HCSGD entry for PTEN
1. General information
Official gene symbol | PTEN |
---|---|
Entrez ID | 5728 |
Gene full name | phosphatase and tensin homolog |
Other gene symbols | 10q23del BZS CWS1 DEC GLM2 MHAM MMAC1 PTEN1 TEP1 |
Links to Entrez Gene | Links to Entrez Gene |
2. Neighbors in the network

This gene isn't in PPI subnetwork.
3. Gene ontology annotation
GO ID | GO term | Evidence | Category |
---|---|---|---|
GO:0000079 | Regulation of cyclin-dependent protein serine/threonine kinase activity | TAS | biological_process |
GO:0000287 | Magnesium ion binding | IEA | molecular_function |
GO:0001525 | Angiogenesis | IEA | biological_process |
GO:0001933 | Negative regulation of protein phosphorylation | IDA | biological_process |
GO:0002902 | Regulation of B cell apoptotic process | IEA | biological_process |
GO:0004438 | Phosphatidylinositol-3-phosphatase activity | IDA | molecular_function |
GO:0004721 | Phosphoprotein phosphatase activity | IDA | molecular_function |
GO:0004722 | Protein serine/threonine phosphatase activity | IDA | molecular_function |
GO:0004725 | Protein tyrosine phosphatase activity | IDA | molecular_function |
GO:0005161 | Platelet-derived growth factor receptor binding | IEA | molecular_function |
GO:0005515 | Protein binding | IPI | molecular_function |
GO:0005634 | Nucleus | IDA | cellular_component |
GO:0005737 | Cytoplasm | IDA TAS | cellular_component |
GO:0005739 | Mitochondrion | IEA | cellular_component |
GO:0005829 | Cytosol | TAS | cellular_component |
GO:0005886 | Plasma membrane | IDA | cellular_component |
GO:0006470 | Protein dephosphorylation | IDA TAS | biological_process |
GO:0006644 | Phospholipid metabolic process | TAS | biological_process |
GO:0006661 | Phosphatidylinositol biosynthetic process | TAS | biological_process |
GO:0006915 | Apoptotic process | ISS | biological_process |
GO:0007092 | Activation of mitotic anaphase-promoting complex activity | IDA | biological_process |
GO:0007173 | Epidermal growth factor receptor signaling pathway | TAS | biological_process |
GO:0007270 | Neuron-neuron synaptic transmission | ISS | biological_process |
GO:0007416 | Synapse assembly | ISS | biological_process |
GO:0007417 | Central nervous system development | ISS | biological_process |
GO:0007507 | Heart development | ISS | biological_process |
GO:0007568 | Aging | IEA | biological_process |
GO:0007584 | Response to nutrient | IEA | biological_process |
GO:0007611 | Learning or memory | ISS | biological_process |
GO:0007613 | Memory | IEA | biological_process |
GO:0007626 | Locomotory behavior | ISS | biological_process |
GO:0008138 | Protein tyrosine/serine/threonine phosphatase activity | IEA | molecular_function |
GO:0008283 | Cell proliferation | TAS | biological_process |
GO:0008284 | Positive regulation of cell proliferation | ISS | biological_process |
GO:0008285 | Negative regulation of cell proliferation | IDA IMP | biological_process |
GO:0008289 | Lipid binding | IEA | molecular_function |
GO:0008543 | Fibroblast growth factor receptor signaling pathway | TAS | biological_process |
GO:0009749 | Response to glucose | IEA | biological_process |
GO:0009898 | Cytoplasmic side of plasma membrane | IDA | cellular_component |
GO:0010043 | Response to zinc ion | IEA | biological_process |
GO:0010975 | Regulation of neuron projection development | ISS | biological_process |
GO:0010997 | Anaphase-promoting complex binding | IPI | molecular_function |
GO:0014067 | Negative regulation of phosphatidylinositol 3-kinase signaling | TAS | biological_process |
GO:0016314 | Phosphatidylinositol-3,4,5-trisphosphate 3-phosphatase activity | IDA TAS | molecular_function |
GO:0016477 | Cell migration | ISS | biological_process |
GO:0016605 | PML body | IEA | cellular_component |
GO:0019899 | Enzyme binding | IPI | molecular_function |
GO:0019901 | Protein kinase binding | IEA | molecular_function |
GO:0021542 | Dentate gyrus development | ISS | biological_process |
GO:0021955 | Central nervous system neuron axonogenesis | ISS | biological_process |
GO:0030165 | PDZ domain binding | IPI | molecular_function |
GO:0030336 | Negative regulation of cell migration | IMP | biological_process |
GO:0031642 | Negative regulation of myelination | IEA | biological_process |
GO:0031647 | Regulation of protein stability | IMP | biological_process |
GO:0031658 | Negative regulation of cyclin-dependent protein serine/threonine kinase activity involved in G1/S transition of mitotic cell cycle | IDA | biological_process |
GO:0032286 | Central nervous system myelin maintenance | ISS | biological_process |
GO:0032355 | Response to estradiol | IEA | biological_process |
GO:0032535 | Regulation of cellular component size | ISS | biological_process |
GO:0033032 | Regulation of myeloid cell apoptotic process | IEA | biological_process |
GO:0033198 | Response to ATP | IEA | biological_process |
GO:0033555 | Multicellular organismal response to stress | ISS | biological_process |
GO:0035176 | Social behavior | ISS | biological_process |
GO:0035335 | Peptidyl-tyrosine dephosphorylation | IDA | biological_process |
GO:0035749 | Myelin sheath adaxonal region | ISS | cellular_component |
GO:0038095 | Fc-epsilon receptor signaling pathway | TAS | biological_process |
GO:0042493 | Response to drug | IEA | biological_process |
GO:0042711 | Maternal behavior | IEA | biological_process |
GO:0042995 | Cell projection | IDA | cellular_component |
GO:0043005 | Neuron projection | ISS | cellular_component |
GO:0043065 | Positive regulation of apoptotic process | IEA | biological_process |
GO:0043066 | Negative regulation of apoptotic process | IEA | biological_process |
GO:0043197 | Dendritic spine | IEA | cellular_component |
GO:0043220 | Schmidt-Lanterman incisure | ISS | cellular_component |
GO:0043491 | Protein kinase B signaling | ISS | biological_process |
GO:0043542 | Endothelial cell migration | IEA | biological_process |
GO:0043647 | Inositol phosphate metabolic process | TAS | biological_process |
GO:0044281 | Small molecule metabolic process | TAS | biological_process |
GO:0045087 | Innate immune response | TAS | biological_process |
GO:0045211 | Postsynaptic membrane | IEA | cellular_component |
GO:0045471 | Response to ethanol | IEA | biological_process |
GO:0045475 | Locomotor rhythm | ISS | biological_process |
GO:0045792 | Negative regulation of cell size | ISS | biological_process |
GO:0046621 | Negative regulation of organ growth | ISS | biological_process |
GO:0046685 | Response to arsenic-containing substance | IEA | biological_process |
GO:0046855 | Inositol phosphate dephosphorylation | IDA | biological_process |
GO:0046856 | Phosphatidylinositol dephosphorylation | IDA IMP | biological_process |
GO:0048008 | Platelet-derived growth factor receptor signaling pathway | IEA | biological_process |
GO:0048011 | Neurotrophin TRK receptor signaling pathway | TAS | biological_process |
GO:0048015 | Phosphatidylinositol-mediated signaling | TAS | biological_process |
GO:0048738 | Cardiac muscle tissue development | IEA | biological_process |
GO:0048853 | Forebrain morphogenesis | ISS | biological_process |
GO:0048854 | Brain morphogenesis | ISS | biological_process |
GO:0050680 | Negative regulation of epithelial cell proliferation | IEA | biological_process |
GO:0050765 | Negative regulation of phagocytosis | IEA | biological_process |
GO:0050771 | Negative regulation of axonogenesis | ISS | biological_process |
GO:0050821 | Protein stabilization | IDA | biological_process |
GO:0050852 | T cell receptor signaling pathway | TAS | biological_process |
GO:0051091 | Positive regulation of sequence-specific DNA binding transcription factor activity | IMP | biological_process |
GO:0051717 | Inositol-1,3,4,5-tetrakisphosphate 3-phosphatase activity | IDA TAS | molecular_function |
GO:0051800 | Phosphatidylinositol-3,4-bisphosphate 3-phosphatase activity | IDA TAS | molecular_function |
GO:0051895 | Negative regulation of focal adhesion assembly | IMP | biological_process |
GO:0051898 | Negative regulation of protein kinase B signaling | IMP | biological_process |
GO:0060024 | Rhythmic synaptic transmission | ISS | biological_process |
GO:0060070 | Canonical Wnt signaling pathway | IDA | biological_process |
GO:0060074 | Synapse maturation | ISS | biological_process |
GO:0060134 | Prepulse inhibition | ISS | biological_process |
GO:0060179 | Male mating behavior | IEA | biological_process |
GO:0060291 | Long-term synaptic potentiation | IEA | biological_process |
GO:0060292 | Long term synaptic depression | IEA | biological_process |
GO:0060736 | Prostate gland growth | IEA | biological_process |
GO:0060997 | Dendritic spine morphogenesis | ISS | biological_process |
GO:0061002 | Negative regulation of dendritic spine morphogenesis | ISS | biological_process |
GO:0090071 | Negative regulation of ribosome biogenesis | IEA | biological_process |
GO:0090344 | Negative regulation of cell aging | IEA | biological_process |
GO:0090394 | Negative regulation of excitatory postsynaptic membrane potential | ISS | biological_process |
GO:0097105 | Presynaptic membrane assembly | ISS | biological_process |
GO:0097107 | Postsynaptic density assembly | ISS | biological_process |
GO:2000060 | Positive regulation of protein ubiquitination involved in ubiquitin-dependent protein catabolic process | IDA | biological_process |
GO:2000134 | Negative regulation of G1/S transition of mitotic cell cycle | IDA | biological_process |
GO:2000463 | Positive regulation of excitatory postsynaptic membrane potential | ISS | biological_process |
GO:2000808 | Negative regulation of synaptic vesicle clustering | ISS | biological_process |
GO:2001235 | Positive regulation of apoptotic signaling pathway | IEA | biological_process |
Entries Per Page
Displaying Page of
4. Expression levels in datasets
- Meta-analysis result
p-value up | p-value down | FDR up | FDR down |
---|---|---|---|
0.9364409521 | 0.1028444436 | 0.9999902473 | 0.6145636034 |
- Individual experiment result
( "-" represent NA in the specific microarray platform )
( "-" represent NA in the specific microarray platform )
Data source | Up or down | Log fold change |
---|---|---|
GSE11954 | Down | -0.3533409490 |
GSE13712_SHEAR | Down | -0.1818629570 |
GSE13712_STATIC | Down | -0.0751387870 |
GSE19018 | Up | 0.3068970661 |
GSE19899_A1 | Up | 0.0816024256 |
GSE19899_A2 | Down | -0.3898145693 |
PubMed_21979375_A1 | Down | -0.4549896670 |
PubMed_21979375_A2 | Down | -0.2228952510 |
GSE35957 | Down | -0.0604933483 |
GSE36640 | Up | 0.0021209995 |
GSE54402 | Down | -0.3217979059 |
GSE9593 | Down | -0.3791067601 |
GSE43922 | Down | -0.1819040012 |
GSE24585 | Down | -0.2274741276 |
GSE37065 | Down | -0.0826974624 |
GSE28863_A1 | Up | 0.2845922839 |
GSE28863_A2 | Down | -0.0432316668 |
GSE28863_A3 | Down | -0.1488636466 |
GSE28863_A4 | Up | 0.0682920488 |
GSE48662 | Up | 0.2235580291 |
5. Regulation relationships with compounds/drugs/microRNAs
- Compounds
Not regulated by compounds
- Drugs
Not regulated by drugs
- MicroRNAs
- mirTarBase
- mirTarBase
MiRNA_name | mirBase ID | miRTarBase ID | Experiment | Support type | References (Pubmed ID) |
---|---|---|---|---|---|
hsa-miR-17-5p | MIMAT0000070 | MIRT000499 | Luciferase reporter assay//Western blot | Functional MTI | 20227518 |
hsa-miR-17-5p | MIMAT0000070 | MIRT000499 | Luciferase reporter assay//qRT-PCR//Western blot | Non-Functional MTI | 20008935 |
hsa-miR-17-5p | MIMAT0000070 | MIRT000499 | Luciferase reporter assay | Functional MTI | 21283765 |
hsa-miR-17-5p | MIMAT0000070 | MIRT000499 | Luciferase reporter assay | Functional MTI | 23418359 |
hsa-miR-217 | MIMAT0000274 | MIRT000533 | Luciferase reporter assay//Western blot | Functional MTI | 20216554 |
hsa-miR-217 | MIMAT0000274 | MIRT000533 | Luciferase reporter assay | Functional MTI | 23471579 |
hsa-miR-216a-5p | MIMAT0000273 | MIRT000534 | Luciferase reporter assay//Western blot | Functional MTI | 20216554 |
hsa-miR-216a-5p | MIMAT0000273 | MIRT000534 | Luciferase reporter assay | Functional MTI | 23471579 |
hsa-miR-214-3p | MIMAT0000271 | MIRT000799 | Western blot//qRT-PCR//Luciferase reporter assay//Reporter assay;Other | Functional MTI | 18199536 |
hsa-miR-214-3p | MIMAT0000271 | MIRT000799 | Luciferase reporter assay//Microarray//qRT-PCR//Western blot | Functional MTI | 20548023 |
hsa-miR-214-3p | MIMAT0000271 | MIRT000799 | Luciferase reporter assay//qRT-PCR//Western blot | Functional MTI | 21228352 |
hsa-miR-26a-5p | MIMAT0000082 | MIRT001095 | Western blot//Luciferase reporter assay | Functional MTI | 19487573 |
hsa-miR-26a-5p | MIMAT0000082 | MIRT001095 | Luciferase reporter assay//Western blot | Functional MTI | 20216554 |
hsa-miR-26a-5p | MIMAT0000082 | MIRT001095 | GFP reporter assay//Luciferase reporter assay//Western blot | Functional MTI | 20080666 |
hsa-miR-21-5p | MIMAT0000076 | MIRT001190 | Western blot | Functional MTI | 19672202 |
hsa-miR-21-5p | MIMAT0000076 | MIRT001190 | Luciferase reporter assay | Functional MTI | 19072831 |
hsa-miR-21-5p | MIMAT0000076 | MIRT001190 | qRT-PCR//Western blot | Functional MTI | 19906824 |
hsa-miR-21-5p | MIMAT0000076 | MIRT001190 | Luciferase reporter assay//qRT-PCR//Western blot | Functional MTI | 18850008 |
hsa-miR-21-5p | MIMAT0000076 | MIRT001190 | Western blot | Functional MTI | 20216554 |
hsa-miR-21-5p | MIMAT0000076 | MIRT001190 | qRT-PCR//Luciferase reporter assay//Western blot | Functional MTI | 20223231 |
hsa-miR-21-5p | MIMAT0000076 | MIRT001190 | Microarray//immunohistochemistry | Functional MTI (Weak) | 19175831 |
hsa-miR-21-5p | MIMAT0000076 | MIRT001190 | Luciferase reporter assay//Western blot//Reporter assay;Western blot;Microarray | Functional MTI | 20048743 |
hsa-miR-21-5p | MIMAT0000076 | MIRT001190 | qRT-PCR//Western blot | Functional MTI | 20813833 |
hsa-miR-21-5p | MIMAT0000076 | MIRT001190 | GFP reporter assay//Northern blot//qRT-PCR//Western blot//ASO assay | Functional MTI | 19641183 |
hsa-miR-21-5p | MIMAT0000076 | MIRT001190 | Luciferase reporter assay | Functional MTI | 19253296 |
hsa-miR-21-5p | MIMAT0000076 | MIRT001190 | Immunocytochemistry//Luciferase reporter assay//Northern blot//qRT-PCR//Western blot//Microarray//Reporter assay | Functional MTI | 17681183 |
hsa-miR-21-5p | MIMAT0000076 | MIRT001190 | Northern blot//qRT-PCR//Western blot//Reporter assay | Functional MTI | 16762633 |
hsa-miR-21-5p | MIMAT0000076 | MIRT001190 | Luciferase reporter assay//qRT-PCR | Functional MTI | 20797623 |
hsa-miR-21-5p | MIMAT0000076 | MIRT001190 | Immunohistochemistry//qRT-PCR//Western blot | Functional MTI | 20978511 |
hsa-miR-21-5p | MIMAT0000076 | MIRT001190 | Luciferase reporter assay//Western blot | Functional MTI | 22267008 |
hsa-miR-21-5p | MIMAT0000076 | MIRT001190 | Immunohistochemistry | Non-Functional MTI (Weak) | 19473551 |
hsa-miR-21-5p | MIMAT0000076 | MIRT001190 | ELISA//Immunoblot//qRT-PCR | Functional MTI (Weak) | 22678116 |
hsa-miR-21-5p | MIMAT0000076 | MIRT001190 | Luciferase reporter assay | Functional MTI | 22956424 |
hsa-miR-21-5p | MIMAT0000076 | MIRT001190 | qRT-PCR//Western blot | Functional MTI | 22761812 |
hsa-miR-21-5p | MIMAT0000076 | MIRT001190 | Immunohistochemistry//Luciferase reporter assay//qRT-PCR//Western blot | Functional MTI | 22770403 |
hsa-miR-21-5p | MIMAT0000076 | MIRT001190 | Luciferase reporter assay//Western blot | Functional MTI | 22879939 |
hsa-miR-21-5p | MIMAT0000076 | MIRT001190 | Luciferase reporter assay | Functional MTI | 23226804 |
hsa-miR-21-5p | MIMAT0000076 | MIRT001190 | Western blot | Functional MTI | 21544242 |
hsa-miR-494-3p | MIMAT0002816 | MIRT001209 | qRT-PCR//Luciferase reporter assay//Western blot | Functional MTI | 20006626 |
hsa-miR-494-3p | MIMAT0002816 | MIRT001209 | Flow//Luciferase reporter assay//Microarray//qRT-PCR//Western blot | Functional MTI | 22544933 |
hsa-miR-519a-3p | MIMAT0002869 | MIRT006233 | Luciferase reporter assay//qRT-PCR//Western blot | Functional MTI | 22262409 |
hsa-miR-519d-3p | MIMAT0002853 | MIRT006196 | Luciferase reporter assay//Western blot | Functional MTI | 22262409 |
hsa-miR-29b-3p | MIMAT0000100 | MIRT006098 | Western blot;Microarray;Other | Functional MTI | 21359530 |
hsa-miR-19a-3p | MIMAT0000073 | MIRT002958 | qRT-PCR//Western blot | Functional MTI | 18460397 |
hsa-miR-19a-3p | MIMAT0000073 | MIRT002958 | Luciferase reporter assay//Reporter assay | Functional MTI | 14697198 |
hsa-miR-19a-3p | MIMAT0000073 | MIRT002958 | Luciferase reporter assay//qRT-PCR//Western blot | Functional MTI | 21853360 |
hsa-miR-141-3p | MIMAT0000432 | MIRT003281 | Luciferase reporter assay//Western blot//Reporter assay;Western blot;Other | Functional MTI | 20053927 |
hsa-miR-19b-3p | MIMAT0000074 | MIRT003371 | Luciferase reporter assay//qRT-PCR//Western blot | Functional MTI | 20008935 |
hsa-miR-18a-5p | MIMAT0000072 | MIRT003370 | Luciferase reporter assay//qRT-PCR//Western blot | Non-Functional MTI | 20008935 |
hsa-miR-20a-5p | MIMAT0000075 | MIRT003369 | Luciferase reporter assay//qRT-PCR//Western blot | Non-Functional MTI | 20008935 |
hsa-miR-20a-5p | MIMAT0000075 | MIRT003369 | Luciferase reporter assay | Functional MTI | 21283765 |
hsa-miR-221-3p | MIMAT0000278 | MIRT005585 | FACS//Flow//Luciferase reporter assay//Northern blot//Western blot | Functional MTI | 20618998 |
hsa-miR-221-3p | MIMAT0000278 | MIRT005585 | Flow//Immunohistochemistry//Luciferase reporter assay//qRT-PCR//Western blot | Functional MTI | 19962668 |
hsa-miR-221-3p | MIMAT0000278 | MIRT005585 | Luciferase reporter assay | Functional MTI | 23372675 |
hsa-miR-222-3p | MIMAT0000279 | MIRT005586 | FACS//Flow//Luciferase reporter assay//Northern blot//Western blot | Functional MTI | 20618998 |
hsa-miR-222-3p | MIMAT0000279 | MIRT005586 | Flow//Immunohistochemistry//Luciferase reporter assay//qRT-PCR//Western blot | Functional MTI | 19962668 |
hsa-miR-222-3p | MIMAT0000279 | MIRT005586 | Western blot | Functional MTI | 23028614 |
hsa-miR-106b-5p | MIMAT0000680 | MIRT005865 | Luciferase reporter assay | Functional MTI | 21283765 |
hsa-miR-93-5p | MIMAT0000093 | MIRT006216 | GFP reporter assay//qRT-PCR//Western blot | Functional MTI | 22465665 |
hsa-miR-519c-3p | MIMAT0002832 | MIRT006640 | Luciferase reporter assay//qRT-PCR//Western blot | Functional MTI | 22262409 |
hsa-miR-103a-3p | MIMAT0000101 | MIRT006693 | Western blot | Non-Functional MTI | 22593189 |
hsa-miR-107 | MIMAT0000104 | MIRT006694 | Western blot | Non-Functional MTI | 22593189 |
hsa-miR-23a-3p | MIMAT0000078 | MIRT006987 | Luciferase reporter assay//qRT-PCR//Western blot | Functional MTI | 23019365 |
hsa-miR-29a-3p | MIMAT0000086 | MIRT007028 | Luciferase reporter assay//Reporter assay | Functional MTI | 21573166 |
hsa-miR-29a-3p | MIMAT0000086 | MIRT007028 | Luciferase reporter assay | Functional MTI | 23426367 |
hsa-miR-23b-3p | MIMAT0000418 | MIRT007096 | Luciferase reporter assay | Functional MTI | 23189187 |
hsa-miR-144-3p | MIMAT0000436 | MIRT007190 | Immunoprecipitaion//Luciferase reporter assay//qRT-PCR//Western blot | Functional MTI | 23125220 |
hsa-miR-193b-3p | MIMAT0002819 | MIRT041436 | CLASH | Functional MTI (Weak) | 23622248 |
hsa-miR-181b-5p | MIMAT0000257 | MIRT047231 | CLASH | Functional MTI (Weak) | 23622248 |
Entries Per Page
Displaying Page of
- mirRecord
- mirRecord
MicroRNA name | mirBase ID | Target site number | MiRNA mature ID | Test method inter | MiRNA regulation site | Reporter target site | Pubmed ID |
---|---|---|---|---|---|---|---|
hsa-miR-19a-3p | MIMAT0000073 | 2 | hsa-miR-19a | 14697198 | |||
hsa-miR-19a-3p | MIMAT0000073 | 1 | hsa-miR-19a | 14697198 | |||
hsa-miR-19a-3p | MIMAT0000073 | 3 | hsa-miR-19a | 14697198 | |||
hsa-miR-214-3p | MIMAT0000271 | 1 | hsa-miR-214 | {Western blot}{Western blot} | {overexpression}{underexpression by 2'-O-Me antisense miRNA oligonucleotides} | 18199536 | |
hsa-miR-22-3p | MIMAT0000077 | NA | hsa-miR-22 | {Western blot} | {overexpression by miRNA precursor transfection} | 0 | |
hsa-miR-21-5p | MIMAT0000076 | NA | hsa-miR-21 | {Western blot} | {overexpression by miRNA precursor transfection} | 19906824 | |
hsa-miR-222-3p | MIMAT0000279 | NA | hsa-miR-222 | {Western blot} | {downregulation by anti-miRNA} | 20618998 | |
hsa-miR-221-3p | MIMAT0000278 | NA | hsa-miR-221 | {Western blot} | {downregulation by anti-miRNA} | 20618998 | |
hsa-miR-29a-3p | MIMAT0000086 | 1 | hsa-miR-29a | {Western blot} | {overexpression by miRNA precursor transfection} | 21573166 | |
hsa-miR-29a-3p | MIMAT0000086 | 2 | hsa-miR-29a | {Western blot} | {overexpression by miRNA precursor transfection} | 21573166 |
Entries Per Page
Displaying Page of
6. Text-mining results about the gene
Gene occurances in abstracts of cellular senescence-associated articles: 43 abstracts the gene occurs.
PubMed ID of the article | Sentenece the gene occurs |
---|---|
28003865 | These data indicate that the signal pathway to ROS generation in replicative aged skin cells can be stimulated by reduced PTEN level |
27009837 | These results suggest that Ad-Bmi-1i not only inhibits tumor growth and stem cell-like phenotype by inducing cellular senescence directly, but also has an indirect anti-tumor activity by anti-angiogenesis effects via regulating PTEN/AKT/VEGF pathway |
26686419 | We previously showed that CSIG plays an important role in regulating cell proliferation and cellular senescence progression through inhibiting PTEN, however, which domain or region of CSIG contributes to this function |
26686419 | The data showed that expression of CSIG potently reduced PTEN expression, increased cell proliferation rates, and reduced the senescent phenotype (lower SA-beta-gal activity) |
26686419 | By contrast, neither the expression of CSIG N- terminal (NT) fragment containing the ribosomal L1 domain nor C-terminal (CT) fragment containing Lys-rich region could significantly altered the levels of PTEN; instead of promoting cell proliferation and delaying cellular senescence, expression of CSIG-NT or CSIG-CT inhibited cell proliferation and accelerated cell senescence (increased SA-beta-gal activity) compared to either CSIG over-expressing or control (empty vector transfected) cells |
26655726 | The PTEN tumor suppressor gene and its role in lymphoma pathogenesis |
26655726 | The phosphatase and tensin homolog gene PTEN is one of the most frequently mutated tumor suppressor genes in human cancer |
26655726 | Loss of PTEN function occurs in a variety of human cancers via its mutation, deletion, transcriptional silencing, or protein instability |
26655726 | PTEN deficiency in cancer has been associated with advanced disease, chemotherapy resistance, and poor survival |
26655726 | In addition to having lipid phosphorylation activity, PTEN has critical roles in the regulation of genomic instability, DNA repair, stem cell self-renewal, cellular senescence, and cell migration |
26655726 | Although PTEN deficiency in solid tumors has been studied extensively, rare studies have investigated PTEN alteration in lymphoid malignancies |
26655726 | However, genomic or epigenomic aberrations of PTEN and dysregulated signaling are likely critical in lymphoma pathogenesis and progression |
26655726 | This review provides updated summary on the role of PTEN deficiency in human cancers, specifically in lymphoid malignancies; the molecular mechanisms of PTEN regulation; and the distinct functions of nuclear PTEN |
26655726 | Therapeutic strategies for rescuing PTEN deficiency in human cancers are proposed |
26511486 | Given that human prostate cancer arises from precancerous lesions such as high-grade prostatic intraepithelial neoplasia (HG-PIN), which frequently have lost phosphatase and tensin homolog (PTEN) tumor suppressor permitting phosphatidylinositol-3-OH kinase (PI3K)-protein kinase B (AKT) oncogenic signaling, we tested the efficacy of MSeA to inhibit HG-PIN progression in Pten prostate-specific knockout (KO) mice and assessed the mechanistic involvement of p53-mediated cellular senescence and of the androgen receptor (AR) |
26511486 | We observed that short-term (4 weeks) oral MSeA treatment significantly increased expression of P53 and P21Cip1 proteins and senescence-associated-beta-galactosidase staining, and reduced Ki67 cell proliferation index in Pten KO prostate epithelium |
26511486 | Long-term (25 weeks) MSeA administration significantly suppressed HG-PIN phenotype, tumor weight, and prevented emergence of invasive carcinoma in Pten KO mice |
26511486 | Mechanistically, the long-term MSeA treatment not only sustained P53-mediated senescence, but also markedly reduced AKT phosphorylation and AR abundance in the Pten KO prostate |
26477312 | NFATc1 promotes prostate tumorigenesis and overcomes PTEN loss-induced senescence |
26477312 | To further characterize interactions between genes involved in prostate tumorigenesis, we generated mice with both NFATc1 activation and Pten inactivation in prostate |
26477312 | We showed that NFATc1 activation led to acceleration of Pten null-driven prostate tumorigenesis by overcoming the PTEN loss-induced cellular senescence through inhibition of p21 activation |
26085373 | Mechanistically, we show that Pten loss increases CK2 levels by activating STAT3 |
26085373 | CK2 upregulation in Pten null tumours affects the stability of Pml, an essential regulator of senescence |
26085373 | However, CK2 inhibition stabilizes Pml levels enhancing senescence in Pten null tumours |
25328137 | Despite NR2E1 regulating targets like p21(CIP1) or PTEN we still lack a full explanation for its role in NSC self-renewal and tumorigenesis |
25156255 | Here we show that at the onset of senescence, PTEN null prostate tumours in mice are massively infiltrated by a population of CD11b(+)Gr-1(+) myeloid cells that protect a fraction of proliferating tumour cells from senescence, thus sustaining tumour growth |
24866151 | Loss of the PTEN and TP53 tumor suppressor genes is commonly observed in prostate cancer, whereas their compound loss is often observed in advanced prostate cancer |
24866151 | Surprisingly, we also find that PARP-induced cellular senescence is morphed into an apoptotic response upon compound loss of PTEN and p53 |
24782600 | Activation of egr-1, in turn, upregulates the dual specificity phosphatase, phosphatase and tensin homologue deleted on chromosome ten (PTEN) resulting in activation of pro-apoptotic caspase-3 and caspase-9 and reduced expression of the anti-apoptosis protein, survivin |
24782600 | This paradigm has human relevance since increased expression of PTEN and reduced expression of survivin were demonstrated in gastric mucosa of aging individuals |
24704020 | Prognostic significance of biallelic loss of PTEN in clear cell renal cell carcinoma |
24704020 | PURPOSE: We investigated the clinical implications of biallelic loss of PTEN in clear cell renal cell carcinoma and whether PTEN biallelic loss would induce p53 dependent cellular senescence |
24704020 | PTEN allelic status was classified into 3 groups, including biallelic PTEN loss (homozygous deletion or combined heterozygous deletion and mutation), monoallelic PTEN loss (heterozygous deletion or mutation) and absent allelic loss |
24704020 | 6%) had biallelic PTEN loss and 69 (16 |
24704020 | PTEN allelic loss was associated with late tumor stage and high histological grade |
24704020 | About half of the patients with PTEN biallelic loss had accompanying TP53 allelic loss |
24704020 | Biallelic loss of PTEN did not increase the expression of genes related to p53 dependent cellular senescence |
24704020 | CONCLUSIONS: PTEN biallelic loss may be a prognostic marker for clear cell renal cell carcinoma |
24270409 | Although such 'escape' from senescence is not sufficient to promote thyroid tumorigenesis in adult mice up to 5 months, the onset of Phosphatase and tensin homolog (Pten)-induced tumor formation is accelerated when Spry1 is concomitantly eliminated |
24074787 | The timing of TP53 mutation also depends on the tumor subtype, being the first important event in luminal tumors but occurring after PTEN loss in basal-like tumors |
24052415 | The expression of p16 and PTEN do not seem to cause synergism of senescence in the benign lesions analyzed in p21p27 double-KO mice |
23936028 | Using the conditional PTEN deletion mouse model, we previously reported that survivin levels increase with prostate tumor growth |
23936028 | We then serially, from about 10-56 weeks of age, evaluated histopathologic changes in the prostate of mice with PTEN deletion combined with survivin mono- or bi-allelic gene deletion |
23936028 | A reduced proliferation index as well as apoptotic and senescent cells were detected in the lesions of mice with compound PTEN/survivin deficiency throughout the time points examined |
23904845 | No pathogenetic mutations in CDKN2A, BRAF, NRAS, KRAS, cKIT, TP53 and PTEN genes were observed |
23826727 | In this model, PTEN loss prevents the decline in proliferation capacity in aged beta-cells and restores the ability of aged beta-cells to respond to injury-induced regeneration |
23826727 | Using several animal and cell models where we can manipulate PTEN expression, we found that PTEN blocks cell cycle re-entry through a novel pathway leading to an increase in p16(ink4a), a cell cycle inhibitor characterized for its role in cellular senescence/aging |
23826727 | A downregulation in p16(ink4a) occurs when PTEN is lost as a result of cyclin D1 induction and the activation of E2F transcription factors |
23727861 | Prostate-specific inactivation of Zbtb7a leads to a marked acceleration of Pten loss-driven prostate tumorigenesis through bypass of Pten loss-induced cellular senescence (PICS) |
23535008 | Therefore, we investigated the role of gene silencing (DNA promoter methylation of LINE-1, PTEN), genetic aberrations (karyotype, KRAS and BRAF mutations) as well as their contribution to the proliferation rate and migratory potential that underlies "initial" and "final" passage sarcoma cells |
23535008 | Increased proliferative potential of final passage STS cells was not associated with significant differences in methylation (LINE-1, PTEN) and mutation status (KRAS, BRAF), but it was dependent on the amount of chromosomal aberrations |
22836754 | B-Raf activation cooperates with PTEN loss to drive c-Myc expression in advanced prostate cancer |
22652801 | Oncogene induced CS can be promoted by the loss of tumor suppressor genes, such as PTEN |
22253608 | Cytoplasmic polyadenylation element binding protein deficiency stimulates PTEN and Stat3 mRNA translation and induces hepatic insulin resistance |
22253608 | An investigation of Cpeb1 knockout mice revealed that the expression of two particular negative regulators of insulin action, PTEN and Stat3, were aberrantly increased |
22253608 | Analysis of HepG2 cells, a human liver cell line, depleted of CPEB demonstrated that this protein directly regulates the translation of PTEN and Stat3 mRNAs |
22120720 | This phenomenon was not restricted to breast cancer cells, as it was also seen in glioblastoma cells in which PKCiota is activated by loss of PTEN |
22037217 | Loss of TGF-beta signaling and PTEN promotes head and neck squamous cell carcinoma through cellular senescence evasion and cancer-related inflammation |
21930937 | HER2 overcomes PTEN (loss)-induced senescence to cause aggressive prostate cancer |
21930937 | In this study, we show that patients who develop prostate tumors with low levels of PTEN and high levels of HER2/3 have a poor prognosis |
21930937 | This is functionally relevant, as targeting Her2 activation to the murine prostate cooperates with Pten loss and drives CaP progression |
21930937 | Taken together, these data suggest that stratification of CaP patients for HER2/3 and PTEN status could identify patients with aggressive CaP who may respond favorably to MEK inhibition |
21909130 | Furthermore, our data imply that chronic activation of AKT signalling provides selective pressure for the loss of p53 function, consistent with observations that PTEN or PIK3CA mutations are significantly associated with p53 mutation in a number of human tumour types |
21695255 | Mammalian target of rapamycin is a therapeutic target for murine ovarian endometrioid adenocarcinomas with dysregulated Wnt/beta-catenin and PTEN |
21695255 | However, the role of WNT/beta-catenin and PTEN/AKT signaling in the etiology and/or progression of this disease is currently unclear |
21695255 | Combining dysregulated beta-catenin with homozygous deletion of PTEN in the OSE resulted in development of significantly more aggressive tumors, which was correlated with inhibition of p53 expression and cellular senescence |
21695255 | Ectopic allotransplants of the mouse ovarian tumor cells with a gain-of-function mutation in beta-catenin and PTEN deletion developed into tumors with OEA histology, the growth of which were significantly inhibited by oral rapamycin treatment |
21695255 | These studies demonstrate that rapamycin might be an effective therapeutic for human ovarian endometrioid patients with dysregulated Wnt/beta-catenin and Pten/PI3K signaling |
21594579 | Induction of cellular senescence by oncogenic insults, such as Ras overexpression or by inactivation of PTEN tumor suppressor, triggers an ARF/p53-dependent tumor-suppressive effect which can significantly restrict cancer progression |
21286718 | The combination could also inhibit the expression of Cyclin D1 and phosphorylated mTOR while had no impact on p53, p16, PTEN, and HIF-1alpha |
21241890 | Nuclear PTEN regulates the APC-CDH1 tumor-suppressive complex in a phosphatase-independent manner |
21241890 | Recently, nuclear compartmentalization of PTEN was found as a key component of its tumor-suppressive activity; however its nuclear function remains poorly defined |
21241890 | Here we show that nuclear PTEN interacts with APC/C, promotes APC/C association with CDH1, and thereby enhances the tumor-suppressive activity of the APC-CDH1 complex |
21241890 | We find that nuclear exclusion but not phosphatase inactivation of PTEN impairs APC-CDH1 |
21241890 | This nuclear function of PTEN provides a straightforward mechanistic explanation for the fail-safe cellular senescence response elicited by acute PTEN loss and the tumor-suppressive activity of catalytically inactive PTEN |
21241890 | Importantly, we demonstrate that PTEN mutant and PTEN null states are not synonymous as they are differentially sensitive to pharmacological inhibition of APC-CDH1 targets such as PLK1 and Aurora kinases |
20938386 | Total RNA isolated from these samples was used to measure the gene expression of p16INK4a, RB, cyclin D1, CDK4, PTEN, p27KIP, p19ARF, p21, TERT, and RAGE by real-time polymerase chain reaction assay |
20938386 | The mRNA levels of p16INK4a, RB, PTEN, p27KIP, p19ARF, and RAGE were upregulated |
20622047 | In addition to the canonical function of dephosphorylation of phosphatidylinositol-3,4,5-trisphosphate (PIP3), recent studies showed the intriguing roles of PTEN in regulating genomic instability, DNA repair, stem cell self-renewal, cellular senescence, and cell migration and/or metastasis |
20622047 | Clinically, PTEN mutations and deficiencies are prevalent in many types of human cancers |
20622047 | Severe PTEN deficiency is also associated with advanced tumor stage and therapeutic resistance, such as the resistance to trastuzumab, an anti-HER2 therapy |
20622047 | In this review, we highlight our current knowledge of PTEN function and the recent discoveries in dissecting the PTEN signaling pathway |
20197621 | We previously demonstrated in a mouse model of prostate cancer that inactivation of the tumor suppressor phosphatase and tensin homolog deleted on chromosome 10 (Pten) elicits a senescence response that opposes tumorigenesis |
20197621 | Using mouse embryonic fibroblasts, we determined that PICS occurs rapidly after Pten inactivation, in the absence of cellular proliferation and DDR |
20197621 | Importantly, we demonstrated that pharmacological inhibition of PTEN drives senescence and inhibits tumorigenesis in vivo in a human xenograft model of prostate cancer |
20174572 | Elevated expression of AKT has been noted in a significant percentage of primary human breast cancers, mainly as a consequence of the PTEN/PI3K pathway deregulation |
19690330 | Acute loss of Pten leads to an increase in the abundance of p19(Arf), p53, and p21 proteins as part of a fail-safe senescence response |
19690330 | In both prostate epithelium and primary mouse embryo fibroblasts (MEFs), the increase in p53 protein abundance found upon loss of Pten was unaffected by the simultaneous loss of p19(Arf) |
19690330 | Consistent with the effect of p19(Arf) loss in Pten-deficient mouse prostate, we found that in human prostate cancers, loss of PTEN was not associated with loss of p14(ARF) (the human equivalent of mouse p19(Arf)) |
19690330 | Collectively, these data reveal differential consequences of p19(Arf) inactivation in prostate cancer and MEFs upon Pten loss that are independent of the p53 pathway |
19647222 | We report that knocking down the expression of inositol polyphosphate 4-phosphatase type II (INPP4B) in human epithelial cells, like knockdown of PTEN, resulted in enhanced Akt activation and anchorage-independent growth and enhanced overall motility |
19647222 | Dual knockdown of INPP4B and PTEN resulted in cellular senescence |
18765664 | S1P-induced Akt and ERK1/2 activation were comparable between ECs of different in vitro ages; however, PTEN (phosphatase and tensin homolog deleted on chromosome 10) activity was significantly elevated and Rac activation was inhibited in senescent ECs |
18765664 | Rac activation and senescent-associated impairments were restored in senescent ECs by the expression of dominant-negative PTEN and by knocking down S1P(2) receptors |
18765664 | These results indicate that the impairment of function in senescent ECs in culture is mediated by an increase in S1P signaling through S1P(2)-mediated activation of the lipid phosphatase PTEN |
18678645 | CSIG inhibits PTEN translation in replicative senescence |
18678645 | Instead, CSIG negatively regulated PTEN and p27(Kip1) expressions, in turn promoting cell proliferation |
18678645 | In PTEN-silenced HEK 293 cells and PTEN-deficient human glioblastoma U87MG cells, the effect of CSIG on p27(Kip1) expression and cell division was abolished, suggesting that PTEN was required for the role of CSIG on p27(Kip1) regulation and cell cycle progression |
18678645 | Investigation into the underlying mechanism revealed that the regulation of PTEN by CSIG was achieved through a translational suppression mechanism |
18678645 | Knockdown of PTEN diminished the effect of CSIG on cellular senescence |
18678645 | Our findings indicate that CSIG acts as a novel regulatory component of replicative senescence, which requires PTEN as a mediator and involves in a translational regulatory mechanism |
18577387 | We propose a stochastic model of p53 regulation, which is based on two feedback loops: the negative, coupling p53 with its immediate downregulator Mdm2, and the positive, which involves PTEN, PIP3 and Akt |
18353141 | Loci with established importance in melanoma, like CDKN2A, BRAF and PTEN, have been joined by some less familiar genes including transcription factor sequences TBX2 and STK11 (LKB) |
16079851 | The PTEN and p53 tumour suppressors are among the most commonly inactivated or mutated genes in human cancer including prostate cancer |
16079851 | Although they are functionally distinct, reciprocal cooperation has been proposed, as PTEN is thought to regulate p53 stability, and p53 to enhance PTEN transcription |
16079851 | Here we show that conditional inactivation of Trp53 in the mouse prostate fails to produce a tumour phenotype, whereas complete Pten inactivation in the prostate triggers non-lethal invasive prostate cancer after long latency |
16079851 | Strikingly, combined inactivation of Pten and Trp53 elicits invasive prostate cancer as early as 2 weeks after puberty and is invariably lethal by 7 months of age |
16079851 | Importantly, acute Pten inactivation induces growth arrest through the p53-dependent cellular senescence pathway both in vitro and in vivo, which can be fully rescued by combined loss of Trp53 |
10832053 | In this review, we describe three senescence-inducing pathways involving these inhibitors, namely, the p16(INK4a)/Rb pathway, the p19(ARF)/p53/p21(Cip1) pathway, and the PTEN/p27(Kip1) pathway |
Entries Per Page
Displaying Page of