Suppression of Insulin Receptor Substrate 1 (IRS-1) Promotes Mammary Tumor Metastasis

Author:

Ma Zhefu1,Gibson Shannon L.1,Byrne Maura A.1,Zhang Junran2,White Morris F.3,Shaw Leslie M.1

Affiliation:

1. Department of Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts 01605

2. Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri 63108

3. Howard Hughes Medical Institute, Children's Hospital, Division of Endocrinology, Harvard Medical School, Boston, Massachusetts 02115

Abstract

ABSTRACT The insulin receptor substrate (IRS) proteins are cytoplasmic adaptors that organize signaling complexes downstream of activated cell surface receptors. Here, we show that IRS-1 and IRS-2, despite significant homology, play critical yet distinct functions in breast cancer, and we identify specific signaling pathways that are influenced by IRS-1 using the polyoma virus middle-T (PyV-MT) transgenic mouse model of mammary carcinoma and Irs-1 null ( Irs1 −/− ) mice. The absence of Irs-1 expression enhanced metastatic spread significantly without a significant effect on primary tumor growth. Orthotopic transplant studies revealed that the increased metastatic potential of Irs1 -deficient tumor cells is cell autonomous. Mammary tumors that developed in PyV-MT:: Irs1 −/− mice exhibited elevated Irs-2 function and enhanced phosphatidylinositol 3-kinase/Akt/mTor activity, suggesting that one mechanism by which Irs-1 impedes metastasis is to suppress Irs-2-dependent signaling. In support of this mechanism, reduction of Irs-2 expression in Irs1 −/− tumor cells restored mTor signaling to wild-type levels. PyV-MT:: Irs1 −/− tumors also exhibited a significant increase in vascular endothelial growth factor expression and microvessel density, which could facilitate their dissemination. The significance of our findings for human breast cancer is heightened by our observation that Irs-1 is inactivated in wild-type, metastatic mammary tumors by serine phosphorylation. Collectively, our findings reveal that inactivation of IRS-1 enhances breast cancer metastasis and support the novel hypothesis that IRS-1 has metastasis suppressor functions for breast cancer.

Publisher

American Society for Microbiology

Subject

Cell Biology,Molecular Biology

Reference87 articles.

1. Aguirre, V., T. Uchida, L. Yenush, R. Davis, and M. F. White. 2000. The c-Jun NH(2)-terminal kinase promotes insulin resistance during association with insulin receptor substrate-1 and phosphorylation of Ser(307). J. Biol. Chem.275:9047-9054.

2. Characterization of a 3-phosphoinositide-dependent protein kinase which phosphorylates and activates protein kinase Bα

3. Araki, E., M. A. Lipes, M. E. Patti, J. C. Bruning, B. Haag III, R. S. Johnson, and C. R. Kahn. 1994. Alternative pathway of insulin signalling in mice with targeted disruption of the IRS-1 gene. Nature372:186-190.

4. Bachelder, R. E., A. Crago, J. Chung, M. A. Wendt, L. M. Shaw, G. Robinson, and A. M. Mercurio. 2001. Vascular endothelial growth factor is an autocrine survival factor for neuropilin-expressing breast carcinoma cells. Cancer Res.61:5736-5740.

5. Bachelder, R. E., M. A. Wendt, and A. M. Mercurio. 2002. Vascular endothelial growth factor promotes breast carcinoma invasion in an autocrine manner by regulating the chemokine receptor CXCR4. Cancer Res.62:7203-7206.

同舟云学术

1.学者识别学者识别

2.学术分析学术分析

3.人才评估人才评估

"同舟云学术"是以全球学者为主线,采集、加工和组织学术论文而形成的新型学术文献查询和分析系统,可以对全球学者进行文献检索和人才价值评估。用户可以通过关注某些学科领域的顶尖人物而持续追踪该领域的学科进展和研究前沿。经过近期的数据扩容,当前同舟云学术共收录了国内外主流学术期刊6万余种,收集的期刊论文及会议论文总量共计约1.5亿篇,并以每天添加12000余篇中外论文的速度递增。我们也可以为用户提供个性化、定制化的学者数据。欢迎来电咨询!咨询电话:010-8811{复制后删除}0370

www.globalauthorid.com

TOP

Copyright © 2019-2024 北京同舟云网络信息技术有限公司
京公网安备11010802033243号  京ICP备18003416号-3