scholarly journals ATM, ATR and DNA-PK: initiators of the cellular genotoxic stress responses

2003 ◽  
Vol 24 (10) ◽  
pp. 1571-1580 ◽  
Author(s):  
J. Yang
2017 ◽  
Vol 37 (13) ◽  
Author(s):  
K. Audrey Audetat ◽  
Matthew D. Galbraith ◽  
Aaron T. Odell ◽  
Thomas Lee ◽  
Ahwan Pandey ◽  
...  

ABSTRACT The human Mediator complex regulates RNA polymerase II transcription genome-wide. A general factor that regulates Mediator function is the four-subunit kinase module, which contains either cyclin-dependent kinase 8 (CDK8) or CDK19. Whereas CDK8 is linked to specific signaling cascades and oncogenesis, the cellular roles of its paralog, CDK19, are poorly studied. We discovered that osteosarcoma cells (SJSA) are naturally depleted of CDK8 protein. Whereas stable CDK19 knockdown was tolerated in SJSA cells, proliferation was reduced. Notably, proliferation defects were rescued upon the reexpression of wild-type or kinase-dead CDK19. Comparative RNA sequencing analyses showed reduced expression of mitotic genes and activation of genes associated with cholesterol metabolism and the p53 pathway in CDK19 knockdown cells. SJSA cells treated with 5-fluorouracil, which induces metabolic and genotoxic stress and activates p53, further implicated CDK19 in p53 target gene expression. To better probe the p53 response, SJSA cells (shCDK19 versus shCTRL) were treated with the p53 activator nutlin-3. Remarkably, CDK19 was required for SJSA cells to return to a proliferative state after nutlin-3 treatment, and this effect was kinase independent. These results implicate CDK19 as a regulator of p53 stress responses and suggest a role for CDK19 in cellular resistance to nutlin-3.


2010 ◽  
Vol 1800 (12) ◽  
pp. 1231-1240 ◽  
Author(s):  
Magdalena J. Laska ◽  
Ulla B. Vogel ◽  
Uffe B. Jensen ◽  
Bjørn A. Nexø

Oncogene ◽  
1999 ◽  
Vol 18 (24) ◽  
pp. 3666-3672 ◽  
Author(s):  
Sally A Amundson ◽  
Mike Bittner ◽  
Yidong Chen ◽  
Jeffrey Trent ◽  
Paul Meltzer ◽  
...  

2021 ◽  
Vol 12 (7) ◽  
Author(s):  
Jun Wang ◽  
Holly R. Thomas ◽  
Zhang Li ◽  
Nan Cher Yeo ◽  
Hannah E. Scott ◽  
...  

AbstractCellular stress can lead to several human disease pathologies due to aberrant cell death. The p53 family (tp53, tp63, and tp73) and downstream transcriptional apoptotic target genes (PUMA/BBC3 and NOXA/PMAIP1) have been implicated as mediators of stress signals. To evaluate the importance of key stress response components in vivo, we have generated zebrafish null alleles in puma, noxa, p53, p63, and p73. Utilizing these genetic mutants, we have deciphered that the apoptotic response to genotoxic stress requires p53 and puma, but not p63, p73, or noxa. We also identified a delayed secondary wave of genotoxic stress-induced apoptosis that is p53/puma independent. Contrary to genotoxic stress, ER stress-induced apoptosis requires p63 and puma, but not p53, p73, or noxa. Lastly, the oxidative stress-induced apoptotic response requires p63, and both noxa and puma. Our data also indicate that while the neural tube is poised for apoptosis due to genotoxic stress, the epidermis is poised for apoptosis due to ER and oxidative stress. These data indicate there are convergent as well as unique molecular pathways involved in the different stress responses. The commonality of puma in these stress pathways, and the lack of gross or tumorigenic phenotypes with puma loss suggest that a inhibitor of Puma may have therapeutic application. In addition, we have also generated a knockout of the negative regulator of p53, mdm2 to further evaluate the p53-induced apoptosis. Our data indicate that the p53 null allele completely rescues the mdm2 null lethality, while the puma null completely rescues the mdm2 null apoptosis but only partially rescues the phenotype. Indicating Puma is the key mediator of p53-dependent apoptosis. Interestingly the p53 homozygous null zebrafish develop tumors faster than the previously described p53 homozygous missense mutant zebrafish, suggesting the missense allele may be hypomorphic allele.


2013 ◽  
Vol 52 (2) ◽  
pp. 272-285 ◽  
Author(s):  
Stephanie Jungmichel ◽  
Florian Rosenthal ◽  
Matthias Altmeyer ◽  
Jiri Lukas ◽  
Michael O. Hottiger ◽  
...  

2002 ◽  
Vol 277 (42) ◽  
pp. 39655-39665 ◽  
Author(s):  
Kohji Noguchi ◽  
Hidesuke Fukazawa ◽  
Yuko Murakami ◽  
Yoshimasa Uehara

Oncotarget ◽  
2017 ◽  
Vol 8 (20) ◽  
pp. 32752-32768 ◽  
Author(s):  
Mingzhu Fang ◽  
Pamela A. Ohman Strickland ◽  
Hwan-Goo Kang ◽  
Helmut Zarbl

2007 ◽  
Vol 27 (6) ◽  
pp. 2189-2201 ◽  
Author(s):  
Peter S. Levitt ◽  
Min Zhu ◽  
Amy Cassano ◽  
Stephanie A. Yazinski ◽  
Houchun Liu ◽  
...  

ABSTRACT Cell cycle checkpoints are evolutionarily conserved signaling pathways that uphold genomic integrity. Complete inactivation of the mouse checkpoint gene Hus1 results in chromosomal instability, genotoxin hypersensitivity, and embryonic lethality. To determine the functional consequences of partial Hus1 impairment, we generated an allelic series in which Hus1 expression was incrementally reduced by combining a hypomorphic Hus1 allele, Hus1 neo , with either wild-type or null (Hus1 Δ1 ) alleles. Primary Hus1 neo/Δ1 embryonic fibroblasts exhibited spontaneous chromosomal abnormalities and underwent premature senescence, while higher Hus1 expression in Hus1 neo/neo cells allowed for normal proliferation. Antioxidant treatment almost fully suppressed premature senescence in Hus1 neo/Δ1 cultures, suggesting a critical role for Hus1 in oxidative stress responses. Treatment of Hus1 neo/neo and Hus1 neo/Δ1 cells with the DNA adducting agent benzo(a)pyrene dihydrodriol epoxide resulted in a loss of cell viability that was associated with S-phase DNA damage checkpoint failure. Likewise, the DNA polymerase inhibitor aphidicolin triggered increased cell death, chromosomal aberrations, and H2AX phosphorylation, a marker for double-stranded DNA breaks, in Hus1 neo/neo and Hus1 neo/Δ1 cultures compared to controls. Despite these pronounced genome maintenance defects in cultured Hus1 neo/Δ1 and Hus1 neo/neo cells, mice of the same genotypes were born at expected frequencies and appeared grossly normal. A significant increase in micronucleus formation was observed in peripheral blood cells from Hus1neo/Δ1 mice, but reduced Hus1 expression did not cause an elevated predisposition to spontaneous tumor development or accelerate tumorigenesis in p53-deficient mice. These results identify differential effects of altered Hus1 gene dosage on genome maintenance during in vitro culture, genotoxic stress responses, embryonic development, and adult homeostasis.


Sign in / Sign up

Export Citation Format

Share Document