeloproliferative neoplasms. Leukemia. 2010;24(7):1290–8. doi: 10.1038/leu.2010.99. [DOI] [PubMed] [Google Scholar]
- 30.Kalitsis P, Fowler KJ, et al. Increased chromosome instability but not cancer predisposition in haploinsufficient Bub3 mice. Genes Chromosomes Cancer. 2005;44(1):29–36. doi: 10.1002/gcc.20215. [DOI] [PubMed] [Google Scholar]
- 31.Kucherlapati M, Yang K, et al. Haploinsufficiency of Flap endonuclease (Fen1) leads to rapid tumor progression. Proc Natl Acad Sci U S A. 2002;99(15):9924–9. doi: 10.1073/pnas.152321699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Kumar MS, Pester RE, et al. Dicer1 functions as a haploinsufficient tumor suppressor. Genes Dev. 2009;23(23):2700–4. doi: 10.1101/gad.1848209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Lam MH, Liu Q, et al. Chk1 is haploinsufficient for multiple functions critical to tumor suppression. Cancer Cell. 2004;6(1):45–59. doi: 10.1016/j.ccr.2004.06.015. [DOI] [PubMed] [Google Scholar]
- 34.Mallakin A, Sugiyama T, et al. Mutually exclusive inactivation of DMP1 and ARF/p53 in lung cancer. Cancer Cell. 2007;12(4):381–94. doi: 10.1016/j.ccr.2007.08.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.McClatchey AI, Saotome I, et al. The Nf2 tumor suppressor gene product is essential for extraembryonic development immediately prior to gastrulation. Genes Dev. 1997;11(10):1253–65. doi: 10.1101/gad.11.10.1253. [DOI] [PubMed] [Google Scholar]
- 36.McPherson JP, Lemmers B, et al. Involvement of mammalian Mus81 in genome integrity and tumor suppression. Science. 2004;304(5678):1822–6. doi: 10.1126/science.1094557. [DOI] [PubMed] [Google Scholar]
- 37.Melo SA, Ropero S, et al. A TARBP2 mutation in human cancer impairs microRNA processing and DICER1 function. Nat Genet. 2009;41(3):365–70. doi: 10.1038/ng.317. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 38.Michel LS, Liberal V, et al. MAD2 haplo-insufficiency causes premature anaphase and chromosome instability in mammalian cells. Nature. 2001;409(6818):355–9. doi: 10.1038/35053094. [DOI] [PubMed] [Google Scholar]
- 39.Mullighan CG, Miller CB, et al. BCR-ABL1 lymphoblastic leukaemia is characterized by the deletion of Ikaros. Nature. 2008;453(7191):110–4. doi: 10.1038/nature06866. [DOI] [PubMed] [Google Scholar]
- 40.Munirajan AK, Ando K, et al. KIF1Bbeta functions as a haploinsufficient tumor suppressor gene mapped to chromosome 1p36.2 by inducing apoptotic cell death. J Biol Chem. 2008;283(36):24426–34. doi: 10.1074/jbc.M802316200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Rio Frio T, Bahubeshi A, et al. DICER1 mutations in familial multinodular goiter with and without ovarian Sertoli-Leydig cell tumors. JAMA. 2011;305(1):68–77. doi: 10.1001/jama.2010.1910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Schofield CM, Hsu R, et al. Monoallelic deletion of the microRNA biogenesis gene Dgcr8 produces deficits in the development of excitatory synaptic transmission in the prefrontal cortex. Neural Dev. 2011;6:11. doi: 10.1186/1749-8104-6-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Shao LJ, Shi HY, et al. Haploinsufficiency of the maspin tumor suppressor gene leads to hyperplastic lesions in prostate. Cancer Res. 2008;68(13):5143–51. doi: 10.1158/0008-5472.CAN-08-0163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Smits R, Ruiz P, et al. E-cadherin and adenomatous polyposis coli mutations are synergistic in intestinal tumor initiation in mice. Gastroenterology. 2000;119(4):1045–53. doi: 10.1053/gast.2000.18162. [DOI] [PubMed] [Google Scholar]
- 45.Song WJ, Sullivan MG, et al. Haploinsufficiency of CBFA2 causes familial thrombocytopenia with propensity to develop acute myelogenous leukaemia. Nat Genet. 1999;23(2):166–75. doi: 10.1038/13793. [DOI] [PubMed] [Google Scholar]
- 46.Su X, Chakravarti D, et al. TAp63 suppresses metastasis through coordinate regulation of Dicer and miRNAs. Nature. 2010;467(7318):986–90. doi: 10.1038/nature09459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Taine L, Goizet C, et al. Submicroscopic deletion of chromosome 16p13.3 in patients with Rubinstein-Taybi syndrome. Am J Med Genet. 1998;78(3):267–70. [PubMed] [Google Scholar]
- 48.Takagi Y, Takahashi M, et al. Roles of MGMT and MLH1 proteins in alkylation-induced apoptosis and mutagenesis. DNA Repair (Amst) 2003;2(10):1135–46. doi: 10.1016/s1568-7864(03)00134-4. [DOI] [PubMed] [Google Scholar]
- 49.Tanaka Y, Naruse I, et al. Abnormal skeletal patterning in embryos lacking a single Cbp allele: a partial similarity with Rubinstein-Taybi syndrome. Proc Natl Acad Sci U S A. 1997;94(19):10215–20. doi: 10.1073/pnas.94.19.10215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Terzian T, Wang Y, et al. Haploinsufficiency of Mdm2 and Mdm4 in tumorigenesis and development. Mol Cell Biol. 2007;27(15):5479–85. doi: 10.1128/MCB.00555-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Umesako S, Fujisawa K, et al. Atm heterozygous deficiency enhances development of mammary carcinomas in p53 heterozygous knockout mice. Breast Cancer Res. 2005;7(1):R164–70. doi: 10.1186/bcr968. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Virely C, Moulin S, et al. Haploinsufficiency of the IKZF1 (IKAROS) tumor suppressor gene cooperates with BCR-ABL in a transgenic model of acute lymphoblastic leukemia. Leukemia. 2010;24(6):1200–4. doi: 10.1038/leu.2010.63. [DOI] [PubMed] [Google Scholar]
- 53.Vooijs M, Berns A. Developmental defects and tumor predisposition in Rb mutant mice. Oncogene. 1999;18(38):5293–303. doi: 10.1038/sj.onc.1202999. [DOI] [PubMed] [Google Scholar]
- 54.Ward IM, Difilippantonio S, et al. 53BP1 cooperates with p53 and functions as a haploinsufficient tumor suppressor in mice. Mol Cell Biol. 2005;25(22):10079–86. doi: 10.1128/MCB.25.22.10079-10086.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Wolfrum S, Teupser D, et al. The protective effect of A20 on atherosclerosis in apolipoprotein E-deficient mice is associated with reduced expression of NF-kappaB target genes. Proc Natl Acad Sci U S A. 2007;104(47):18601–6. doi: 10.1073/pnas.0709011104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Xu X, Brodie SG, et al. Haploid loss of the tumor suppressor Smad4/Dpc4 initiates gastric polyposis and cancer in mice. Oncogene. 2000;19(15):1868–74. doi: 10.1038/sj.onc.1203504. [DOI] [PubMed] [Google Scholar]
- 57.Zheng L, Flesken-Nikitin A, et al. Deficiency of Retinoblastoma gene in mouse embryonic stem cells leads to genetic instability. Cancer Res. 2002;62(9):2498–502. [PubMed] [Google Scholar]
- 58.Zhou XZ, Huang P, et al. The telomerase inhibitor PinX1 is a major haploin-sufficient tumor suppressor essential for chromosome stability in mice. J Clin Invest. 2011;121(4):1266–82. doi: 10.1172/JCI43452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Zhu Y, Ghosh P, et al. Neurofibromas in NF1: Schwann cell origin and role of tumor environment. Science. 2002;296(5569):920–2. doi: 10.1126/science.1068452. [DOI] [PMC free article] [PubMed] [Google Scholar]
Footnotes
Author Contributions
AKM conceived and designed the study. MM and GR performed the experiments. AKM, MM, and GR analyzed the data. MM and AKM prepared the manuscript. All authors read and approved the final manuscript.
Competing Interests
Author(s) disclose no potential conflicts of interest.
Disclosures and Ethics
As a requirement of publication author(s) have provided to the publisher signed confirmation of compliance with legal and ethical obligations including but not limited to the following: authorship and contributorship, conflicts of interest, privacy and confidentiality and (where applicable) protection of human and animal research subjects. The authors have read and confirmed their agreement with the ICMJE authorship and conflict of interest criteria. The authors have also confirmed that this article is unique and not under consideration or published in any other publication, and that they have permission from rights holders to reproduce any copyrighted material. Any disclosures are made in this section. The external blind peer reviewers report no conflicts of interest.
Funding Sources
This research was supported in part by a grant from the Department of Biotechnology (DBT), New Delhi (Grant Number BT/PR10023/AGR/36/27/2007) to AKM. MM is a recipient of research fellowship from Council of Scientific and Industrial Research (CSIR), New Delhi. We also thank DST-FIST and UGC-SAP for the infrastructure provided by grants to the department.
References
- 1.Nesbit CE, Tersak JM, Prochownik EV. MYC oncogenes and human neoplastic disease. Oncogene. 1999;18(19):3004–16. doi: 10.1038/sj.onc.1202746. [DOI] [PubMed] [Google Scholar]
- 2.Caron de Fromentel C, Soussi T. TP53 tumor suppressor gene: a model for investigating human mutagenesis. Genes Chromosomes Cancer. 1992;4(1):1–15. doi: 10.1002/gcc.2870040102. [DOI] [PubMed] [Google Scholar]
- 3.Hainaut P, Hernandez T, Robinson A, et al. Nucleic Acids Res. 1. Vol. 26. IARC; 1998. Database of p53 gene mutations in human tumors and cell lines: updated compilation, revised formats and new visualisation tools; pp. 205–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Santarosa M, Ashworth A. Haploinsufficiency for tumor suppressor genes: when you don’t need to go all the way. Biochim Biophys Acta. 2004;1654(2):105–22. doi: 10.1016/j.bbcan.2004.01.001. [DOI] [PubMed] [Google Scholar]
- 5.Payne SR, Kemp CJ. Tumor suppressor genetics. Carcinogenesis. 2005;26(12):2031–45. doi: 10.1093/carcin/bgi223. [DOI] [PubMed] [Google Scholar]
- 6.Solimini NL, Xu Q, Mermel CH, et al. Recurrent hemizygous feletions in cancers may optimize proliferative potential. Science. 2012;337(6090):104–9. doi: 10.1126/science.1219580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Smilenov LB, Lieberman HB, Mitchell SA, Baker RA, Hopkins KM, Hall EJ. Combined haploinsufficiency for ATM and RAD9 as a factor in cell transformation, apoptosis, and DNA lesion repair dynamics. Cancer Res. 2005;65(3):933–8. [PubMed] [Google Scholar]
- 8.Iwakuma T, Tochigi Y, Van Pelt CS, et al. Mtbp haploinsufficiency in mice increases tumor metastasis. Oncogene. 2008;27(13):1813–20. doi: 10.1038/sj.onc.1210827. [DOI] [PubMed] [Google Scholar]
- 9.Alimonti A, Carracedo A, Clohessy JG, et al. Subtle variations in Pten dose determine cancer susceptibility. Nat Genet. 2010;42(5):454–8. doi: 10.1038/ng.556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Hemann MT, Fridman JS, Zilfou JT, et al. An epi-allelic series of p53 hypomorphs created by stable RNAi produces distinct tumor phenotypes in vivo. Nat Genet. 2003;33(3):396–400. doi: 10.1038/ng1091. [DOI] [PubMed] [Google Scholar]
- 11.Berger AH, Knudson AG, Pandolfi PP. A continuum model for tumor suppression. Nature. 2011;476(7359):163–9. doi: 10.1038/nature10275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Ebert BL. Deletion 5q in myelodysplastic syndrome: a paradigm for the study of hemizygous deletions in cancer. Leukemia. 2009;23(7):1252–6. doi: 10.1038/leu.2009.53. [DOI] [PubMed] [Google Scholar]
- 13.Xue W, Kitzing T, Roessler S, et al. A cluster of cooperating tumor-suppressor gene candidates in chromosomal deletions. Proc Natl Acad Sci U S A. 2012;109(21):8212–7. doi: 10.1073/pnas.1206062109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.de Pontual L, Yao E, Callier P, et al. Germline deletion of the miR-17 ~92 cluster causes skeletal and growth defects in humans. Nat Genet. 2011;43(10):1026–30. doi: 10.1038/ng.915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Kumar MS, Pester RE, Chen CY, et al. Dicer1 functions as a haploinsufficient tumor suppressor. Genes Dev. 2009;23(23):2700–4. doi: 10.1101/gad.1848209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Melo SA, Ropero S, Moutinho C, et al. A TARBP2 mutation in human cancer impairs microRNA processing and DICER1 function. Nat Genet. 2009;41(3):365–70. doi: 10.1038/ng.317. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 17.Grosshans H, Bussing I. MicroRNA biogenesis takes another single hit from microsatellite instability. Cancer Cell. 2010;18(4):295–7. doi: 10.1016/j.ccr.2010.10.005. [DOI] [PubMed] [Google Scholar]
- 18.Lewis BP, Burge CB, Bartel DP. Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell. 2005;120(1):15–20. doi: 10.1016/j.cell.2004.12.035. [DOI] [PubMed] [Google Scholar]
- 19.Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell. 2009;136(2):215–33. doi: 10.1016/j.cell.2009.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Krol J, Loedige I, Filipowicz W. The widespread regulation of microRNA biogenesis, function and decay. Nat Rev Genet. 2010;11(9):597–610. doi: 10.1038/nrg2843. [DOI] [PubMed] [Google Scholar]
- 21.Fabbri M, Croce CM, Calin GA. MicroRNAs. Cancer J. 2008;14(1):1–6. doi: 10.1097/PPO.0b013e318164145e. [DOI] [PubMed] [Google Scholar]
- 22.Liu J, Rivas FV, Wohlschlegel J, Yates JR, 3rd, Parker R, Hannon GJ. A role for the P-body component GW182 in microRNA function. Nat Cell Biol. 2005;7(12):1261–6. doi: 10.1038/ncb1333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Abelson JF, Kwan KY, O’Roak BJ, et al. Sequence variants in SLITRK1 are associated with Tourette’s syndrome. Science. 2005;310(5746):317–20. doi: 10.1126/science.1116502. [DOI] [PubMed] [Google Scholar]
- 24.Calin GA, Ferracin M, Cimmino A, et al. A MicroRNA signature associated with prognosis and progression in chronic lymphocytic leukemia. N Engl J Med. 2005;353(17):1793–801. doi: 10.1056/NEJMoa050995. [DOI] [PubMed] [Google Scholar]
- 25.Wojcik SE, Rossi S, Shimizu M, et al. Non-codingRNA sequence variations in human chronic lymphocytic leukemia and colorectal cancer. Carcinogenesis. 2010;31(2):208–15. doi: 10.1093/carcin/bgp209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Slaby O, Bienertova-Vasku J, Svoboda M, Vyzula R. Genetic polymorphisms and microRNAs: new direction in molecular epidemiology of solid cancer. J Cell Mol Med. 2012;16(1):8–21. doi: 10.1111/j.1582-4934.2011.01359.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Kruglyak L, Nickerson DA. Variation is the spice of life. Nat Genet. 2001;27(3):234–6. doi: 10.1038/85776. [DOI] [PubMed] [Google Scholar]
- 28.Ryan BM, Robles AI, Harris CC. Genetic variation in microRNA networks: the implications for cancer research. Nat Rev Cancer. 2010;10(6):389–402. doi: 10.1038/nrc2867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Georges M, Coppieters W, Charlier C. Polymorphic miRNA-mediated gene regulation: contribution to phenotypic variation and disease. Curr Opin Genet Dev. 2007;17(3):166–76. doi: 10.1016/j.gde.2007.04.005. [DOI] [PubMed] [Google Scholar]
- 30.Nicoloso MS, Sun H, Spizzo R, et al. Single-nucleotide polymorphisms inside microRNA target sites influence tumor susceptibility. Cancer Res. 2010;70(7):2789–98. doi: 10.1158/0008-5472.CAN-09-3541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Brendle A, Lei H, Brandt A, et al. Polymorphisms in predicted microRNA-binding sites in integrin genes and breast cancer: ITGB4 as prognostic marker. Carcinogenesis. 2008;29(7):1394–9. doi: 10.1093/carcin/bgn126. [DOI] [PubMed] [Google Scholar]
- 32.Chin LJ, Ratner E, Leng S, et al. A SNP in a let-7 microRNA complementary site in the KRAS 3′untranslated region increases non-small cell lung cancer risk. Cancer Res. 2008;68(20):8535–40. doi: 10.1158/0008-5472.CAN-08-2129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Saetrom P, Biesinger J, Li SM, et al. A risk variant in an miR-125b binding site in BMPR1B is associated with breast cancer pathogenesis. Cancer Res. 2009 Sep 15;69(18):7459–65. doi: 10.1158/0008-5472.CAN-09-1201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Dang VT, Kassahn KS, Marcos AE, Ragan MA. Identification of human haploinsufficient genes and their genomic proximity to segmental duplications. Eur J Hum Genet. 2008;16(11):1350–7. doi: 10.1038/ejhg.2008.111. [DOI] [PubMed] [Google Scholar]
- 35.Ziebarth JD, Bhattacharya A, Chen A, Cui Y. PolymiRTS Database 2.0: linking polymorphisms in microRNA target sites with human diseases and complex traits. Nucleic Acids Res. 2012;40(Database issue):D216–21. doi: 10.1093/nar/gkr1026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Gong J, Tong Y, Zhang HM, et al. Genome-wide identification of SNPs in microRNA genes and the SNP effects on microRNA target binding and biogenesis. Hum Mutat. 2012;33(1):254–63. doi: 10.1002/humu.21641. [DOI] [PubMed] [Google Scholar]
- 37.Warde-Farley D, Donaldson SL, Comes O, et al. The GeneMANIA prediction server: biological network integration for gene prioritization and predicting gene function. Nucleic Acids Res. 2010;38(Web Server issue):W214–20. doi: 10.1093/nar/gkq537. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Montojo J, Zuberi K, Rodriguez H, et al. GeneMANIA Cytoscape plug-in: fast gene function predictions on the desktop. Bioinformatics. 2010;26(22):2927–8. doi: 10.1093/bioinformatics/btq562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Smoot ME, Ono K, Ruscheinski J, Wang PL, Ideker T. Cytoscape 2.8: new features for data integration and network visualization. Bioinformatics. 2011;27(3):431–2. doi: 10.1093/bioinformatics/btq675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Munirajan AK, Ando K, Mukai A, et al. KIF1Bbeta functions as a haploinsufficient tumor suppressor gene mapped to chromosome 1p36.2 by inducing apoptotic cell death. J Biol Chem. 2008;283(36):24426–34. doi: 10.1074/jbc.M802316200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Jones RG, Thompson CB. Tumor suppressors and cell metabolism: a recipe for cancer growth. Genes Dev. 2009;23(5):537–48. doi: 10.1101/gad.1756509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Scuoppo C, Miething C, Lindqvist L, et al. A tumor suppressor network relying on the polyamine-hypusine axis. Nature. 2012;487(7406):244–8. doi: 10.1038/nature11126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Chong MM, Rasmussen JP, Rudensky AY, Littman DR. The RNAseIII enzyme Drosha is critical in T cells for preventing lethal inflammatory disease. J Exp Med. 2008;205(9):2005–17. doi: 10.1084/jem.20081219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Moser MJ, Kamath-Loeb AS, Jacob JE, Bennett SE, Oshima J, Monnat RJ., Jr WRN helicase expression in Werner syndrome cell lines. Nucleic Acids Res. 2000;28(2):648–54. doi: 10.1093/nar/28.2.648. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Depienne C, Bouteiller D, Meneret A, et al. RAD51 haploinsufficiency causes congenital mirror movements in humans. Am J Hum Genet. 2012;90(2):301–7. doi: 10.1016/j.ajhg.2011.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Kim DH, Park SE, Kim M, et al. A functional single nucleotide polymorphism at the promoter region of cyclin A2 is associated with increased risk of colon, liver, and lung cancers. Cancer. 2011;117(17):4080–91. doi: 10.1002/cncr.25930. [DOI] [PubMed] [Google Scholar]
- 47.Izeradjene K, Combs C, Best M, et al. Kras(G12D) and Smad4/Dpc4 haplo-insufficiency cooperate to induce mucinous cystic neoplasms and invasive adenocarcinoma of the pancreas. Cancer Cell. 2007;11(3):229–43. doi: 10.1016/j.ccr.2007.01.017. [DOI] [PubMed] [Google Scholar]
- 48.Stefani G, Slack FJ. A ‘pivotal’ new rule for microRNA-mRNA interactions. Nat Struct Mol Biol. 2012;19(3):265–6. doi: 10.1038/nsmb.2256. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Supplement I.
Haploinsufficient genes | References |
---|---|
ANX7, APC, ARF, ATM, BAG1, BECN1, BRCA1, BRCA2, BUBR1, CAMTA1, CAV1, CDKN1B, CDKN1C, CDKN2B, CSN5, CTCF, DFFB, DIRC2, EEF1E1, ETV6, FBXW7, FOXP1, H2 AX, INI1, INK4C, KLF6, LIG4, LIS1, LKB1, MEL-18, MYH, NHERF1, NKX3–1, NPM1, P53, PAX5, PLK4, PPARA, PPARG, PRKAR1 A, PTCH1, PTEN, RAD50, RASSF1 A, ROR2, RPRM, SAM68, SERCA2, SMAD5, SOCS1, SOX9, SPRED1, ST7, TGFB1, TP53BP2, TP73, TRAPPC2, TSC1, TSC2, VHL, WT1, WWOX, YWHAE | 11 |
ATR | 18 |
BIM (BCL2 L11) | 17 |
BLM | 21 |
BUB3 | 30 |
CDH1 | 44 |
CDKN1 A | 28 |
CHK1 (CHEK1) | 33 |
CLU | 9 |
CREBBP (CBP) | 49, 47, 5 |
DDB2 | 26 |
DGCR8 | 42 |
DICER1 | 32, 2, 41 |
DLL4 | 19, 23 |
DMP1 | 25, 51, 34 |
DOK2 | 4 |
FBXO11 | 14 |
FEN1 | 31 |
IKZF1 (IKAROS) | 39, 29, 52 |
KIF1Bβ | 40 |
KLF5 | 10 |
MAD2 L1 | 38 |
MDM2 and MDM4 | 50 |
MLH1 | 48 |
MSH2 | 12, 13, 6 |
MUS81 | 36 |
NBN | 15 |
NF1 | 27, 59 |
NF2 | 35 |
PINX1 | 58 |
RAD17 | 8 |
RB1 | 53, 57 |
Ribosomal proteins (RPL35, RPL37 A, RPS19 and RPS8) | 1 |
RPS14 | 16, 3 |
RUNX1 | 45 |
RXRA | 24 |
SERPINB5 (Maspin) | 43 |
SMAD4/DPC4 | 56 |
TARBP2 | 37 |
TIP60 | 20 |
TNFAIP3 | 55, 7 |
TP53BP1 | 54 |
TP63 (TAp63) | 46 |
XPO5 | 22 |
Supplement 2.
Gene | Location | SNP ID | Allele change | miR ID |
---|---|---|---|---|
ATM | 108237837 | rs227091 | C to T | hsa-miR-3664-3p, hsa-miR-4433-3p, hsa-miR-4768-3p, hsa-miR-512-5p |
CDH1 | 68867609 | rs35942505 | C to T | hsa-miR-548ae, hsa-miR-548ah-3p, hsa-miR-548aj-3p, hsa-miR-548am-3p, hsa-miR-548aq-3p, hsa-miR-548x-3p |
DFFB | 3800570 | rs140704651 | C to T | hsa-miR-3664-3p, hsa-miR-4433-3p, hsa-miR-4768-3p, hsa-miR-512-5p |
KIF1B | 10367348 | rs142468272 | C to A | hsa-miR-4435, hsa-miR-4701-5p, hsa-miR-548s, hsa-miR-588 |
10367422 | rs2004034 | G to A | hsa-miR-25-3p, hsa-miR-32-5p, hsa-miR-363-3p, hsa-miR-367-3p, hsa-miR-92a-3p, hsa-miR-92b-3p | |
10437247 | rs2155760 | C to T | hsa-miR-125a-5p, hsa-miR-125b-5p, hsa-miR-4319, hsa-miR-4446-5p, hsa-miR-4732-3p, hsa-miR-4755-5p, hsa-miR-5006-3p, hsa-miR-670 | |
MDM2 | 69236548 | rs1690917 | G to T | hsa-miR-548ac, hsa-miR-548d-3p, hsa-miR-548h-3p, hsa-miR-548z |
69238940 | rs184278637 | G to A | hsa-miR-3609, hsa-miR-4796-3p, hsa-miR-519a-3p, hsa-miR-519b-3p, hsa-miR-519c-3p, hsa-miR-548ah-5p | |
RAD50 | 131980270 | rs75939007 | A to G | hsa-miR-199a-3p, hsa-miR-199b-3p, hsa-miR-3129-5p, hsa-miR-936 |
RXRA | 137330802 | rs10119893 | G to A | hsa-miR-1254, hsa-miR-1271-3p, hsa-miR-3116, hsa-miR-550a-3-5p, hsa-miR-550a-5p |
SMAD4 | 48610254 | rs146551171 | T to C | hsa-miR-142-5p, hsa-miR-548a-5p, hsa-miR-548ab, hsa-miR-548ak, hsa-miR-548am-5p, hsa-miR-548ap-5p, hsa-miR-548aq-5p, hsamiR-548ar-5p, hsa-miR-548as-5p, hsa-miR-548au-5p, hsa-miR-548av-5p, hsa-miR-548b-5p, hsa-miR-548c-5p, hsa-miR-548d-5p, hsa-miR-548h-5p, hsa-miR-548i, hsa-miR-548j, hsa-miR-548k, hsamiR-548o-5p, hsa-miR-548w, hsa-miR-548y, hsa-miR-559, hsa-miR- 5590-3p |
TP53 | 7574015 | rs121912664 | G to A | hsa-miR-302a-3p, hsa-miR-302b-3p, hsa-miR-302c-3p, hsa-miR-302d-3p, hsa-miR-302e, hsa-miR-372, hsa-miR-373-3p, hsa-miR-520a-3p, hsa-miR-520b, hsa-miR-520c-3p, hsa-miR- 520d-3p, hsa-miR-520e |