scholarly journals Regulation of p53-MDMX Interaction by Casein Kinase 1 Alpha

2005 ◽  
Vol 25 (15) ◽  
pp. 6509-6520 ◽  
Author(s):  
Lihong Chen ◽  
Changgong Li ◽  
Yu Pan ◽  
Jiandong Chen

ABSTRACT MDMX is a homolog of MDM2 that is critical for regulating p53 function during mouse development. MDMX degradation is regulated by MDM2-mediated ubiquitination. Whether there are other mechanisms of MDMX regulation is largely unknown. We found that MDMX binds to the casein kinase 1 alpha isoform (CK1α) and is phosphorylated by CK1α. Expression of CK1α stimulates the ability of MDMX to bind to p53 and inhibit p53 transcriptional function. Regulation of MDMX-p53 interaction requires CK1α binding to the central region of MDMX and phosphorylation of MDMX on serine 289. Inhibition of CK1α expression by isoform-specific small interfering RNA (siRNA) activates p53 and further enhances p53 activity after ionizing irradiation. CK1α siRNA also cooperates with DNA damage to induce apoptosis. These results suggest that CK1α is a functionally relevant MDMX-binding protein and plays an important role in regulating p53 activity in the absence or presence of stress.

2011 ◽  
Vol 22 (16) ◽  
pp. 2834-2847 ◽  
Author(s):  
Zheng Meng ◽  
Luisa Capalbo ◽  
David M. Glover ◽  
William G. Dunphy

The mediator protein Claspin is critical for the activation of the checkpoint kinase Chk1 during checkpoint responses to stalled replication forks. This function involves the Chk1-activating domain (CKAD) of Claspin, which undergoes phosphorylation on multiple conserved sites. These phosphorylations promote binding of Chk1 to Claspin and ensuing activation of Chk1 by ATR. However, despite the importance of this regulatory process, the kinase responsible for these phosphorylations has remained unknown. By using a multifaceted approach, we have found that casein kinase 1 gamma 1 (CK1γ1) carries out this function. CK1γ1 phosphorylates the CKAD of Claspin efficiently in vitro, and depletion of CK1γ1 from human cells by small interfering RNA (siRNA) results in dramatically diminished phosphorylation of Claspin. Consequently, the siRNA-treated cells display impaired activation of Chk1 and resultant checkpoint defects. These results indicate that CK1γ1 is a novel component of checkpoint responses that controls the interaction of a key checkpoint effector kinase with its cognate mediator protein.


RSC Advances ◽  
2019 ◽  
Vol 9 (56) ◽  
pp. 32781-32781
Author(s):  
Ye Cao ◽  
Jiajia Zheng ◽  
Chentao Lv

Correction for ‘miR-199a-3p knockdown inhibits dedifferentiated liposarcoma (DDLPS) cell viability and enhances apoptosis through targeting casein kinase-1 alpha (CK1α)’ by Ye Cao et al., RSC Adv., 2019, 9, 22755–22763.


Author(s):  
Morvarid Siri ◽  
Hamid Behrouj ◽  
Sanaz Dastghaib ◽  
Mozhdeh Zamani ◽  
Wirginia Likus ◽  
...  

AbstractAdjuvant chemotherapy with 5-fluorouracil (5-FU) does not improve survival of patients suffering from a form of colorectal cancer (CRC) characterized by high level of microsatellite instability (MSI-H). Given the importance of autophagy and multi-drug-resistant (MDR) proteins in chemotherapy resistance, as well as the role of casein kinase 1-alpha (CK1α) in the regulation of autophagy, we tested the combined effect of 5-FU and CK1α inhibitor (D4476) on HCT116 cells as a model of MSI-H colorectal cancer. To achieve this goal, the gene expression of Beclin1 and MDR genes, ABCG2 and ABCC3 were analyzed using quantitative real-time polymerase chain reaction. We used immunoblotting to measure autophagy flux (LC3, p62) and flow cytometry to detect apoptosis. Our findings showed that combination treatment with 5-FU and D4476 inhibited autophagy flux. Moreover, 5-FU and D4476 combination therapy induced G2, S and G1 phase arrests and it depleted mRNA of both cell proliferation-related genes and MDR-related genes (ABCG2, cyclin D1 and c-myc). Hence, our data indicates that targeting of CK1α may increase the sensitivity of HCT116 cells to 5-FU. To our knowledge, this is the first description of sensitization of CRC cells to 5-FU chemotherapy by CK1α inhibitor. Graphic abstract


Nature ◽  
2009 ◽  
Vol 459 (7244) ◽  
pp. 274-277 ◽  
Author(s):  
Heng-Chi Lee ◽  
Shwu-Shin Chang ◽  
Swati Choudhary ◽  
Antti P. Aalto ◽  
Mekhala Maiti ◽  
...  

PLoS ONE ◽  
2017 ◽  
Vol 12 (1) ◽  
pp. e0170903 ◽  
Author(s):  
Yoshimi Endo Greer ◽  
Bo Gao ◽  
Yingzi Yang ◽  
Andre Nussenzweig ◽  
Jeffrey S. Rubin

Author(s):  
Hamid Behrouj ◽  
Atefeh Seghatoleslam ◽  
Pooneh Mokarram ◽  
Saeid Ghavami

The Wnt/β-catenin pathway, which interferes with cell proliferation, differentiation, and autophagy, is commonly dysregulated in colorectal cancer (CRC). Mutation of the RAS oncogene is the most prevalent genetic alteration in CRC and has been linked to activation of protein kinase B (AKT) signaling. Phosphorylation of β-catenin at Ser 552 by AKT contributes to β-catenin stability, transcriptional activity, and increase of cell proliferation. Casein kinase 1 alpha (CK1α) is an enzyme that simultaneously regulates Wnt/β-catenin and AKT. The link of the AKT and Wnt pathway to autophagy in RAS-mutated CRC cells has not well identified. Therefore, we investigated how pharmacological CK1α inhibition (D4476) is involved in regulation of autophagy, Wnt/β-catenin, and AKT pathways in RAS-mutated CRC cell lines. qRT-PCR and immunoblotting experiments revealed that phospho-AKT (S473) and phospho-β-catenin (S552) are constitutively increased in RAS-mutated CRC cell lines, in parallel with augmented CK1α expression. The results also showed that D4476 significantly reduced the AKT/phospho-β-catenin (S552) axis concomitantly with autophagy flux inhibition in RAS-mutated CRC cells. Furthermore, D4476 significantly induced apoptosis in RAS-mutated CRC cells. In conclusion, our results indicate that CK1α inhibition reduces autophagy flux and promotes apoptosis by interfering with the AKT/phospho-β-catenin (S552) axis in RAS-mutated CRC cells.


Sign in / Sign up

Export Citation Format

Share Document