66 results on '"Jer-Yen Yang"'
Search Results
2. Supplementary Figures S1-S5 from Leptin–STAT3–G9a Signaling Promotes Obesity-Mediated Breast Cancer Progression
- Author
-
Chun-Ju Chang, Ignacio G. Camarillo, Jer-Yen Yang, Meng-Ju Wu, and Chao-Ching Chang
- Abstract
Supplementary Figures S1-S5. Leptin induces STAT3-G9a interaction and their co-occupancy on targeted gene promoters (S1); Leptin induces EMT and stem cell-like traits via down-regulation of miR-200c (S2); STAT3 and G9a expression is required for OBR induction by leptin (S3); Distinct tumor phenotypes in Western obese animals compared to lean animals (S4); STAT3 activation and OBR overexpression in diet-induced obesity rats (S5).
- Published
- 2023
3. Supplementary Figure 1 from Down-regulation of Myeloid Cell Leukemia-1 through Inhibiting Erk/Pin 1 Pathway by Sorafenib Facilitates Chemosensitization in Breast Cancer
- Author
-
Mien-Chie Hung, Chang-Hai Tsai, Long-Yuan Li, Fuu-Jen Tsai, Chun-Yi Lin, Hsu-Ping Kuo, Jung-Mao Hsu, Yun-Ju Rita Chen, Chia-Jui Yen, Dung-Fang Lee, Chien-Chen Lai, Yan Yang, Chun-Ju Chang, Yong Liao, Yongkun Wei, Weiya Xia, Jer-Yen Yang, Longfei Huo, and Qingqing Ding
- Abstract
Supplementary Figure 1 from Down-regulation of Myeloid Cell Leukemia-1 through Inhibiting Erk/Pin 1 Pathway by Sorafenib Facilitates Chemosensitization in Breast Cancer
- Published
- 2023
4. Supplemental Figure S1 from H3K9 Histone Methyltransferase, KMT1E/SETDB1, Cooperates with the SMAD2/3 Pathway to Suppress Lung Cancer Metastasis
- Author
-
Tsai-Yu Tzeng, C.-K. James Shen, Yueh-Min Lin, Liang-In Lin, Chiun Hsu, Yih-Leh Huang, Wan-Ping Wang, Wen-Feng Huang, Kuan-Hsien Chou, Yu-Hsiang Lin, Da-Liang Ou, I-Hsuan Lin, Jer-Yen Yang, Jeng-Wei Lu, and Pei-Chun Wu
- Abstract
Cell viability effects of KMT1E in lung cancer.
- Published
- 2023
5. Supplemental Materials and Methods from H3K9 Histone Methyltransferase, KMT1E/SETDB1, Cooperates with the SMAD2/3 Pathway to Suppress Lung Cancer Metastasis
- Author
-
Tsai-Yu Tzeng, C.-K. James Shen, Yueh-Min Lin, Liang-In Lin, Chiun Hsu, Yih-Leh Huang, Wan-Ping Wang, Wen-Feng Huang, Kuan-Hsien Chou, Yu-Hsiang Lin, Da-Liang Ou, I-Hsuan Lin, Jer-Yen Yang, Jeng-Wei Lu, and Pei-Chun Wu
- Abstract
revised supporting materials and methods
- Published
- 2023
6. Supplementary Figure Legends 1-3 from Activation of Murine Double Minute 2 by Akt in Mammary Epithelium Delays Mammary Involution and Accelerates Mammary Tumorigenesis
- Author
-
Mien-Chie Hung, Dihua Yu, William J. Muller, Gordon B. Mills, Shannon L. Wyszomierski, Hui-Lung Sun, Yi Du, Chao-Kai Chou, Jing-Yu Lang, Jennifer L. Hsu, Jer-Yen Yang, Weiya Xia, and Xiaoyun Cheng
- Abstract
Supplementary Figure Legends 1-3 from Activation of Murine Double Minute 2 by Akt in Mammary Epithelium Delays Mammary Involution and Accelerates Mammary Tumorigenesis
- Published
- 2023
7. Supplemental Figure Legends from H3K9 Histone Methyltransferase, KMT1E/SETDB1, Cooperates with the SMAD2/3 Pathway to Suppress Lung Cancer Metastasis
- Author
-
Tsai-Yu Tzeng, C.-K. James Shen, Yueh-Min Lin, Liang-In Lin, Chiun Hsu, Yih-Leh Huang, Wan-Ping Wang, Wen-Feng Huang, Kuan-Hsien Chou, Yu-Hsiang Lin, Da-Liang Ou, I-Hsuan Lin, Jer-Yen Yang, Jeng-Wei Lu, and Pei-Chun Wu
- Abstract
Supplemental Figure Legends
- Published
- 2023
8. Supplemental Figure S8 from H3K9 Histone Methyltransferase, KMT1E/SETDB1, Cooperates with the SMAD2/3 Pathway to Suppress Lung Cancer Metastasis
- Author
-
Tsai-Yu Tzeng, C.-K. James Shen, Yueh-Min Lin, Liang-In Lin, Chiun Hsu, Yih-Leh Huang, Wan-Ping Wang, Wen-Feng Huang, Kuan-Hsien Chou, Yu-Hsiang Lin, Da-Liang Ou, I-Hsuan Lin, Jer-Yen Yang, Jeng-Wei Lu, and Pei-Chun Wu
- Abstract
ANXA2 is highly expressed in metastatic cancer cells.
- Published
- 2023
9. Supplemental Table S1 from H3K9 Histone Methyltransferase, KMT1E/SETDB1, Cooperates with the SMAD2/3 Pathway to Suppress Lung Cancer Metastasis
- Author
-
Tsai-Yu Tzeng, C.-K. James Shen, Yueh-Min Lin, Liang-In Lin, Chiun Hsu, Yih-Leh Huang, Wan-Ping Wang, Wen-Feng Huang, Kuan-Hsien Chou, Yu-Hsiang Lin, Da-Liang Ou, I-Hsuan Lin, Jer-Yen Yang, Jeng-Wei Lu, and Pei-Chun Wu
- Abstract
KMT1E expression in 192 lung cancer as evaluated by tissue microarray-based immunohistochemistry
- Published
- 2023
10. Supplementary Tables S1-S6 from Leptin–STAT3–G9a Signaling Promotes Obesity-Mediated Breast Cancer Progression
- Author
-
Chun-Ju Chang, Ignacio G. Camarillo, Jer-Yen Yang, Meng-Ju Wu, and Chao-Ching Chang
- Abstract
Supplementary Tables S1-S6. List of proteins with expression levels significantly changed in MCF7 cells under leptin treatment using antibody array (S1); G9a is a new STAT3-associated protein under leptin treatment (S2); STAT3-G9a regulates a cohort of genes involved in cell differentiation process (S3); Body composition analysis from DEXA scans (S4); Categorization of tumor number and tumor type in each group of animals (S5); Elevated LEP, p-STAT3, and OBR levels are correlated with reduced miR-200c expression in diet-induced obesity and breast cancer rats (S6).
- Published
- 2023
11. Supplementary Figure 2 from Down-regulation of Myeloid Cell Leukemia-1 through Inhibiting Erk/Pin 1 Pathway by Sorafenib Facilitates Chemosensitization in Breast Cancer
- Author
-
Mien-Chie Hung, Chang-Hai Tsai, Long-Yuan Li, Fuu-Jen Tsai, Chun-Yi Lin, Hsu-Ping Kuo, Jung-Mao Hsu, Yun-Ju Rita Chen, Chia-Jui Yen, Dung-Fang Lee, Chien-Chen Lai, Yan Yang, Chun-Ju Chang, Yong Liao, Yongkun Wei, Weiya Xia, Jer-Yen Yang, Longfei Huo, and Qingqing Ding
- Abstract
Supplementary Figure 2 from Down-regulation of Myeloid Cell Leukemia-1 through Inhibiting Erk/Pin 1 Pathway by Sorafenib Facilitates Chemosensitization in Breast Cancer
- Published
- 2023
12. Supplementary Figure 3 from Down-regulation of Myeloid Cell Leukemia-1 through Inhibiting Erk/Pin 1 Pathway by Sorafenib Facilitates Chemosensitization in Breast Cancer
- Author
-
Mien-Chie Hung, Chang-Hai Tsai, Long-Yuan Li, Fuu-Jen Tsai, Chun-Yi Lin, Hsu-Ping Kuo, Jung-Mao Hsu, Yun-Ju Rita Chen, Chia-Jui Yen, Dung-Fang Lee, Chien-Chen Lai, Yan Yang, Chun-Ju Chang, Yong Liao, Yongkun Wei, Weiya Xia, Jer-Yen Yang, Longfei Huo, and Qingqing Ding
- Abstract
Supplementary Figure 3 from Down-regulation of Myeloid Cell Leukemia-1 through Inhibiting Erk/Pin 1 Pathway by Sorafenib Facilitates Chemosensitization in Breast Cancer
- Published
- 2023
13. Supplemental Table S3 from H3K9 Histone Methyltransferase, KMT1E/SETDB1, Cooperates with the SMAD2/3 Pathway to Suppress Lung Cancer Metastasis
- Author
-
Tsai-Yu Tzeng, C.-K. James Shen, Yueh-Min Lin, Liang-In Lin, Chiun Hsu, Yih-Leh Huang, Wan-Ping Wang, Wen-Feng Huang, Kuan-Hsien Chou, Yu-Hsiang Lin, Da-Liang Ou, I-Hsuan Lin, Jer-Yen Yang, Jeng-Wei Lu, and Pei-Chun Wu
- Abstract
KMT1E DNA binding sequences identified by ChIP-Seq.
- Published
- 2023
14. Supplementary Methods, Figure 1, Table 1 from Blockade of Mitogen-Activated Protein Kinase/Extracellular Signal-Regulated Kinase Kinase and Murine Double Minute Synergistically Induces Apoptosis in Acute Myeloid Leukemia via BH3-Only Proteins Puma and Bim
- Author
-
Michael Andreeff, Mien-Chie Hung, Teresa J. McQueen, Jer-Yen Yang, Wendy D. Schober, Karen C. Dywer, Jared K. Burks, Marina Konopleva, and Weiguo Zhang
- Abstract
Supplementary Methods, Figure 1, Table 1 from Blockade of Mitogen-Activated Protein Kinase/Extracellular Signal-Regulated Kinase Kinase and Murine Double Minute Synergistically Induces Apoptosis in Acute Myeloid Leukemia via BH3-Only Proteins Puma and Bim
- Published
- 2023
15. Supplemental Table S2 from H3K9 Histone Methyltransferase, KMT1E/SETDB1, Cooperates with the SMAD2/3 Pathway to Suppress Lung Cancer Metastasis
- Author
-
Tsai-Yu Tzeng, C.-K. James Shen, Yueh-Min Lin, Liang-In Lin, Chiun Hsu, Yih-Leh Huang, Wan-Ping Wang, Wen-Feng Huang, Kuan-Hsien Chou, Yu-Hsiang Lin, Da-Liang Ou, I-Hsuan Lin, Jer-Yen Yang, Jeng-Wei Lu, and Pei-Chun Wu
- Abstract
Clinicopathological characteristics of patients with lung cancer.
- Published
- 2023
16. Supplementary Figure Legends 1-10 from Activation of FOXO3a Is Sufficient to Reverse Mitogen-Activated Protein/Extracellular Signal-Regulated Kinase Kinase Inhibitor Chemoresistance in Human Cancer
- Author
-
Mien-Chie Hung, Gabriel N. Hortobagyi, Michael Andreeff, Yi Du, Jeffrey A. Engelman, Kwok-Kin Wong, Yan Wang, Weiya Xia, Chun-Ju Chang, and Jer-Yen Yang
- Abstract
Supplementary Figure Legends 1-10 from Activation of FOXO3a Is Sufficient to Reverse Mitogen-Activated Protein/Extracellular Signal-Regulated Kinase Kinase Inhibitor Chemoresistance in Human Cancer
- Published
- 2023
17. Supplementary Figures 1-10 from Activation of FOXO3a Is Sufficient to Reverse Mitogen-Activated Protein/Extracellular Signal-Regulated Kinase Kinase Inhibitor Chemoresistance in Human Cancer
- Author
-
Mien-Chie Hung, Gabriel N. Hortobagyi, Michael Andreeff, Yi Du, Jeffrey A. Engelman, Kwok-Kin Wong, Yan Wang, Weiya Xia, Chun-Ju Chang, and Jer-Yen Yang
- Abstract
Supplementary Figures 1-10 from Activation of FOXO3a Is Sufficient to Reverse Mitogen-Activated Protein/Extracellular Signal-Regulated Kinase Kinase Inhibitor Chemoresistance in Human Cancer
- Published
- 2023
18. Supplemental Figure S4 from H3K9 Histone Methyltransferase, KMT1E/SETDB1, Cooperates with the SMAD2/3 Pathway to Suppress Lung Cancer Metastasis
- Author
-
Tsai-Yu Tzeng, C.-K. James Shen, Yueh-Min Lin, Liang-In Lin, Chiun Hsu, Yih-Leh Huang, Wan-Ping Wang, Wen-Feng Huang, Kuan-Hsien Chou, Yu-Hsiang Lin, Da-Liang Ou, I-Hsuan Lin, Jer-Yen Yang, Jeng-Wei Lu, and Pei-Chun Wu
- Abstract
ANXA2 is required for rescue filopodia formation in lung cancer cells.
- Published
- 2023
19. Supplemental Figure S5 from H3K9 Histone Methyltransferase, KMT1E/SETDB1, Cooperates with the SMAD2/3 Pathway to Suppress Lung Cancer Metastasis
- Author
-
Tsai-Yu Tzeng, C.-K. James Shen, Yueh-Min Lin, Liang-In Lin, Chiun Hsu, Yih-Leh Huang, Wan-Ping Wang, Wen-Feng Huang, Kuan-Hsien Chou, Yu-Hsiang Lin, Da-Liang Ou, I-Hsuan Lin, Jer-Yen Yang, Jeng-Wei Lu, and Pei-Chun Wu
- Abstract
ANXA2 and SMAD3 promotes F-actin polymerization formation in lung cancer cells.
- Published
- 2023
20. Supplemental Figure S3 from H3K9 Histone Methyltransferase, KMT1E/SETDB1, Cooperates with the SMAD2/3 Pathway to Suppress Lung Cancer Metastasis
- Author
-
Tsai-Yu Tzeng, C.-K. James Shen, Yueh-Min Lin, Liang-In Lin, Chiun Hsu, Yih-Leh Huang, Wan-Ping Wang, Wen-Feng Huang, Kuan-Hsien Chou, Yu-Hsiang Lin, Da-Liang Ou, I-Hsuan Lin, Jer-Yen Yang, Jeng-Wei Lu, and Pei-Chun Wu
- Abstract
KMT1E inhibits filopodia formation in sublines cells.
- Published
- 2023
21. Supplemental Figure S7 from H3K9 Histone Methyltransferase, KMT1E/SETDB1, Cooperates with the SMAD2/3 Pathway to Suppress Lung Cancer Metastasis
- Author
-
Tsai-Yu Tzeng, C.-K. James Shen, Yueh-Min Lin, Liang-In Lin, Chiun Hsu, Yih-Leh Huang, Wan-Ping Wang, Wen-Feng Huang, Kuan-Hsien Chou, Yu-Hsiang Lin, Da-Liang Ou, I-Hsuan Lin, Jer-Yen Yang, Jeng-Wei Lu, and Pei-Chun Wu
- Abstract
KMT1E modulates ANXA2 expression to suppress cancer cell invasion in lung cancer cells.
- Published
- 2023
22. Supplementary Methods from Leptin–STAT3–G9a Signaling Promotes Obesity-Mediated Breast Cancer Progression
- Author
-
Chun-Ju Chang, Ignacio G. Camarillo, Jer-Yen Yang, Meng-Ju Wu, and Chao-Ching Chang
- Abstract
Supplementary Methods. Description of additional methods and procedures used in the study.
- Published
- 2023
23. PRMT1 is an important factor for medulloblastoma cell proliferation and survival
- Author
-
Xiao Gu, Miao He, Timofey Lebedev, Cheng-Han Lin, Zhong-Yan Hua, Y. George Zheng, Zhi-Jie Li, Jer-Yen Yang, and Xing-Guo Li
- Subjects
Biophysics ,Biochemistry - Abstract
Aberrant expression of protein arginine methyltransferases (PRMTs) has been implicated in a number of brain tumors, but the role of PRMT1 in medulloblastoma, the most common malignant pediatric brain tumor, remains unexplored
- Published
- 2022
24. Hedgehog Pathway Inhibitors against Tumor Microenvironment
- Author
-
Silpa Gampala and Jer-Yen Yang
- Subjects
QH301-705.5 ,Angiogenesis ,medicine.medical_treatment ,Antineoplastic Agents ,Review ,HH pathway inhibitors ,Immune system ,hedgehog pathway ,Glioma ,medicine ,Tumor Microenvironment ,Animals ,Humans ,cancer ,Hedgehog Proteins ,Biology (General) ,Hedgehog ,Immune Checkpoint Inhibitors ,Tumor microenvironment ,drug resistance ,business.industry ,Immunity ,Cancer ,General Medicine ,Immunotherapy ,medicine.disease ,Hedgehog signaling pathway ,Cancer research ,immunotherapy ,business ,Signal Transduction - Abstract
Targeting the hedgehog (HH) pathway to treat aggressive cancers of the brain, breast, pancreas, and prostate has been ongoing for decades. Gli gene amplifications have been long discovered within malignant glioma patients, and since then, inhibitors against HH pathway-associated molecules have successfully reached the clinical stage where several of them have been approved by the FDA. Albeit this success rate implies suitable progress, clinically used HH pathway inhibitors fail to treat patients with metastatic or recurrent disease. This is mainly due to heterogeneous tumor cells that have acquired resistance to the inhibitors along with the obstacle of effectively targeting the tumor microenvironment (TME). Severe side effects such as hyponatremia, diarrhea, fatigue, amenorrhea, nausea, hair loss, abnormal taste, and weight loss have also been reported. Furthermore, HH signaling is known to be involved in the regulation of immune cell maturation, angiogenesis, inflammation, and polarization of macrophages and myeloid-derived suppressor cells. It is critical to determine key mechanisms that can be targeted at different levels of tumor development and progression to address various clinical issues. Hence current research focus encompasses understanding how HH controls TME to develop TME altering and combinatorial targeting strategies. In this review, we aim to discuss the pros and cons of targeting HH signaling molecules, understand the mechanism involved in treatment resistance, reveal the role of the HH pathway in anti-tumor immune response, and explore the development of potential combination treatment of immune checkpoint inhibitors with HH pathway inhibitors to target HH-driven cancers.
- Published
- 2021
25. TET2 directs mammary luminal cell differentiation and endocrine response
- Author
-
Jer-Yen Yang, Chun Ju Chang, Mi Ran Kim, Meng-Ju Wu, and Yingsheng Zhang
- Subjects
0301 basic medicine ,Mammary stem cells ,Science ,Cellular differentiation ,General Physics and Astronomy ,Breast Neoplasms ,Endocrine System ,Biology ,medicine.disease_cause ,Article ,General Biochemistry, Genetics and Molecular Biology ,Dioxygenases ,Epigenesis, Genetic ,Mice ,03 medical and health sciences ,Mammary Glands, Animal ,Breast cancer ,0302 clinical medicine ,Proto-Oncogene Proteins ,medicine ,Animals ,Humans ,Cell Lineage ,lcsh:Science ,Mice, Knockout ,Mammary tumor ,Multidisciplinary ,Estradiol ,Estrogen Receptor alpha ,GATA3 ,Cell Differentiation ,Estrogens ,General Chemistry ,DNA Methylation ,Cell biology ,DNA-Binding Proteins ,030104 developmental biology ,DNA demethylation ,030220 oncology & carcinogenesis ,DNA methylation ,lcsh:Q ,Female ,Stem cell ,FOXA1 ,Carcinogenesis - Abstract
Epigenetic regulation plays an important role in governing stem cell fate and tumorigenesis. Lost expression of a key DNA demethylation enzyme TET2 is associated with human cancers and has been linked to stem cell traits in vitro; however, whether and how TET2 regulates mammary stem cell fate and mammary tumorigenesis in vivo remains to be determined. Here, using our recently established mammary specific Tet2 deletion mouse model, the data reveals that TET2 plays a pivotal role in mammary gland development and luminal lineage commitment. We show that TET2 and FOXP1 form a chromatin complex that mediates demethylation of ESR1, GATA3, and FOXA1, three key genes that are known to coordinately orchestrate mammary luminal lineage specification and endocrine response, and also are often silenced by DNA methylation in aggressive breast cancers. Furthermore, Tet2 deletion-PyMT breast cancer mouse model exhibits enhanced mammary tumor development with deficient ERα expression that confers tamoxifen resistance in vivo. As a result, this study elucidates a role for TET2 in governing luminal cell differentiation and endocrine response that underlies breast cancer resistance to anti-estrogen treatments., TET2 loss is associated with human cancers but its role in the mammary gland development and tumorigenesis is unclear. Here, the authors show that TET2–FOXP1 complex mediates demethylation of genes involved in luminal lineage commitment and endocrine response, underlying a role of TET2 loss in endocrine resistant breast cancer.
- Published
- 2020
26. Identification of RECK as an evolutionarily conserved tumor suppressor gene for zebrafish malignant peripheral nerve sheath tumors
- Author
-
Suresh K. Mittal, Rashmi Kumari, Yava L. Jones-Hall, Martin R. Silic, Alexandra Nin-Velez, Jer Yen Yang, and GuangJun Zhang
- Subjects
0303 health sciences ,Tumor suppressor gene ,Oncogenomics ,Gene mutation ,Biology ,medicine.disease_cause ,biology.organism_classification ,3. Good health ,03 medical and health sciences ,0302 clinical medicine ,Oncology ,CDKN2A ,030220 oncology & carcinogenesis ,Genetic model ,medicine ,Cancer research ,Carcinogenesis ,Gene ,Zebrafish ,030304 developmental biology - Abstract
Malignant peripheral nerve sheath tumors (MPNSTs) are a type of sarcoma with poor prognosis due to their complex genetic changes, invasive growth, and insensitivity to chemo- and radiotherapies. One of the most frequently lost chromosome arms in human MPNSTs is chromosome 9p. However, the cancer driver genes located on it remain largely unknown, except the tumor suppressor gene, p16 (INK4)/CDKN2A. Previously, we identified RECK as a tumor suppressor gene candidate on chromosome 9p using zebrafish-human comparative oncogenomics. In this study, we investigated the tumorigenesis of the reck gene using zebrafish genetic models in both tp53 and ribosomal protein gene mutation background. We also examined the biological effects of RECK gene restoration in human MPNST cell lines. These results provide the first genetic evidence that reck is a bona fide tumor suppressor gene for MPNSTs in zebrafish. In addition, restoration of the RECK gene in human MPNST cells leads to growth inhibition suggesting that the reactivation of RECK could serve as a potential therapeutic strategy for MPNSTs.
- Published
- 2018
27. Dual degradation signals destruct GLI1: AMPK inhibits GLI1 through β-TrCP-mediated proteasome degradation
- Author
-
Yen Hsing Li, Chun Ju Chang, Jer Yen Yang, Rui Zhang, GuangJun Zhang, Wei Hsuan Hsu, Kay Ka Wai Li, and Sherri Y. Huang
- Subjects
AMPK ,0301 basic medicine ,Proteasome Endopeptidase Complex ,medicine.medical_specialty ,Cellular pathology ,Beta-Transducin Repeat-Containing Proteins ,GLI1 ,Active Transport, Cell Nucleus ,Gene Expression ,AMP-Activated Protein Kinases ,medulloblastoma ,Zinc Finger Protein GLI1 ,Mice ,03 medical and health sciences ,Ubiquitin ,AMP-activated protein kinase ,Cell Line, Tumor ,Neoplasms ,Internal medicine ,β-transducin repeat containing protein (β-TrCP) ,medicine ,Animals ,Humans ,Phosphorylation ,Cell Proliferation ,integumentary system ,biology ,Ubiquitination ,beta-Transducin Repeat-Containing Proteins ,Hedgehog signaling pathway ,3. Good health ,Cell biology ,Protein Transport ,030104 developmental biology ,Endocrinology ,Oncology ,Proteolysis ,biology.protein ,Signal transduction ,Hedgehog ,Research Paper ,Signal Transduction - Abstract
// Rui Zhang 1, * , Sherri Y. Huang 1, * , Kay Ka-Wai Li 2 , Yen-Hsing Li 1 , Wei-Hsuan Hsu 1 , Guang Jun Zhang 3, 4 , Chun-Ju Chang 1, 3 and Jer-Yen Yang 1, 3 1 Department of Basic Medical Sciences, West Lafayette, Indiana, USA 2 5/F of Cancer Centre, Prince of Wales Hospital, Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Shatin, Hong Kong 3 Center for Cancer Research, Purdue University, West Lafayette, Indiana, USA 4 Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana, USA * These authors contributed equally to this work Correspondence to: Jer-Yen Yang, email: jyyang@purdue.edu Keywords: AMPK, β-transducin repeat containing protein (β-TrCP), Hedgehog, GLI1, medulloblastoma Received: December 15, 2016 Accepted: April 05, 2017 Published: May 10, 2017 ABSTRACT Overexpression of the GLI1 gene has frequently been found in various cancer types, particularly in brain tumors, in which aberrant GLI1 induction promotes cancer cell growth. Therefore, identifying the molecular players controlling GLI1 expression is of clinical importance. Previously, we reported that AMPK directly phosphorylated and destabilized GLI1, resulting in the suppression of the Hedgehog signaling pathway. The current study not only demonstrates that AMPK inhibits GLI1 nuclear localization, but further reveals that β-TrCP plays an essential role in AMPK-induced GLI1 degradation. We found that activation of AMPK promotes the interaction between β-TrCP and GLI1, and induces β-TrCP-mediated GLI1-ubiquitination and degradation. Inhibiting AMPK activity results in the dissociation of the β-TrCP and GLI1 interaction, and diminishes β-TrCP-mediated-GLI1 ubiquitination and degradation. On GLI1, substitution of AMPK phosphorylation sites to aspartic acid (GLI1 3E ) results in stronger binding affinity of GLI1 with β-TrCP, accompanied by enhanced GLI1 ubiquitination and later degradation. In contrast, the GLI1 alanine mutant (GLI1 3A ) shows weaker binding with β-TrCP, which is accompanied by reduced β-TrCP-mediated ubiquitination and degradation. Together, these results demonstrate that AMPK regulates GLI1 interaction with β-TrCP by phosphorylating GLI1 and thus both post-translational modifications by AMPK and β-TrCP ultimately impact GLI1 degradation.
- Published
- 2017
28. Targeting the Sonic Hedgehog Pathway in Brain Cancers: Advances, Limitations, and Future Directions
- Author
-
Jer Yen Yang and Sherri Y. Huang
- Subjects
animal structures ,biology ,business.industry ,Malignant brain tumor ,Brain tumor ,Vismodegib ,Cancer ,medicine.disease ,Hedgehog signaling pathway ,Sonidegib ,nervous system diseases ,Brain cancer ,chemistry.chemical_compound ,chemistry ,embryonic structures ,biology.protein ,Cancer research ,Medicine ,Sonic hedgehog ,business ,medicine.drug - Abstract
Aberrant signaling of the sonic hedgehog (SHH) pathway can result in increased cellular proliferation and cancer. SHH subgroup medulloblastomas (MBs) comprise about a third of the molecular types of this malignant brain tumor, and unfortunately drug resistance limits the efficacy of vismodegib and sonidegib, the only FDA-approved therapies directly targeting the SHH pathway. Emerging literature also suggests important roles of SHH signaling in meningiomas and in glioblastoma multiforme (GBM), the latter being an extremely aggressive tumor and the most common primary brain tumor in adults. This chapter illustrates the roles of the SHH pathway in the pathogenesis of MB, GBM, and meningiomas. In addition, we will cover the existing challenges with current drug therapies and detail recent findings in the regulation of the SHH pathway, findings which advance the discovery of novel drug therapies.
- Published
- 2018
29. Contributors
- Author
-
Rolla Abu-Arja, Ana Aguilar-Bonilla, Manmeet S. Ahluwalia, Sheikh C. Ali, Deborah H. Allen, Iyad Alnahhas, Gregory D. Arnone, Pascal Auquier, Nicholas G. Avgeropoulos, Kristen A. Batich, Karine Baumstarck, Susan D. Bell, Viviana Benitez, Deborah T. Blumenthal, Miriam Bornhorst, Laurent Boyer, Alexa N. Bramall, Sarah Braun, Henry Brem, William C. Broaddus, Alberto Broniscer, Nicholas Butowski, Soonmee Cha, Marc C. Chamberlain, Susan Chang, Alain Charest, Zhi-Jian Chen, Sajeel Chowdhary, Jennifer L. Clarke, Adam L. Cohen, Howard Colman, Michael Cooper, Jennifer Cotter, Sunit Das, Kuntal De, Macarena I. De la Fuente, Anand K. Deo, Annick Desjardins, Girish Dhall, Nancy D. Doolittle, Kira M. Downey, Gavin P. Dunn, Herbert H. Engelhard, Emilie Ernoult, Richard G. Everson, Robert A. Fenstermaker, Isabelle Ferry, Sheila Figel, Karen L. Fink, Jonathan L. Finlay, Joanna E. Gernsback, Pierre Giglio, Noah Gorelick, Daphne Haas-Kogan, Jan T. Hachmann, Hasan Hashem, John W. Henson, Sherri Y. Huang, Mai Anh Huynh, Eugene I. Hwang, Eric Jackson, Tanner M. Johanns, Liron Kahlon, Deepak Kanojia, Matthias A. Karajannis, Balveen Kaur, Santosh Kesari, Lindsay B. Kilburn, Julius W. Kim, Cassie Kline, John Paul G. Kolcun, Ricardo J. Komotar, Matthew Koshy, Claudia M. Kuzan-Fischer, Sajani S. Lakka, Autumn Lanoye, George P. Lee, Eudocia Q. Lee, Maciej S. Lesniak, Linda M. Liau, Ashlee R. Loughan, Mark G. Malkin, Brendan J. McCullough, John M. McGregor, Ankit I. Mehta, Julie J. Miller, Nimish Mohile, Paul L. Moots, Sabine Mueller, Joydeep Mukherjee, Tulio P. Murillo, Megan E. Muroski, Edward A. Neuwelt, Herbert B. Newton, Martin K. Nicholas, Nancyann Oberheim Bush, Hideho Okada, Antonio M. Omuro, Roger J. Packer, Nina A. Paleologos, Wojciech K. Panek, Sebastian P. Pernal, Emily Owens Pickle, Katarzyna C. Pituch, Ian F. Pollack, Jeffrey Raizer, Aida Rashidi, David A. Reardon, Alyssa T. Reddy, J. Robert Kane, James T. Rutka, John H. Sampson, Mithun Sattur, Adrienne C. Scheck, Brian J. Scott, Wendy L. See, Nawal Shaikh, Mayur Sharma, Tolou Shokuhfar, Seema Shroff, Amy A. Smith, Subanandhini Subramaniam, Carter M. Suryadevara, Kenneth D. Swanson, Benita Tamrazi, Shervin Taslimi, Beverly Teicher, Hung N. Tran, Betty Tyler, Santhosh A. Upadhyaya, Rafael A. Vega, Monica Venere, Michael Vogelbaum, Diane Von Ah, Barliz Waissengrin, Emily S. Waite, Eric T. Wong, Annie Xiao, Jer-Yen Yang, Jacob S. Young, Ji Young Yoo, and Wafik Zaky
- Published
- 2018
30. Targeting the Hedgehog Pathway in Pediatric Medulloblastoma
- Author
-
Jer Yen Yang and Sherri Y. Huang
- Subjects
Medulloblastoma ,Cancer Research ,Pathology ,medicine.medical_specialty ,Chemotherapy ,Oncogene ,hedgehog ,Gli1 ,medicine.medical_treatment ,Review ,Biology ,medulloblastoma ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,lcsh:RC254-282 ,Hedgehog signaling pathway ,Crosstalk (biology) ,Oncology ,GLI1 ,Cancer research ,medicine ,biology.protein ,Hedgehog ,Transcription factor - Abstract
Medulloblastoma (MB), a primitive neuroectomal tumor of the cerebellum, is the most common malignant pediatric brain tumor. The cause of MB is largely unknown, but aberrant activation of Hedgehog (Hh) pathway is responsible for ~30% of MB. Despite aggressive treatment with surgical resection, radiation and chemotherapy, 70%–80% of pediatric medulloblastoma cases can be controlled, but most treated patients suffer devastating side effects. Therefore, developing a new effective treatment strategy is urgently needed. Hh signaling controls transcription of target genes by regulating activities of the three Glioma-associated oncogene (Gli1-3) transcription factors. In this review, we will focus on current clinical treatment options of MB and discuss mechanisms of drug resistance. In addition, we will describe current known molecular pathways which crosstalk with the Hedgehog pathway both in the context of medulloblastoma and non-medulloblastoma cancer development. Finally, we will introduce post-translational modifications that modulate Gli1 activity and summarize the positive and negative regulations of the Hh/Gli1 pathway. Towards developing novel combination therapies for medulloblastoma treatment, current information on interacting pathways and direct regulation of Hh signaling should prove critical.
- Published
- 2015
31. AMP-Activated Protein Kinase Directly Phosphorylates and Destabilizes Hedgehog Pathway Transcription Factor GLI1 in Medulloblastoma
- Author
-
Anne Brunet, Matthew P. Scott, Jer Yen Yang, Chun Ju Chang, Shihuan Kuang, GuangJun Zhang, Jen-Leih Wu, Yu-Kuo Wang, Jia Luo, Yen-Hsing Li, Victoria Hedrick, Julia Chang, Max R. Banko, Lake N. Paul, and Yung-Yi C. Mosley
- Subjects
Medical Sciences ,Cellular differentiation ,Molecular Sequence Data ,AMP-Activated Protein Kinases ,Zinc Finger Protein GLI1 ,Article ,General Biochemistry, Genetics and Molecular Biology ,Mice ,AMP-activated protein kinase ,GLI1 ,Cell Line, Tumor ,Animals ,Humans ,Amino Acid Sequence ,Phosphorylation ,Protein kinase A ,lcsh:QH301-705.5 ,Transcription factor ,Zebrafish ,biology ,Protein Stability ,AMPK ,3T3 Cells ,Hedgehog signaling pathway ,HEK293 Cells ,lcsh:Biology (General) ,Biochemistry ,biology.protein ,Protein Processing, Post-Translational ,Medulloblastoma ,Transcription Factors - Abstract
The Hedgehog (Hh) pathway regulates cell differen- tiation and proliferation during development by controlling the Gli transcription factors. Cell fate de- cisions and progression toward organ and tissue maturity must be coordinated, and how an energy sensor regulates the Hh pathway is not clear. AMP- activated protein kinase (AMPK) is an important sensor of energy stores and controls protein synthe- sis and other energy-intensive processes. AMPK is directly responsive to intracellular AMP levels, inhib- iting a wide range of cell activities if ATP is low and AMP is high. Thus, AMPK can affect development by influencing protein synthesis and other processes needed for growth and differentiation. Activation of AMPK reduces GLI1 protein levels and stability, thus blocking Sonic-hedgehog-induced transcrip- tional activity. AMPK phosphorylates GLI1 at serines 102 and 408 and threonine 1074. Mutation of these three sites into alanine prevents phosphorylation by AMPK. This leads to increased GLI1 protein stability, transcriptional activity, and oncogenic potency.
- Published
- 2015
- Full Text
- View/download PDF
32. H3K9 Histone Methyltransferase, KMT1E/SETDB1, Cooperates with the SMAD2/3 Pathway to Suppress Lung Cancer Metastasis
- Author
-
Wen-Feng Huang, Kuan-Hsien Chou, Jer Yen Yang, Da-Liang Ou, C. K. James Shen, Pei-Chun Wu, Chiun Hsu, Liang-In Lin, I-Hsuan Lin, Tsai-Yu Tzeng, Yu Hsiang Lin, Yih-Leh Huang, Jeng-Wei Lu, Yueh-Min Lin, and Wan-Ping Wang
- Subjects
Cancer Research ,Lung Neoplasms ,Methyltransferase ,Smad2 Protein ,Methylation ,Epigenesis, Genetic ,Metastasis ,Cell Line, Tumor ,Histone methylation ,medicine ,Animals ,Humans ,Metastasis suppressor ,Gene Silencing ,Protein Methyltransferases ,Smad3 Protein ,Neoplasm Metastasis ,Promoter Regions, Genetic ,Lung cancer ,Annexin A2 ,Zebrafish ,biology ,Histone-Lysine N-Methyltransferase ,medicine.disease ,Xenograft Model Antitumor Assays ,Histone ,Oncology ,Histone methyltransferase ,Cancer cell ,Cancer research ,biology.protein ,Signal Transduction - Abstract
Aberrant histone methylation is a frequent event during tumor development and progression. KMT1E (also known as SETDB1) is a histone H3K9 methyltransferase that contributes to epigenetic silencing of both oncogenes and tumor suppressor genes in cancer cells. In this report, we demonstrate that KMT1E acts as a metastasis suppressor that is strongly downregulated in highly metastatic lung cancer cells. Restoring KMT1E expression in this setting suppressed filopodia formation, migration, and invasive behavior. Conversely, loss of KMT1E in lung cancer cells with limited metastatic potential promoted migration in vitro and restored metastatic prowess in vivo. Mechanistic investigations indicated that KMT1E cooperates with the TGFβ-regulated complex SMAD2/3 to repress metastasis through ANXA2. Together, our findings defined an essential role for the KMT1E/SMAD2/3 repressor complex in TGFβ-mediated lung cancer metastasis. Cancer Res; 74(24); 7333–43. ©2014 AACR.
- Published
- 2014
33. Corrigendum: KANK1 inhibits cell growth by inducing apoptosis through regulating CXXC5 in human malignant peripheral nerve sheath tumors
- Author
-
Kenny H Chen, Suresh K. Mittal, Zhibin Cui, GuangJun Zhang, Jer Yen Yang, and Yingjia Shen
- Subjects
0301 basic medicine ,03 medical and health sciences ,030104 developmental biology ,Multidisciplinary ,business.industry ,Apoptosis ,Cell growth ,Cancer research ,Peripheral Nerve Sheath Tumors ,Medicine ,business ,Article - Abstract
Malignant peripheral nerve sheath tumors (MPNSTs) are a type of rare sarcomas with a poor prognosis due to its highly invasive nature and limited treatment options. Currently there is no targeted-cancer therapy for this type of malignancy. Thus, it is important to identify more cancer driver genes that may serve as targets of cancer therapy. Through comparative oncogenomics, we have found that KANK1 was a candidate tumor suppressor gene (TSG) for human MPNSTs. Although KANK1 is known as a cytoskeleton regulator, its tumorigenic function in MPNSTs remains largely unknown. In this study, we report that restoration of KANK1 in human MPNST cells inhibits cell growth both in human cell culture and xenograft mice by increasing apoptosis. Consistently, knockdown of KANK1 in neurofibroma cells promoted cell growth. Using RNA-seq analysis, we identified CXXC5 and other apoptosis-related genes, and demonstrated that CXXC5 is regulated by KANK1. Knockdown of CXXC5 was found to diminish KANK1-induced apoptosis in MPNST cells. Thus, KANK1 inhibits MPNST cell growth though CXXC5 mediated apoptosis. Our results suggest that KANK1 may function as a tumor suppressor in human MPNSTs, and thus it may be useful for targeted therapy.
- Published
- 2017
34. UV-induced effects on toxicity of model disinfection byproducts
- Author
-
Ernest R. Blatchley, Jer Yen Yang, Shih Chi Weng, and Yen-Hsing Li
- Subjects
chemistry.chemical_classification ,Chlorinated water ,Environmental Engineering ,0208 environmental biotechnology ,02 engineering and technology ,010501 environmental sciences ,medicine.disease_cause ,01 natural sciences ,Pollution ,Organic compound ,020801 environmental engineering ,Potable water ,chemistry ,Environmental chemistry ,Toxicity ,medicine ,Environmental Chemistry ,Water treatment ,Photodegradation ,Cytotoxicity ,Waste Management and Disposal ,Genotoxicity ,0105 earth and related environmental sciences - Abstract
UV (Ultraviolet)-based treatment has been demonstrated to be effective for removal of some disinfection byproducts (DBPs) and to be beneficial for reduction of genotoxicity and cytotoxicity in chlorinated water. However, to a large extent, UV-induced effects on chemistry and toxicology have been treated as a black box, in the sense that little or no UV dose-dependent behavior has been reported. To address this issue, the effects of UV254 irradiation on 1,4-dibenzoquinone (BQ), 2,6-dichloro-1,4-benzoquinone (DCBQ), and chlorocreatinine (Cl-Cre) as model DBPs were examined, both in terms of photodegradation and cytotoxicity. These compounds have been identified as DBPs that are relevant in swimming pool settings; however, these compounds will be relevant in other water treatment settings, including drinking water production and wastewater reuse. UV254 irradiation was shown to promote photodecay of all three compounds. However, for BQ and DCBQ, the corresponding cytotoxicity of the UV-irradiated samples remained essentially unchanged, even when the compound was completely photodegraded. These results indicate that the photodegradation products of BQ and DCBQ carry similar cytotoxicity as their respective parent compounds. On the other hand, UV254-irradiation of Cl-Cre yielded a decrease in cytotoxicity that correlated with photodechlorination of Cl-Cre. These experiments also demonstrated a reduction in cytotoxicity in connection with photodechlorination of an N-chlorinated organic compound. Overall, the results of these experiments indicate the importance of defining products of UV photodecay processes, both in terms of chemistry and toxicity; these attributes are expected to be important in many UV-based applications, including potable water production, water reuse, and recreational water settings.
- Published
- 2017
35. Activation of AMPK inhibits medulloblastoma cell growth and Gli1 activity
- Author
-
Sherri Y Huang, Jer Yen Yang, and Shau-Kwuan Chen
- Subjects
Medulloblastoma ,biology ,GLI1 ,Cell growth ,Chemistry ,biology.protein ,medicine ,Cancer research ,AMPK ,General Medicine ,medicine.disease - Published
- 2017
36. Abstract 798: Epithelial-mesenchymal transition directs stem cell polarity via regulation of mitofusin
- Author
-
Chun Ju Chang, Silpa Gampala, Yueyang Wang, Meng-Ju Wu, Yingsheng Zhang, Jer Yen Yang, and Mi Ran Kim
- Subjects
Cancer Research ,Oncology ,mitochondrial fusion ,Stem cell fate determination ,Chemistry ,Organelle ,NUMB ,MFN1 ,Epithelial–mesenchymal transition ,Stem cell ,Mitochondrion ,Cell biology - Abstract
Mitochondria are dynamic organelles that have been linked to stem cell homeostasis. However, the mechanism involved in mitochondrial regulation of stem cell fate determination remain elusive. Here we discover that epithelial-mesenchymal transition (EMT), a key process in cancer progression, induces mitochondrial fusion through regulation of miR200c-PGC1a-MFN1 pathway. EMT-activated MFN1 forms a complex with PKCz and is required for PKCz-mediated NUMB phosphorylation and dissociation from the cortical membrane to direct asymmetric division of mammary stem cells, where fused mitochondria are tethered by MFN1-PKCz to the cortical membrane and asymmetrically segregated to the stem cell-like progeny with enhanced glutathione synthesis and reactive oxygen species scavenging capacities, allowing sustaining of a self-renewing stem cell pool. Suppression of MFN1 expression leads to equal distribution of the fragmented mitochondria in both progenies that undergo symmetric luminal cell differentiation. Together, this study elucidates a novel role of mitofusin in stem cell fate determination to mediate EMT-associated stemness and provides therapeutic implications for targeting EMT-induced tumor stem cell populations. Citation Format: Meng-Ju Wu, Mi Ran Kim, Silpa Gampala, Yingsheng Zhang, Yueyang Wang, Jer-Yen Yang, Chun-Ju Chang. Epithelial-mesenchymal transition directs stem cell polarity via regulation of mitofusin [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 798.
- Published
- 2019
37. Epithelial-Mesenchymal Transition Directs Stem Cell Polarity via Regulation of Mitofusin
- Author
-
Mi Ran Kim, Yu Syuan Chen, Silpa Gampala, Meng-Ju Wu, Jer Yen Yang, Chun Ju Chang, Chih Yu Chang, Chao Ching Chang, Yingsheng Zhang, and Yueyang Wang
- Subjects
0301 basic medicine ,Epithelial-Mesenchymal Transition ,Physiology ,Mitochondrion ,Mitochondrial Membrane Transport Proteins ,Cell Line ,GTP Phosphohydrolases ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Organelle ,Animals ,Humans ,MFN1 ,Epithelial–mesenchymal transition ,Molecular Biology ,Mice, Knockout ,Chemistry ,Stem Cells ,Cell Polarity ,Cell Biology ,Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha ,Mitochondria ,Cell biology ,MicroRNAs ,030104 developmental biology ,Stem cell fate determination ,mitochondrial fusion ,NUMB ,Female ,Stem cell ,030217 neurology & neurosurgery - Abstract
Summary Mitochondria are dynamic organelles that have been linked to stem cell homeostasis. However, the mechanisms involved in mitochondrial regulation of stem cell fate determination remain elusive. Here we discover that epithelial-mesenchymal transition (EMT), a key process in cancer progression, induces mitochondrial fusion through regulation of the miR200c-PGC1α-MFN1 pathway. EMT-activated MFN1 forms a complex with PKCζ and is required for PKCζ-mediated NUMB phosphorylation and dissociation from the cortical membrane to direct asymmetric division of mammary stem cells, where fused mitochondria are tethered by MFN1-PKCζ to the cortical membrane and asymmetrically segregated to the stem cell-like progeny with enhanced glutathione synthesis and reactive oxygen species scavenging capacities, allowing sustaining of a self-renewing stem cell pool. Suppression of MFN1 expression leads to equal distribution of the fragmented mitochondria in both progenies that undergo symmetric luminal cell differentiation. Together, this study elucidates an essential role of mitofusin in stem cell fate determination to mediate EMT-associated stemness.
- Published
- 2019
38. Chlorine/UV Process for Decomposition and Detoxification of Microcystin-LR
- Author
-
Jer-Yen Yang, Karl V. Wood, Arlene P. Rothwell, Ernest R. Blatchley, Xinran Zhang, Weiguang Li, and Jing Li
- Subjects
Cyanobacteria ,0208 environmental biotechnology ,Quantum yield ,chemistry.chemical_element ,Microcystin-LR ,02 engineering and technology ,010501 environmental sciences ,Photochemistry ,01 natural sciences ,Mass Spectrometry ,chemistry.chemical_compound ,Water Supply ,polycyclic compounds ,Chlorine ,Environmental Chemistry ,Moiety ,0105 earth and related environmental sciences ,biology ,Advanced oxidation process ,Hepatotoxin ,General Chemistry ,biology.organism_classification ,Decomposition ,020801 environmental engineering ,chemistry ,Oxidation-Reduction - Abstract
Microcystin-LR (MC-LR) is a potent hepatotoxin that is often associated with blooms of cyanobacteria. Experiments were conducted to evaluate the efficiency of the chlorine/UV process for MC-LR decomposition and detoxification. Chlorinated MC-LR was observed to be more photoactive than MC-LR. LC/MS analyses confirmed that the arginine moiety represented an important reaction site within the MC-LR molecule for conditions of chlorination below the chlorine demand of the molecule. Prechlorination activated MC-LR toward UV254 exposure by increasing the product of the molar absorption coefficient and the quantum yield of chloro-MC-LR, relative to the unchlorinated molecule. This mechanism of decay is fundamentally different than the conventional view of chlorine/UV as an advanced oxidation process. A toxicity assay based on human liver cells indicated MC-LR degradation byproducts in the chlorine/UV process possessed less cytotoxicity than those that resulted from chlorination or UV254 irradiation applied separately. MC-LR decomposition and detoxification in this combined process were more effective at pH 8.5 than at pH 7.5 or 6.5. These results suggest that the chlorine/UV process could represent an effective strategy for control of microcystins and their associated toxicity in drinking water supplies.
- Published
- 2016
39. EZH2 Promotes Expansion of Breast Tumor Initiating Cells through Activation of RAF1-β-Catenin Signaling
- Author
-
Xiaoming Xie, Weiya Xia, Jer Yen Yang, Chun Te Chen, Gabriel N. Hortobagyi, Wendy A. Woodward, Chi-Hong Chao, Mien Chie Hung, Chun Ju Chang, and Jung Mao Hsu
- Subjects
Cancer Research ,Cellular pathology ,Pyridines ,Cell ,Gene Expression ,Mice, SCID ,Mice ,0302 clinical medicine ,Mice, Inbred NOD ,DNA Breaks, Double-Stranded ,Phosphorylation ,Extracellular Signal-Regulated MAP Kinases ,beta Catenin ,0303 health sciences ,biology ,Benzenesulfonates ,Polycomb Repressive Complex 2 ,Sorafenib ,Cell Hypoxia ,3. Good health ,DNA-Binding Proteins ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Neoplastic Stem Cells ,Female ,Stem cell ,Signal Transduction ,medicine.drug ,Niacinamide ,Beta-catenin ,Cell Survival ,Transplantation, Heterologous ,Breast Neoplasms ,macromolecular substances ,Models, Biological ,Article ,03 medical and health sciences ,Breast cancer ,Cell Line, Tumor ,Spheroids, Cellular ,medicine ,Animals ,Humans ,Enhancer of Zeste Homolog 2 Protein ,Cell Proliferation ,030304 developmental biology ,Centrosome ,Chromosome Aberrations ,Cell growth ,Phenylurea Compounds ,Cancer ,Cell Biology ,medicine.disease ,Xenograft Model Antitumor Assays ,Proto-Oncogene Proteins c-raf ,biology.protein ,Cancer research ,Benzimidazoles ,Rad51 Recombinase ,DNA Damage ,Transcription Factors - Abstract
SummaryIt has been proposed that an aggressive secondary cancer stem cell population arises from a primary cancer stem cell population through acquisition of additional genetic mutations and drives cancer progression. Overexpression of Polycomb protein EZH2, essential in stem cell self-renewal, has been linked to breast cancer progression. However, critical mechanism linking increased EZH2 expression to BTIC (breast tumor initiating cell) regulation and cancer progression remains unclear. Here, we identify a mechanism in which EZH2 expression-mediated downregulation of DNA damage repair leads to accumulation of recurrent RAF1 gene amplification in BTICs, which activates p-ERK-β-catenin signaling to promote BTIC expansion. We further reveal that AZD6244, a clinical trial drug that inhibits RAF1-ERK signaling, could prevent breast cancer progression by eliminating BTICs.
- Published
- 2011
40. Activation of p21(CIP1/WAF1) in mammary epithelium accelerates mammary tumorigenesis and promotes lung metastasis
- Author
-
Gordon B. Mills, Xiaoyun Cheng, Hui-Lung Sun, Dihua Yu, Weiya Xia, William J. Muller, Shannon L. Wyszomierski, Jennifer L. Hsu, Mien Chie Hung, Chao Kai Chou, and Jer Yen Yang
- Subjects
Cyclin-Dependent Kinase Inhibitor p21 ,Genetically modified mouse ,Lung Neoplasms ,Transgene ,Biophysics ,Breast Neoplasms ,Mice, Transgenic ,Biology ,medicine.disease_cause ,Biochemistry ,Epithelium ,Article ,Metastasis ,Mice ,In vivo ,medicine ,Animals ,Humans ,Molecular Biology ,Mammary Neoplasms, Experimental ,Cell Biology ,medicine.disease ,In vitro ,Cell Transformation, Neoplastic ,Mammary Epithelium ,Cytoplasm ,Cancer research ,Female ,Carcinogenesis ,Proto-Oncogene Proteins c-akt - Abstract
While p21 is well known to inhibit cyclin-CDK activity in the nucleus and it has also been demonstrated to have oncogenic properties in different types of human cancers. In vitro studies showed that the oncogenic function of p21is closely related to its cytoplasmic localization. However, it is unclear whether cytoplasmic p21 contributes to tumorigenesis in vivo. To address this question, we generated transgenic mice expressing the Akt-phosphorylated form of p21 (p21T145D) in the mammary epithelium. The results showed that Akt-activated p21 was expressed in the cytoplasm of mammary epithelium. Overexpression of Akt-activated p21 accelerated tumor onset and promoted lung metastasis in MMTV/neu mice, providing evidence that p21, especially cytoplasmic phosphorylated p21, has an oncogenic role in promoting mammary tumorigenesis and metastasis.
- Published
- 2010
41. Blockade of Mitogen-Activated Protein Kinase/Extracellular Signal-Regulated Kinase Kinase and Murine Double Minute Synergistically Induces Apoptosis in Acute Myeloid Leukemia via BH3-Only Proteins Puma and Bim
- Author
-
Mien Chie Hung, Jared K. Burks, Michael Andreeff, Karen C. Dywer, Teresa McQueen, Marina Konopleva, Jer Yen Yang, Weiguo Zhang, and Wendy D. Schober
- Subjects
MAPK/ERK pathway ,Cancer Research ,MAP Kinase Signaling System ,Apoptosis ,HL-60 Cells ,Biology ,Piperazines ,Article ,Proto-Oncogene Proteins ,hemic and lymphatic diseases ,Puma ,Humans ,Phosphorylation ,Extracellular Signal-Regulated MAP Kinases ,Protein kinase A ,neoplasms ,Bcl-2-Like Protein 11 ,Kinase ,Forkhead Box Protein O3 ,Imidazoles ,Membrane Proteins ,Myeloid leukemia ,Drug Synergism ,Forkhead Transcription Factors ,Proto-Oncogene Proteins c-mdm2 ,U937 Cells ,biology.organism_classification ,Myeloid Cell Leukemia Sequence 1 Protein ,Proto-Oncogene Proteins c-bcl-2 ,Oncology ,Leukemia, Myeloid ,Mitogen-activated protein kinase ,Acute Disease ,biology.protein ,Cancer research ,Benzimidazoles ,biological phenomena, cell phenomena, and immunity ,Signal transduction ,Apoptosis Regulatory Proteins - Abstract
Molecular aberrations of the Ras/Raf/mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK) kinase (MEK)/ERK and/or Murine double minute (MDM2)/p53 signaling pathways have been reported in 80% and 50% of primary acute myeloid leukemia (AML) samples and confer poor outcome. In this study, antileukemic effects of combined MEK inhibition by AZD6244 and nongenotoxic p53 activation by MDM2 antagonist Nutlin-3a were investigated. Simultaneous blockade of MEK and MDM2 signaling by AZD6244 and Nutlin-3a triggered synergistic proapoptotic responses in AML cell lines [combination index (CI) = 0.06 ± 0.03 and 0.43 ± 0.03 in OCI/AML3 and MOLM13 cells, respectively] and in primary AML cells (CI = 0.52 ± 0.01). Mechanistically, the combination upregulated levels of BH3-only proteins Puma and Bim, in part via transcriptional upregulation of the FOXO3a transcription factor. Suppression of Puma and Bim by short interfering RNA rescued OCI/AML3 cells from AZD/Nutlin-induced apoptosis. These results strongly indicate the therapeutic potential of combined MEK/MDM2 blockade in AML and implicate Puma and Bim as major regulators of AML cell survival. Cancer Res; 70(6); 2424–34
- Published
- 2010
42. A New Fork for Clinical Application: Targeting Forkhead Transcription Factors in Cancer
- Author
-
Mien Chie Hung and Jer Yen Yang
- Subjects
endocrine system ,Cancer Research ,Cell cycle checkpoint ,IκB kinase ,Biology ,medicine.disease_cause ,Models, Biological ,Article ,Drug Delivery Systems ,Forkhead Transcription Factors ,Neoplasms ,medicine ,Humans ,Protein kinase B ,Transcription factor ,Cell Nucleus ,Cell growth ,Forkhead Box Protein O3 ,fungi ,Cancer ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Oncology ,embryonic structures ,Immunology ,Cancer research ,biological phenomena, cell phenomena, and immunity ,Carcinogenesis ,Protein Kinases ,hormones, hormone substitutes, and hormone antagonists ,Signal Transduction - Abstract
Forkhead O transcription factors (FOXO) play a pivotal role in the regulation of a myriad of cellular functions including cell cycle arrest, cell death, and protection from stress stimuli. Activation of cell survival pathways such as phosphoinositide-3-kinase/AKT/IKK or RAS/mitogen-activated protein kinase are known to phosphorylate FOXOs at different sites which cause FOXOs nuclear exclusion and degradation, resulting in the suppression of FOXO's transcriptional activity. Perturbation of FOXO's function leads to deregulated cell proliferation and accumulation of DNA damage, resulting in diseases such as cancer. Emerging evidence shows that active FOXO proteins are crucial for keeping cells in check; and inactivation of FOXO proteins is associated with tumorigenesis, including breast cancer, prostate cancer, glioblastoma, rhabdomyosarcoma, and leukemia. Moreover, clinically used drugs like paclitaxel, imatinib, and doxorubicin have been shown to achieve their therapeutic effects through activation of FOXO3a and FOXO3a targets. In this review, we will focus the novel functions of FOXOs revealed in recent studies and further highlight FOXOs as new therapeutic targets in a broad spectrum of cancers.
- Published
- 2009
43. Down-regulation of Myeloid Cell Leukemia-1 through Inhibiting Erk/Pin 1 Pathway by Sorafenib Facilitates Chemosensitization in Breast Cancer
- Author
-
Dung Fang Lee, Mien Chie Hung, Longfei Huo, Chien-Chen Lai, Chun Ju Chang, Jer Yen Yang, Chia Jui Yen, Chun Yi Lin, Yong Liao, Weiya Xia, Yun Ju Rita Chen, Long Yuan Li, Qingqing Ding, Fuu Jen Tsai, Yan Yang, Chang Hai Tsai, Hsu-Ping Kuo, Jung Mao Hsu, and Yongkun Wei
- Subjects
Niacinamide ,MAPK/ERK pathway ,Sorafenib ,Cancer Research ,MAP Kinase Signaling System ,Pyridines ,Cell ,Down-Regulation ,Antineoplastic Agents ,Breast Neoplasms ,Biology ,Article ,Cell Line, Tumor ,hemic and lymphatic diseases ,medicine ,Humans ,Phosphorylation ,neoplasms ,Cell growth ,Phenylurea Compounds ,Benzenesulfonates ,Cancer ,Peptidylprolyl Isomerase ,medicine.disease ,NIMA-Interacting Peptidylprolyl Isomerase ,Myeloid Cell Leukemia Sequence 1 Protein ,medicine.anatomical_structure ,Proto-Oncogene Proteins c-bcl-2 ,Oncology ,Tumor progression ,Mutagenesis, Site-Directed ,PIN1 ,Cancer research ,medicine.drug - Abstract
Myeloid cell leukemia-1 (Mcl-1), a Bcl-2–like antiapoptotic protein, plays a role in cell immortalization and chemoresistance in a number of human malignancies. A peptidyl-prolyl cis/trans isomerase, Pin1 is involved in many cellular events, such as cell cycle progression, cell proliferation, and differentiation through isomerizing prophosphorylated substrates. It has been reported that down-regulation of Pin1 induces apoptosis, and that Erk phosphorylates and up-regulates Mcl-1; however, the underlying mechanisms for the two phenomena are not clear yet. Here, we showed that Pin 1 stabilizes Mcl-1, which is required for Mcl-1 posphorylation by Erk. First, we found expression of Mcl-1 and Pin1 were positively correlated and associated with poor survival in human breast cancer. We then showed that Erk could phosphorylate Mcl-1 at two consensus residues, Thr 92 and 163, which is required for the association of Mcl-1 and Pin1, resulting in stabilization of Mcl-1. Moreover, Pin1 is also required for the up-regulation of Mcl-1 by Erk activation. Based on this newly identified mechanism of Mcl-1 stabilization, two strategies were used to overcome Mcl-1–mediated chemoresistance: inhibiting Erk by Sorafenib, an approved clinical anticancer drug, or knocking down Pin1 by using a SiRNA technique. In conclusion, the current report not only unravels a novel mechanism to link Erk/Pin1 pathway and Mcl-1–mediated chemoresistance but also provides a plausible combination therapy, Taxol (Paclitaxel) plus Sorafenib, which was shown to be effective in killing breast cancer cells. [Cancer Res 2008;68(15):6109–17]
- Published
- 2008
44. Crystal structure of the human FOXO3a-DBD/DNA complex suggests the effects of post-translational modification
- Author
-
Cheng-Yang Huang, Jer Yen Yang, Mien Chie Hung, Y. Sun, Chwan-Deng Hsiao, and Kuang-Lei Tsai
- Subjects
Molecular Sequence Data ,Crystal structure ,Biology ,Crystallography, X-Ray ,Biochemistry ,Structural Biology ,Transcription (biology) ,Genetics ,Consensus sequence ,Humans ,Amino Acid Sequence ,Binding site ,Molecular Biology ,Transcription factor ,Peptide sequence ,Chemistry ,Forkhead Box Protein O3 ,fungi ,FOXO Family ,Forkhead Transcription Factors ,DNA ,Cell Biology ,Molecular biology ,Protein Structure, Tertiary ,Computer Science Applications ,Cell biology ,Posttranslational modification ,Biophysics ,Phosphorylation ,Signal transduction ,Dna complex ,Protein Processing, Post-Translational ,Sequence Alignment ,hormones, hormone substitutes, and hormone antagonists - Abstract
FOXO3a is a transcription factor of the FOXO family. The FOXO proteins participate in multiple signaling pathways, and their transcriptional activity is regulated by several post-translational mechanisms, including phosphorylation, acetylation and ubiquitination. Because these post-translational modification sites are located within the C-terminal basic region of the FOXO DNA-binding domain (FOXO-DBD), it is possible that these post-translational modifications could alter the DNA-binding characteristics. To understand how FOXO mediate transcriptional activity, we report here the 2.7 Å crystal structure of the DNA-binding domain of FOXO3a (FOXO3a-DBD) bound to a 13-bp DNA duplex containing a FOXO consensus binding sequence (GTAAACA). Based on a unique structural feature in the C-terminal region and results from biochemical and mutational studies, our studies may explain how FOXO-DBD C-terminal phosphorylation by protein kinase B (PKB) or acetylation by cAMP-response element binding protein (CBP) can attenuate the DNA-binding activity and thereby reduce transcriptional activity of FOXO proteins. In addition, we demonstrate that the methyl groups of specific thymine bases within the consensus sequence are important for FOXO3a-DBD recognition of the consensus binding site.
- Published
- 2007
45. Low-density lipoprotein receptor-related protein 5 (LRP5) mediates the prostate cancer-induced formation of new bone
- Author
-
Zhongyou Li, Adriana Lopez, Elba Vazquez, Christopher J. Logothetis, Jer Yen Yang, Funda Vakar-Lopez, D. Rose, Paul Mathew, Sue-Hwa Lin, and Nora M. Navone
- Subjects
Male ,musculoskeletal diseases ,Cancer Research ,medicine.medical_specialty ,WNT7B ,Bone Neoplasms ,Osteolysis ,Biology ,PROSTATE CANCER ,WNT ,Mice ,Prostate cancer ,OSTEOBLASTS ,Cell Line, Tumor ,Internal medicine ,Genetics ,medicine ,Animals ,Humans ,Molecular Biology ,LDL-Receptor Related Proteins ,BONE METASTASES ,Osteoblasts ,DKK1 ,Otras Ciencias Químicas ,Ciencias Químicas ,Wnt signaling pathway ,Prostatic Neoplasms ,Bone metastasis ,Cancer ,LRP5 ,Osteoblast ,medicine.disease ,Wnt Proteins ,Low Density Lipoprotein Receptor-Related Protein-5 ,Phenotype ,Endocrinology ,medicine.anatomical_structure ,Cancer cell ,Cancer research ,Intercellular Signaling Peptides and Proteins ,CIENCIAS NATURALES Y EXACTAS ,Signal Transduction - Abstract
The tendency of prostate cancer to produce osteoblastic bone metastases suggests that cancer cells and osteoblasts interact in ways that contribute to cancer progression. To identify factors that mediate these interactions, we compared gene expression patterns between two bone-derived prostate cancer cell lines that produce osteoblastic (MDA PCa 2b) or osteolytic lesions (PC-3). Both cell lines expressed Wnt ligands, including WNT7b, a canonical Wnt implicated in osteogenesis. PC-3 cells expressed 50 times more Dickkopf-1 (DKK1), an inhibitor of Wnt pathways, than did MDA PCa 2b cells. Evaluation of the functional role of these factors (in cocultures of prostate cancer cells with primary mouse osteoblasts (PMOs) or in bone organ cultures) showed that MDA PCa 2b cells activated Wnt canonical signaling in PMOs and that DKK1 blocked osteoblast proliferation and new bone formation induced by MDA PCa 2b cells. MDA PCa 2b cells did not induce bone formation in calvaria from mice lacking the Wnt co-receptor Lrp5. In human specimens, WNT7b was not expressed in normal prostate but was expressed in areas of high-grade prostate intraepithelial neoplasia, in three of nine primary prostate tumor specimens and in 16 of 38 samples of bone metastases from prostate cancer. DKK1 was not expressed in normal or cancerous tissue but was expressed in two of three specimens of osteolytic bone metastases (P=0.0119). We conclude that MDA PCa 2b induces new bone formation through Wnt canonical signaling, that LRP5 mediates this effect, and that DKK1 is involved in the balance between bone formation and resorption that determines lesion phenotype. Fil: Li, Z. G.. University of Texas; Estados Unidos Fil: Yang, J.. University of Texas; Estados Unidos Fil: Vazquez, Elba Susana. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Ciudad Universitaria. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales. Universidad de Buenos Aires. Facultad de Ciencias Exactas y Naturales. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales; Argentina Fil: Rose, D.. University of Texas; Estados Unidos Fil: Vakar Lopez, F.. University of Texas; Estados Unidos Fil: Mathew, P.. University of Texas; Estados Unidos Fil: Lopez, A.. University of Texas; Estados Unidos Fil: Logothetis, C. J.. University of Texas; Estados Unidos Fil: Lin, S. H.. University of Texas; Estados Unidos Fil: Navone, N. M.. University of Texas; Estados Unidos
- Published
- 2007
46. New insight into cancer aneuploidy in zebrafish
- Author
-
GuangJun, Zhang, Jer-Yen, Yang, and Zhibin, Cui
- Subjects
Fish Diseases ,Genome ,Animals ,Humans ,Neoplasms, Experimental ,Aneuploidy ,Zebrafish - Abstract
Aneuploidy is one of the most common genetic alterations in cancer cell genomes. It greatly contributes to the heterogeneity of cancer cell genomes, and its roles in tumorigenesis are attracting more and more attentions. Zebrafish is emerging as a new genetic model for many human diseases including cancer. The zebrafish cancer model has shown an equivalent degree of aneuploidy as found in corresponding human cancers, thus it provides a great tool for us to study cancer aneuploidy and, in general, cancer biology. Here, we discuss some new advances of aneuploidy and the potential usages of this cancer model system.
- Published
- 2015
47. New Insight into Cancer Aneuploidy in Zebrafish
- Author
-
Zhibin Cui, GuangJun Zhang, and Jer-Yen Yang
- Subjects
Genetics ,biology ,Cancer Model ,Aneuploidy ,Cancer ,Synthetic lethality ,Computational biology ,medicine.disease ,biology.organism_classification ,medicine.disease_cause ,Genetic model ,Cancer cell ,medicine ,Carcinogenesis ,Zebrafish - Abstract
Aneuploidy is one of the most common genetic alterations in cancer cell genomes. It greatly contributes to the heterogeneity of cancer cell genomes, and its roles in tumorigenesis are attracting more and more attentions. Zebrafish is emerging as a new genetic model for many human diseases including cancer. The zebrafish cancer model has shown an equivalent degree of aneuploidy as found in corresponding human cancers, thus it provides a great tool for us to study cancer aneuploidy and, in general, cancer biology. Here, we discuss some new advances of aneuploidy and the potential usages of this cancer model system.
- Published
- 2015
48. Erk Associates with and Primes GSK-3β for Its Inactivation Resulting in Upregulation of β-Catenin
- Author
-
Geoffrey Bartholomeusz, Jun Qin, Ralf C. Bargou, Mien Chie Hung, Dung Fang Lee, Qingqing Ding, Jer Yen Yang, Chang Hai Tsai, Zheng Li, Jaw Ching Liu, Yong Pan, Chien-Chen Lai, Yan Li, Weiya Xia, Fuu Jen Tsai, and Jiahong Xia
- Subjects
MAPK/ERK pathway ,Carcinoma, Hepatocellular ,Amino Acid Motifs ,Breast Neoplasms ,macromolecular substances ,Biology ,Receptor tyrosine kinase ,Glycogen Synthase Kinase 3 ,Downregulation and upregulation ,medicine ,Tumor Cells, Cultured ,Humans ,Viral Regulatory and Accessory Proteins ,Insulin-Like Growth Factor I ,Phosphorylation ,Extracellular Signal-Regulated MAP Kinases ,Molecular Biology ,beta Catenin ,Oncogene ,Liver Neoplasms ,Cell Biology ,Genes, erbB-2 ,medicine.disease ,digestive system diseases ,Up-Regulation ,Enzyme Activation ,HBx ,Cytoskeletal Proteins ,Hepatocellular carcinoma ,Catenin ,biology.protein ,Cancer research ,Trans-Activators ,Female - Abstract
Beta-catenin is upregulated in many human cancers and considered to be an oncogene. Hepatocellular carcinoma (HCC) is one of the most prevalent human malignancies, and individuals who are chronic hepatitis B virus (HBV) carriers have a greater than 100-fold increased relative risk of developing HCC. Here we report a mechanism by which HBV-X protein (HBX) upregulates beta-catenin. Erk, which is activated by HBX, associates with GSK-3beta through a docking motif ((291)FKFP) of GSK-3beta and phosphorylates GSK-3beta at the (43)Thr residue, which primes GSK-3beta for its subsequent phosphorylation at Ser9 by p90RSK, resulting in inactivation of GSK-3beta and upregulation of beta-catenin. This pathway is a general signal, as it was also observed in cell lines in which Erk-primed inactivation of GSK-3beta was regulated by IGF-1, TGF-beta, and receptor tyrosine kinase HER2, and is further supported by immunohistochemical staining in different human tumors, including cancers of the liver, breast, kidney, and stomach.
- Published
- 2005
- Full Text
- View/download PDF
49. Adenoviral E1A Targets Mdm4 to Stabilize Tumor Suppressor p53
- Author
-
Hsiu Ming Shih, Guillermina Lozano, Mien Chie Hung, Wen Bin Tsai, Chi-Ping Day, Jer Yen Yang, and Zheng Li
- Subjects
Cancer Research ,Tumor suppressor gene ,Ubiquitin-Protein Ligases ,viruses ,Genetic enhancement ,Biology ,medicine.disease_cause ,Virus ,law.invention ,Mice ,law ,Cell Line, Tumor ,Proto-Oncogene Proteins ,Two-Hybrid System Techniques ,Tumor Suppressor Protein p14ARF ,medicine ,Animals ,Humans ,Cell Nucleus ,Ubiquitin ,Nuclear Proteins ,Proto-Oncogene Proteins c-mdm2 ,Yeast ,Adenoviridae ,medicine.anatomical_structure ,Oncology ,Cancer research ,biology.protein ,Mdm2 ,Suppressor ,Adenovirus E1A Proteins ,Tumor Suppressor Protein p53 ,Nucleus ,Protein Binding - Abstract
The adenoviral protein E1A associates with multiple anticancer activities, including stabilization of p53 tumor suppressor, and has been tested through gene therapy approaches in clinical trials. To identify potential E1A-binding proteins involved in E1A’s anticancer activities, we screened a yeast two-hybrid library and identified Mdm4, an Mdm2-related p53-binding protein, as a novel E1A-binding protein. The NH2-terminal region of Mdm4 and the CR1 domain of E1A were required for the interaction between E1A and Mdm4. E1A preferentially bound to Mdm4 rather than Mdm2 and formed a complex with p53 in the presence of Mdm4, resulting in the stabilization of p53 in a p14ARF-independent manner. E1A failed to stabilize p53 in the absence of Mdm4, showing that Mdm4 was required for p53 stabilization by E1A. Moreover, E1A-mediated stabilization of p53 occurred in nucleus. Although it had no effect on the p53-Mdm2 interaction, E1A facilitated Mdm4 binding to p53 and inhibited Mdm2 binding to Mdm4, resulting in decreased nuclear exportation of p53. Thus, our findings highlighted a novel mechanism, whereby E1A stabilized the p53 tumor suppressor through Mdm4.
- Published
- 2004
50. Leptin-STAT3-G9a Signaling Promotes Obesity-Mediated Breast Cancer Progression
- Author
-
Ignacio G. Camarillo, Chao-Ching Chang, Meng-Ju Wu, Chun Ju Chang, and Jer Yen Yang
- Subjects
Oncology ,Leptin ,STAT3 Transcription Factor ,Cancer Research ,medicine.medical_specialty ,Carcinogenesis ,Population ,Adipokine ,Breast Neoplasms ,Mammary Neoplasms, Animal ,Biology ,medicine.disease_cause ,Article ,Mice ,Breast cancer ,Internal medicine ,Histocompatibility Antigens ,microRNA ,medicine ,Animals ,Humans ,Obesity ,education ,education.field_of_study ,Leptin receptor ,Gene Expression Profiling ,Notices ,Histone-Lysine N-Methyltransferase ,medicine.disease ,Rats ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,Tumor progression ,Histone methyltransferase ,Cancer research ,MCF-7 Cells ,Neoplastic Stem Cells ,Receptors, Leptin ,Female - Abstract
Obesity has been linked to breast cancer progression but the underlying mechanisms remain obscure. Here we report how leptin, an obesity-associated adipokine, regulates a transcriptional pathway to silence a genetic program of epithelial homeostasis in breast cancer stem–like cells (CSC) that promotes malignant progression. Using genome-wide ChIP-seq and RNA expression profiling, we defined a role for activated STAT3 and G9a histone methyltransferase in epigenetic silencing of miR-200c, which promotes the formation of breast CSCs defined by elevated cell surface levels of the leptin receptor (OBRhi). Inhibiting the STAT3/G9a pathway restored expression of miR-200c, which in turn reversed the CSC phenotype to a more differentiated epithelial phenotype. In a rat model of breast cancer driven by diet-induced obesity, STAT3 blockade suppressed the CSC-like OBRhi population and abrogated tumor progression. Together, our results show how targeting STAT3-G9a signaling regulates CSC plasticity during obesity-related breast cancer progression, suggesting a novel therapeutic paradigm to suppress CSC pools and limit breast malignancy. Cancer Res; 75(11); 2375–86. ©2015 AACR.
- Published
- 2014
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.