Graduation Year

2014

Document Type

Dissertation

Degree

Ph.D

Degree Name

Doctor of Philosophy (Ph.D.)

Department

Microbiology, Molecular Biology and Biochemistry

Degree Granting Department

Cell Biology, Microbiology and Molecular Biology

Major Professor

Meera Nanjundan, Ph.D.

Committee Member

Richard Pollenz, Ph.D.

Committee Member

Patrick Bradshaw, Ph.D.

Committee Member

Sandy Westerheide, Ph.D.

Keywords

Chemotherapeutics, dsDNA, Interferons, Ovarian Cancer, Phospholipid Srcamblase, SnoN

Abstract

Ovarian cancer is one of the most common causes of gynecological cancer related deaths in women. In 2014, the estimated number of deaths due to ovarian cancer is 14,270 with occurrence of over 22, 240 new cases (National Cancer Institute, http://seer.cancer.gov/statfacts/html/ovary.html). Despite improvement in treatment strategies, the 5-year survival rate is still below 50% mainly due to chemoresistance and relapse. Amplification of chromosomal region 3q26 is a common characteristic in various epithelial cancers including ovarian cancer. This region harbors various oncogenes including the TGFβ signaling mediators EVI1 and SnoN/SkiL, PKCι and PIK3CA amplified at 3q26.2 and 3q26.3, respectively, in ovarian cancers. Previous studies indicate that these genes can exhibit cooperative oncogenicity by cross-regulating one another and facilitating cancer development. Our earlier studies demonstrated that treatment of ovarian cancer cells with arsenic trioxide (As2O3) promotes cytoprotective autophagy regulated by induction of SnoN to antagonize the cytotoxic effects of As2O3. Since exact mechanisms underlying As2O3-induced SnoN expression and cytoprotective responses were unclear, we hypothesized that SnoN may be regulated by signaling pathways involving genes amplified at the 3q26 locus.

Phospholipid scramblase 1 (PLSCR1) is located at 3q23 proximal to the amplified 3q26 region. It had been implicated in disruption of plasma membrane asymmetry by mediating phospholipid scrambling, a process critical for cellular events such as blood coagulation and apoptosis. However, recent findings have led to more investigations on the role and regulation of PLSCR1 in cancer development and immune responses. PLSCR1 expression is regulated by various stimuli including growth factors (EGF, G-CSF, and SCF), cytokines (IFN), and differentiation-inducing agents (ATRA). Despite these studies, transcriptional regulation of PLSCR1 remains incompletely understood. Numerous studies have suggested a critical role for PLSCR1 in the pathophysiology of various cancers including leukemia, ovarian cancer, colorectal cancer, and metastatic liver cancer. However, the precise contribution of PLSCR1 and its regulation in ovarian cancer development is unclear. Since PLSCR1 (at 3q23) is located in close proximity to SnoN/SkiL (at 3q26.2), we hypothesized that PLSCR1 expression in ovarian cancer cells could be regulated by SnoN. Herein, we present studies that primarily focus on understanding the role and regulation of SnoN/SkiL (a TGFβ pathway regulator) and PLSCR1 (an interferon-regulated gene), which are located at 3q26.2 and 3q23, respectively, in epithelial ovarian cancer.

In Chapter 3, we determined that activation of the PI3K signaling pathway mediates SnoN expression and cytoprotective responses upon stimulation of ovarian cancer cells with As2O3. We first identified that As2O3 stimulation leads to activation of EGFR and its downstream signaling mediators as well as modulates its interaction with the adaptor proteins, ShcA and Grb2. Interestingly, while treatment with a general SFK inhibitor (PP2), reduced the As2O3-induced EGFR activation and SnoN induction, a more specific inhibitor SU6656 did not alter SnoN expression. Further, via studies utilizing specific inhibitors and siRNA targeting PI3K, we determined that inhibition of PI3K signaling pathway decreases SnoN induction and increases apoptosis in ovarian cancer cells in response to As2O3. This suggests that PI3K (PIK3CA) activity is required for the As2O3-mediated SnoN induction and the cell survival responses in ovarian cancer cells. Finally, we determined by siRNA-mediated knockdown that EGFR and MAPK1 alter As2O3-induced cell death response independently of SnoN induction.

In Chapter 4, via bioinformatic analyses, we identified that PLSCR1 DNA copy number and mRNA expression is elevated in ovarian cancer patients and cell lines relative to immortalized (Tag/hTERT) normal ovarian surface epithelial (OSE) cells. Interestingly, altered PLSCR1 DNA and mRNA levels were correlated with SnoN in ovarian cancers. We next identified that SnoN knockdown leads to a significant (~35%, P2O3 transcriptionally downregulates PLSCR1 in a ROS-independent mechanism. Furthermore, PLSCR1 knockdown, similar to SnoN knockdown increases ovarian cancer cell sensitivity to As2O3. PLSCR1 knockdown increases cleaved PARP (marker of apoptosis) with a consequent reduction in LC3-II levels (marker of autophagosomes). Collectively, these studies implicate PLSCR1 in the pathophysiology of ovarian cancers and in altering the chemotherapeutic responses in ovarian cancer cells.

PLSCR1 is an IFN-regulated gene and mediates antiviral/immune responses. More recent studies in plasmacytoid dendritic cells have implicated PLSCR1 in regulating TLR9 signaling upon stimulation with CpG ODN. However, whether PLSCR1 could mediate the innate immune responses upon stimulation with dsDNA remained unclear. In Chapter 5, we identified that stimulation of normal ovarian and mammary epithelial cells with dsDNA (empty plasmid) markedly induces PLSCR1 consequent with activation of IRF3, a downstream mediator of TLR signaling that transcriptionally regulates the expression of type 1 IFNs. Interestingly, IRF3 knockdown ablates the dsDNA-induced PLSCR1 expression suggesting that PLSCR1 induction in response to dsDNA could be mediated by IRF3. Additionally, we have determined that dsDNA stimulation induces nucleic acid sensing TLRs, TLR9 and TLR4 as well as IFN-α and IFN-β mRNAs. Interestingly, dsDNA stimulation did not induce PLSCR1 or IRF3 activation in ovarian cancer cells suggesting that the mechanisms of IRF3 activation and PLSCR1 induction in response to dsDNA might be dysregulated in ovarian cancers.

Collectively, our studies demonstrate a possible synergistic role of SnoN and PLSCR1 in ovarian cancer pathophysiology and suggest a potentially dysregulated role of PLSCR1 in the dsDNA-induced immune responses of malignant epithelial cells relative to normal epithelial cells. These studies could potentially lead to development of a novel combinatorial therapeutic strategy that targets both these molecules for improving treatment of patients with ovarian carcinoma.

Share

COinS