American Association for Cancer Research
Browse
10780432ccr151275-sup-150575_1_supp_3171445_nvq4vv.docx (21.46 MB)

Supplementary data from Adrenergic Stimulation of DUSP1 Impairs Chemotherapy Response in Ovarian Cancer

Download (21.46 MB)
journal contribution
posted on 2023-03-31, 18:40 authored by Yu Kang, Archana S. Nagaraja, Guillermo N. Armaiz-Pena, Piotr L. Dorniak, Wei Hu, Rajesha Rupaimoole, Tao Liu, Kshipra M. Gharpure, Rebecca A. Previs, Jean M. Hansen, Cristian Rodriguez-Aguayo, Cristina Ivan, Prahlad Ram, Vasudha Sehgal, Gabriel Lopez-Berestein, Susan K. Lutgendorf, Steven W. Cole, Anil K. Sood

Supplementary Figure 1 Functional genomic analysis of HeyA8 and SKOV3ip ovarian cancer cells 4 h after exposure to norepinephrine and compared to control cells. Supplementary Figure 2 A, Western blot analysis of ADRB1, ADRB2, ADRB3, and DUSP1 expression in 10 epithelial ovarian cancer cell lines and one breast cancer cell line (MDA-231), using non-transformed ovarian epithelium cells (HIO-180) as control. B, DUSP1 mRNA levels of cells exposed to norepinephrine 10 μM for 4 h. Supplementary Figure 3 A, DUSP1 silencing. Two commercially available siRNAs that directly target the DUSP1 gene were transfected to norepinephrine-treated HeyA8 cells. B, DUSP1 overexpression. SKOV3ip1 cells were transiently transfected with a DUSP1-myc/DDK-tagged expression vector (Origene). Supplementary Figure 4 SKOV3ip1 cells were treated with paclitaxel alone (10nM) or in combination with norepinephrine (NE; 10μM) or vascular endothelial growth factor (VEGF; 10ng/ml or 50ng/ml). NE or VEGF was administered 30 minutes prior to paclitaxel exposure.

Funding

NIH

National Natural Science Foundation of China

NCI

DHHS

Cancer Prevention and Research Institute of Texas

History

ARTICLE ABSTRACT

Purpose: Chronic adrenergic activation has been shown to associate with adverse clinical outcomes in cancer patients, but the underlying mechanisms are not well understood. The focus of the current study was to determine the functional and biologic effects of adrenergic pathways on response to chemotherapy in the context of ovarian cancer.Experimental Design: Increased DUSP1 production by sympathetic nervous system mediators (e.g., norepinephrine) was analyzed by real-time quantitative RT-PCR and by Western blotting. In vitro chemotherapy-induced cell apoptosis was examined by flow cytometry. For in vivo therapy, a well-characterized model of chronic stress was used.Results: Catecholamines significantly inhibited paclitaxel- and cisplatin-induced apoptosis in ovarian cancer cells. Genomic analyses of cells treated with norepinephrine identified DUSP1 as a potential mediator. DUSP1 overexpression resulted in reduced paclitaxel-induced apoptosis in ovarian cancer cells compared with control; conversely, DUSP1 gene silencing resulted in increased apoptosis compared with control cells. DUSP1 gene silencing in vivo significantly enhanced response to paclitaxel and increased apoptosis. In vitro analyses indicated that norepinephrine-induced DUSP1 gene expression was mediated through ADRB2 activation of cAMP–PLC–PKC–CREB signaling, which inhibits JNK-mediated phosphorylation of c-Jun and protects ovarian cancer cells from apoptosis. Moreover, analysis of The Cancer Genome Atlas data showed that increased DUSP1 expression was associated with decreased overall (P = 0.049) and progression-free (P = 0.0005) survival.Conclusions: These findings provide a new understanding of the mechanisms by which adrenergic pathways can impair response to chemotherapy and have implications for cancer management. Clin Cancer Res; 22(7); 1713–24. ©2015 AACR.