Visible node/edge number:
Node colors:
Node labels:

show label of nodes with degree >=

Node sizes:
Edge colors:
Edge widths factor:
Layout:

gravity >=

edgeLength =

GRN in network:

Notice: IE browser need to manually refresh (F5) this page after resetting the network.

This regulatory network was inferred from the input dataset. The miRNAs and mRNAs are presented as round and rectangle nodes respectively. The numerical value popped up upon mouse over the gene node is the log2 transformed fold-change of the gene expression between the two groups. All of the nodes are clickable, and the detailed information of the miRNAs/mRNAs and related cancer pathway will be displayed in another window. The edges between nodes are supported by both interactions (predicted or experimentally verified) and correlations learnt from cancer dataset. The numerical value popped up upon mouse over the edge is the correlation beat value (effect size) between the two nodes. The experimental evidences of the edges reported in previous cancer studies are highlighted by red/orange color. All of these information can be accessed by the "mouse-over" action. This network shows a full map of the miRNA-mRNA regulation of the input gene list(s), and the hub miRNAs (with the high network degree/betweenness centrality) would be the potential cancer drivers or tumor suppressors. The full result table can be accessed in the "Regulations" tab.

"miRNACancerMAP" is also a network visualization tool for users to draw their regulatory network by personal customization. Users can set the complexity of the network by limiting the number of nodes or edges. And the color of the nodes can be defined by different categories of the mRNAs and miRNAs, such as Gene-Ontology, pathway, and expression status. Users can also select to use network degree or network betweenness centrality to define the node size. And edges can be black or colored by the correlation. Purple edge means negative correlation (mostly found between miRNA and mRNA), and blue edge means positive correlation (found in PPI or miRNA-miRNA sponge effect). We can also add the protein-protein interactions (PPI) into the network. This result will show the cluster of genes regulated by some specific miRNAs. Additionally, miRNA-miRNA edges can be added by the "miRNA sponge" button, presenting some clusters of miRNAs that have the interactions via sponge effect.

miRNA-gene regulations

(Download full result)

Num microRNA           Gene miRNA log2FC miRNA pvalue Gene log2FC Gene pvalue Interaction Correlation beta Correlation P-value PMID Reported in cancer studies
1 hsa-miR-29b-3p ABL1 0.27 0.74029 -0.21 0.734 miRNATAP -0.15 0 NA
2 hsa-miR-30e-5p ABL1 0.74 0.51197 -0.21 0.734 MirTarget; miRNATAP -0.12 0 NA
3 hsa-miR-140-3p ANAPC1 0.56 0.53812 0.3 0.50716 MirTarget -0.1 4.0E-5 NA
4 hsa-miR-100-5p ANAPC11 0.16 0.88382 0.03 0.95793 miRNAWalker2 validate -0.3 0 NA
5 hsa-miR-146b-5p ANAPC13 0.88 0.28494 -0.32 0.55225 miRanda -0.13 0 NA
6 hsa-miR-139-5p ANAPC7 -0.29 0.60387 0.04 0.93977 miRanda -0.12 0 NA
7 hsa-miR-181a-5p ATM 0.21 0.83306 -0.05 0.92324 miRNAWalker2 validate; miRTarBase; MirTarget -0.21 0 21102523; 23656790; 24531888; 27150990; 26113450 Ataxia telangiectasia mutated ATM a target gene of miR-181 exhibited reduced expression in mammospheres and upon TGF-β treatment;We report that miR-181a and miR-181b were overexpressed in more aggressive breast cancers and their expression correlates inversely with ATM levels;Ataxia-telangiectasia mutation ATM was predicted as a target gene of miR-181a with bioinformatics analysis and was verified by lucifersae reporter assay; A luciferase reporter assay demonstrated that ATM was a direct target of miR-181a miR-181a mimics transfection down regulated ATM mRNA and protein expression; There was inverse correlation between miR-181a and ATM protein expression in gastric cancer and normal gastric tissues; Our study demonstrates that over-expression of miR-181a might be involved in development of gastric cancer by promoting proliferation and inhibiting apoptosis probably through directly targeting ATM miR-181a modulation may be a potential strategy for the development of miRNA-based therapy of gastric cancer;MiR 181a Promotes Proliferation of Human Acute Myeloid Leukemia Cells by Targeting ATM; Dual luciferase reporter gene assay showed that miR-181a significantly suppressed the reporter gene activity containing ATM 3'-UTR by about 56.8% P < 0.05 but it didn't suppress the reporter gene activity containing 3'-UTR ATM mutation; Western blot showed that miR-181a significantly downregulated the expression of ATM in human leukemia cells; It is also found that miR-181a was significantly increased in AML which showed a negative correlation with ATM expression; miR-181a promotes cell proliferation in AML by regulating the tumor suppressor ATM thus it plays the role as oncogene in pathogenesis of AML;miR 181a promotes G1/S transition and cell proliferation in pediatric acute myeloid leukemia by targeting ATM; Pediatric AML patients and healthy controls were enrolled and the expression of miR-181a and ataxia telangiectasia mutated ATM in tissues were examined using quantitative PCR; Moreover cell proliferation and cell cycle were evaluated in several cell lines HL60 NB4 and K562 by using flow cytometry after transfected with miR-181a mimics and inhibitors or ATM siRNA and control siRNA; Finally ATM as the potential target protein of miR-181a was examined; We found that miR-181a was significantly increased in pediatric AML which showed an inverse association with ATM expression; Luciferase activity of the reporter construct identified ATM as the direct molecular target of miR-181a; The results revealed novel mechanism through which miR-181a regulates G1/S transition and cell proliferation in pediatric AML by regulating the tumor suppressor ATM providing insights into the molecular mechanism in pediatric AML
8 hsa-miR-181b-5p ATM 0.52 0.50566 -0.05 0.92324 MirTarget -0.22 0 21102523; 23656790 Ataxia telangiectasia mutated ATM a target gene of miR-181 exhibited reduced expression in mammospheres and upon TGF-β treatment;We report that miR-181a and miR-181b were overexpressed in more aggressive breast cancers and their expression correlates inversely with ATM levels
9 hsa-miR-181d-5p ATM 0.55 0.20485 -0.05 0.92324 MirTarget -0.12 0 NA
10 hsa-miR-18a-5p ATM 2.3 0 -0.05 0.92324 miRNAWalker2 validate; miRTarBase; MirTarget -0.12 0 23857602; 23437304; 25963391; 23229340 Furthermore we used antisense oligonucleotides against micro RNAs miRNA or miRNA overexpression plasmids to study the role of miR-18a and -106a on ATM expression; Furthermore we identified that ERα activates miR-18a and -106a to downregulate ATM expression; We reveal a novel mechanism involving ERα and miR-18a and -106a regulation of ATM in breast cancer;MicroRNA 18a attenuates DNA damage repair through suppressing the expression of ataxia telangiectasia mutated in colorectal cancer; Through in silico search the 3'UTR of Ataxia telangiectasia mutated ATM contains a conserved miR-18a binding site; Expression of ATM was down-regulated in CRC tumors p<0.0001 and inversely correlated with miR-18a expression r = -0.4562 p<0.01; This was further confirmed by the down-regulation of ATM protein by miR-18a; As ATM is a key enzyme in DNA damage repair we evaluated the effect of miR-18a on DNA double-strand breaks; miR-18a attenuates cellular repair of DNA double-strand breaks by directly suppressing ATM a key enzyme in DNA damage repair;However the upregulation of miR-18a suppressed the level of ataxia-telangiectasia mutated and attenuated DNA double-strand break repair after irradiation which re-sensitized the cervical cancer cells to radiotherapy by promoting apoptosis;MicroRNA 18a upregulates autophagy and ataxia telangiectasia mutated gene expression in HCT116 colon cancer cells; Previous studies showed that certain microRNAs including miR-18a potentially regulate ATM in cancer cells; However the mechanisms behind the modulation of ATM by miR-18a remain to be elucidated in colon cancer cells; In the present study we explored the impact of miR-18a on the autophagy process and ATM expression in HCT116 colon cancer cells; Western blotting and luciferase assays were implemented to explore the impact of miR-18a on ATM gene expression in HCT116 cells; Moreover miR-18a overexpression led to the upregulation of ATM expression and suppression of mTORC1 activity; Results of the present study pertaining to the role of miR-18a in regulating autophagy and ATM gene expression in colon cancer cells revealed a novel function for miR-18a in a critical cellular event and on a crucial gene with significant impacts in cancer development progression treatment and in other diseases
11 hsa-miR-21-5p ATM -0.14 0.92885 -0.05 0.92324 mirMAP -0.23 0 26289851 MiR-21 is an oncomiR that is overexpressed in nearly all cancers including ATC; Hence suppression of miR-21 could pave the way for ATC therapy
12 hsa-miR-140-3p ATR 0.56 0.53812 0.18 0.71274 PITA -0.14 0 NA
13 hsa-miR-361-5p ATR 0.3 0.67981 0.18 0.71274 miRanda -0.24 0 NA
14 hsa-miR-374a-5p ATR 0.47 0.30955 0.18 0.71274 mirMAP -0.11 0.00014 NA
15 hsa-miR-10b-3p BUB1 -0.78 0.05898 1.35 0.00016 MirTarget -0.88 0 NA
16 hsa-miR-139-5p BUB1 -0.29 0.60387 1.35 0.00016 miRanda -0.68 0 NA
17 hsa-miR-199a-5p BUB1 -0.59 0.53489 1.35 0.00016 miRanda -0.38 0 NA
18 hsa-miR-199b-5p BUB1 -0.49 0.51436 1.35 0.00016 miRanda -0.59 0 NA
19 hsa-miR-495-3p BUB1 0.04 0.88707 1.35 0.00016 MirTarget -0.42 0 NA
20 hsa-miR-653-5p BUB1 -0.29 0.40635 1.35 0.00016 MirTarget -0.19 5.0E-5 NA
21 hsa-miR-215-5p BUB1B 0.47 0.06524 0.95 0.00484 miRNAWalker2 validate -0.12 0.00478 NA
22 hsa-miR-22-3p BUB1B 0.05 0.97344 0.95 0.00484 miRNAWalker2 validate -0.41 0 NA
23 hsa-miR-486-5p BUB1B 0.22 0.72625 0.95 0.00484 miRanda -0.33 0 NA
24 hsa-miR-139-5p BUB3 -0.29 0.60387 -0.36 0.57227 miRanda -0.19 0 NA
25 hsa-let-7b-3p CCNA2 -0.11 0.8248 1.39 0.00016 MirTarget -0.24 0 NA
26 hsa-let-7b-5p CCNA2 -0.33 0.80106 1.39 0.00016 miRNAWalker2 validate; miRTarBase -0.52 0 NA
27 hsa-miR-199a-5p CCNA2 -0.59 0.53489 1.39 0.00016 miRanda -0.38 0 NA
28 hsa-miR-199b-5p CCNA2 -0.49 0.51436 1.39 0.00016 miRanda -0.5 0 NA
29 hsa-miR-218-5p CCNA2 0.01 0.99053 1.39 0.00016 MirTarget -0.49 0 NA
30 hsa-miR-22-3p CCNA2 0.05 0.97344 1.39 0.00016 MirTarget -0.25 0.00339 25596928 The sequence of miR-22 which is conserved in mice rats humans and other mammalians aligns with the sequence of 3'-UTR of CCNA2; Chenodeoxycholic acid treatment and miR-22 mimics reduced CCNA2 protein and increased the number of G0/G1 Huh7 and HCT116 cells; In humans the expression levels of miR-22 and CCNA2 are inversely correlated in liver and colon cancers
31 hsa-miR-29a-3p CCNA2 0.46 0.68374 1.39 0.00016 MirTarget -0.45 0 NA
32 hsa-miR-29c-3p CCNA2 -0.64 0.52285 1.39 0.00016 MirTarget -0.43 0 NA
33 hsa-miR-320a CCNA2 0.24 0.77369 1.39 0.00016 miRanda -0.21 0.0016 NA
34 hsa-miR-34c-5p CCNA2 -0.74 0.01835 1.39 0.00016 miRanda -0.29 0 NA
35 hsa-miR-486-5p CCNA2 0.22 0.72625 1.39 0.00016 miRanda -0.26 0 NA
36 hsa-let-7b-5p CCNB1 -0.33 0.80106 0.67 0.15126 miRNAWalker2 validate -0.39 0 NA
37 hsa-miR-139-5p CCNB1 -0.29 0.60387 0.67 0.15126 miRanda -0.49 0 NA
38 hsa-miR-140-5p CCNB1 0.78 0.10115 0.67 0.15126 miRanda -0.15 0.00114 NA
39 hsa-let-7a-5p CCNB2 -0.25 0.85781 1.39 0.00014 miRNAWalker2 validate -0.62 0 NA
40 hsa-let-7b-5p CCNB2 -0.33 0.80106 1.39 0.00014 miRNAWalker2 validate -0.54 0 NA
41 hsa-let-7c-5p CCNB2 0.07 0.94922 1.39 0.00014 miRNAWalker2 validate -0.66 0 NA
42 hsa-let-7i-5p CCND1 0.49 0.5398 -1.64 0.03536 miRNATAP -0.76 0 NA
43 hsa-miR-106a-5p CCND1 1.16 0.00089 -1.64 0.03536 MirTarget; miRNATAP -0.17 0 NA
44 hsa-miR-106b-5p CCND1 1.05 0.13909 -1.64 0.03536 miRNAWalker2 validate; miRTarBase; MirTarget; miRNATAP -0.26 0 NA
45 hsa-miR-142-3p CCND1 1.18 0.19612 -1.64 0.03536 miRanda -0.11 0.00024 23619912 Transfection of miR-142-3p mimics in colon cancer cells downregulated cyclin D1 expression induced G1 phase cell cycle arrest and elevated the sensitivity of the cells to 5-fluorouracil
46 hsa-miR-142-5p CCND1 1.23 0.01501 -1.64 0.03536 PITA -0.21 0 NA
47 hsa-miR-150-5p CCND1 1.17 0.17525 -1.64 0.03536 mirMAP -0.19 0 NA
48 hsa-miR-155-5p CCND1 1.13 0.13193 -1.64 0.03536 miRNAWalker2 validate -0.3 0 26955820 MicroRNA 155 expression inversely correlates with pathologic stage of gastric cancer and it inhibits gastric cancer cell growth by targeting cyclin D1
49 hsa-miR-15b-5p CCND1 0.69 0.30443 -1.64 0.03536 miRNAWalker2 validate; miRTarBase; MirTarget; miRNATAP -0.13 0.00922 NA
50 hsa-miR-17-5p CCND1 1.81 0.01774 -1.64 0.03536 miRNAWalker2 validate; MirTarget; TargetScan; miRNATAP -0.24 0 26431674 Bioinformatics Prediction and In Vitro Analysis Revealed That miR 17 Targets Cyclin D1 mRNA in Triple Negative Breast Cancer Cells; In this study using bioinformatic analyses miR-17 was selected as it targets the 3'UTR of CCND1 gene with the highest score; After lentiviral transduction of miR-17 to the target cells gene expression analysis showed decreased expression of CCND1 gene
51 hsa-miR-186-5p CCND1 1.04 0.14871 -1.64 0.03536 mirMAP -0.49 0 NA
52 hsa-miR-19a-3p CCND1 1.47 0.00036 -1.64 0.03536 miRNAWalker2 validate; miRTarBase; miRNATAP -0.22 0 25985117 Moreover miR-19a might play inhibitory roles in HCC malignancy via regulating Cyclin D1 expression
53 hsa-miR-19b-1-5p CCND1 1.37 0 -1.64 0.03536 miRNAWalker2 validate; miRTarBase -0.21 0 NA
54 hsa-miR-19b-3p CCND1 1.16 0.06285 -1.64 0.03536 miRNATAP -0.25 0 NA
55 hsa-miR-20a-5p CCND1 1.44 0.04652 -1.64 0.03536 miRNAWalker2 validate; miRTarBase; MirTarget; miRNATAP -0.22 0 NA
56 hsa-miR-23b-3p CCND1 0.33 0.69728 -1.64 0.03536 miRNATAP -0.21 0.00024 NA
57 hsa-miR-29a-3p CCND1 0.46 0.68374 -1.64 0.03536 mirMAP -0.36 0 NA
58 hsa-miR-340-5p CCND1 0.36 0.35065 -1.64 0.03536 mirMAP -0.16 0.00211 NA
59 hsa-miR-374a-5p CCND1 0.47 0.30955 -1.64 0.03536 MirTarget -0.56 0 27191497 microRNA 374a suppresses colon cancer progression by directly reducing CCND1 to inactivate the PI3K/AKT pathway; Furthermore luciferase reporter assays confirmed that miR-374a could directly reduce CCND1; We examined miR-374a levels by in situ hybridization and its correlation with CCND1 expression in CRC tumor tissues; High miR-374a expression with low level of CCND1 was protective factor in CRC; Together these findings indicate that miR-374a inactivates the PI3K/AKT axis by inhibiting CCND1 suppressing of colon cancer progression
60 hsa-miR-374b-5p CCND1 0.48 0.36577 -1.64 0.03536 miRNAWalker2 validate; MirTarget -0.53 0 NA
61 hsa-miR-424-5p CCND1 0.58 0.32687 -1.64 0.03536 miRNAWalker2 validate; miRTarBase; MirTarget; miRNATAP -0.17 1.0E-5 NA
62 hsa-miR-495-3p CCND1 0.04 0.88707 -1.64 0.03536 MirTarget -0.27 0 NA
63 hsa-miR-511-5p CCND1 1.26 7.0E-5 -1.64 0.03536 MirTarget -0.22 0 NA
64 hsa-miR-589-3p CCND1 0.87 0 -1.64 0.03536 MirTarget -0.12 0.00155 NA
65 hsa-miR-590-3p CCND1 1.01 0 -1.64 0.03536 mirMAP -0.22 0 NA
66 hsa-miR-9-5p CCND1 2.31 0.01107 -1.64 0.03536 miRNAWalker2 validate -0.16 0 NA
67 hsa-miR-92a-3p CCND1 1.23 0.30313 -1.64 0.03536 miRNAWalker2 validate -0.36 0 NA
68 hsa-miR-93-5p CCND1 0.94 0.3985 -1.64 0.03536 miRNAWalker2 validate; MirTarget; miRNATAP -0.16 0.00197 NA
69 hsa-miR-942-5p CCND1 1.66 0 -1.64 0.03536 MirTarget -0.26 0 NA
70 hsa-let-7a-3p CCND2 0.31 0.49951 -0.07 0.88928 mirMAP -0.22 0 20418948 MicroRNA let 7a inhibits proliferation of human prostate cancer cells in vitro and in vivo by targeting E2F2 and CCND2
71 hsa-let-7e-5p CCND2 -0.08 0.93271 -0.07 0.88928 miRNATAP -0.17 0.00031 NA
72 hsa-miR-106a-5p CCND2 1.16 0.00089 -0.07 0.88928 miRNATAP -0.12 1.0E-5 NA
73 hsa-miR-106b-5p CCND2 1.05 0.13909 -0.07 0.88928 miRNAWalker2 validate; miRTarBase; miRNATAP -0.32 0 NA
74 hsa-miR-130b-5p CCND2 1.39 0 -0.07 0.88928 mirMAP -0.11 1.0E-5 NA
75 hsa-miR-141-3p CCND2 0.52 0.54365 -0.07 0.88928 MirTarget; TargetScan -0.3 0 NA
76 hsa-miR-151a-3p CCND2 0.49 0.62227 -0.07 0.88928 mirMAP -0.18 0.00011 NA
77 hsa-miR-15a-5p CCND2 0.34 0.60504 -0.07 0.88928 miRNAWalker2 validate; miRTarBase; miRNATAP -0.38 0 NA
78 hsa-miR-15b-5p CCND2 0.69 0.30443 -0.07 0.88928 miRNATAP -0.2 0 NA
79 hsa-miR-16-5p CCND2 0.61 0.44106 -0.07 0.88928 miRNAWalker2 validate; miRNATAP -0.13 0.0015 NA
80 hsa-miR-17-5p CCND2 1.81 0.01774 -0.07 0.88928 miRNAWalker2 validate; miRTarBase; TargetScan; miRNATAP -0.22 0 NA
81 hsa-miR-182-5p CCND2 -0.35 0.79413 -0.07 0.88928 miRNAWalker2 validate; miRTarBase; miRNATAP -0.33 0 NA
82 hsa-miR-183-5p CCND2 0.2 0.86335 -0.07 0.88928 miRNATAP -0.3 0 NA
83 hsa-miR-185-5p CCND2 0.41 0.43546 -0.07 0.88928 MirTarget; miRNATAP -0.25 0 NA
84 hsa-miR-191-5p CCND2 -0.18 0.82875 -0.07 0.88928 MirTarget -0.19 0 NA
85 hsa-miR-19a-3p CCND2 1.47 0.00036 -0.07 0.88928 MirTarget; miRNATAP -0.19 0 NA
86 hsa-miR-19b-3p CCND2 1.16 0.06285 -0.07 0.88928 miRNAWalker2 validate; MirTarget; miRNATAP -0.17 0 NA
87 hsa-miR-200a-3p CCND2 0.3 0.67136 -0.07 0.88928 MirTarget -0.26 0 NA
88 hsa-miR-20a-5p CCND2 1.44 0.04652 -0.07 0.88928 miRNAWalker2 validate; miRTarBase; miRNATAP -0.24 0 NA
89 hsa-miR-20b-5p CCND2 0.53 0.21452 -0.07 0.88928 miRNATAP -0.1 0 NA
90 hsa-miR-21-3p CCND2 0.12 0.90444 -0.07 0.88928 mirMAP -0.13 2.0E-5 NA
91 hsa-miR-29b-3p CCND2 0.27 0.74029 -0.07 0.88928 MirTarget; miRNATAP -0.17 0 22330340; 24130168 In addition to CCND2 miR-29 also targets E2F7 another cell cycle regulator;We further demonstrated that miR-29 family acted as tumor suppressors through targeting CCND2 and matrix metalloproteinase-2 genes in GC
92 hsa-miR-301a-3p CCND2 0.97 0.00057 -0.07 0.88928 miRNAWalker2 validate -0.18 0 NA
93 hsa-miR-3065-3p CCND2 -0.22 0.65713 -0.07 0.88928 MirTarget; miRNATAP -0.22 0 NA
94 hsa-miR-30d-3p CCND2 0.27 0.27013 -0.07 0.88928 mirMAP -0.28 0 NA
95 hsa-miR-320b CCND2 0.71 0.0026 -0.07 0.88928 mirMAP; miRNATAP -0.11 0.00066 NA
96 hsa-miR-324-3p CCND2 0.75 0.04368 -0.07 0.88928 miRNAWalker2 validate -0.19 0 NA
97 hsa-miR-331-5p CCND2 0.4 0.0121 -0.07 0.88928 miRNATAP -0.22 0 NA
98 hsa-miR-342-3p CCND2 -0.72 0.32809 -0.07 0.88928 miRNAWalker2 validate; mirMAP -0.1 0.00019 NA
99 hsa-miR-429 CCND2 0.45 0.43718 -0.07 0.88928 miRNATAP -0.23 0 NA
100 hsa-miR-590-3p CCND2 1.01 0 -0.07 0.88928 miRanda; mirMAP -0.14 1.0E-5 NA
101 hsa-miR-590-5p CCND2 1.4 0 -0.07 0.88928 mirMAP -0.15 0 NA
102 hsa-miR-660-5p CCND2 0.79 0.10863 -0.07 0.88928 mirMAP -0.11 0.0034 NA
103 hsa-miR-7-1-3p CCND2 -0.14 0.72688 -0.07 0.88928 mirMAP -0.14 0 NA
104 hsa-miR-93-5p CCND2 0.94 0.3985 -0.07 0.88928 miRNATAP -0.4 0 NA
105 hsa-miR-96-5p CCND2 0.1 0.81662 -0.07 0.88928 TargetScan; miRNATAP -0.31 0 NA
106 hsa-miR-125b-5p CCNE1 0.09 0.91774 2.38 0 miRNAWalker2 validate -0.38 0 NA
107 hsa-miR-195-5p CCNE1 -0.13 0.8022 2.38 0 miRNAWalker2 validate; MirTarget; miRNATAP -0.62 0 24402230 Furthermore through qPCR and western blot assays we showed that overexpression of miR-195-5p reduced CCNE1 mRNA and protein levels respectively
108 hsa-miR-26a-5p CCNE1 -0.05 0.95817 2.38 0 miRNAWalker2 validate; miRTarBase; miRNATAP -0.62 0 22094936 Cell cycle regulation and CCNE1 and CDC2 were the only significant overlapping pathway and genes differentially expressed between tumors with high and low levels of miR-26a and EZH2 respectively; Low mRNA levels of EZH2 CCNE1 and CDC2 and high levels of miR-26a are associated with favorable outcome on tamoxifen
109 hsa-miR-26b-5p CCNE1 -0.02 0.9773 2.38 0 miRNAWalker2 validate; miRTarBase; miRNATAP -0.32 0 NA
110 hsa-miR-342-5p CCNE1 -1 0.00867 2.38 0 miRNAWalker2 validate -0.14 0.00142 NA
111 hsa-miR-497-5p CCNE1 0.02 0.96778 2.38 0 MirTarget; miRNATAP -0.38 0 24112607; 25909221; 24909281 Western blot assays confirmed that overexpression of miR-497 reduced cyclin E1 protein levels; Inhibited cellular growth suppressed cellular migration and invasion and G1 cell cycle arrest were observed upon overexpression of miR-497 in cells possibly by targeting cyclin E1;The effect of simultaneous overexpression of miR-497 and miR-34a on the inhibition of cell proliferation colony formation and tumor growth and the downregulation of cyclin E1 was stronger than the effect of each miRNA alone; The synergistic actions of miR-497 and miR-34a partly correlated with cyclin E1 levels; These results indicate cyclin E1 is downregulated by both miR-497 and miR-34a which synergistically retard the growth of human lung cancer cells;miR 497 suppresses proliferation of human cervical carcinoma HeLa cells by targeting cyclin E1; Furthermore the target effect of miR-497 on the CCNE1 was identified by dual-luciferase reporter assay system qRT-PCR and Western blotting; Over-expressed miR-497 in HeLa cells could suppress cell proliferation by targeting CCNE1
112 hsa-miR-26a-5p CCNE2 -0.05 0.95817 0.56 0.05682 miRNAWalker2 validate; miRTarBase; miRNATAP -0.63 0 24116110; 21901171 The loss of miR 26a mediated post transcriptional regulation of cyclin E2 in pancreatic cancer cell proliferation and decreased patient survival; The in vitro and in vivo assays showed that overexpression of miR-26a resulted in cell cycle arrest inhibited cell proliferation and decreased tumor growth which was associated with cyclin E2 downregulation;We also show that enforced expression of miR-26a in AML cells is able to inhibit cell cycle progression by downregulating cyclin E2 expression
113 hsa-miR-28-3p CCNE2 0.57 0.60357 0.56 0.05682 PITA; miRNATAP -0.33 4.0E-5 NA
114 hsa-miR-30a-5p CCNE2 -0.49 0.73014 0.56 0.05682 miRNATAP -0.39 0 NA
115 hsa-miR-30c-5p CCNE2 0.29 0.7388 0.56 0.05682 miRNATAP -0.25 2.0E-5 NA
116 hsa-miR-335-3p CCNE2 0.21 0.73517 0.56 0.05682 mirMAP -0.17 0 NA
117 hsa-miR-34c-5p CCNE2 -0.74 0.01835 0.56 0.05682 miRNAWalker2 validate; miRTarBase; PITA; miRanda; miRNATAP -0.25 0 NA
118 hsa-miR-1301-3p CCNH 1.13 0.00036 -0.62 0.15896 miRNAWalker2 validate -0.1 0 NA
119 hsa-miR-142-5p CCNH 1.23 0.01501 -0.62 0.15896 PITA -0.11 0 NA
120 hsa-miR-590-3p CCNH 1.01 0 -0.62 0.15896 miRanda -0.11 0 NA
121 hsa-miR-141-3p CDC14A 0.52 0.54365 -0.47 0.15652 TargetScan; miRNATAP -0.14 1.0E-5 NA
122 hsa-miR-148a-3p CDC14A 0.72 0.59727 -0.47 0.15652 miRNATAP -0.2 1.0E-5 NA
123 hsa-miR-148a-5p CDC14A 0.75 0.11901 -0.47 0.15652 mirMAP -0.19 1.0E-5 NA
124 hsa-miR-200a-3p CDC14A 0.3 0.67136 -0.47 0.15652 miRNATAP -0.1 0.00072 NA
125 hsa-miR-429 CDC14A 0.45 0.43718 -0.47 0.15652 miRanda -0.15 0 NA
126 hsa-miR-493-5p CDC14A -0.31 0.25802 -0.47 0.15652 miRNATAP -0.11 0.00331 NA
127 hsa-miR-590-3p CDC14A 1.01 0 -0.47 0.15652 PITA; miRanda; mirMAP; miRNATAP -0.15 0.00059 NA
128 hsa-let-7a-3p CDC14B 0.31 0.49951 0.46 0.21275 miRNATAP -0.19 4.0E-5 NA
129 hsa-miR-141-3p CDC14B 0.52 0.54365 0.46 0.21275 mirMAP -0.35 0 NA
130 hsa-miR-15a-5p CDC14B 0.34 0.60504 0.46 0.21275 miRNAWalker2 validate; MirTarget -0.48 0 NA
131 hsa-miR-15b-5p CDC14B 0.69 0.30443 0.46 0.21275 MirTarget -0.2 0 NA
132 hsa-miR-16-5p CDC14B 0.61 0.44106 0.46 0.21275 miRNAWalker2 validate; MirTarget -0.16 0.0003 NA
133 hsa-miR-17-3p CDC14B 1.47 0.04186 0.46 0.21275 MirTarget -0.18 0 NA
134 hsa-miR-183-5p CDC14B 0.2 0.86335 0.46 0.21275 MirTarget -0.37 0 NA
135 hsa-miR-185-5p CDC14B 0.41 0.43546 0.46 0.21275 MirTarget -0.39 0 NA
136 hsa-miR-200a-3p CDC14B 0.3 0.67136 0.46 0.21275 mirMAP -0.29 0 NA
137 hsa-miR-21-3p CDC14B 0.12 0.90444 0.46 0.21275 mirMAP -0.35 0 NA
138 hsa-miR-30d-3p CDC14B 0.27 0.27013 0.46 0.21275 MirTarget -0.18 2.0E-5 NA
139 hsa-miR-429 CDC14B 0.45 0.43718 0.46 0.21275 PITA; miRanda; miRNATAP -0.31 0 NA
140 hsa-miR-590-3p CDC14B 1.01 0 0.46 0.21275 miRanda; mirMAP -0.21 0 NA
141 hsa-miR-590-5p CDC14B 1.4 0 0.46 0.21275 miRanda -0.13 8.0E-5 NA
142 hsa-miR-215-5p CDC20 0.47 0.06524 1.86 1.0E-5 miRNAWalker2 validate -0.15 0.00257 NA
143 hsa-miR-30a-5p CDC20 -0.49 0.73014 1.86 1.0E-5 miRNAWalker2 validate -0.42 0 NA
144 hsa-let-7a-5p CDC25A -0.25 0.85781 1.56 0 MirTarget; TargetScan; miRNATAP -0.56 0 NA
145 hsa-let-7b-5p CDC25A -0.33 0.80106 1.56 0 miRNAWalker2 validate; miRTarBase; MirTarget; miRNATAP -0.49 0 NA
146 hsa-let-7c-5p CDC25A 0.07 0.94922 1.56 0 MirTarget -0.48 0 25909324 MicroRNA let 7c Inhibits Cell Proliferation and Induces Cell Cycle Arrest by Targeting CDC25A in Human Hepatocellular Carcinoma; The aim of the present study was to determine whether the cell cycle regulator CDC25A is involved in the antitumor effect of let-7c in HCC; The luciferase reporter assay showed that CDC25A was a direct target of let-7c and that let-7c inhibited the expression of CDC25A protein by directly targeting its 3' UTR; In conclusion this study indicates that let-7c suppresses HCC progression possibly by directly targeting the cell cycle regulator CDC25A and indirectly affecting its downstream target molecules
147 hsa-miR-195-5p CDC25A -0.13 0.8022 1.56 0 MirTarget; miRNATAP -0.53 0 NA
148 hsa-miR-337-3p CDC25A -0.5 0.30557 1.56 0 mirMAP -0.35 0 NA
149 hsa-miR-34c-5p CDC25A -0.74 0.01835 1.56 0 miRNATAP -0.31 0 21321636 Ectopic expression of miR-449b and miR-34c resulted in lowered adhesion activities by 28%-34% and in cell cycle arrests with increased cell number of 15.62% and 15.71% in G1 and with decreased cell number of 15.96% and 16.56% in S Cell cycle related proteins CDK6 and CDC25A were down-regulated; The decreases of CDK6 and CDC25A by miR-449b were 39% and 22% respecyively; 49% and 32% by miR-34c respectively
150 hsa-miR-497-5p CDC25A 0.02 0.96778 1.56 0 MirTarget; miRNATAP -0.28 0 NA
NumGOOverlapSizeP ValueAdj. P Value
1 CELL CYCLE 76 1316 1.197e-76 5.569e-73
2 CELL CYCLE PROCESS 72 1081 1.853e-75 4.311e-72
3 MITOTIC CELL CYCLE 65 766 7.593e-73 1.178e-69
4 REGULATION OF CELL CYCLE 66 949 1.98e-68 2.303e-65
5 REGULATION OF MITOTIC CELL CYCLE 46 468 2.081e-51 1.937e-48
6 CELL CYCLE PHASE TRANSITION 37 255 4.533e-47 3.515e-44
7 REGULATION OF CELL CYCLE PHASE TRANSITION 39 321 1.282e-46 8.523e-44
8 REGULATION OF CELL CYCLE PROCESS 45 558 3.236e-46 1.673e-43
9 NEGATIVE REGULATION OF CELL CYCLE 42 433 3.006e-46 1.673e-43
10 CELL CYCLE CHECKPOINT 32 194 2.545e-42 1.184e-39
11 NEGATIVE REGULATION OF CELL CYCLE PROCESS 31 214 4.321e-39 1.828e-36
12 CELL DIVISION 38 460 8.424e-39 3.267e-36
13 NEGATIVE REGULATION OF MITOTIC CELL CYCLE 30 199 2.427e-38 8.688e-36
14 MITOTIC CELL CYCLE CHECKPOINT 26 139 9.548e-36 3.173e-33
15 REGULATION OF TRANSFERASE ACTIVITY 44 946 1.273e-34 3.484e-32
16 CELL CYCLE G1 S PHASE TRANSITION 24 111 1.241e-34 3.484e-32
17 G1 S TRANSITION OF MITOTIC CELL CYCLE 24 111 1.241e-34 3.484e-32
18 REGULATION OF PROTEIN MODIFICATION PROCESS 53 1710 5.71e-34 1.476e-31
19 NEGATIVE REGULATION OF CELL CYCLE PHASE TRANSITION 24 146 1.582e-31 3.874e-29
20 POSITIVE REGULATION OF CELL CYCLE 30 332 1.995e-31 4.641e-29
21 MITOTIC NUCLEAR DIVISION 30 361 2.529e-30 5.603e-28
22 POSITIVE REGULATION OF CELL CYCLE PROCESS 26 247 6.602e-29 1.396e-26
23 DNA INTEGRITY CHECKPOINT 22 146 5.082e-28 1.028e-25
24 ORGANELLE FISSION 31 496 1.509e-27 2.926e-25
25 NEGATIVE REGULATION OF PROTEIN METABOLIC PROCESS 40 1087 3.259e-27 6.066e-25
26 NEGATIVE REGULATION OF PROTEIN MODIFICATION PROCESS 32 616 5.831e-26 1.044e-23
27 REGULATION OF ORGANELLE ORGANIZATION 39 1178 9.039e-25 1.558e-22
28 CELLULAR RESPONSE TO DNA DAMAGE STIMULUS 32 720 7.195e-24 1.196e-21
29 REGULATION OF CELL DIVISION 23 272 2.432e-23 3.901e-21
30 POSITIVE REGULATION OF PROTEIN METABOLIC PROCESS 41 1492 4.207e-23 6.525e-21
31 REGULATION OF CELL CYCLE G1 S PHASE TRANSITION 19 147 6.364e-23 9.553e-21
32 NEGATIVE REGULATION OF CELL CYCLE G1 S PHASE TRANSITION 17 98 7.18e-23 1.044e-20
33 REGULATION OF PHOSPHORUS METABOLIC PROCESS 42 1618 8.677e-23 1.223e-20
34 NEGATIVE REGULATION OF TRANSFERASE ACTIVITY 24 351 3.806e-22 5.209e-20
35 REGULATION OF CELL CYCLE ARREST 17 108 4.181e-22 5.558e-20
36 POSITIVE REGULATION OF PROTEIN MODIFICATION PROCESS 36 1135 4.59e-22 5.933e-20
37 REGULATION OF NUCLEAR DIVISION 19 163 4.863e-22 6.116e-20
38 CHROMOSOME ORGANIZATION 34 1009 1.406e-21 1.722e-19
39 G1 DNA DAMAGE CHECKPOINT 15 73 1.819e-21 2.171e-19
40 SIGNAL TRANSDUCTION IN RESPONSE TO DNA DAMAGE 16 96 2.849e-21 3.314e-19
41 MITOTIC DNA INTEGRITY CHECKPOINT 16 100 5.711e-21 6.481e-19
42 REGULATION OF KINASE ACTIVITY 30 776 1.368e-20 1.516e-18
43 POSITIVE REGULATION OF CELL CYCLE ARREST 15 85 2.158e-20 2.335e-18
44 CELL CYCLE G2 M PHASE TRANSITION 17 138 3.291e-20 3.427e-18
45 REGULATION OF SISTER CHROMATID SEGREGATION 14 67 3.315e-20 3.427e-18
46 POSITIVE REGULATION OF TRANSFERASE ACTIVITY 27 616 7.132e-20 7.214e-18
47 REGULATION OF PROTEIN CATABOLIC PROCESS 23 393 1.056e-19 1.045e-17
48 REGULATION OF CYCLIN DEPENDENT PROTEIN KINASE ACTIVITY 15 97 1.781e-19 1.726e-17
49 CELL CYCLE ARREST 17 154 2.247e-19 2.134e-17
50 POSITIVE REGULATION OF PROTEOLYSIS 22 363 3.5e-19 3.257e-17
51 REGULATION OF LIGASE ACTIVITY 16 130 4.648e-19 4.081e-17
52 REGULATION OF CELLULAR PROTEIN CATABOLIC PROCESS 20 274 4.602e-19 4.081e-17
53 REGULATION OF CELL PROLIFERATION 37 1496 4.565e-19 4.081e-17
54 REGULATION OF CHROMOSOME ORGANIZATION 20 278 6.132e-19 5.284e-17
55 NEGATIVE REGULATION OF CATALYTIC ACTIVITY 29 829 1.056e-18 8.935e-17
56 DNA METABOLIC PROCESS 28 758 1.167e-18 9.695e-17
57 REGULATION OF CHROMOSOME SEGREGATION 14 85 1.208e-18 9.861e-17
58 CELLULAR RESPONSE TO STRESS 37 1565 2.046e-18 1.641e-16
59 REGULATION OF PROTEOLYSIS 27 711 2.765e-18 2.181e-16
60 REGULATION OF PROTEIN SERINE THREONINE KINASE ACTIVITY 23 470 5.627e-18 4.364e-16
61 POSITIVE REGULATION OF MITOTIC CELL CYCLE 15 123 7.369e-18 5.621e-16
62 POSITIVE REGULATION OF CELL DEATH 25 605 8.766e-18 6.579e-16
63 NEGATIVE REGULATION OF MOLECULAR FUNCTION 31 1079 1.35e-17 9.97e-16
64 ANAPHASE PROMOTING COMPLEX DEPENDENT CATABOLIC PROCESS 13 77 1.572e-17 1.143e-15
65 CHROMOSOME SEGREGATION 18 272 1.842e-16 1.315e-14
66 NUCLEAR CHROMOSOME SEGREGATION 17 228 1.866e-16 1.315e-14
67 POSITIVE REGULATION OF CELLULAR PROTEIN CATABOLIC PROCESS 16 192 2.646e-16 1.838e-14
68 PROTEIN PHOSPHORYLATION 28 944 3.4e-16 2.327e-14
69 POSITIVE REGULATION OF CATALYTIC ACTIVITY 34 1518 4.3e-16 2.858e-14
70 SIGNAL TRANSDUCTION BY P53 CLASS MEDIATOR 14 127 4.248e-16 2.858e-14
71 REGULATION OF CATABOLIC PROCESS 25 731 7.265e-16 4.761e-14
72 REGULATION OF PROTEIN UBIQUITINATION INVOLVED IN UBIQUITIN DEPENDENT PROTEIN CATABOLIC PROCESS 13 103 8.35e-16 5.396e-14
73 POSITIVE REGULATION OF CELL PROLIFERATION 26 814 8.644e-16 5.51e-14
74 PROTEIN UBIQUITINATION INVOLVED IN UBIQUITIN DEPENDENT PROTEIN CATABOLIC PROCESS 14 134 9.144e-16 5.75e-14
75 POSITIVE REGULATION OF CELLULAR PROTEIN LOCALIZATION 19 360 1.589e-15 9.729e-14
76 POSITIVE REGULATION OF BIOSYNTHETIC PROCESS 36 1805 1.584e-15 9.729e-14
77 SISTER CHROMATID SEGREGATION 15 176 1.741e-15 1.052e-13
78 POSITIVE REGULATION OF PROTEIN CATABOLIC PROCESS 17 263 2.037e-15 1.2e-13
79 POSITIVE REGULATION OF LIGASE ACTIVITY 13 110 2.018e-15 1.2e-13
80 RESPONSE TO ABIOTIC STIMULUS 28 1024 2.675e-15 1.556e-13
81 NEGATIVE REGULATION OF CHROMOSOME SEGREGATION 9 28 2.911e-15 1.672e-13
82 POSITIVE REGULATION OF GENE EXPRESSION 35 1733 3.18e-15 1.804e-13
83 REGULATION OF PROTEIN MODIFICATION BY SMALL PROTEIN CONJUGATION OR REMOVAL 17 280 5.759e-15 3.228e-13
84 POSITIVE REGULATION OF MOLECULAR FUNCTION 35 1791 8.621e-15 4.763e-13
85 POSITIVE REGULATION OF PROTEIN MODIFICATION BY SMALL PROTEIN CONJUGATION OR REMOVAL 15 196 8.7e-15 4.763e-13
86 POSITIVE REGULATION OF CELL CYCLE PHASE TRANSITION 11 68 8.92e-15 4.826e-13
87 REGULATION OF CELL DEATH 32 1472 1e-14 5.349e-13
88 NEGATIVE REGULATION OF PHOSPHORUS METABOLIC PROCESS 21 541 1.912e-14 9.995e-13
89 NEGATIVE REGULATION OF PHOSPHATE METABOLIC PROCESS 21 541 1.912e-14 9.995e-13
90 DNA REPLICATION 15 208 2.101e-14 1.086e-12
91 NEGATIVE REGULATION OF MITOTIC NUCLEAR DIVISION 9 34 2.163e-14 1.106e-12
92 REGULATION OF CELLULAR PROTEIN LOCALIZATION 21 552 2.835e-14 1.434e-12
93 POSITIVE REGULATION OF FIBROBLAST PROLIFERATION 10 53 3.017e-14 1.509e-12
94 RESPONSE TO OXYGEN LEVELS 17 311 3.244e-14 1.606e-12
95 POSITIVE REGULATION OF MITOCHONDRIAL OUTER MEMBRANE PERMEABILIZATION INVOLVED IN APOPTOTIC SIGNALING PATHWAY 9 36 3.854e-14 1.888e-12
96 NEGATIVE REGULATION OF PROTEIN MODIFICATION BY SMALL PROTEIN CONJUGATION OR REMOVAL 13 139 4.469e-14 2.166e-12
97 POSITIVE REGULATION OF ORGANELLE ORGANIZATION 21 573 5.874e-14 2.818e-12
98 PROTEASOMAL PROTEIN CATABOLIC PROCESS 16 271 6.042e-14 2.869e-12
99 SISTER CHROMATID COHESION 12 111 7.716e-14 3.627e-12
100 REGULATION OF PROTEASOMAL UBIQUITIN DEPENDENT PROTEIN CATABOLIC PROCESS 13 148 1.015e-13 4.721e-12
101 NEGATIVE REGULATION OF CELL DIVISION 10 60 1.137e-13 5.24e-12
102 REGULATION OF MITOCHONDRIAL OUTER MEMBRANE PERMEABILIZATION INVOLVED IN APOPTOTIC SIGNALING PATHWAY 9 43 2.251e-13 1.027e-11
103 PHOSPHORYLATION 28 1228 2.447e-13 1.105e-11
104 DNA REPAIR 19 480 2.829e-13 1.266e-11
105 PROTEIN MODIFICATION BY SMALL PROTEIN CONJUGATION OR REMOVAL 24 873 3.637e-13 1.612e-11
106 NEGATIVE REGULATION OF PHOSPHORYLATION 18 422 3.684e-13 1.617e-11
107 POSITIVE REGULATION OF CELLULAR COMPONENT ORGANIZATION 27 1152 3.857e-13 1.677e-11
108 REGULATION OF PROTEIN INSERTION INTO MITOCHONDRIAL MEMBRANE INVOLVED IN APOPTOTIC SIGNALING PATHWAY 8 29 4.432e-13 1.858e-11
109 POSITIVE REGULATION OF PROTEIN INSERTION INTO MITOCHONDRIAL MEMBRANE INVOLVED IN APOPTOTIC SIGNALING PATHWAY 8 29 4.432e-13 1.858e-11
110 DNA REPLICATION INITIATION 8 29 4.432e-13 1.858e-11
111 NEGATIVE REGULATION OF NUCLEAR DIVISION 9 46 4.351e-13 1.858e-11
112 REGULATION OF PROTEASOMAL PROTEIN CATABOLIC PROCESS 13 181 1.367e-12 5.68e-11
113 POSITIVE REGULATION OF CATABOLIC PROCESS 17 395 1.556e-12 6.409e-11
114 REGULATION OF DNA METABOLIC PROCESS 16 340 1.966e-12 8.023e-11
115 INTRACELLULAR SIGNAL TRANSDUCTION 30 1572 2.605e-12 1.054e-10
116 REGULATION OF FIBROBLAST PROLIFERATION 10 81 2.627e-12 1.054e-10
117 NEGATIVE REGULATION OF CELL PROLIFERATION 20 643 5.174e-12 2.058e-10
118 CELL DEATH 24 1001 6.665e-12 2.628e-10
119 POSITIVE REGULATION OF INTRACELLULAR TRANSPORT 16 370 7.055e-12 2.759e-10
120 PEPTIDYL AMINO ACID MODIFICATION 22 841 1.122e-11 4.35e-10
121 REGULATION OF TRANSCRIPTION FROM RNA POLYMERASE II PROMOTER 31 1784 1.132e-11 4.353e-10
122 SPINDLE CHECKPOINT 7 25 1.224e-11 4.667e-10
123 CELLULAR RESPONSE TO UV 9 66 1.36e-11 5.144e-10
124 POSITIVE REGULATION OF RESPONSE TO STIMULUS 32 1929 1.576e-11 5.912e-10
125 REGULATION OF MEMBRANE PERMEABILITY 9 70 2.355e-11 8.767e-10
126 CELLULAR RESPONSE TO RADIATION 11 137 2.404e-11 8.879e-10
127 RESPONSE TO LIPID 22 888 3.221e-11 1.18e-09
128 POSITIVE REGULATION OF CELL COMMUNICATION 28 1532 4.904e-11 1.783e-09
129 POSITIVE REGULATION OF TRANSCRIPTION FROM RNA POLYMERASE II PROMOTER 23 1004 5.025e-11 1.813e-09
130 POSITIVE REGULATION OF STEM CELL DIFFERENTIATION 8 50 5.139e-11 1.84e-09
131 NEGATIVE REGULATION OF PROTEIN CATABOLIC PROCESS 10 109 5.39e-11 1.915e-09
132 RESPONSE TO DRUG 16 431 6.87e-11 2.422e-09
133 PROTEIN CATABOLIC PROCESS 18 579 7.055e-11 2.468e-09
134 NEGATIVE REGULATION OF KINASE ACTIVITY 13 250 8.062e-11 2.799e-09
135 POSITIVE REGULATION OF PHOSPHATE METABOLIC PROCESS 23 1036 9.376e-11 3.208e-09
136 POSITIVE REGULATION OF PHOSPHORUS METABOLIC PROCESS 23 1036 9.376e-11 3.208e-09
137 POSITIVE REGULATION OF ESTABLISHMENT OF PROTEIN LOCALIZATION 17 514 9.837e-11 3.341e-09
138 REGULATION OF PROTEIN LOCALIZATION 22 950 1.176e-10 3.966e-09
139 POSITIVE REGULATION OF EPITHELIAL TO MESENCHYMAL TRANSITION 7 34 1.326e-10 4.407e-09
140 NEGATIVE REGULATION OF CELLULAR COMPONENT ORGANIZATION 19 684 1.317e-10 4.407e-09
141 REGULATION OF DNA REPLICATION 11 161 1.387e-10 4.576e-09
142 PHOSPHATE CONTAINING COMPOUND METABOLIC PROCESS 31 1977 1.55e-10 5.079e-09
143 NEGATIVE REGULATION OF NITROGEN COMPOUND METABOLIC PROCESS 27 1517 2.198e-10 7.101e-09
144 REGULATION OF INTRACELLULAR TRANSPORT 18 621 2.193e-10 7.101e-09
145 RESPONSE TO UV 10 126 2.291e-10 7.302e-09
146 NEGATIVE REGULATION OF PROTEIN SERINE THREONINE KINASE ACTIVITY 10 126 2.291e-10 7.302e-09
147 CELLULAR RESPONSE TO LIGHT STIMULUS 9 91 2.631e-10 8.272e-09
148 MITOTIC SISTER CHROMATID SEGREGATION 9 91 2.631e-10 8.272e-09
149 POSITIVE REGULATION OF APOPTOTIC SIGNALING PATHWAY 11 171 2.65e-10 8.277e-09
150 PROTEIN UBIQUITINATION 18 629 2.694e-10 8.358e-09
151 MITOCHONDRIAL MEMBRANE ORGANIZATION 9 92 2.906e-10 8.954e-09
152 CELLULAR RESPONSE TO ENDOGENOUS STIMULUS 22 1008 3.62e-10 1.108e-08
153 RESPONSE TO RADIATION 15 413 3.9e-10 1.186e-08
154 NEGATIVE REGULATION OF CELLULAR PROTEIN CATABOLIC PROCESS 8 64 4.028e-10 1.217e-08
155 RESPONSE TO ORGANIC CYCLIC COMPOUND 21 917 4.146e-10 1.245e-08
156 NEGATIVE REGULATION OF CHROMOSOME ORGANIZATION 9 96 4.275e-10 1.275e-08
157 RESPONSE TO ENDOGENOUS STIMULUS 26 1450 4.476e-10 1.326e-08
158 RESPONSE TO STEROID HORMONE 16 497 5.536e-10 1.63e-08
159 DNA DEPENDENT DNA REPLICATION 9 99 5.645e-10 1.652e-08
160 MEIOTIC CELL CYCLE 11 186 6.506e-10 1.892e-08
161 CELL PROLIFERATION 18 672 7.759e-10 2.242e-08
162 POSITIVE REGULATION OF CHROMOSOME SEGREGATION 6 25 1.083e-09 3.11e-08
163 DIGESTIVE SYSTEM DEVELOPMENT 10 148 1.119e-09 3.193e-08
164 REPLICATIVE SENESCENCE 5 12 1.203e-09 3.406e-08
165 REGULATION OF CELLULAR RESPONSE TO STRESS 18 691 1.208e-09 3.406e-08
166 POSITIVE REGULATION OF CHROMOSOME ORGANIZATION 10 150 1.276e-09 3.575e-08
167 NEGATIVE REGULATION OF ORGANELLE ORGANIZATION 14 387 1.672e-09 4.659e-08
168 PROTEIN K11 LINKED UBIQUITINATION 6 27 1.797e-09 4.977e-08
169 CELLULAR RESPONSE TO ORGANIC CYCLIC COMPOUND 15 465 1.98e-09 5.45e-08
170 CELLULAR RESPONSE TO ABIOTIC STIMULUS 12 263 2.013e-09 5.51e-08
171 NEGATIVE REGULATION OF DNA REPLICATION 7 55 4.638e-09 1.262e-07
172 REGULATION OF ESTABLISHMENT OF PROTEIN LOCALIZATION TO MITOCHONDRION 9 128 5.599e-09 1.515e-07
173 G2 DNA DAMAGE CHECKPOINT 6 33 6.581e-09 1.77e-07
174 POSITIVE REGULATION OF CELL DIVISION 9 132 7.345e-09 1.964e-07
175 REGULATION OF CELL CYCLE G2 M PHASE TRANSITION 7 59 7.686e-09 2.044e-07
176 REGULATION OF MICROTUBULE BASED PROCESS 11 243 1.078e-08 2.841e-07
177 RESPONSE TO HORMONE 19 893 1.081e-08 2.841e-07
178 POSITIVE REGULATION OF CYCLIN DEPENDENT PROTEIN KINASE ACTIVITY 6 36 1.145e-08 2.993e-07
179 POSITIVE REGULATION OF TRANSMEMBRANE RECEPTOR PROTEIN SERINE THREONINE KINASE SIGNALING PATHWAY 8 100 1.487e-08 3.866e-07
180 REGULATION OF EPITHELIAL TO MESENCHYMAL TRANSITION 7 67 1.904e-08 4.921e-07
181 MACROMOLECULE CATABOLIC PROCESS 19 926 1.935e-08 4.975e-07
182 RESPONSE TO GROWTH FACTOR 14 475 2.252e-08 5.758e-07
183 NEGATIVE REGULATION OF CATABOLIC PROCESS 10 203 2.355e-08 5.988e-07
184 NEGATIVE REGULATION OF PROTEOLYSIS 12 329 2.457e-08 6.213e-07
185 MEIOTIC CELL CYCLE PROCESS 9 152 2.525e-08 6.351e-07
186 POSITIVE REGULATION OF KINASE ACTIVITY 14 482 2.704e-08 6.764e-07
187 REGULATION OF CELLULAR LOCALIZATION 22 1277 2.798e-08 6.961e-07
188 REGULATION OF TRANSMEMBRANE RECEPTOR PROTEIN SERINE THREONINE KINASE SIGNALING PATHWAY 10 207 2.834e-08 7.014e-07
189 NEGATIVE REGULATION OF DNA METABOLIC PROCESS 8 111 3.392e-08 8.35e-07
190 REGULATION OF STEM CELL DIFFERENTIATION 8 113 3.903e-08 9.558e-07
191 POSITIVE REGULATION OF CELL MORPHOGENESIS INVOLVED IN DIFFERENTIATION 9 162 4.387e-08 1.069e-06
192 PROTEIN SUMOYLATION 8 115 4.479e-08 1.081e-06
193 REGULATION OF CYTOSKELETON ORGANIZATION 14 502 4.483e-08 1.081e-06
194 REGULATION OF MITOCHONDRION ORGANIZATION 10 218 4.623e-08 1.098e-06
195 RESPONSE TO ESTROGEN 10 218 4.623e-08 1.098e-06
196 REGULATION OF PROTEIN LOCALIZATION TO NUCLEUS 10 218 4.623e-08 1.098e-06
197 REGULATION OF BINDING 11 283 5.154e-08 1.217e-06
198 PROTEOLYSIS 21 1208 5.328e-08 1.252e-06
199 POSITIVE REGULATION OF MITOCHONDRION ORGANIZATION 9 167 5.704e-08 1.334e-06
200 APOPTOTIC SIGNALING PATHWAY 11 289 6.379e-08 1.484e-06
201 CELLULAR RESPONSE TO ORGANIC SUBSTANCE 26 1848 7.009e-08 1.615e-06
202 RESPONSE TO ALCOHOL 12 362 7.002e-08 1.615e-06
203 ORGAN REGENERATION 7 83 8.581e-08 1.967e-06
204 MITOCHONDRIAL TRANSPORT 9 177 9.403e-08 2.145e-06
205 NEGATIVE REGULATION OF GENE EXPRESSION 23 1493 9.81e-08 2.227e-06
206 REGULATION OF MICROTUBULE POLYMERIZATION OR DEPOLYMERIZATION 9 178 9.868e-08 2.229e-06
207 POSITIVE REGULATION OF PROTEIN LOCALIZATION TO NUCLEUS 8 129 1.097e-07 2.467e-06
208 REGULATION OF TRANSCRIPTION INVOLVED IN G1 S TRANSITION OF MITOTIC CELL CYCLE 5 27 1.164e-07 2.604e-06
209 PROTEIN POLYUBIQUITINATION 10 243 1.279e-07 2.847e-06
210 POSITIVE REGULATION OF DNA METABOLIC PROCESS 9 185 1.372e-07 3.041e-06
211 SMAD PROTEIN SIGNAL TRANSDUCTION 6 56 1.778e-07 3.902e-06
212 REPRODUCTION 21 1297 1.77e-07 3.902e-06
213 GLAND DEVELOPMENT 12 395 1.8e-07 3.931e-06
214 MITOTIC CELL CYCLE ARREST 4 13 2.54e-07 5.524e-06
215 REGULATION OF RESPONSE TO DNA DAMAGE STIMULUS 8 145 2.706e-07 5.856e-06
216 REGULATION OF TRANSFORMING GROWTH FACTOR BETA RECEPTOR SIGNALING PATHWAY 7 99 2.905e-07 6.229e-06
217 REGULATION OF CELLULAR RESPONSE TO TRANSFORMING GROWTH FACTOR BETA STIMULUS 7 99 2.905e-07 6.229e-06
218 REGULATION OF CELL MORPHOGENESIS INVOLVED IN DIFFERENTIATION 11 337 2.991e-07 6.383e-06
219 PEPTIDYL SERINE MODIFICATION 8 148 3.166e-07 6.726e-06
220 REGULATION OF CELL AGING 5 33 3.351e-07 7.087e-06
221 POSITIVE REGULATION OF MITOTIC SISTER CHROMATID SEPARATION 4 14 3.544e-07 7.396e-06
222 POSITIVE REGULATION OF MITOTIC METAPHASE ANAPHASE TRANSITION 4 14 3.544e-07 7.396e-06
223 POSITIVE REGULATION OF METAPHASE ANAPHASE TRANSITION OF CELL CYCLE 4 14 3.544e-07 7.396e-06
224 INTRINSIC APOPTOTIC SIGNALING PATHWAY 8 152 3.883e-07 8.065e-06
225 CELLULAR RESPONSE TO REACTIVE OXYGEN SPECIES 7 104 4.072e-07 8.42e-06
226 NEGATIVE REGULATION OF CELLULAR CATABOLIC PROCESS 8 156 4.734e-07 9.704e-06
227 RESPONSE TO LIGHT STIMULUS 10 280 4.733e-07 9.704e-06
228 ACTIVATION OF ANAPHASE PROMOTING COMPLEX ACTIVITY 4 15 4.817e-07 9.83e-06
229 RESPONSE TO OXYGEN CONTAINING COMPOUND 21 1381 5.006e-07 1.017e-05
230 REGULATION OF EPITHELIAL CELL PROLIFERATION 10 285 5.564e-07 1.126e-05
231 REGULATION OF NUCLEOCYTOPLASMIC TRANSPORT 9 220 5.938e-07 1.196e-05
232 REGENERATION 8 161 6.019e-07 1.207e-05
233 REGULATION OF APOPTOTIC SIGNALING PATHWAY 11 363 6.241e-07 1.246e-05
234 POSITIVE REGULATION OF PROTEIN SERINE THREONINE KINASE ACTIVITY 10 289 6.319e-07 1.251e-05
235 REGULATION OF SIGNAL TRANSDUCTION BY P53 CLASS MEDIATOR 8 162 6.309e-07 1.251e-05
236 POSITIVE REGULATION OF TRANSPORT 17 936 6.765e-07 1.334e-05
237 MESENCHYME MORPHOGENESIS 5 38 6.963e-07 1.367e-05
238 INTRINSIC APOPTOTIC SIGNALING PATHWAY IN RESPONSE TO DNA DAMAGE 6 71 7.434e-07 1.447e-05
239 CHROMATIN MODIFICATION 13 539 7.411e-07 1.447e-05
240 NEGATIVE REGULATION OF EPITHELIAL CELL PROLIFERATION 7 115 8.073e-07 1.565e-05
241 NEGATIVE REGULATION OF CELL AGING 4 17 8.342e-07 1.606e-05
242 RHYTHMIC PROCESS 10 298 8.352e-07 1.606e-05
243 CELLULAR RESPONSE TO LIPID 12 457 8.476e-07 1.623e-05
244 NEGATIVE REGULATION OF TRANSCRIPTION FROM RNA POLYMERASE II PROMOTER 15 740 8.654e-07 1.65e-05
245 NEGATIVE REGULATION OF CELL GROWTH 8 170 9.089e-07 1.726e-05
246 REGULATION OF DNA DEPENDENT DNA REPLICATION 5 41 1.029e-06 1.946e-05
247 RESPONSE TO NITROGEN COMPOUND 16 859 1.088e-06 2.05e-05
248 REGULATION OF CELLULAR RESPONSE TO HEAT 6 76 1.115e-06 2.092e-05
249 POSITIVE REGULATION OF NUCLEOCYTOPLASMIC TRANSPORT 7 121 1.139e-06 2.128e-05
250 PEPTIDYL LYSINE MODIFICATION 10 312 1.265e-06 2.355e-05
251 REGULATION OF CELL GROWTH 11 391 1.292e-06 2.395e-05
252 CHROMATIN ORGANIZATION 14 663 1.297e-06 2.395e-05
253 NEGATIVE REGULATION OF CELL DEATH 16 872 1.324e-06 2.435e-05
254 REGULATION OF RESPONSE TO STRESS 21 1468 1.351e-06 2.474e-05
255 RESPONSE TO INORGANIC SUBSTANCE 12 479 1.386e-06 2.529e-05
256 RESPONSE TO KETONE 8 182 1.519e-06 2.761e-05
257 PROTEIN LOCALIZATION TO CHROMOSOME 5 45 1.654e-06 2.994e-05
258 GROWTH 11 410 2.046e-06 3.69e-05
259 POSITIVE REGULATION OF DNA REPLICATION 6 86 2.314e-06 4.119e-05
260 NEGATIVE REGULATION OF DEVELOPMENTAL PROCESS 15 801 2.31e-06 4.119e-05
261 POSITIVE REGULATION OF DEVELOPMENTAL PROCESS 18 1142 2.319e-06 4.119e-05
262 POSITIVE REGULATION OF PATHWAY RESTRICTED SMAD PROTEIN PHOSPHORYLATION 5 48 2.294e-06 4.119e-05
263 CELLULAR RESPONSE TO NITROGEN COMPOUND 12 505 2.397e-06 4.241e-05
264 MEIOSIS I 6 88 2.648e-06 4.667e-05
265 AGING 9 264 2.688e-06 4.719e-05
266 POSITIVE REGULATION OF MULTICELLULAR ORGANISMAL METABOLIC PROCESS 4 23 3.041e-06 5.299e-05
267 POSITIVE REGULATION OF COLLAGEN METABOLIC PROCESS 4 23 3.041e-06 5.299e-05
268 POSITIVE REGULATION OF MITOTIC NUCLEAR DIVISION 5 51 3.114e-06 5.406e-05
269 MICROTUBULE CYTOSKELETON ORGANIZATION 10 348 3.362e-06 5.815e-05
270 CELLULAR RESPONSE TO OXYGEN LEVELS 7 143 3.483e-06 6.002e-05
271 RESPONSE TO TRANSFORMING GROWTH FACTOR BETA 7 144 3.648e-06 6.263e-05
272 RESPONSE TO IONIZING RADIATION 7 145 3.819e-06 6.533e-05
273 RESPONSE TO ESTRADIOL 7 146 3.997e-06 6.812e-05
274 TRANSFORMING GROWTH FACTOR BETA RECEPTOR SIGNALING PATHWAY 6 95 4.142e-06 7.034e-05
275 HISTONE PHOSPHORYLATION 4 25 4.315e-06 7.301e-05
276 REGULATION OF GROWTH 13 633 4.369e-06 7.366e-05
277 CELLULAR CATABOLIC PROCESS 19 1322 4.477e-06 7.521e-05
278 POSITIVE REGULATION OF PROTEASOMAL PROTEIN CATABOLIC PROCESS 6 98 4.964e-06 8.308e-05
279 REGULATION OF CELLULAR SENESCENCE 4 26 5.082e-06 8.475e-05
280 CELLULAR RESPONSE TO STEROID HORMONE STIMULUS 8 218 5.802e-06 9.642e-05
281 CELLULAR RESPONSE TO HORMONE STIMULUS 12 552 5.955e-06 9.825e-05
282 REGULATION OF CELL MORPHOGENESIS 12 552 5.955e-06 9.825e-05
283 REGULATION OF CELL DIFFERENTIATION 20 1492 6.813e-06 0.000112
284 REGULATION OF DNA DAMAGE RESPONSE SIGNAL TRANSDUCTION BY P53 CLASS MEDIATOR 4 28 6.913e-06 0.0001133
285 REGULATION OF PATHWAY RESTRICTED SMAD PROTEIN PHOSPHORYLATION 5 60 7.015e-06 0.0001145
286 CELLULAR RESPONSE TO HYDROGEN PEROXIDE 5 61 7.615e-06 0.0001239
287 NEGATIVE REGULATION OF PRODUCTION OF MOLECULAR MEDIATOR OF IMMUNE RESPONSE 4 29 7.992e-06 0.0001287
288 REGULATION OF EXTRACELLULAR MATRIX ORGANIZATION 4 29 7.992e-06 0.0001287
289 REGULATION OF HEART MORPHOGENESIS 4 29 7.992e-06 0.0001287
290 SOMITOGENESIS 5 62 8.254e-06 0.000132
291 POSITIVE REGULATION OF NUCLEAR DIVISION 5 62 8.254e-06 0.000132
292 REGULATION OF CELLULAR RESPONSE TO GROWTH FACTOR STIMULUS 8 229 8.32e-06 0.0001326
293 REGULATION OF PROTEIN ACETYLATION 5 64 9.658e-06 0.0001529
294 POSITIVE REGULATION OF RESPONSE TO DNA DAMAGE STIMULUS 5 64 9.658e-06 0.0001529
295 IMMUNE SYSTEM DEVELOPMENT 12 582 1.015e-05 0.0001601
296 NEGATIVE REGULATION OF GROWTH 8 236 1.036e-05 0.0001629
297 LENS DEVELOPMENT IN CAMERA TYPE EYE 5 66 1.124e-05 0.0001761
298 SENSORY ORGAN DEVELOPMENT 11 493 1.179e-05 0.0001841
299 NOTCH SIGNALING PATHWAY 6 114 1.188e-05 0.0001849
300 NEGATIVE REGULATION OF CYCLIN DEPENDENT PROTEIN KINASE ACTIVITY 4 32 1.198e-05 0.0001858
301 CELL AGING 5 67 1.211e-05 0.0001871
302 NEGATIVE REGULATION OF CELLULAR SENESCENCE 3 11 1.37e-05 0.0002103
303 ACTIVATION OF MAPKKK ACTIVITY 3 11 1.37e-05 0.0002103
304 MITOTIC SPINDLE ORGANIZATION 5 69 1.399e-05 0.0002141
305 PROTEIN DESTABILIZATION 4 34 1.534e-05 0.0002341
306 EPITHELIUM DEVELOPMENT 15 945 1.684e-05 0.000256
307 RESPONSE TO MINERALOCORTICOID 4 35 1.726e-05 0.0002608
308 RESPONSE TO IRON ION 4 35 1.726e-05 0.0002608
309 RESPONSE TO METAL ION 9 333 1.742e-05 0.0002615
310 DEVELOPMENTAL GROWTH 9 333 1.742e-05 0.0002615
311 REGULATION OF PROTEIN IMPORT 7 183 1.753e-05 0.0002623
312 REGULATION OF MACROPHAGE CYTOKINE PRODUCTION 3 12 1.82e-05 0.0002689
313 POSITIVE REGULATION OF SMAD PROTEIN IMPORT INTO NUCLEUS 3 12 1.82e-05 0.0002689
314 MITOTIC SISTER CHROMATID COHESION 3 12 1.82e-05 0.0002689
315 CELLULAR RESPONSE TO OXIDATIVE STRESS 7 184 1.816e-05 0.0002689
316 TRANSCRIPTION COUPLED NUCLEOTIDE EXCISION REPAIR 5 73 1.844e-05 0.0002715
317 MESENCHYME DEVELOPMENT 7 190 2.233e-05 0.0003278
318 RESPONSE TO REACTIVE OXYGEN SPECIES 7 191 2.31e-05 0.0003379
319 CELLULAR RESPONSE TO EXTERNAL STIMULUS 8 264 2.326e-05 0.0003393
320 POSITIVE REGULATION OF DNA DAMAGE RESPONSE SIGNAL TRANSDUCTION BY P53 CLASS MEDIATOR 3 13 2.359e-05 0.000343
321 REGULATION OF MULTICELLULAR ORGANISMAL METABOLIC PROCESS 4 38 2.409e-05 0.0003492
322 CATABOLIC PROCESS 21 1773 2.533e-05 0.000366
323 SOMITE DEVELOPMENT 5 78 2.546e-05 0.0003668
324 CELLULAR MACROMOLECULE LOCALIZATION 17 1234 2.683e-05 0.0003853
325 POSITIVE REGULATION OF P38MAPK CASCADE 3 14 2.993e-05 0.0004284
326 DNA GEOMETRIC CHANGE 5 81 3.058e-05 0.0004365
327 TISSUE DEVELOPMENT 19 1518 3.163e-05 0.0004501
328 TUBE DEVELOPMENT 11 552 3.343e-05 0.0004742
329 PROTEIN COMPLEX SUBUNIT ORGANIZATION 19 1527 3.431e-05 0.0004852
330 REGULATION OF MULTICELLULAR ORGANISMAL DEVELOPMENT 20 1672 3.591e-05 0.0005063
331 INTERSPECIES INTERACTION BETWEEN ORGANISMS 12 662 3.616e-05 0.0005068
332 SYMBIOSIS ENCOMPASSING MUTUALISM THROUGH PARASITISM 12 662 3.616e-05 0.0005068
333 POSITIVE REGULATION OF CYTOPLASMIC TRANSPORT 8 282 3.721e-05 0.00052
334 POSITIVE REGULATION OF CHROMATIN MODIFICATION 5 85 3.861e-05 0.0005379
335 HEART DEVELOPMENT 10 466 4.205e-05 0.0005841
336 REGULATION OF SMAD PROTEIN IMPORT INTO NUCLEUS 3 16 4.574e-05 0.0006297
337 NEGATIVE REGULATION OF DNA DEPENDENT DNA REPLICATION 3 16 4.574e-05 0.0006297
338 CELLULAR RESPONSE TO ANTIBIOTIC 3 16 4.574e-05 0.0006297
339 REGULATION OF CYSTEINE TYPE ENDOPEPTIDASE ACTIVITY 7 213 4.632e-05 0.0006358
340 SEGMENTATION 5 89 4.818e-05 0.0006594
341 REGULATION OF INTRACELLULAR PROTEIN TRANSPORT 9 381 4.991e-05 0.000681
342 CELLULAR RESPONSE TO OXYGEN CONTAINING COMPOUND 13 799 5.094e-05 0.000691
343 MESONEPHROS DEVELOPMENT 5 90 5.084e-05 0.000691
344 REGULATION OF PROTEIN COMPLEX DISASSEMBLY 7 217 5.211e-05 0.0007048
345 REGULATION OF CYTOPLASMIC TRANSPORT 10 481 5.484e-05 0.0007397
346 POSITIVE REGULATION OF SIGNAL TRANSDUCTION BY P53 CLASS MEDIATOR 3 17 5.537e-05 0.0007403
347 POSITIVE REGULATION OF EXTRACELLULAR MATRIX ORGANIZATION 3 17 5.537e-05 0.0007403
348 REGULATION OF SISTER CHROMATID COHESION 3 17 5.537e-05 0.0007403
349 REGULATION OF PROTEIN STABILITY 7 221 5.848e-05 0.0007796
350 REGULATION OF CHROMATIN ORGANIZATION 6 152 6.034e-05 0.0008022
351 REGULATION OF LIPID KINASE ACTIVITY 4 48 6.138e-05 0.0008114
352 HOMOLOGOUS CHROMOSOME SEGREGATION 4 48 6.138e-05 0.0008114
353 REGULATION OF DNA BIOSYNTHETIC PROCESS 5 94 6.262e-05 0.0008254
354 POSITIVE REGULATION OF CELL CYCLE G2 M PHASE TRANSITION 3 18 6.623e-05 0.000868
355 REGULATION OF UBIQUITIN PROTEIN LIGASE ACTIVITY 3 18 6.623e-05 0.000868
356 REGULATION OF PROTEIN TARGETING 8 307 6.765e-05 0.0008842
357 HEAD DEVELOPMENT 12 709 6.997e-05 0.000912
358 RESPONSE TO PROGESTERONE 4 50 7.216e-05 0.0009353
359 RESPONSE TO GAMMA RADIATION 4 50 7.216e-05 0.0009353
360 HEMATOPOIETIC PROGENITOR CELL DIFFERENTIATION 5 98 7.639e-05 0.0009874
361 RESPONSE TO AMMONIUM ION 4 51 7.804e-05 0.001006
362 REGULATION OF CELL DEVELOPMENT 13 836 8.064e-05 0.001037
363 NEGATIVE REGULATION OF CELL DIFFERENTIATION 11 609 8.093e-05 0.001037
364 POSITIVE REGULATION OF CELLULAR COMPONENT BIOGENESIS 9 406 8.127e-05 0.001039
365 CELLULAR RESPONSE TO IONIZING RADIATION 4 52 8.425e-05 0.001074
366 TRANSCRIPTION FROM RNA POLYMERASE II PROMOTER 12 724 8.537e-05 0.001085
367 INTRINSIC APOPTOTIC SIGNALING PATHWAY BY P53 CLASS MEDIATOR 4 53 9.082e-05 0.001151
368 NEGATIVE REGULATION OF CELL CYCLE ARREST 3 20 9.193e-05 0.001162
369 NEGATIVE REGULATION OF TRANSMEMBRANE RECEPTOR PROTEIN SERINE THREONINE KINASE SIGNALING PATHWAY 5 102 9.24e-05 0.001165
370 RESPONSE TO TOXIC SUBSTANCE 7 241 0.0001006 0.001266
371 POSITIVE REGULATION OF PROTEIN IMPORT 5 104 0.0001013 0.001271
372 EYE DEVELOPMENT 8 326 0.0001027 0.001284
373 CRANIAL SKELETAL SYSTEM DEVELOPMENT 4 55 0.0001051 0.001311
374 MICROTUBULE BASED PROCESS 10 522 0.0001079 0.001343
375 NEGATIVE REGULATION OF PROTEIN COMPLEX DISASSEMBLY 6 170 0.000112 0.00139
376 EPITHELIAL TO MESENCHYMAL TRANSITION 4 56 0.0001128 0.001395
377 NEGATIVE REGULATION OF CYTOKINE PRODUCTION INVOLVED IN IMMUNE RESPONSE 3 22 0.0001234 0.001515
378 ENDOCARDIAL CUSHION MORPHOGENESIS 3 22 0.0001234 0.001515
379 POSITIVE REGULATION OF MULTICELLULAR ORGANISMAL PROCESS 17 1395 0.0001232 0.001515
380 MACROMOLECULAR COMPLEX ASSEMBLY 17 1398 0.0001265 0.001545
381 RESPONSE TO HYDROGEN PEROXIDE 5 109 0.0001265 0.001545
382 POSITIVE REGULATION OF DNA BIOSYNTHETIC PROCESS 4 59 0.0001383 0.001685
383 RESPONSE TO INCREASED OXYGEN LEVELS 3 23 0.0001414 0.001714
384 RESPONSE TO HYPEROXIA 3 23 0.0001414 0.001714
385 REGULATION OF CELLULAR COMPONENT BIOGENESIS 12 767 0.0001468 0.001774
386 NUCLEOTIDE EXCISION REPAIR 5 113 0.0001499 0.001806
387 COVALENT CHROMATIN MODIFICATION 8 345 0.0001516 0.001823
388 POSITIVE REGULATION OF INTRACELLULAR PROTEIN TRANSPORT 7 258 0.0001535 0.001841
389 EMBRYO DEVELOPMENT 13 894 0.0001574 0.001883
390 POSITIVE REGULATION OF CELLULAR RESPONSE TO TRANSFORMING GROWTH FACTOR BETA STIMULUS 3 24 0.0001611 0.001908
391 POSITIVE REGULATION OF G1 S TRANSITION OF MITOTIC CELL CYCLE 3 24 0.0001611 0.001908
392 POSITIVE REGULATION OF TRANSFORMING GROWTH FACTOR BETA RECEPTOR SIGNALING PATHWAY 3 24 0.0001611 0.001908
393 CELL DEVELOPMENT 17 1426 0.0001606 0.001908
394 EMBRYO DEVELOPMENT ENDING IN BIRTH OR EGG HATCHING 10 554 0.0001752 0.002069
395 LENS FIBER CELL DIFFERENTIATION 3 25 0.0001825 0.002123
396 CELLULAR RESPONSE TO TOXIC SUBSTANCE 3 25 0.0001825 0.002123
397 REGULATION OF TRANSFORMING GROWTH FACTOR BETA PRODUCTION 3 25 0.0001825 0.002123
398 POSITIVE REGULATION OF PEPTIDYL THREONINE PHOSPHORYLATION 3 25 0.0001825 0.002123
399 EPITHELIAL CELL APOPTOTIC PROCESS 3 25 0.0001825 0.002123
400 PROTEIN LOCALIZATION TO ORGANELLE 10 556 0.0001804 0.002123
401 REGULATION OF CYTOKINE PRODUCTION 10 563 0.0001995 0.002315
402 MEIOTIC CHROMOSOME SEGREGATION 4 65 0.0002016 0.002334
403 RESPONSE TO CORTICOSTERONE 3 26 0.0002057 0.002363
404 REGULATION OF P38MAPK CASCADE 3 26 0.0002057 0.002363
405 TRANSMEMBRANE RECEPTOR PROTEIN SERINE THREONINE KINASE SIGNALING PATHWAY 6 190 0.0002053 0.002363
406 NEGATIVE REGULATION OF CELLULAR RESPONSE TO GROWTH FACTOR STIMULUS 5 121 0.0002064 0.002366
407 NEGATIVE REGULATION OF CELLULAR RESPONSE TO TRANSFORMING GROWTH FACTOR BETA STIMULUS 4 66 0.0002139 0.00244
408 NEGATIVE REGULATION OF TRANSFORMING GROWTH FACTOR BETA RECEPTOR SIGNALING PATHWAY 4 66 0.0002139 0.00244
409 DNA CONFORMATION CHANGE 7 273 0.000217 0.002469
410 DEVELOPMENTAL MATURATION 6 193 0.0002234 0.002535
411 POSITIVE REGULATION OF NEURON DEATH 4 67 0.0002267 0.002561
412 REGULATION OF DNA TEMPLATED TRANSCRIPTION IN RESPONSE TO STRESS 4 67 0.0002267 0.002561
413 ANTERIOR POSTERIOR PATTERN SPECIFICATION 6 194 0.0002297 0.002588
414 KIDNEY EPITHELIUM DEVELOPMENT 5 125 0.0002401 0.002699
415 POSITIVE REGULATION OF CELL DEVELOPMENT 9 472 0.0002515 0.00282
416 NEGATIVE REGULATION OF IMMUNE SYSTEM PROCESS 8 372 0.0002529 0.002828
417 NEGATIVE REGULATION OF DENDRITE DEVELOPMENT 3 28 0.0002575 0.002873
418 POSITIVE REGULATION OF BINDING 5 127 0.0002585 0.002878
419 POSITIVE REGULATION OF CELL DIFFERENTIATION 12 823 0.0002809 0.00312
420 INTRACELLULAR STEROID HORMONE RECEPTOR SIGNALING PATHWAY 4 71 0.0002835 0.003134
421 ENDODERM DEVELOPMENT 4 71 0.0002835 0.003134
422 NUCLEOTIDE EXCISION REPAIR PREINCISION COMPLEX ASSEMBLY 3 29 0.0002862 0.003149
423 POSITIVE REGULATION OF CELL CYCLE G1 S PHASE TRANSITION 3 29 0.0002862 0.003149
424 CELL MATURATION 5 131 0.0002984 0.003275
425 NEGATIVE REGULATION OF RESPONSE TO STIMULUS 16 1360 0.0002992 0.003275
426 MITOCHONDRION ORGANIZATION 10 594 0.000306 0.003343
427 REGULATION OF TRANSPORT 19 1804 0.0003108 0.003387
428 REGULATION OF INTRACELLULAR SIGNAL TRANSDUCTION 18 1656 0.0003149 0.003423
429 ORGAN MORPHOGENESIS 12 841 0.0003418 0.003706
430 LYMPHOCYTE DIFFERENTIATION 6 209 0.0003425 0.003706
431 RESPONSE TO ETHANOL 5 136 0.0003547 0.003829
432 REGULATION OF PEPTIDASE ACTIVITY 8 392 0.0003591 0.003868
433 SALIVARY GLAND DEVELOPMENT 3 32 0.0003848 0.004116
434 ENDOCARDIAL CUSHION DEVELOPMENT 3 32 0.0003848 0.004116
435 REGULATION OF TRANSCRIPTION FROM RNA POLYMERASE II PROMOTER IN RESPONSE TO HYPOXIA 3 32 0.0003848 0.004116
436 NEUROGENESIS 16 1402 0.0004196 0.004478
437 REGULATION OF PROTEIN EXPORT FROM NUCLEUS 3 33 0.0004219 0.004493
438 RESPONSE TO PEPTIDE 8 404 0.0004387 0.004661
439 RAS PROTEIN SIGNAL TRANSDUCTION 5 143 0.0004466 0.004733
440 PROTEIN TARGETING 8 406 0.0004533 0.004794
441 NEGATIVE REGULATION OF CYTOSKELETON ORGANIZATION 6 221 0.0004607 0.004856
442 HEART VALVE DEVELOPMENT 3 34 0.0004613 0.004856
443 IN UTERO EMBRYONIC DEVELOPMENT 7 311 0.0004763 0.005003
444 POSITIVE REGULATION OF INTRACELLULAR SIGNAL TRANSDUCTION 12 876 0.0004927 0.005164
445 RESPONSE TO MONOAMINE 3 35 0.0005029 0.005259
446 RESPONSE TO TRANSITION METAL NANOPARTICLE 5 148 0.0005224 0.00545
447 POSITIVE REGULATION OF OSSIFICATION 4 84 0.0005389 0.005609
448 POSITIVE REGULATION OF PROTEIN ACETYLATION 3 36 0.0005469 0.005667
449 HEAD MORPHOGENESIS 3 36 0.0005469 0.005667
450 CHROMATIN REMODELING 5 150 0.0005553 0.005734
451 REGULATION OF GENE EXPRESSION EPIGENETIC 6 229 0.0005558 0.005734
452 REGULATION OF ANATOMICAL STRUCTURE MORPHOGENESIS 13 1021 0.0005653 0.00582
453 REGULATION OF PEPTIDYL THREONINE PHOSPHORYLATION 3 37 0.0005932 0.006093
454 MULTICELLULAR ORGANISM REPRODUCTION 11 768 0.000595 0.006098
455 TRANSCRIPTION INITIATION FROM RNA POLYMERASE II PROMOTER 5 153 0.0006076 0.006213
456 TISSUE REMODELING 4 87 0.0006154 0.006279
457 MEMBRANE ORGANIZATION 12 899 0.0006199 0.006312
458 OVULATION CYCLE PROCESS 4 88 0.0006425 0.006499
459 POSITIVE REGULATION OF BIOMINERAL TISSUE DEVELOPMENT 3 38 0.000642 0.006499
460 OOCYTE DIFFERENTIATION 3 38 0.000642 0.006499
461 CELLULAR RESPONSE TO INORGANIC SUBSTANCE 5 156 0.0006634 0.006696
462 B CELL DIFFERENTIATION 4 89 0.0006705 0.006753
463 CELLULAR RESPONSE TO NUTRIENT 3 39 0.0006933 0.006952
464 SPLEEN DEVELOPMENT 3 39 0.0006933 0.006952
465 REGULATION OF CELL MATRIX ADHESION 4 90 0.0006993 0.006997
466 REGULATION OF DEPHOSPHORYLATION 5 158 0.0007028 0.007017
467 REGULATION OF ORGAN MORPHOGENESIS 6 242 0.0007419 0.007392
468 POSITIVE REGULATION OF CELL MATRIX ADHESION 3 40 0.0007471 0.007412
469 RESPONSE TO CADMIUM ION 3 40 0.0007471 0.007412
470 NEGATIVE REGULATION OF CELL COMMUNICATION 14 1192 0.0007589 0.007513
471 REGULATION OF DNA BINDING 4 93 0.000791 0.007814
472 NEGATIVE REGULATION OF FAT CELL DIFFERENTIATION 3 41 0.0008035 0.007904
473 ANDROGEN RECEPTOR SIGNALING PATHWAY 3 41 0.0008035 0.007904
474 LYMPHOCYTE ACTIVATION 7 342 0.0008344 0.00819
475 POSTTRANSCRIPTIONAL REGULATION OF GENE EXPRESSION 8 448 0.0008623 0.008447
476 PROTEIN LOCALIZATION 18 1805 0.000878 0.008582
477 RESPONSE TO CARBOHYDRATE 5 168 0.0009261 0.009015
478 BETA CATENIN TCF COMPLEX ASSEMBLY 3 43 0.0009243 0.009015
479 CELL ACTIVATION 9 568 0.0009535 0.009262
480 RESPONSE TO VITAMIN 4 98 0.0009623 0.009328
481 BODY MORPHOGENESIS 3 44 0.0009887 0.009544
482 RESPONSE TO OXIDATIVE STRESS 7 352 0.0009868 0.009544
483 EXTRINSIC APOPTOTIC SIGNALING PATHWAY 4 99 0.0009994 0.009628
484 REGULATION OF SMOOTH MUSCLE CELL PROLIFERATION 4 100 0.001038 0.009954
485 PEPTIDYL TYROSINE DEPHOSPHORYLATION 4 100 0.001038 0.009954
NumGOOverlapSizeP ValueAdj. P Value
1 CYCLIN DEPENDENT PROTEIN SERINE THREONINE KINASE REGULATOR ACTIVITY 11 28 1.442e-19 1.34e-16
2 ENZYME BINDING 37 1737 6.307e-17 2.93e-14
3 KINASE BINDING 22 606 1.619e-14 5.014e-12
4 PROTEIN KINASE ACTIVITY 22 640 4.911e-14 1.141e-11
5 PROTEIN SERINE THREONINE KINASE ACTIVITY 18 445 9.021e-13 1.676e-10
6 KINASE ACTIVITY 23 842 1.425e-12 1.831e-10
7 PROTEIN COMPLEX BINDING 24 935 1.576e-12 1.831e-10
8 TRANSCRIPTION FACTOR BINDING 19 524 1.33e-12 1.831e-10
9 KINASE REGULATOR ACTIVITY 13 186 1.937e-12 2e-10
10 CYCLIN DEPENDENT PROTEIN SERINE THREONINE KINASE INHIBITOR ACTIVITY 6 12 5.917e-12 5.497e-10
11 MACROMOLECULAR COMPLEX BINDING 27 1399 3.529e-11 2.981e-09
12 TRANSFERASE ACTIVITY TRANSFERRING PHOSPHORUS CONTAINING GROUPS 23 992 3.953e-11 3.06e-09
13 PROTEIN SERINE THREONINE KINASE INHIBITOR ACTIVITY 7 30 5.09e-11 3.638e-09
14 CYCLIN DEPENDENT PROTEIN KINASE ACTIVITY 6 34 7.963e-09 5.284e-07
15 ADENYL NUCLEOTIDE BINDING 23 1514 1.263e-07 7.823e-06
16 KINASE INHIBITOR ACTIVITY 7 89 1.394e-07 8.093e-06
17 ENZYME REGULATOR ACTIVITY 18 959 1.85e-07 1.011e-05
18 RNA POLYMERASE II TRANSCRIPTION FACTOR BINDING 7 104 4.072e-07 2.101e-05
19 CYCLIN BINDING 4 19 1.349e-06 6.597e-05
20 MOLECULAR FUNCTION REGULATOR 20 1353 1.542e-06 7.164e-05
21 STEROID HORMONE RECEPTOR BINDING 6 81 1.626e-06 7.192e-05
22 UBIQUITIN LIKE PROTEIN LIGASE BINDING 9 264 2.688e-06 0.0001135
23 TRANSFORMING GROWTH FACTOR BETA RECEPTOR BINDING 5 50 2.818e-06 0.0001138
24 RIBONUCLEOTIDE BINDING 23 1860 4.571e-06 0.0001769
25 CORE PROMOTER BINDING 7 152 5.216e-06 0.0001938
26 ACTIVATING TRANSCRIPTION FACTOR BINDING 5 57 5.435e-06 0.0001942
27 NF KAPPAB BINDING 4 30 9.19e-06 0.0003162
28 P53 BINDING 5 67 1.211e-05 0.0004016
29 SMAD BINDING 5 72 1.723e-05 0.0005521
30 CULLIN FAMILY PROTEIN BINDING 3 12 1.82e-05 0.0005637
31 RNA POLYMERASE II ACTIVATING TRANSCRIPTION FACTOR BINDING 4 36 1.935e-05 0.00058
32 CHROMATIN BINDING 10 435 2.347e-05 0.0006813
33 ANDROGEN RECEPTOR BINDING 4 39 2.675e-05 0.0007402
34 DNA DEPENDENT ATPASE ACTIVITY 5 79 2.709e-05 0.0007402
35 LIGASE REGULATOR ACTIVITY 3 14 2.993e-05 0.0007943
36 PEROXISOME PROLIFERATOR ACTIVATED RECEPTOR BINDING 3 15 3.729e-05 0.0009622
37 PROTEIN HETERODIMERIZATION ACTIVITY 10 468 4.359e-05 0.001095
38 TRANSCRIPTION FACTOR ACTIVITY PROTEIN BINDING 11 588 5.916e-05 0.001446
39 NUCLEIC ACID BINDING TRANSCRIPTION FACTOR ACTIVITY 16 1199 7.011e-05 0.00167
40 HISTONE KINASE ACTIVITY 3 19 7.839e-05 0.001821
41 DNA HELICASE ACTIVITY 4 53 9.082e-05 0.002058
42 HORMONE RECEPTOR BINDING 6 168 0.0001049 0.002321
43 R SMAD BINDING 3 23 0.0001414 0.003056
44 PROTEIN C TERMINUS BINDING 6 186 0.0001829 0.003863
45 RECEPTOR BINDING 17 1476 0.0002421 0.004998
46 BHLH TRANSCRIPTION FACTOR BINDING 3 28 0.0002575 0.0052
47 REGULATORY REGION NUCLEIC ACID BINDING 12 818 0.0002657 0.005253
48 ENZYME INHIBITOR ACTIVITY 8 378 0.0002816 0.00545
49 PROTEIN DOMAIN SPECIFIC BINDING 10 624 0.000451 0.008551
50 ATP DEPENDENT DNA HELICASE ACTIVITY 3 34 0.0004613 0.008571
51 RNA POLYMERASE II TRANSCRIPTION FACTOR ACTIVITY SEQUENCE SPECIFIC DNA BINDING 10 629 0.00048 0.008744
52 BETA CATENIN BINDING 4 84 0.0005389 0.009627
NumGOOverlapSizeP ValueAdj. P Value
1 CHROMOSOME 32 880 3.177e-21 1.856e-18
2 CHROMOSOMAL REGION 21 330 9.223e-19 2.693e-16
3 TRANSCRIPTION FACTOR COMPLEX 19 298 4.873e-17 9.485e-15
4 TRANSFERASE COMPLEX 25 703 2.942e-16 4.295e-14
5 NUCLEAR CHROMOSOME 22 523 7.824e-16 9.138e-14
6 CYCLIN DEPENDENT PROTEIN KINASE HOLOENZYME COMPLEX 9 31 8.404e-15 8.18e-13
7 ANAPHASE PROMOTING COMPLEX 8 22 3.386e-14 2.825e-12
8 CHROMATIN 19 441 6.249e-14 4.562e-12
9 NUCLEAR UBIQUITIN LIGASE COMPLEX 9 42 1.786e-13 1.159e-11
10 CATALYTIC COMPLEX 25 1038 1.972e-12 1.151e-10
11 CULLIN RING UBIQUITIN LIGASE COMPLEX 12 150 2.935e-12 1.558e-10
12 PROTEIN KINASE COMPLEX 10 90 7.746e-12 3.77e-10
13 MICROTUBULE CYTOSKELETON 24 1068 2.58e-11 1.159e-09
14 SPINDLE 14 289 3.657e-11 1.525e-09
15 CHROMOSOME CENTROMERIC REGION 11 174 3.193e-10 1.243e-08
16 CONDENSED CHROMOSOME 11 195 1.074e-09 3.921e-08
17 UBIQUITIN LIGASE COMPLEX 12 262 1.928e-09 6.624e-08
18 CHROMOSOME TELOMERIC REGION 10 162 2.699e-09 8.757e-08
19 CONDENSED NUCLEAR CHROMOSOME 8 85 4.06e-09 1.248e-07
20 CENTROSOME 14 487 3.075e-08 8.979e-07
21 SPINDLE POLE 8 126 9.14e-08 2.542e-06
22 MICROTUBULE ORGANIZING CENTER 15 623 9.655e-08 2.563e-06
23 TRANSFERASE COMPLEX TRANSFERRING PHOSPHORUS CONTAINING GROUPS 10 237 1.013e-07 2.571e-06
24 MCM COMPLEX 4 11 1.18e-07 2.873e-06
25 NUCLEAR CHROMOSOME TELOMERIC REGION 8 132 1.311e-07 3.063e-06
26 CONDENSED CHROMOSOME OUTER KINETOCHORE 4 12 1.765e-07 3.964e-06
27 CYTOSKELETAL PART 22 1436 2.2e-07 4.758e-06
28 CYTOSKELETON 26 1967 2.422e-07 5.051e-06
29 CONDENSED CHROMOSOME CENTROMERIC REGION 7 102 3.565e-07 7.179e-06
30 KINETOCHORE 7 120 1.077e-06 2.028e-05
31 CONDENSED NUCLEAR CHROMOSOME CENTROMERIC REGION 4 18 1.069e-06 2.028e-05
32 RNA POLYMERASE II TRANSCRIPTION FACTOR COMPLEX 6 101 5.913e-06 0.0001079
33 COHESIN COMPLEX 3 11 1.37e-05 0.0002424
34 NUCLEAR TRANSCRIPTION FACTOR COMPLEX 6 127 2.199e-05 0.0003669
35 NUCLEOLUS 14 848 2.161e-05 0.0003669
36 CARBOXY TERMINAL DOMAIN PROTEIN KINASE COMPLEX 3 22 0.0001234 0.002002
37 SPINDLE MIDZONE 3 27 0.0002306 0.00364
38 CYTOPLASMIC VESICLE PART 10 601 0.0003358 0.00516
39 SCF UBIQUITIN LIGASE COMPLEX 3 34 0.0004613 0.006908

Over-represented Pathway

NumPathwayPathviewOverlapSizeP ValueAdj. P Value
1 hsa04110_Cell_cycle 89 128 3.716e-214 6.689e-212
2 hsa04114_Oocyte_meiosis 26 114 3.279e-38 2.951e-36
3 hsa04115_p53_signaling_pathway 18 69 1.082e-27 6.494e-26
4 hsa04350_TGF.beta_signaling_pathway 16 85 3.516e-22 1.582e-20
5 hsa04914_Progesterone.mediated_oocyte_maturation 15 87 3.136e-20 1.129e-18
6 hsa04390_Hippo_signaling_pathway 16 154 7.515e-18 2.255e-16
7 hsa04151_PI3K_AKT_signaling_pathway 19 351 9.999e-16 2.571e-14
8 hsa04120_Ubiquitin_mediated_proteolysis 11 139 2.817e-11 6.338e-10
9 hsa04310_Wnt_signaling_pathway 11 151 6.934e-11 1.387e-09
10 hsa04722_Neurotrophin_signaling_pathway 8 127 9.719e-08 1.749e-06
11 hsa03030_DNA_replication 5 36 5.267e-07 8.618e-06
12 hsa04520_Adherens_junction 5 73 1.844e-05 0.0002766
13 hsa04010_MAPK_signaling_pathway 8 268 2.59e-05 0.0003587
14 hsa04012_ErbB_signaling_pathway 5 87 4.319e-05 0.0005553
15 hsa03420_Nucleotide_excision_repair 4 45 4.748e-05 0.0005698
16 hsa04144_Endocytosis 6 203 0.0002931 0.003298
17 hsa04630_Jak.STAT_signaling_pathway 5 155 0.0006444 0.006823
18 hsa04330_Notch_signaling_pathway 3 47 0.001199 0.01199
19 hsa04710_Circadian_rhythm_._mammal 2 23 0.004662 0.04416
20 hsa04916_Melanogenesis 3 101 0.01037 0.09335
21 hsa04720_Long.term_potentiation 2 70 0.0389 0.3334
22 hsa04510_Focal_adhesion 3 200 0.05991 0.4739
23 hsa04660_T_cell_receptor_signaling_pathway 2 108 0.08361 0.6271
24 hsa04380_Osteoclast_differentiation 2 128 0.1112 0.7608
25 hsa04360_Axon_guidance 2 130 0.1141 0.7608
26 hsa04141_Protein_processing_in_endoplasmic_reticulum 2 168 0.1719 1

lncRNA-mediated sponge

(Download full result)

Num lncRNA miRNAs           miRNAs count     Gene Sponge regulatory network lncRNA log2FC lncRNA pvalue Gene log2FC Gene pvalue lncRNA-gene Pearson correlation

Quest ID: c329e339621cea8ee56f9aa3928f9a8f