Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Chao Kai Chou is active.

Publication


Featured researches published by Chao Kai Chou.


Cell | 2007

IKKβ Suppression of TSC1 Links Inflammation and Tumor Angiogenesis via the mTOR Pathway

Dung Fang Lee; Hsu Ping Kuo; Chun Te Chen; Jung Mao Hsu; Chao Kai Chou; Yongkun Wei; Hui Lung Sun; Long Yuan Li; Bo Ping; Wei Chien Huang; Xianghuo He; Jen Yu Hung; Chien-Chen Lai; Qingqing Ding; Jen Liang Su; Jer Yen Yang; Aysegul A. Sahin; Gabriel N. Hortobagyi; Fuu Jen Tsai; Chang Hai Tsai; Mien Chie Hung

TNFalpha has recently emerged as a regulator linking inflammation to cancer pathogenesis, but the detailed cellular and molecular mechanisms underlying this link remain to be elucidated. The tuberous sclerosis 1 (TSC1)/TSC2 tumor suppressor complex serves as a repressor of the mTOR pathway, and disruption of TSC1/TSC2 complex function may contribute to tumorigenesis. Here we show that IKKbeta, a major downstream kinase in the TNFalpha signaling pathway, physically interacts with and phosphorylates TSC1 at Ser487 and Ser511, resulting in suppression of TSC1. The IKKbeta-mediated TSC1 suppression activates the mTOR pathway, enhances angiogenesis, and results in tumor development. We further find that expression of activated IKKbeta is associated with TSC1 Ser511 phosphorylation and VEGF production in multiple tumor types and correlates with poor clinical outcome of breast cancer patients. Our findings identify a pathway that is critical for inflammation-mediated tumor angiogenesis and may provide a target for clinical intervention in human cancer.


Molecular Cell | 2009

KEAP1 E3 Ligase-Mediated Downregulation of NF-κB Signaling by Targeting IKKβ

Dung Fang Lee; Hsu Ping Kuo; Mo Liu; Chao Kai Chou; Weiya Xia; Yi Du; Jia Shen; Chun Te Chen; Longfei Huo; Ming Chuan Hsu; Chia Wei Li; Qingqing Ding; Tsai Lien Liao; Chien-Chen Lai; Ann Chi Lin; Ya Hui Chang; Shih-Feng Tsai; Long Yuan Li; Mien Chie Hung

IkappaB kinase beta (IKKbeta) is involved in tumor development and progression through activation of the nuclear factor (NF)-kappaB pathway. However, the molecular mechanism that regulates IKKbeta degradation remains largely unknown. Here, we show that a Cullin 3 (CUL3)-based ubiquitin ligase, Kelch-like ECH-associated protein 1 (KEAP1), is responsible for IKKbeta ubiquitination. Depletion of KEAP1 led to the accumulation and stabilization of IKKbeta and to upregulation of NF-kappaB-derived tumor angiogenic factors. A systematic analysis of the CUL3, KEAP1, and RBX1 genomic loci revealed a high percentage of genome loss and missense mutations in human cancers that failed to facilitate IKKbeta degradation. Our results suggest that the dysregulation of KEAP1-mediated IKKbeta ubiquitination may contribute to tumorigenesis.


Science Signaling | 2010

ARD1 Stabilization of TSC2 Suppresses Tumorigenesis Through the mTOR Signaling Pathway

Hsu Ping Kuo; Dung Fang Lee; Chun Te Chen; Mo Liu; Chao Kai Chou; Hong Jen Lee; Yi Du; Xiaoming Xie; Yongkun Wei; Weiya Xia; Zhang Weihua; Jer Yen Yang; Chia Jui Yen; Tzu Hsuan Huang; Minjia Tan; Gang Xing; Yingming Zhao; Chien Hsing Lin; Shih-Feng Tsai; Isaiah J. Fidler; Mien Chie Hung

Acetylation by ARD1 of an inhibitor of mammalian target of rapamycin may inhibit cancer growth. Inhibiting mTOR and Tumor Growth The acetylase arrest-defective protein 1 (ARD1) has been implicated in mammalian cell proliferation and apoptosis, and the dysfunction of these processes contributes to cancer development. Kuo et al. searched an online database for a possible relationship between ARD1 and cancer and found that increased ARD1 mRNA abundance was associated with various measures of improved clinical outcome in patients with breast cancer, suggesting that ARD1 might act as a tumor suppressor. They uncovered ARD1 loss of heterozygosity (LOH) in primary breast cancer and several different cancer cell lines. Further analysis indicated that ARD1 acetylated—and thereby stabilized—tuberous sclerosis 2 (TSC2), part of a complex that inhibits signaling through the mammalian target of rapamycin (mTOR) pathway, a master regulator of cellular metabolism. ARD1-dependent inhibition of mTOR signaling decreased cell proliferation and increased autophagy, thereby suppressing tumor growth in nude mice. Moreover, ARD1 abundance correlated with that of TSC2 in various types of cancer. The authors thus propose that ARD1 suppressed development of multiple types of cancer through its stabilization of TSC2 and the consequent inhibition of mTOR signaling. Mammalian target of rapamycin (mTOR) regulates various cellular functions, including tumorigenesis, and is inhibited by the tuberous sclerosis 1 (TSC1)–TSC2 complex. Here, we demonstrate that arrest-defective protein 1 (ARD1) physically interacts with, acetylates, and stabilizes TSC2, thereby repressing mTOR activity. The inhibition of mTOR by ARD1 inhibits cell proliferation and increases autophagy, thereby inhibiting tumorigenicity. Correlation between ARD1 and TSC2 abundance was apparent in multiple tumor types. Moreover, evaluation of loss of heterozygosity at Xq28 revealed allelic loss in 31% of tested breast cancer cell lines and tumor samples. Together, our findings suggest that ARD1 functions as an inhibitor of the mTOR pathway and that dysregulation of the ARD1-TSC2-mTOR axis may contribute to cancer development.


Molecular Cell | 2012

IKKα Activation of NOTCH Links Tumorigenesis via FOXA2 Suppression

Mo Liu; Dung Fang Lee; Chun Te Chen; Chia Jui Yen; Long Yuan Li; Hong Jen Lee; Chun-Ju Chang; Wei Chao Chang; Jung Mao Hsu; Hsu Ping Kuo; Weiya Xia; Yongkun Wei; Pei Chun Chiu; Chao Kai Chou; Yi Du; Debanjan Dhar; Michael Karin; Chung-Hsuan Chen; Mien Chie Hung

Proinflammatory cytokine TNFα plays critical roles in promoting malignant cell proliferation, angiogenesis, and tumor metastasis in many cancers. However, the mechanism of TNFα-mediated tumor development remains unclear. Here, we show that IKKα, an important downstream kinase of TNFα, interacts with and phosphorylates FOXA2 at S107/S111, thereby suppressing FOXA2 transactivation activity and leading to decreased NUMB expression, and further activates the downstream NOTCH pathway and promotes cell proliferation and tumorigenesis. Moreover, we found that levels of IKKα, pFOXA2 (S107/111), and activated NOTCH1 were significantly higher in hepatocellular carcinoma tumors than in normal liver tissues and that pFOXA2 (S107/111) expression was positively correlated with IKKα and activated NOTCH1 expression in tumor tissues. Therefore, dysregulation of NUMB-mediated suppression of NOTCH1 by TNFα/IKKα-associated FOXA2 inhibition likely contributes to inflammation-mediated cancer pathogenesis. Here, we report a TNFα/IKKα/FOXA2/NUMB/NOTCH1 pathway that is critical for inflammation-mediated tumorigenesis and may provide a target for clinical intervention in human cancer.


Applied Physics Letters | 2006

Electrospinning of silica nanochannels for single molecule detection

Miao Wang; Nan Jing; Chin B. Su; Jun Kameoka; Chao Kai Chou; Mien Chie Hung; Kuang-An Chang

We have fabricated silica nanochannels with inner diameter as small as 20nm using a scanned coaxial electrospinning and demonstrated their application for single molecule detection. A coaxial jet, with the use of motor oil as the core and silica sol-gel solution as the shell, is extruded through a coaxial source and deposited on the rotating collector as oriented nanofibers. They are then annealed to cross-link silica and eliminate motor oil, thereby forming nanochannels. Subsequently, a fluorescent dye was injected into the individual nanochannels via a capillary force and single molecule detection was performed by monitoring the photon signals from 5-Iodoacetamidofluorescein.


Cancer Research | 2008

Bile Acid Exposure Up-regulates Tuberous Sclerosis Complex 1/Mammalian Target of Rapamycin Pathway in Barrett's-Associated Esophageal Adenocarcinoma

Chia Jui Yen; Julie Izzo; Dung Fang Lee; Sushovan Guha; Yongkun Wei; Tsung Teh Wu; Chun Te Chen; Hsu Ping Kuo; Jung Mao Hsu; Hui Lung Sun; Chao Kai Chou; Navtej Buttar; Kenneth K. Wang; Peng Huang; Jaffer A. Ajani; Mien Chie Hung

Barretts esophagus, a columnar metaplasia of the lower esophagus epithelium related to gastroesophageal reflux disease, is the strongest known risk factor for the development of esophageal adenocarcinoma (EAC). Understanding the signal transduction events involved in esophageal epithelium carcinogenesis may provide insights into the origins of EAC and may suggest new therapies. To elucidate the molecular pathways of bile acid-induced tumorigenesis, the newly identified inflammation-associated signaling pathway involving I kappaB kinases beta (IKK beta), tuberous sclerosis complex 1 (TSC1), and mammalian target of rapamycin (mTOR) downstream effector S6 kinase (S6K1) was confirmed to be activated in immortalized Barretts CPC-A and CPC-C cells and esophageal cancer SEG-1 and BE3 cells. Phosphorylation of TSC1 and S6K1 was induced in response to bile acid stimulation. Treatment of these cells with the mTOR inhibitor rapamycin or the IKK beta inhibitor Bay 11-7082 suppressed bile acid-induced cell proliferation and anchorage-independent growth. We next used an orthotopic rat model to evaluate the role of bile acid in the progression of Barretts esophagus to EAC. Of interest, we found high expression of phosphorylated IKK beta (pIKK beta) and phosphorylated S6K1 (pS6K1) in tumor tissues and the Barretts epithelium compared with normal epithelium. Furthermore, immunostaining of clinical EAC tissue specimens revealed that pIKK beta expression was strongly correlated with pS6K1 level. Together, these results show that bile acid can deregulate TSC1/mTOR through IKK beta signaling, which may play a critical role in EAC progression. In addition, Bay 11-7082 and rapamycin may potentially be chemopreventive drugs against Barretts esophagus-associated EAC.


Clinical Cancer Research | 2017

PARP inhibitor upregulates PD-L1 expression and enhances cancer-associated immunosuppression

Shiping Jiao; Weiya Xia; Hirohito Yamaguchi; Yongkun Wei; Mei Kuang Chen; Jung Mao Hsu; Jennifer L. Hsu; Wen Hsuan Yu; Yi Du; Heng Huan Lee; Chia Wei Li; Chao Kai Chou; Seung Oe Lim; Shih Shin Chang; Jennifer K. Litton; Banu Arun; Gabriel N. Hortobagyi; Mien Chie Hung

Purpose: To explore whether a cross-talk exists between PARP inhibition and PD-L1/PD-1 immune checkpoint axis, and determine whether blockade of PD-L1/PD-1 potentiates PARP inhibitor (PARPi) in tumor suppression. Experimental Design: Breast cancer cell lines, xenograft tumors, and syngeneic tumors treated with PARPi were assessed for PD-L1 expression by immunoblotting, IHC, and FACS analyses. The phospho-kinase antibody array screen was used to explore the underlying mechanism of PARPi-induced PD-L1 upregulation. The therapeutic efficacy of PARPi alone, PD-L1 blockade alone, or their combination was tested in a syngeneic tumor model. The tumor-infiltrating lymphocytes and tumor cells isolated from syngeneic tumors were analyzed by CyTOF and FACS to evaluate the activity of antitumor immunity in the tumor microenvironment. Results: PARPi upregulated PD-L1 expression in breast cancer cell lines and animal models. Mechanistically, PARPi inactivated GSK3β, which in turn enhanced PARPi-mediated PD-L1 upregulation. PARPi attenuated anticancer immunity via upregulation of PD-L1, and blockade of PD-L1 resensitized PARPi-treated cancer cells to T-cell killing. The combination of PARPi and anti-PD-L1 therapy compared with each agent alone significantly increased the therapeutic efficacy in vivo. Conclusions: Our study demonstrates a cross-talk between PARPi and tumor-associated immunosuppression and provides evidence to support the combination of PARPi and PD-L1 or PD-1 immune checkpoint blockade as a potential therapeutic approach to treat breast cancer. Clin Cancer Res; 23(14); 3711–20. ©2017 AACR.


Journal of Clinical Investigation | 2015

PRMT1-mediated methylation of the EGF receptor regulates signaling and cetuximab response

Hsin Wei Liao; Jung Mao Hsu; Weiya Xia; Hung Ling Wang; Ying Nai Wang; Wei Chao Chang; Stefan T. Arold; Chao Kai Chou; Pei Hsiang Tsou; Hirohito Yamaguchi; Yueh Fu Fang; Hong Jen Lee; Heng Huan Lee; Shyh Kuan Tai; Mhu Hwa Yang; Maria Pia Morelli; Malabika Sen; John E. Ladbury; Chung-Hsuan Chen; Jennifer R. Grandis; Scott Kopetz; Mien Chie Hung

Posttranslational modifications to the intracellular domain of the EGFR are known to regulate EGFR functions; however, modifications to the extracellular domain and their effects remain relatively unexplored. Here, we determined that methylation at R198 and R200 of the EGFR extracellular domain by protein arginine methyltransferase 1 (PRMT1) enhances binding to EGF and subsequent receptor dimerization and signaling activation. In a mouse orthotopic colorectal cancer xenograft model, expression of a methylation-defective EGFR reduced tumor growth. Moreover, increased EGFR methylation sustained signaling activation and cell proliferation in the presence of the therapeutic EGFR monoclonal antibody cetuximab. In colorectal cancer patients, EGFR methylation level also correlated with a higher recurrence rate after cetuximab treatment and reduced overall survival. Together, these data indicate that R198/R200 methylation of the EGFR plays an important role in regulating EGFR functionality and resistance to cetuximab treatment.


PLOS ONE | 2013

Inhibition of type I insulin-like growth factor receptor signaling attenuates the development of breast cancer brain metastasis.

Sandra M. Saldana; Heng Huan Lee; Frank J. Lowery; Yekaterina B. Khotskaya; Weiya Xia; Chenyu Zhang; Shih Shin Chang; Chao Kai Chou; Patricia S. Steeg; Dihua Yu; Mien Chie Hung

Brain metastasis is a common cause of mortality in cancer patients, yet potential therapeutic targets remain largely unknown. The type I insulin-like growth factor receptor (IGF-IR) is known to play a role in the progression of breast cancer and is currently being investigated in the clinical setting for various types of cancer. The present study demonstrates that IGF-IR is constitutively autophosphorylated in brain-seeking breast cancer sublines. Knockdown of IGF-IR results in a decrease of phospho-AKT and phospho-p70s6k, as well as decreased migration and invasion of MDA-MB-231Br brain-seeking cells. In addition, transient ablation of IGFBP3, which is overexpressed in brain-seeking cells, blocks IGF-IR activation. Using an in vivo experimental brain metastasis model, we show that IGF-IR knockdown brain-seeking cells have reduced potential to establish brain metastases. Finally, we demonstrate that the malignancy of brain-seeking cells is attenuated by pharmacological inhibition with picropodophyllin, an IGF-IR-specific tyrosine kinase inhibitor. Together, our data suggest that the IGF-IR is an important mediator of brain metastasis and its ablation delays the onset of brain metastases in our model system.


Biochemical and Biophysical Research Communications | 2010

Activation of p21(CIP1/WAF1) in mammary epithelium accelerates mammary tumorigenesis and promotes lung metastasis

Xiaoyun Cheng; Weiya Xia; Jer Yen Yang; Jennifer L. Hsu; Chao Kai Chou; Hui Lung Sun; Shannon L. Wyszomierski; Gordon B. Mills; William J. Muller; Dihua Yu; Mien Chie Hung

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.

Collaboration


Dive into the Chao Kai Chou's collaboration.

Top Co-Authors

Avatar

Mien Chie Hung

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chun Te Chen

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Weiya Xia

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Hirohito Yamaguchi

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jung Mao Hsu

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Yongkun Wei

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Heng Huan Lee

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jennifer L. Hsu

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge