Network


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

Hotspot


Dive into the research topics where Jennifer L. Hsu is active.

Publication


Featured researches published by Jennifer L. Hsu.


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.


Nature | 2013

EGFR modulates microRNA maturation in response to hypoxia through phosphorylation of AGO2

Jia Shen; Weiya Xia; Yekaterina B. Khotskaya; Longfei Huo; Kotaro Nakanishi; Seung Oe Lim; Yi Du; Yan Wang; Wei Chao Chang; Chung-Hsuan Chen; Jennifer L. Hsu; Yun Wu; Yung Carmen Lam; Brian P. James; Xiuping Liu; Chang Gong Liu; Dinshaw J. Patel; Mien Chie Hung

MicroRNAs (miRNAs) are generated by two-step processing to yield small RNAs that negatively regulate target gene expression at the post-transcriptional level. Deregulation of miRNAs has been linked to diverse pathological processes, including cancer. Recent studies have also implicated miRNAs in the regulation of cellular response to a spectrum of stresses, such as hypoxia, which is frequently encountered in the poorly angiogenic core of a solid tumour. However, the upstream regulators of miRNA biogenesis machineries remain obscure, raising the question of how tumour cells efficiently coordinate and impose specificity on miRNA expression and function in response to stresses. Here we show that epidermal growth factor receptor (EGFR), which is the product of a well-characterized oncogene in human cancers, suppresses the maturation of specific tumour-suppressor-like miRNAs in response to hypoxic stress through phosphorylation of argonaute 2 (AGO2) at Tyr 393. The association between EGFR and AGO2 is enhanced by hypoxia, leading to elevated AGO2-Y393 phosphorylation, which in turn reduces the binding of Dicer to AGO2 and inhibits miRNA processing from precursor miRNAs to mature miRNAs. We also identify a long-loop structure in precursor miRNAs as a critical regulatory element in phospho-Y393-AGO2-mediated miRNA maturation. Furthermore, AGO2-Y393 phosphorylation mediates EGFR-enhanced cell survival and invasiveness under hypoxia, and correlates with poorer overall survival in breast cancer patients. Our study reveals a previously unrecognized function of EGFR in miRNA maturation and demonstrates how EGFR is likely to function as a regulator of AGO2 through novel post-translational modification. These findings suggest that modulation of miRNA biogenesis is important for stress response in tumour cells and has potential clinical implications.


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.


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.


Journal of Medicinal Chemistry | 2010

Antitumor agents. 272. Structure-activity relationships and in vivo selective anti-breast cancer activity of novel neo-tanshinlactone analogues.

Yizhou Dong; Qian Shi; Huei Chen Pai; Chieh Yu Peng; Shiow Lin Pan; Che-Ming Teng; Kyoko Nakagawa-Goto; Donglei Yu; Yi Nan Liu; Pei Chi Wu; Kenneth F. Bastow; Susan L. Morris-Natschke; Arnold Brossi; Jing Yu Lang; Jennifer L. Hsu; Mien Chie Hung; Eva Y.-H. P. Lee; Kuo Hsiung Lee

Neo-tanshinlactone (1) and its previously reported analogues, such as 2, are potent and selective in vitro antibreast cancer agents. The synthetic pathway to 2 was optimized from seven to five steps, with a better overall yield. Structure-activity relationships studies on these compounds revealed some key molecular determinants for this family of antibreast agents. Several derivatives (19-21 and 24) exerted potent and selective antibreast cancer activity with IC(50) values of 0.3, 0.2, 0.1, and 0.1 microg/mL, respectively, against the ZR-75-1 cell lines. Compound 24 was 2- to 3-fold more potent than 1 against SK-BR-3 and ZR-75-1. Importantly, 21 exhibited high selectivity; it was 23 times more active against ZR-75-1 than MCF-7. Compound 20 had an approximately 12-fold ratio of SK-BR-3/MCF-7 selectivity. In addition, analogue 2 showed potent activity against a ZR-75-1 xenograft model, but not PC-3 and MDA-MB-231 xenografts, as well as high selectivity against breast cancer cell line compared with normal breast tissue-derived cell lines. Further development of lead compounds 19-21 and 24 as clinical trial candidates is warranted.


Journal of Biological Chemistry | 2006

Functional Divergence of a Unique C-terminal Domain of Leucyl-tRNA Synthetase to Accommodate Its Splicing and Aminoacylation Roles

Jennifer L. Hsu; Seung Bae Rho; Kevin M. Vannella; Susan A. Martinis

Leucyl-tRNA synthetase (LeuRS) performs dual essential roles in group I intron RNA splicing as well as protein synthesis within the yeast mitochondria. Deletions of the C terminus differentially impact the two functions of the enzyme in splicing and aminoacylation in vivo. Herein, we determined that a fiveamino acid C-terminal deletion of LeuRS, which does not complement a null strain, can form a ternary complex with the bI4 intron and its maturase splicing partner. However, the complex fails to stimulate splicing activity. The x-ray co-crystal structure of LeuRS showed that a C-terminal extension of about 60 amino acids forms a discrete domain, which is unique among the LeuRSs and interacts with the corner of the L-shaped tRNALeu. Interestingly, deletion of the entire yeast mitochondrial LeuRS C-terminal domain enhanced its aminoacylation and amino acid editing activities. In striking contrast, deletion of the corresponding C-terminal domain of Escherichia coli LeuRS abolished aminoacylation of tRNALeu and also amino acid editing of mischarged tRNA molecules. These results suggest that the role of the leucine-specific C-terminal domain in tRNA recognition for aminoacylation and amino acid editing has adapted differentially and with surprisingly opposite effects. We propose that the secondary role of yeast mitochondrial LeuRS in RNA splicing has impacted the functional evolution of this critical C-terminal domain.


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.


Nature Medicine | 2016

Blocking c-Met-mediated PARP1 phosphorylation enhances anti-tumor effects of PARP inhibitors.

Yi Du; Hirohito Yamaguchi; Yongkun Wei; Jennifer L. Hsu; Hung Ling Wang; Yi Hsin Hsu; Wan Chi Lin; Wen Hsuan Yu; Paul G. Leonard; Gilbert R. Lee; Mei Kuang Chen; Katsuya Nakai; Ming Chuan Hsu; Chun Te Chen; Ye Sun; Yun Wu; Wei Chao Chang; Wen Chien Huang; Chien Liang Liu; Yuan Ching Chang; Chung-Hsuan Chen; Morag Park; Philip Jones; Gabriel N. Hortobagyi; Mien Chie Hung

Poly (ADP-ribose) polymerase (PARP) inhibitors have emerged as promising therapeutics for many diseases, including cancer, in clinical trials. One PARP inhibitor, olaparib (Lynparza, AstraZeneca), was recently approved by the FDA to treat ovarian cancer with mutations in BRCA genes. BRCA1 and BRCA2 have essential roles in repairing DNA double-strand breaks, and a deficiency of BRCA proteins sensitizes cancer cells to PARP inhibition. Here we show that the receptor tyrosine kinase c-Met associates with and phosphorylates PARP1 at Tyr907 (PARP1 pTyr907 or pY907). PARP1 pY907 increases PARP1 enzymatic activity and reduces binding to a PARP inhibitor, thereby rendering cancer cells resistant to PARP inhibition. The combination of c-Met and PARP1 inhibitors synergized to suppress the growth of breast cancer cells in vitro and xenograft tumor models, and we observed similar synergistic effects in a lung cancer xenograft tumor model. These results suggest that the abundance of PARP1 pY907 may predict tumor resistance to PARP inhibitors, and that treatment with a combination of c-Met and PARP inhibitors may benefit patients whose tumors show high c-Met expression and who do not respond to PARP inhibition alone.

Collaboration


Dive into the Jennifer L. Hsu's collaboration.

Top Co-Authors

Avatar

Mien Chie Hung

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

Chia Wei Li

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

Yan Wang

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Yi Du

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Gabriel N. Hortobagyi

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

Longfei Huo

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
Researchain Logo
Decentralizing Knowledge