Abstract

The high-mobility group A (HMGA) proteins are a family of non-histone chromatin factors, encoded by the HMGA1 and HMGA2 genes. Several studies demonstrate that HMGA proteins have a critical role in neoplastic transformation, and their overexpression is mainly associated with a highly malignant phenotype, also representing a poor prognostic index. Even though a cytoplasmic localization of these proteins has been previously reported in some highly malignant neoplasias, a clear role for this localization has not been defined. Here, we first confirm the localization of the HMGA1 proteins in the cytoplasm of cancer cells, and then we report a novel mechanism through which HMGA1 inhibits p53-mitochondrial apoptosis by counteracting the binding of p53 to the anti-apoptotic factor Bcl-2. Indeed, we demonstrate a physical and functional interaction between HMGA1 and Bcl-2 proteins. This interaction occurs at mitochondria interfering with the ability of p53 protein to bind Bcl-2, thus counteracting p53-mediated mitochondrial apoptosis. This effect is associated with the inhibition of cytochrome c release and activation of caspases. Consistent with this mechanism, a strong correlation between HMGA1 cytoplasmic localization and a more aggressive histotype of thyroid, breast and colon carcinomas has been observed. Therefore, cytoplasmic localization of HMGA1 proteins in malignant tissues is a novel mechanism of inactivation of p53 apoptotic function.

Highlights

  • HMGA1a, HMGA1b and HMGA1c, derived from an alternative splicing of the same gene, HMGA1,1,2 and HMGA2, encoded by the homonymous gene.[3]

  • We have previously demonstrated that overexpression of high-mobility group A (HMGA) proteins is required for cell transformation, as the blockage of their synthesis prevents tumorigenic transformation of rat thyroid cells by murine-transforming retroviruses,[16] and infection with a recombinant adenovirus carrying the HMGA1b complementary DNA in antisense orientation led several carcinoma cell lines to death.[17]

  • HMGA1b recombinant protein displaced B-cell lymphoma gene-2 (Bcl-2) from the binding to p53. These findings demonstrate that HMGA1b directly interferes with the p53/Bcl-2 binding

Read more

Summary

Introduction

HMGA1a, HMGA1b and HMGA1c, derived from an alternative splicing of the same gene, HMGA1,1,2 and HMGA2, encoded by the homonymous gene.[3]. HMGA proteins do not have transcriptional activity per se, by interacting with the transcriptional machinery, they alter the chromatin structure and, thereby, regulate the transcriptional activity of several genes.[4,5] The expression of HMGA proteins is high during embryogenesis and low or undetectable in normal adult tissues.[6,7] high HMGA expression has been observed in human malignant neoplasias, including thyroid,[8] colon,[9] prostate,[10] pancreas,[11] cervix,[12] ovary,[13] and breast[14] carcinomas Their overexpression is mainly associated with a highly malignant phenotype, representing a poor prognostic index, as HMGA overexpression often correlates with the presence of metastases, and with a reduced survival.[15] A functional role of this aberrant HMGA overexpression in cancer has been previously demonstrated by in vitro and in vivo studies.

Methods
Results
Conclusion
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.