Abstract

Inappropriate activation of cell cycle proteins, in particular cyclin D/Cdk4, is implicated in neuronal death induced by various pathologic stresses, including DNA damage and ischemia. Key targets of Cdk4 in proliferating cells include members of the E2F transcription factors, which mediate the expression of cell cycle proteins as well as death-inducing genes. However, the presence of multiple E2F family members complicates our understanding of their role in death. We focused on whether E2F4, an E2F member believed to exhibit crucial control over the maintenance of a differentiated state of neurons, may be critical in ischemic neuronal death. We observed that, in contrast to E2F1 and E2F3, which sensitize to death, E2F4 plays a crucial protective role in neuronal death evoked by DNA damage, hypoxia, and global ischemic insult both in vitro and in vivo. E2F4 occupies promoter regions of proapoptotic factors, such as B-Myb, under basal conditions. Following stress exposure, E2F4-p130 complexes are lost rapidly along with the presence of E2F4 at E2F-containing B-Myb promoter sites. In contrast, the presence of E2F1 at B-Myb sites increases with stress. Furthermore, B-Myb and C-Myb expression increases with ischemic insult. Taken together, we propose a model by which E2F4 plays a protective role in neurons from ischemic insult by forming repressive complexes that prevent prodeath factors such as Myb from being expressed.

Highlights

  • The contribution of E2F4 to hypoxic/ischemic neuronal death is understood poorly

  • In contrast to E2F1 and E2F3, which sensitize to death, E2F4 plays a crucial protective role in neuronal death evoked by DNA damage, hypoxia, and global ischemic insult both in vitro and in vivo

  • To begin to examine the involvement of different E2F members, we focused initially on the effect of select activating E2Fs (E2F1 and E2F3) and the repressive E2F4 members in neuronal death

Read more

Summary

Background

The contribution of E2F4 to hypoxic/ischemic neuronal death is understood poorly. Results: Loss of E2F4 leads to an increase in B-Myb and contributes to hypoxic/ischemic neuronal death. We propose a model by which E2F4 plays a protective role in neurons from ischemic insult by forming repressive complexes that prevent prodeath factors such as Myb from being expressed. Expression of a dominant negative form of Cdk, an important regulator of the G1/S phase of the cell cycle, is protective following DNA damage and global cerebral ischemia [5, 9] Together, these studies reveal a crucial role for Cdk in neuronal death induced by DNA damage and ischemic insult. E2F4-p130 can recruit chromatin modification factors, such as histone deacetylases (HDACs), to promoters of target genes to form an active repression complex [13,14,15,16] In this scheme, phosphorylation of p130 by Cdks disrupts its associa-

The abbreviations used are
EXPERIMENTAL PROCEDURES
RESULTS
DISCUSSION
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.