Faculty Opinions recommendation of Interplay between Ino80 and Swr1 chromatin remodeling enzymes regulates cell cycle checkpoint adaptation in response to DNA damage.

Author(s):  
Tim Humphrey
Biomolecules ◽  
2021 ◽  
Vol 11 (5) ◽  
pp. 750
Author(s):  
Kiyohiro Ando ◽  
Akira Nakagawara

Unrestrained proliferation is a common feature of malignant neoplasms. Targeting the cell cycle is a therapeutic strategy to prevent unlimited cell division. Recently developed rationales for these selective inhibitors can be subdivided into two categories with antithetical functionality. One applies a “brake” to the cell cycle to halt cell proliferation, such as with inhibitors of cell cycle kinases. The other “accelerates” the cell cycle to initiate replication/mitotic catastrophe, such as with inhibitors of cell cycle checkpoint kinases. The fate of cell cycle progression or arrest is tightly regulated by the presence of tolerable or excessive DNA damage, respectively. This suggests that there is compatibility between inhibitors of DNA repair kinases, such as PARP inhibitors, and inhibitors of cell cycle checkpoint kinases. In the present review, we explore alterations to the cell cycle that are concomitant with altered DNA damage repair machinery in unfavorable neuroblastomas, with respect to their unique genomic and molecular features. We highlight the vulnerabilities of these alterations that are attributable to the features of each. Based on the assessment, we offer possible therapeutic approaches for personalized medicine, which are seemingly antithetical, but both are promising strategies for targeting the altered cell cycle in unfavorable neuroblastomas.


2014 ◽  
Vol 34 (12) ◽  
pp. 2264-2282 ◽  
Author(s):  
A. Nagarajan ◽  
S. K. Dogra ◽  
A. Y. Liu ◽  
M. R. Green ◽  
N. Wajapeyee

2020 ◽  
Vol 22 ◽  
Author(s):  
Hannah L. Smith ◽  
Harriet Southgate ◽  
Deborah A. Tweddle ◽  
Nicola J. Curtin

Abstract DNA damage response (DDR) pathway prevents high level endogenous and environmental DNA damage being replicated and passed on to the next generation of cells via an orchestrated and integrated network of cell cycle checkpoint signalling and DNA repair pathways. Depending on the type of damage, and where in the cell cycle it occurs different pathways are involved, with the ATM-CHK2-p53 pathway controlling the G1 checkpoint or ATR-CHK1-Wee1 pathway controlling the S and G2/M checkpoints. Loss of G1 checkpoint control is common in cancer through TP53, ATM mutations, Rb loss or cyclin E overexpression, providing a stronger rationale for targeting the S/G2 checkpoints. This review will focus on the ATM-CHK2-p53-p21 pathway and the ATR-CHK1-WEE1 pathway and ongoing efforts to target these pathways for patient benefit.


2007 ◽  
Vol 27 (19) ◽  
pp. 6852-6862 ◽  
Author(s):  
Aimin Peng ◽  
Andrea L. Lewellyn ◽  
James L. Maller

ABSTRACT In Xenopus laevis embryos, the midblastula transition (MBT) at the 12th cell division marks initiation of critical developmental events, including zygotic transcription and the abrupt inclusion of gap phases into the cell cycle. Interestingly, although an ionizing radiation-induced checkpoint response is absent in pre-MBT embryos, introduction of a threshold amount of undamaged plasmid or sperm DNA allows a DNA damage checkpoint response to be activated. We show here that undamaged threshold DNA directly participates in checkpoint signaling, as judged by several dynamic changes, including H2AX phosphorylation, ATM phosphorylation and loading onto chromatin, and Chk1/Chk2 phosphorylation and release from nuclear DNA. These responses on physically separate threshold DNA require γ-H2AX and are triggered by an ATM-dependent soluble signal initiated by damaged DNA. The signal persists in egg extracts even after damaged DNA is removed from the system, indicating that the absence of damaged DNA is not sufficient to end the checkpoint response. The results identify a novel mechanism by which undamaged DNA enhances checkpoint signaling and provide an example of how the transition to cell cycle checkpoint activation during development is accomplished by maternally programmed increases in the DNA-to-cytoplasm ratio.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1604-1604
Author(s):  
Philip O. Saunders ◽  
Kenneth F. Bradstock ◽  
Linda J. Bendall

Abstract Five year survival for patients with relapsed pre-B ALL remains less than 10%, requiring new approaches to therapy. We sought to evaluate the potential of mTOR inhibitor RAD001 to enhance pre-B ALL cell killing by agents that induce DNA damage or microtubule disruption and identify interactions that may indicate novel approaches to therapy. Combining 16μM RAD001 with agents that cause DNA damage or microtubule disruption in vitro, enhanced caspase-dependent killing (p<0.05) of pre-B ALL cells. We observed 16μM RAD001 suppressed p53 and markedly attenuated p21 responses to DNA damage or vincristine. Lentiviral siRNA knock down of p53 in Nalm6 cells led to significantly increased (p<0.05) cell kill by vincristine relative to luciferase knockdown cells with an intact p53 response. This data indicates enhanced killing by combining RAD001 with DNA damage or vincristine does not require p53. Intracellular flow cytometry revealed that combining 16μM RAD001 with DNA damage or vincristine activates the JNK pathway. c-Jun has been reported to promote proliferation, apoptosis, suppress p53 and p21 promoters and prolong the half-life of p53 analogue, p73. Concordantly, we observed up regulation of p73, puma, bax, bim and cleaved caspase 3, associated with enhanced cell death. This data indicates that p73 provides an alternate pathway to apoptosis. We hypothesized that 16μM RAD001 enhances chemosensitivity through a JNK dependent suppression of cell cycle checkpoint regulation. We observed 1.5μM RAD001 inhibited pRb, Ki67 and PCNA expression, increasing G0/1 cell cycle arrest in response to DNA damage or vincristine, however 16μM RAD001 increased pRb, cyclin D1, Ki67, active CDC2 and PCNA expression. Increased DNA content, BrdU uptake and PCNA expression indicate cell cycle progression occurs in the presence of DNA damage or vincristine, when combined with 16μM RAD001. To validate the role of the JNK pathway in enhancing chemosensitivity we evaluated the impact of JNK inhibition on cell cycle regulation and cell survival. We observed enhanced cell cycle checkpoint regulation, indicated by reduced expression of c-jun, pRb, PCNA and Ki67 in Nalm6 cells. Furthermore, JNK inhibition enhanced G0/1 or G2 arrest in response to DNA damage in Nalm6 and REH cell lines respectively and enhanced G2 arrest in response to vincristine. JNK inhibition led to reduced cell kill by DNA damage or microtubule disruption in Nalm6 and REH cell lines. This data strongly suggests that impaired cell cycle regulation by 16μM RAD001 is mediated through a JNK dependent mechanism. We conclude that dose escalated RAD001 enhances chemosensitivity independently of p53, through a JNK dependent impairment of cell cycle regulation, in response to DNA damage or microtubule disruption. Our data indicates that dose escalated RAD001 has the potential to enhance chemosensitivity in patients with pre-B ALL and provides a rationale for combining agents which induce JNK activation with DNA damage or microtubule disruption, as a therapeutic strategy in pre-B ALL.


2003 ◽  
Vol 23 (3) ◽  
pp. 791-803 ◽  
Author(s):  
Robert S. Weiss ◽  
Philip Leder ◽  
Cyrus Vaziri

ABSTRACT Mouse Hus1 encodes an evolutionarily conserved DNA damage response protein. In this study we examined how targeted deletion of Hus1 affects cell cycle checkpoint responses to genotoxic stress. Unlike hus1− fission yeast (Schizosaccharomyces pombe) cells, which are defective for the G2/M DNA damage checkpoint, Hus1-null mouse cells did not inappropriately enter mitosis following genotoxin treatment. However, Hus1-deficient cells displayed a striking S-phase DNA damage checkpoint defect. Whereas wild-type cells transiently repressed DNA replication in response to benzo(a)pyrene dihydrodiol epoxide (BPDE), a genotoxin that causes bulky DNA adducts, Hus1-null cells maintained relatively high levels of DNA synthesis following treatment with this agent. However, when treated with DNA strand break-inducing agents such as ionizing radiation (IR), Hus1-deficient cells showed intact S-phase checkpoint responses. Conversely, checkpoint-mediated inhibition of DNA synthesis in response to BPDE did not require NBS1, a component of the IR-responsive S-phase checkpoint pathway. Taken together, these results demonstrate that Hus1 is required specifically for one of two separable mammalian checkpoint pathways that respond to distinct forms of genome damage during S phase.


2002 ◽  
Vol 22 (22) ◽  
pp. 7831-7841 ◽  
Author(s):  
Eugene S. Kandel ◽  
Jennifer Skeen ◽  
Nathan Majewski ◽  
Antonio Di Cristofano ◽  
Pier Paolo Pandolfi ◽  
...  

ABSTRACT Activation of Akt, or protein kinase B, is frequently observed in human cancers. Here we report that Akt activation via overexpression of a constitutively active form or via the loss of PTEN can overcome a G2/M cell cycle checkpoint that is induced by DNA damage. Activated Akt also alleviates the reduction in CDC2 activity and mitotic index upon exposure to DNA damage. In addition, we found that PTEN null embryonic stem (ES) cells transit faster from the G2/M to the G1 phase of the cell cycle when compared to wild-type ES cells and that inhibition of phosphoinositol-3-kinase (PI3K) in HEK293 cells elicits G2 arrest that is alleviated by activated Akt. Furthermore, the transition from the G2/M to the G1 phase of the cell cycle in Akt1 null mouse embryo fibroblasts (MEFs) is attenuated when compared to that of wild-type MEFs. These results indicate that the PI3K/PTEN/Akt pathway plays a role in the regulation of G2/M transition. Thus, cells expressing activated Akt continue to divide, without being eliminated by apoptosis, in the presence of continuous exposure to mutagen and accumulate mutations, as measured by inactivation of an exogenously expressed herpes simplex virus thymidine kinase (HSV-tk) gene. This phenotype is independent of p53 status and cannot be reproduced by overexpression of Bcl-2 or Myc and Bcl-2 but seems to counteract a cell cycle checkpoint mediated by DNA mismatch repair (MMR). Accordingly, restoration of the G2/M cell cycle checkpoint and apoptosis in MMR-deficient cells, through reintroduction of the missing component of MMR, is alleviated by activated Akt. We suggest that this new activity of Akt in conjunction with its antiapoptotic activity may contribute to genetic instability and could explain its frequent activation in human cancers.


Sign in / Sign up

Export Citation Format

Share Document