Interferon regulatory factor-1 (IRF-1) exhibits tumor suppressor activities in breast cancer associated with caspase activation and induction of apoptosis

Kerrie B. Bouker, Todd C. Skaar, Rebecca B. Riggins, David S. Harburger, David R. Fernandez, Alan Zwart, Antai Wang, Robert Clarke

Research output: Contribution to journalArticlepeer-review

111 Scopus citations

Abstract

We have directly assessed the ability of interferon regulatory factor-1 (IRF-1) to act as a tumor suppressor gene in human breast cancer cells and explored whether this suppressor function is mechanistically conferred by affecting cell cycle transition, apoptosis and/or caspase activation. We have used a dual approach, measuring whether overexpression of wild-type IRF-1 or a dominant negative IRF-1 (dnIRF-1) produce opposing effects on breast cancer cell proliferation in vitro or tumorigenicity in athymic nude mice. Mechanistic studies determined the effects of blocking endogenous IRF-1 expression on cell cycle transition by flow cytometry, on apoptosis by Annexin V staining, and on caspase activation by fluorescent substrate cleavage. IRF-1 mRNA (P ≤ 0.001) and protein (P ≤ 0.001) are highly expressed in non-tumorigenic, normal, mammary epithelial cells, with intermediate expression in tumorigenic, but non-metastatic, cells and very low expression in metastatic cell lines. In MCF-7 cells transfected with a wild-type IRF-1 (MCF-7/IRF-1), IRF-1 mRNA expression inversely correlates with the rate of cell proliferation (r = -0.91; P = 0.002). Conversely, expression of dnIRF-1 in both MCF-7 (MCF-7/dnIRF-1; p53 wild-type) and T47D cells (T47D/dnIRF-1; p53 mutant) increases cell proliferation (P ≤ 0.001). In athymic nude mice, the incidence of MCF-7/IRF-1 xenografts is reduced (P = 0.045), whereas MCF-7/dnIRF-1 xenografts exhibit a significantly higher tumor incidence (P ≤ 0.001). Effects of IRF-1/dnIRF-1 are mediated through changes in the rates of apoptosis and not through cell cycle regulation. MCF-7/dnIRF-1 cells exhibit a 50% decrease in basal apoptosis (P = 0.007) and a significant reduction in caspase 8 activity (P = 0.03); similar effects occur in T47D/dnIRF-1 cells, where the effects on apoptosis appear to be mediated through inhibition of caspases 3/7 (P < 0.001) and caspase 8 (P = 0.03). These data establish a functional role for IRF-1 in the growth suppression of breast cancer cells and strongly implicate IRF-1 as a tumor suppressor gene in breast cancer that acts, independent of p53, to control apoptosis.

Original languageEnglish (US)
Pages (from-to)1527-1535
Number of pages9
JournalCarcinogenesis
Volume26
Issue number9
DOIs
StatePublished - Sep 2005
Externally publishedYes

Bibliographical note

Funding Information:
This work was supported in part by the American Cancer Society award IRG-97-1520-01-IRG (T.S.), the Ella and Charles O. Lathum Charitable Trust (T.S.) Public Health Service awards R01-CA/AG58022–10 and R01-CA096483–01 (R.C.), Department of Defense awards DAMD17–99–9189 (K.B.B.), BC030280 and BC010619 (R.C.) from the United States Army Medical Research and Material Command. Technical services also were provided by the Animal Shared Resource (R.Clarke) and Flow Cytometry and Cell Sorting and Macromolecular Shared Resources funded through Public Health Service award P30-CA51008–14 (Lombardi Comprehensive Cancer Center Support Grant).

Fingerprint

Dive into the research topics of 'Interferon regulatory factor-1 (IRF-1) exhibits tumor suppressor activities in breast cancer associated with caspase activation and induction of apoptosis'. Together they form a unique fingerprint.

Cite this