Date of Graduation

5-2015

Document Type

Dissertation (PhD)

Program Affiliation

Cancer Biology

Degree Name

Doctor of Philosophy (PhD)

Advisor/Committee Chair

Khandan Keyomarsi, Ph.D.

Committee Member

Jill Schumacher, Ph.D.

Committee Member

Wei Zhang, Ph.D.

Committee Member

Jeffrey Frost, Ph.D.

Committee Member

Zhimin Lu, Ph.D.

Abstract

Cyclin E is altered or overexpressed in approximately one-third of tumors from patients with invasive breast cancer and is a powerful independent predictor for survival in women with stage I-III breast cancer. Full-length cyclin E (EL) is post-translationally cleaved into two low-molecular-weight isoforms, LMW-E (T1) and LMW-E (T2). LMW-E have been shown to exhibit greater binding affinity for cyclin-dependent kinase 2 (CDK2) , cyclin dependent kinase inhibitors (CKIs), p21 and p27, but are resistant to p21 and p27 inhibition. In addition, transgenic mice expressing LMW-E have increased mammary tumor development and metastasis compared to EL transgenic mice. Therefore, LMW-E are more aggressive in cell cycle abrogation and mammary tumor development. The LMW-E isoforms are tumor specific and accumulate in the cytoplasm due to lack of a nuclear localization sequence (NLS). Therefore, we hypothesized that aberrant localization of LMW-E isoforms leads to molecular events that ultimately contribute to LMW-E breast cancer tumorigenicity. To address this hypothesis, we used a retrovirus-based protein complementation assay (RePCA) to identify LMW-E (T1) protein-protein interactions in breast cancer. Using this methodology, we found ATP-citrate lyase (ACLY) as a novel interacting protein of LMW-E (T1) in the cytoplasm. ACLY is a 125kDa homotetrameric enzyme that catalyzes cytoplasmic citrate to acetyl-CoA and oxaloacetate in the de novo lipogenesis pathway. End products of this pathway consist of complex fatty acids that fuel membrane production of highly proliferating cells and lipid-based post-translational modifications that mediate protein-protein interactions. Additionally, we found that LMW-E upregulates ACLY enzymatic activity which leads to lipid droplet formation; thereby providing cells with essential building blocks to support growth. ACLY is also required for LMW-E mediated transformation, migration and invasion in vitro, as well as tumor growth in vivo. Taken together these data suggest a novel interplay between LMW-E and ACLY and how metabolic pathways and the cell cycle are linked in breast cancer tumorigenesis.

Keywords

Breast cancer Low molecular weight cyclin E ATP-citrate lyase Retrovirus-based protein complementation assay Lipid droplets

Share

COinS
 
 

To view the content in your browser, please download Adobe Reader or, alternately,
you may Download the file to your hard drive.

NOTE: The latest versions of Adobe Reader do not support viewing PDF files within Firefox on Mac OS and if you are using a modern (Intel) Mac, there is no official plugin for viewing PDF files within the browser window.