« Home « Kết quả tìm kiếm

Novel chemotherapeutic agent FX-9 activates NF-κB signaling and induces G1 phase arrest by activating CDKN1A in a human prostate cancer cell line


Tóm tắt Xem thử

- The 6-h samples showed activation of the cell cycle inhibitor CDKN1A which might be involved in the secondary response in 12-h samples..
- This secondary response predominantly consisted of cell cycle-related changes, with further activation of CDKN1A and inhibition of the transcription factor E2F1 , including downstream target genes, resulting in G1-phase arrest..
- Conclusions: FX-9 induced G1-phase arrest of PC-3 cells through activation of the cell cycle inhibitor CDKN1A , which was initiated by an inflammatory response of noncanonical NF- κ B signaling..
- To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/..
- The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data..
- Antimitotic drugs disrupt the cell cycle progression of tumor cells.
- The purpose of the cell cycle is to du- plicate the DNA accurately and to segregate the cop- ies into two identical daughter cells.
- In brief, the cell cycle is divided into G1-phase, S-phase, G2-phase and M-phase.
- If a checkpoint is activated, for example, due to DNA damage, the cell cycle is arrested until either DNA repair or programmed cell death occurs [10].
- It develops in 10–20% of patients with prostate cancer where androgen deprivation therapy has failed [17].
- FX-9 induced Reactome-pathways of the immune system after 6-h exposure and in cell cycle pathways after 12-h exposure.
- Two signaling pathways of the super- pathway “ cell cycle ” were enriched by upregulation of CDKN1A (FC 3.88, Fig.
- The pathway “ TP53 regulates transcription of genes involved in G1 cell cycle arrest ” belongs to the superpathway gene expression, describes the involvement of genes in G1-phase cell cycle arrest and was enriched by activation of E2F7 (FC 2.56).
- Seven signaling pathways assigned to the superpathway “cell cycle” were enriched by DEGs..
- The two pathways be- longing to the superpathway “gene expression” are also involved in the cell cycle.
- The signaling pathways “DNA replication” and “tran- scriptional regulation of granulopoiesis” were enriched by DEGs similar to those in the superpathway “cell cycle”.
- In the superpathway “RNA metabolism”, the sig- naling pathway “Insulin-like Growth Factor-2 mRNA Binding Proteins (IGF2BPs/IMPs/VICKZs) bind RNA”.
- FX-9 induced time-dependent differences in the biological processes of inflammatory response and G1/S transition of mitotic cell cycle.
- The biological processes “G1/S transition of mitotic cell cycle” and “positive regulation of endothelial cell proliferation” were only enriched in the 12-h.
- “G1/S transition of mi- totic cell cycle” was enriched with an FDR-value of 0.0006 by nine genes, including CDC6, CDT1, CDKN1A, CCNE2, MCM3, and MCM6..
- 6 GE TP53 Regulates Transcription of Genes Involved in G1 Cell Cycle Arrest .
- 9 CC Transcriptional activation of cell cycle inhibitor p .
- CC “cell cycle”, ST “signal transduction”, and CR “cellular response to external stimuli”.
- and “ Cell Cycle ” after 6-h and 12-h FX-9 exposure.
- Due to the enrichment of DEGs in cell cycle signaling pathways from 12-h exposure, this time point was chosen to verify the transcriptome data of CDKN1A, E2F1, SERPINE1, and NFKB2 at protein level by im- munocytochemistry.
- CDKN1A is a cell cycle inhibi- tor, E2F1 is an important transcription factor for entry in the S-phase, SERPINE1 plays a role in senescence, and NFKB2 is a marker for DNA damage [23–26].
- As CDKN1A, E2F1 and NFKB2 are assigned to the signal- ing pathways “immune system” or “cell cycle”, their gene expressions were visualized in Fig.
- 1 GE TP53 Regulates Transcription of Genes Involved in G1 Cell Cycle Arrest e-09.
- 5 GE TP53 Regulates Transcription of Cell Cycle Genes e-06.
- 6 CC Cell Cycle e-06.
- 7 CC Cell Cycle Checkpoints e-06.
- CC “ cell cycle.
- positive regulation of angiogenesis 10 4.80E-04 9 8.40E-04.
- extracellular matrix organization 10 1.40E-02 10 4.80E-03.
- G1/S transition of mitotic cell cycle 9 6.00E-04.
- the 12-h exposure, cell cycle disturbance.
- Interestingly, the potent cell cycle inhibitor CDKN1A and transcrip- tion factor E2F7 were activated in the 6-h samples, po- tentially causing the cell cycle-related response in the 12-h samples..
- After 12-h FX-9 exposure, signaling pathways of cell cycle regulation were enriched by DEGs.
- FX-9 exposure increases the cell cycle inhibitor CDKN1A, which en- codes protein p21, providing multiple functions, for ex- ample, cell cycle arrest in response to drug induced DNA-damage [35].
- Inhi- biting these can provide a G1 cell cycle arrest and suppression of DNA replication [45, 46].
- Tumor cells could escape from cell cycle arrest and re-enter the cell cycle (mitotic slippage), usually resulting in those senescent cells or cell death in the G1-phase [53].
- Specifically, the enrichment of cell cycle associated DEGs regulating the transition from G1-phase to S-phase supports the thesis of an FX-9 in- duced G1-phase arrest.
- Described effects of FX-9 like in- duction of cell cycle inhibitor CDKN1A, activation of NF-κB signaling pathway and senescence can be caused by DNA damage .
- org/10.1186/s y..
- https://doi..
- org/10.1002/adsc.201600169..
- https://doi.org/10.1016/j.bmcl .
- https://doi.org/10.1016/S .
- https://doi.org/1 0.1016/j.ejmech .
- https://doi.org/10.1093/carcin/bgt133..
- https://doi.org/10.1016/j.ejmech .
- https://doi.org/10.4062/biomolther.2018.199..
- org/10.3390/ijms20225567..
- https://doi.org/10.1038/nm0103-140..
- Regulation of DNA repair throughout the cell cycle.
- https://doi.org/10.1038/nrm2351..
- Tumor cell-specific cytotoxicity by targeting cell cycle checkpoints.
- https://doi.org/10.1096/fj.02-1003fje..
- Cell-cycle therapeutics come of age.
- https://doi.org/10.1200/JCO .
- https://doi.org .
- https://doi.org P..
- https://doi.org/10.3390/cancers11091220..
- Biology of the cell cycle inhibitor p21 CDKN1A: molecular mechanisms and relevance in chemical toxicology.
- https://doi.org/10.1007/s .
- https://doi.org/10.1111/j x..
- https://doi.org/10.1093/nar/gkz1031..
- https://doi.org/10.1093/bioinformatics/.
- https://doi.org/10.1016/j.rvsc .
- Cell Cycle.
- https://doi.org/10.41 61/cc.5.23.3510..
- https://doi.org/10.1016/j.bcp .
- Emerging roles of E2Fs in cancer: an exit from cell cycle control.
- https://doi.org/10.103 8/nrc2696..
- https://doi.org/10.1016/j.dnarep .
- https://doi.org/10.1016/j.cell .
- https://doi.org/10.1038/nri.2017.52..
- https://doi.org/10.1073/pnas.95.7.3792..
- https://doi.org/10.1038/cdd.2017.46..
- https://doi.org/10.1038/ni0302-221..
- https://doi.org/10.1 002/jcb.20477..
- Multiple roles of the cell cycle inhibitor p21CDKN1A in the DNA damage response.
- https://doi.org/10.1016/j.mrrev .
- https://doi.org/10.1101/gad .
- https://doi.org/10.11 01/gad .
- https://doi.org/10.1371/journal.pgen.1003726..
- Inhibition of E2F1 activity and cell cycle progression by arsenic via retinoblastoma protein.
- regulation of cell growth- dependent expression of mammalian CDC6 gene by the cell cycle transcription factor E2F.
- https://doi.org/10.1 038/sj.onc.1202105..
- https://doi.org/10.103 8/sj.onc.1207488..
- https://doi.org/10.1093/nar/30.8.1859..
- https://doi.org/10.1074/jbc.M311933200..
- https://doi.org/10.1593/neo.81048..
- org/10.3389/fonc .
- https://doi.org/10.1128/mcb .
- https://doi.org/10.1128/MCB.1 8.2.753..
- https://doi.org/10.103 8/nrm2233..
- https://doi.org/10.1038/ncb1448..
- https://doi.org/1 0.1111/acel.12875..
- https://doi.org/10.1 530/ERC-17-0147.

Xem thử không khả dụng, vui lòng xem tại trang nguồn
hoặc xem Tóm tắt