Published in

National Academy of Sciences, Proceedings of the National Academy of Sciences, 11(106), p. 4390-4395, 2009

DOI: 10.1073/pnas.0809080106

Links

Tools

Export citation

Search in Google Scholar

Apoptosis-stimulating protein of p53 (ASPP2) heterozygous mice are tumor-prone and have attenuated cellular damage–response thresholds

This paper is made freely available by the publisher.
This paper is made freely available by the publisher.

Full text: Download

Red circle
Preprint: archiving forbidden
Green circle
Postprint: archiving allowed
Red circle
Published version: archiving forbidden
Data provided by SHERPA/RoMEO

Abstract

The expression of ASPP2 (53BP2L), a proapoptotic member of a family of p53-binding proteins, is frequently suppressed in many human cancers. Accumulating evidence suggests that ASPP2 inhibits tumor growth; however, the mechanisms by which ASPP2 suppresses tumor formation remain to be clarified. To study this, we targeted the ASPP2 allele in a mouse by replacing exons 10–17 with a neoR gene. ASPP2 −/− mice were not viable because of an early embryonic lethal event. Although ASPP2 +/− mice appeared developmentally normal, they displayed an increased incidence of a variety of spontaneous tumors as they aged. Moreover, γ-irradiated 6-week-old ASPP2 +/− mice developed an increased incidence of high-grade T cell lymphomas of thymic origin compared with ASPP2 +/+ mice. Primary thymocytes derived from ASPP2 +/− mice exhibited an attenuated apoptotic response to γ-irradiation compared with ASPP2 +/+ thymocytes. Additionally, ASPP2 +/− primary mouse embryonic fibroblasts demonstrated a defective G 0 /G 1 cell cycle checkpoint after γ-irradiation. Our results demonstrate that ASPP2 is a haploinsufficient tumor suppressor and, importantly, open new avenues for investigation into the mechanisms by which disruption of ASPP2 pathways could play a role in tumorigenesis and response to therapy.