Network


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

Hotspot


Dive into the research topics where Chia Wei Li is active.

Publication


Featured researches published by Chia Wei Li.


Nature Cell Biology | 2011

p53 regulates epithelial–mesenchymal transition and stem cell properties through modulating miRNAs

Chun-Ju Chang; Chi Hong Chao; Weiya Xia; Jer Yen Yang; Yan Xiong; Chia Wei Li; Wen Hsuan Yu; Sumaiyah K. Rehman; Jennifer L. Hsu; Heng Huan Lee; Mo Liu; Chun Te Chen; Dihua Yu; Mien Chie Hung

The epithelial–mesenchymal transition (EMT) has recently been linked to stem cell phenotype. However, the molecular mechanism underlying EMT and regulation of stemness remains elusive. Here, using genomic approaches, we show that tumour suppressor p53 has a role in regulating both EMT and EMT-associated stem cell properties through transcriptional activation of the microRNA miR-200c. p53 transactivates miR-200c through direct binding to the miR-200c promoter. Loss of p53 in mammary epithelial cells leads to decreased expression of miR-200c and activates the EMT programme, accompanied by an increased mammary stem cell population. Re-expressing miR-200c suppresses genes that mediate EMT and stemness properties and thereby reverts the mesenchymal and stem-cell-like phenotype caused by loss of p53 to a differentiated epithelial cell phenotype. Furthermore, loss of p53 correlates with a decrease in the level of miR-200c, but an increase in the expression of EMT and stemness markers, and development of a high tumour grade in a cohort of breast tumours. This study elucidates a role for p53 in regulating EMT–MET (mesenchymal–epithelial transition) and stemness or differentiation plasticity, and reveals a potential therapeutic implication to suppress EMT-associated cancer stem cells through activation of the p53–miR-200c pathway.


Cancer Research | 2012

Epithelial–Mesenchymal Transition Induced by TNF-α Requires NF-κB–Mediated Transcriptional Upregulation of Twist1

Chia Wei Li; Weiya Xia; Longfei Huo; Seung Oe Lim; Yun Wu; Jennifer L. Hsu; Chi Hong Chao; Hirohito Yamaguchi; Neng Kai Yang; Qingqing Ding; Yan Wang; Yun Ju Lai; Adam M. LaBaff; Ting Jung Wu; Been-Ren Lin; Muh Hwa Yang; Gabriel N. Hortobagyi; Mien Chie Hung

Proinflammatory cytokines produced in the tumor microenvironment facilitate tumor development and metastatic progression. In particular, TNF-α promotes cancer invasion and angiogenesis associated with epithelial-mesenchymal transition (EMT); however, the mechanisms underlying its induction of EMT in cancer cells remain unclear. Here we show that EMT and cancer stemness properties induced by chronic treatment with TNF-α are mediated by the upregulation of the transcriptional repressor Twist1. Exposure to TNF-α rapidly induced Twist1 mRNA and protein expression in normal breast epithelial and breast cancer cells. Both IKK-β and NF-κB p65 were required for TNF-α-induced expression of Twist1, suggesting the involvement of canonical NF-κB signaling. In support of this likelihood, we defined a functional NF-κB-binding site in the Twist1 promoter, and overexpression of p65 was sufficient to induce transcriptional upregulation of Twist1 along with EMT in mammary epithelial cells. Conversely, suppressing Twist1 expression abrogated p65-induced cell migration, invasion, EMT, and stemness properties, establishing that Twist1 is required for NF-κB to induce these aggressive phenotypes in breast cancer cells. Taken together, our results establish a signaling axis through which the tumor microenvironment elicits Twist1 expression to promote cancer metastasis. We suggest that targeting NF-κB-mediated Twist1 upregulation may offer an effective a therapeutic strategy for breast cancer treatment.


Cancer Cell | 2012

The Crosstalk of mTOR/S6K1 and Hedgehog Pathways

Yan Wang; Qingqing Ding; Chia Jui Yen; Weiya Xia; Julie Izzo; Jing Yu Lang; Chia Wei Li; Jennifer L. Hsu; Stephanie A. Miller; Xuemei Wang; Dung Fang Lee; Jung Mao Hsu; Longfei Huo; Adam M. LaBaff; Dongping Liu; Tzu Hsuan Huang; Chien-Chen Lai; Fuu Jen Tsai; Wei Chao Chang; Chung-Hsuan Chen; Tsung Teh Wu; Navtej Buttar; Kenneth K. Wang; Yun Wu; Huamin Wang; Jaffer A. Ajani; Mien Chie Hung

Esophageal adenocarcinoma (EAC) is the most prevalent esophageal cancer type in the United States. The TNF-α/mTOR pathway is known to mediate the development of EAC. Additionally, aberrant activation of Gli1, downstream effector of the Hedgehog (HH) pathway, has been observed in EAC. In this study, we found that an activated mTOR/S6K1 pathway promotes Gli1 transcriptional activity and oncogenic function through S6K1-mediated Gli1 phosphorylation at Ser84, which releases Gli1 from its endogenous inhibitor, SuFu. Moreover, elimination of S6K1 activation by an mTOR pathway inhibitor enhances the killing effects of the HH pathway inhibitor. Together, our results established a crosstalk between the mTOR/S6K1 and HH pathways, which provides a mechanism for SMO-independent Gli1 activation and also a rationale for combination therapy for EAC.


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.


Oncogene | 2011

Tumor-suppressor role for the SPOP ubiquitin ligase in signal-dependent proteolysis of the oncogenic co-activator SRC-3/AIB1.

Chia Wei Li; Ao J; Fu J; Dung Fang Lee; Jianming Xu; David M. Lonard; Bert W. O'Malley

Steroid receptor co-activator-3 (SRC-3/AIB1) is an oncogene that is amplified and overexpressed in many human cancers. However, the molecular mechanisms that regulate ‘activated SRC-3 oncoprotein’ turnover during tumorigenesis remain to be elucidated. Here, we report that speckle-type POZ protein (SPOP), a cullin 3 (CUL3)-based ubiquitin ligase, is responsible for SRC-3 ubiquitination and proteolysis. SPOP interacts directly with an SRC-3 phospho-degron in a phosphorylation-dependent manner. Casein kinase Iɛ phosphorylates the S102 in this degron and promotes SPOP-dependent turnover of SRC-3. Short hairpin RNA knockdown and overexpression experiments substantiated that the SPOP/CUL3/Rbx1 ubiquitin ligase complex promotes SRC-3 turnover. A systematic analysis of the SPOP genomic locus revealed that a high percentage of genomic loss or loss of heterozygosity occurs at this locus in breast cancers. Furthermore, we demonstrate that restoration of SPOP expression inhibited SRC-3-mediated oncogenic signaling and tumorigenesis, thus positioning SPOP as a tumor suppressor.


Journal of Clinical Investigation | 2011

APOBEC3G promotes liver metastasis in an orthotopic mouse model of colorectal cancer and predicts human hepatic metastasis

Qingqing Ding; Chun-Ju Chang; Xiaoming Xie; Weiya Xia; Jer Yen Yang; Shao Chun Wang; Yan Wang; Jiahong Xia; Libo Chen; Changchung Cai; Huabin Li; Chia Jui Yen; Hsu Ping Kuo; Dung Fang Lee; Jing Yu Lang; Longfei Huo; Xiaoyun Cheng; Yun Ju Chen; Chia Wei Li; Long Bin Jeng; Jennifer L. Hsu; Long Yuan Li; Alai Tan; Steven A. Curley; Lee M. Ellis; Raymond N. DuBois; Mien Chie Hung

Colorectal cancer is the second leading cause of death from cancer in the United States. Metastases in the liver, the most common metastatic site for colorectal cancer, are found in one-third of the patients who die of colorectal cancer. Currently, the genes and molecular mechanisms that are functionally critical in modulating colorectal cancer hepatic metastasis remain unclear. Here, we report our studies using functional selection in an orthotopic mouse model of colorectal cancer to identify a set of genes that play an important role in mediating colorectal cancer liver metastasis. These genes included APOBEC3G, CD133, LIPC, and S100P. Clinically, we found these genes to be highly expressed in a cohort of human hepatic metastasis and their primary colorectal tumors, suggesting that it might be possible to use these genes to predict the likelihood of hepatic metastasis. We have further revealed what we believe to be a novel mechanism in which APOBEC3G promotes colorectal cancer hepatic metastasis through inhibition of miR-29-mediated suppression of MMP2. Together, our data elucidate key factors and mechanisms involved in colorectal cancer liver metastasis, which could be potential targets for diagnosis and treatment.


Nature Communications | 2016

Glycosylation and stabilization of programmed death ligand-1 suppresses T-cell activity

Chia Wei Li; Seung Oe Lim; Weiya Xia; Heng Huan Lee; Li Chuan Chan; Chu Wei Kuo; Kay Hooi Khoo; Shih Shin Chang; Jong Ho Cha; Taewan Kim; Jennifer L. Hsu; Yun Wu; Jung Mao Hsu; Hirohito Yamaguchi; Qingqing Ding; Yan Wang; Jun Yao; Cheng Chung Lee; Hsing Ju Wu; Aysegul A. Sahin; James P. Allison; Dihua Yu; Gabriel N. Hortobagyi; Mien Chie Hung

Extracellular interaction between programmed death ligand-1 (PD-L1) and programmed cell death protein-1 (PD-1) leads to tumour-associated immune escape. Here we show that the immunosuppression activity of PD-L1 is stringently modulated by ubiquitination and N-glycosylation. We show that glycogen synthase kinase 3β (GSK3β) interacts with PD-L1 and induces phosphorylation-dependent proteasome degradation of PD-L1 by β-TrCP. In-depth analysis of PD-L1 N192, N200 and N219 glycosylation suggests that glycosylation antagonizes GSK3β binding. In this regard, only non-glycosylated PD-L1 forms a complex with GSK3β and β-TrCP. We also demonstrate that epidermal growth factor (EGF) stabilizes PD-L1 via GSK3β inactivation in basal-like breast cancer. Inhibition of EGF signalling by gefitinib destabilizes PD-L1, enhances antitumour T-cell immunity and therapeutic efficacy of PD-1 blockade in syngeneic mouse models. Together, our results link ubiquitination and glycosylation pathways to the stringent regulation of PD-L1, which could lead to potential therapeutic strategies to enhance cancer immune therapy efficacy.


Urological Research | 2000

Distributions of p53 codon 72 polymorphism in bladder cancer - Proline form is prominent in invasive tumor

Wen-Chi Chen; Fuu Jen Tsai; Jer-Yuarn Wu; Hsi Chin Wu; Hsueh-Fu Lu; Chia Wei Li

Abstract Abnormal function of p53 is commonly associated with various cancer formations. High-grade and late-stage bladder cancers have been reported to have mutated or become inactive p53 when using immunohistochemical stains. Recently, p53 codon 72 polymorphism was extensively studied to determine the risk factors responsible for cancer formation. There was a general population of codon 72 sequence polymorphism of the wild type p53. A single base change from G to C caused the alteration of amino acid residue 72 from arginine to proline. The arginine form is considered to be a significant risk factor in the development of cancer. However, various reports had indicated discrepancies with regard to this polymorphism; some showed no significant difference between the control and cancer groups, while other series were associated with high risks in the proline form homozygotes. To resolve the undefined distribution of this polymorphism in bladder cancers, 58 patients with bladder cancer were enrolled onto this study. When checked using the Chi-squared test (P=0.952) there were no differences between the control subjects and bladder cancer patients in the distribution of polymorphism. However, proline form homozygotes were more frequently found in the invasive group than the non-invasive group by Fishers exact test (25% and 2.9%, respectively, P < 0.001). More than 70% of the non-invasive bladder cancers were the arginine form homozygotes. This result is consistent with those reported for hepatocellular carcinoma that showed a history of chronic liver disease and proline form homozygotes in a report by Yu et al. Our data suggest that proline form homozygotes are associated with invasive bladder cancer.


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.


Science Signaling | 2014

MDM2-mediated degradation of SIRT6 phosphorylated by AKT1 promotes tumorigenesis and trastuzumab resistance in breast cancer

Umadevi Thirumurthi; Jia Shen; Weiya Xia; Adam M. LaBaff; Yongkun Wei; Chia Wei Li; Wei Chao Chang; Chung-Hsuan Chen; Hui Kuan Lin; Dihua Yu; Mien Chie Hung

Activating a deacetylase may overcome resistance to a receptor-targeted inhibitor in some breast cancer patients. Connecting SIRT6 to Cancer Drug Resistance A subtype of breast cancer responds to trastuzumab therapy, which is known as Herceptin and targets a specific growth factor receptor, and some breast tumors have less of the deacetylase SIRT6. Thirumurthi et al. investigated if SIRT6 affected trastuzumab resistance. Breast cancer patient survival positively correlated with SIRT6 abundance; SIRT6 negatively correlated with that of the active form of the kinase AKT in tumor biopsies and in trastuzumab-resistant breast cancer cells. The phosphorylation of SIRT6 by AKT induced its degradation in cells. Thus, increasing SIRT6 restores trastuzumab-mediated cell death to cells that were initially resistant, suggesting a potential strategy to improve therapeutic outcome. Sirtuin 6 (SIRT6) is associated with longevity and is also a tumor suppressor. Identification of molecular regulators of SIRT6 might enable its activation therapeutically in cancer patients. In various breast cancer cell lines, we found that SIRT6 was phosphorylated at Ser338 by the kinase AKT1, which induced the interaction and ubiquitination of SIRT6 by MDM2, targeting SIRT6 for protease-dependent degradation. The survival of breast cancer patients positively correlated with the abundance of SIRT6 and inversely correlated with the phosphorylation of SIRT6 at Ser338. In a panel of breast tumor biopsies, SIRT6 abundance inversely correlated with the abundance of phosphorylated AKT. Inhibiting AKT or preventing SIRT6 phosphorylation by mutating Ser338 prevented the degradation of SIRT6 mediated by MDM2, suppressed the proliferation of breast cancer cells in culture, and inhibited the growth of breast tumor xenografts in mice. Overexpressing MDM2 decreased the abundance of SIRT6 in cells, whereas overexpressing an E3 ligase–deficient MDM2 or knocking down endogenous MDM2 increased SIRT6 abundance. Trastuzumab (known as Herceptin) is a drug that targets a specific receptor common in some breast cancers, and knocking down SIRT6 increased the survival of a breast cancer cell exposed to trastuzumab. Overexpression of a nonphosphorylatable SIRT6 mutant increased trastuzumab sensitivity in a resistant breast cancer cell line. Thus, stabilizing SIRT6 may be a clinical strategy for overcoming trastuzumab resistance in breast cancer patients.

Collaboration


Dive into the Chia Wei Li's collaboration.

Top Co-Authors

Avatar

Mien Chie Hung

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
Top Co-Authors

Avatar

Weiya Xia

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Yan Wang

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

Seung Oe Lim

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Longfei Huo

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Qingqing Ding

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Shih Shin Chang

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
Researchain Logo
Decentralizing Knowledge