4.7 Article

VGLL4 Selectively Represses YAP-Dependent Gene Induction and Tumorigenic Phenotypes in Breast Cancer

期刊

SCIENTIFIC REPORTS
卷 7, 期 -, 页码 -

出版社

NATURE PUBLISHING GROUP
DOI: 10.1038/s41598-017-06227-7

关键词

-

资金

  1. China Scholarship Council [201403170287]
  2. Roswell Park Cancer Institute
  3. National Cancer Institute (NCI) [P30 CA016056, R21 CA179693]
  4. Roswell Park Alliance Foundation
  5. American Cancer Society [RSG-14-214-01-TBE]

向作者/读者索取更多资源

Members of the mammalian Vestigial-like (VGLL) family of transcriptional cofactors activate genes in response to a wide variety of environmental cues. Recently, VGLL proteins have been proposed to regulate key signaling networks involved in cancer development and progression. However, the biological and clinical significance of VGLL dysregulation in human breast cancer pathogenesis remains unknown. Here, we report that diminished VGLL4 expression, but not VGLL1-3, correlated with both shorter relapse-free survival and shorter disease-specific survival of cancer patients with different molecular subtypes of breast cancer. Additionally, we further demonstrate that overexpression of VGLL4 reduces breast cancer cell proliferation, migration, intravasation/extravasation potential, favors cell death, and suppresses tumor growth in vivo. Mechanistically, VGLL4 negatively regulates the TEAD1-YAP1 transcriptional complex and exerts its growth inhibitory control through its evolutionary conserved TDU2 domain at its C-terminus. The results suggest that VGLL4 is a candidate tumor suppressor gene which acts by selectively antagonizing YAP-dependent tumor growth. VGLL4 may be a promising therapeutic target in breast cancer.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.7
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

Meeting Abstract Oncology

Off-target analysis of RNAi screens identifies microRNA seed sequence contributing to metastatic invasion

Henry G. Withers, Ryan Ransom, Irwin H. Gelman

CANCER RESEARCH (2016)

Article Biochemistry & Molecular Biology

Hypoxia and the metabolic phenotype of prostate cancer cells

L. H. Higgins, H. G. Withers, A. Garbens, H. D. Love, L. Magnoni, S. W. Hayward, C. D. Moyes

BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS (2009)

Article Oncology

Identification of Genes Regulating Breast Cancer Dormancy in 3D Bone Endosteal Niche Cultures

Julie McGrath, Louis Panzica, Ryan Ransom, Henry G. Withers, Irwin H. Gelman

MOLECULAR CANCER RESEARCH (2019)

Article Oncology

MUC1-C integrates type II interferon and chromatin remodeling pathways in immunosuppression of prostate cancer

Masayuki Hagiwara, Atsushi Fushimi, Atrayee Bhattacharya, Nami Yamashita, Yoshihiro Morimoto, Mototsugu Oya, Henry G. Withers, Qiang Hu, Tao Liu, Song Liu, Kwok K. Wong, Mark D. Long, Donald Kufe

Summary: The oncogenic MUC1-C protein plays a crucial role in driving the dedifferentiation of castrate resistant prostate cancer cells. It activates the type II interferon-gamma (IFN-gamma) pathway and induces chromatin remodeling, leading to the suppression of immune response and promotion of cancer stem cell state.

ONCOIMMUNOLOGY (2022)

Article Oncology

Dependence on the MUC1-C Oncoprotein in Classic, Variant, and Non-neuroendocrine Small Cell Lung Cancer

Atsushi Fushimi, Yoshihiro Morimoto, Satoshi Ishikawa, Nami Yamashita, Atrayee Bhattacharya, Tatsuaki Daimon, Hasan Rajabi, Caining Jin, Masayuki Hagiwara, Yota Yasumizu, Zhou Luan, Wenhao Suo, Kwok-Kin Wong, Henry Withers, Song Liu, Mark D. Long, Donald Kufe

Summary: MUC1-C protein is activated in pulmonary epithelial cells and plays a crucial role in promoting the growth and development of small cell lung cancer (SCLC) cells through activation of the MYC pathway. Furthermore, the MUC1-C->MYC->NOTCH2 network is essential for self-renewal capacity and tumorigenicity of SCLC cells.

MOLECULAR CANCER RESEARCH (2022)

Article Oncology

Phase IIa Study of SurVaxM Plus Adjuvant Temozolomide for Newly Diagnosed Glioblastoma

Manmeet S. Ahluwalia, David A. Reardon, Ajay P. Abad, William T. Curry, Eric T. Wong, Sheila A. Figel, Laszlo L. Mechtler, David M. Peereboom, Alan D. Hutson, Henry G. Withers, Song Liu, Ahmed N. Belal, Jingxin Qiu, Kathleen M. Mogensen, Sanam S. Dharma, Andrew Dhawan, Meaghan T. Birkemeier, Danielle M. Casucci, Michael J. Ciesielski, Robert A. Fenstermaker

Summary: SurVaxM is a peptide vaccine that activates the immune system against glioblastoma cells. The combination of SurVaxM and adjuvant chemotherapy can extend the progression-free survival of newly diagnosed glioblastoma patients.

JOURNAL OF CLINICAL ONCOLOGY (2023)

Article Oncology

MUC1-C Dictates PBRM1-Mediated Chronic Induction of Interferon Signaling, DNA Damage Resistance, and Immunosuppression in Triple-Negative Breast Cancer

Nami Yamashita, Yoshihiro Morimoto, Atsushi Fushimi, Rehan Ahmad, Atrayee Bhattacharya, Tatsuaki Daimon, Naoki Haratake, Yuka Inoue, Satoshi Ishikawa, Masaaki Yamamoto, Tsuyoshi Hata, Sayuri Akiyoshi, Qiang Hu, Tao Liu, Henry Withers, Song Liu, Geoffrey I. Shapiro, Tomoharu Yoshizumi, Mark D. Long, Donald Kufe

Summary: In certain cancer cells, the chromatin remodeling complex SWI/SNF PBAF's subunit polybromo-1 (PBRM1) drives DNA damage resistance and immune evasion through unclear mechanisms. This study found that MUC1-C is necessary for PBRM1 expression in triple-negative breast cancer (TNBC) cells, and the two proteins form a nuclear complex. Transcriptional and chromatin accessibility analysis showed that MUC1-C and PBRM1 increase the expression of STAT1 and IRF1 by enhancing chromatin accessibility on their respective genes, as well as other genes involved in DNA damage resistance and immune evasion.

MOLECULAR CANCER RESEARCH (2023)

Article Multidisciplinary Sciences

MUC1-C integrates aerobic glycolysis with suppression of oxidative phosphorylation in triple-negative breast cancer stem cells

Nami Yamashita, Henry Withers, Yoshihiro Morimoto, Atrayee Bhattacharya, Naoki Haratake, Tatsuaki Daimon, Atsushi Fushimi, Ayako Nakashoji, Aaron R. Thorner, Emily Isenhart, Spencer Rosario, Mark D. Long, Donald Kufe

Summary: Activation of MUC1-C protein is crucial for the development and state of TNBC CSCs, as it regulates self-renewal and oxidative phosphorylation inhibition by integrating activation of glycolytic pathway and repression of mtDNA genes. These findings have significant implications for understanding the pathogenesis and treatment of breast cancer.

ISCIENCE (2023)

Article Chemistry, Medicinal

Inhibition of cellular respiration by doxorubicin

Zhimin Tao, Henry G. Withers, Harvey S. Penefsky, Jerry Goodisman, Abdul-Kader Souid

CHEMICAL RESEARCH IN TOXICOLOGY (2006)

暂无数据