Network


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

Hotspot


Dive into the research topics where Wei Chao Chang is active.

Publication


Featured researches published by Wei Chao Chang.


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 Biological Chemistry | 2011

Nuclear translocation of epidermal growth factor receptor by Akt-dependent phosphorylation enhances breast cancer-resistant protein expression in gefitinib-resistant cells

Wei Chien Huang; Yun Ju Chen; Long Yuan Li; Ya Ling Wei; Sheng Chieh Hsu; Shing Ling Tsai; Pei Chun Chiu; Wei-Pang Huang; Ying Nai Wang; Chung-Hsuan Chen; Wei Chao Chang; Wen Chang Chang; Andy Jer En Chen; Chang Hai Tsai; Mien Chie Hung

Epidermal growth factor receptor (EGFR), an aberrantly overexpressed or activated receptor-tyrosine kinase in many cancers, plays a pivotal role in cancer progression and has been an attractive target for cancer therapy. Gefitinib and erlotinib, two EGFR-tyrosine kinase inhibitors, have been approved for non-small cell lung cancer. However, durable clinical efficacy of these EGFR inhibitors is severely limited by the emergence of acquired resistance. For example, the expression of breast cancer-resistant protein (BCRP/ABCG2) has been shown to confer acquired resistance of wild-type EGFR (wtEGFR)-expressing cancer cells to gefitinib. However, the underlying molecular mechanisms still remain unclear. Here, we show that wtEGFR expression is elevated in the nucleus of acquired gefitinib-resistant cancer cells. Moreover, nuclear translocation of EGFR requires phosphorylation at Ser-229 by Akt. In the nucleus, EGFR then targets the proximal promoter of BCRP/ABCG2 and thereby enhances its gene transcription. The nuclear EGFR-mediated BCRP/ABCG2 expression may contribute at least in part to the acquired resistance of wtEGFR-expressing cancer cells to gefitinib. Our findings shed light on the role of nuclear EGFR in the sensitivity of wtEGFR-expressing cancer cells to EGFR tyrosine kinase inhibitors and also deciphered a putative molecular mechanism contributing to gefitinib resistance through BCRP/ABCG2 expression.


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.


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.


Journal of Biological Chemistry | 2012

Nuclear EGFR Suppresses Ribonuclease Activity of Polynucleotide Phosphorylase through DNAPK-mediated Phosphorylation at Serine 776

Yung Luen Yu; Ruey Hwang Chou; Chia Han Wu; Ying Nai Wang; Wei Jung Chang; Yen Ju Tseng; Wei Chao Chang; Chien-Chen Lai; Hong Jen Lee; Longfei Huo; Chung-Hsuan Chen; Mien Chie Hung

Background: Nuclear translocation and activity of EGFR are correlated with radioresistance of cancer. Results: PNPase, identified as a novel EGFR interacting partner, is inactivated by DNAPK-mediated Ser-776 phosphorylation and contributes to radiosensitivity of cancer. Conclusion: An entirely new mechanism of nuclear EGFR in radioresistance is discovered. Significance: This study provides new insight into resistance of breast cancer to radiotherapy. Nuclear existence of epidermal growth factor receptor (EGFR) has been documented for more than two decades. Resistance of cancer to radiotherapy is frequently correlated with elevated EGFR expression, activity, and nuclear translocation. However, the role of nuclear EGFR (nEGFR) in radioresistance of cancers remains elusive. In the current study, we identified a novel nEGFR-associated protein, polynucleotide phosphorylase (PNPase), which possesses 3′ to 5′ exoribonuclease activity toward c-MYC mRNA. Knockdown of PNPase increased radioresistance. Inactivation or knock-down of EGFR enhanced PNPase-mediated c-MYC mRNA degradation in breast cancer cells, and also increased its radiosensitivity. Interestingly, the association of nEGFR with PNPase and DNA-dependent protein kinase (DNAPK) increased significantly in breast cancer cells after exposure to ionizing radiation (IR). We also demonstrated that DNAPK phosphorylates PNPase at Ser-776, which is critical for its ribonuclease activity. The phospho-mimetic S776D mutant of PNPase impaired its ribonuclease activity whereas the nonphosphorylatable S776A mutant effectively degraded c-MYC mRNA. Here, we uncovered a novel role of nEGFR in radioresistance, and that is, upon ionizing radiation, nEGFR inactivates the ribonuclease activity of PNPase toward c-MYC mRNA through DNAPK-mediated Ser-776 phosphorylation, leading to increase of c-MYC mRNA, which contributes to radioresistance of cancer cells.


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.


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.


Developmental Cell | 2014

EGFR Modulates DNA Synthesis and Repair through Tyr Phosphorylation of Histone H4

Ruey Hwang Chou; Ying Nai Wang; Yi-Hsien Hsieh; Long Yuan Li; Weiya Xia; Wei Chao Chang; Ling Chu Chang; Chien Chia Cheng; Chien-Chen Lai; Jennifer L. Hsu; Wei Jung Chang; Shu Ya Chiang; Hong Jen Lee; Hsin Wei Liao; Pei Huan Chuang; Hui Yu Chen; Hung Ling Wang; Sheng Chu Kuo; Chung-Hsuan Chen; Yung Luen Yu; Mien Chie Hung

Posttranslational modifications of histones play fundamental roles in many biological functions. Specifically, histone H4-K20 methylation is critical for DNA synthesis and repair. However, little is known about how these functions are regulated by the upstream stimuli. Here, we identify a tyrosine phosphorylation site at Y72 of histone H4, which facilitates recruitment of histone methyltransferases (HMTases), SET8 and SUV4-20H, to enhance its K20 methylation, thereby promoting DNA synthesis and repair. Phosphorylation-defective histone H4 mutant is deficient in K20 methylation, leading to reduced DNA synthesis, delayed cell cycle progression, and decreased DNA repair ability. Disrupting the interaction between epidermal growth factor receptor (EGFR) and histone H4 by Y72 peptide significantly reduced tumor growth. Furthermore, EGFR expression clinically correlates with histone H4-Y72 phosphorylation, H4-K20 monomethylation, and the Ki-67 proliferation marker. These findings uncover a mechanism by which EGFR transduces signal to chromatin to regulate DNA synthesis and repair.


Cancer Cell | 2013

Epidermal growth factor receptor potentiates MCM7-mediated DNA replication through tyrosine phosphorylation of Lyn kinase in human cancers.

Tzu Hsuan Huang; Longfei Huo; Ying Nai Wang; Weiya Xia; Yongkun Wei; Shih Shin Chang; Wei Chao Chang; Yueh Fu Fang; Chun Te Chen; Jing Yu Lang; Chun Tu; Yan Wang; Ming Chuan Hsu; Hsu Ping Kuo; How Wen Ko; Jia Shen; Heng Huan Lee; Pei Chih Lee; Yun Wu; Chung-Hsuan Chen; Mien Chie Hung

Epidermal growth factor receptor (EGFR) initiates a signaling cascade that leads to DNA synthesis and cell proliferation, but its role in regulating DNA replication licensing is unclear. Here, we show that activated EGFR phosphorylates the p56 isoform of Lyn, p56(Lyn), at Y32, which then phosphorylates MCM7, a licensing factor critical for DNA replication, at Y600 to increase its association with other minichromosome maintenance complex proteins, thereby promoting DNA synthesis complex assembly and cell proliferation. Both p56(Lyn) Y32 and MCM7 Y600 phosphorylation are enhanced in proliferating cells and correlated with poor survival of breast cancer patients. These results establish a signaling cascade in which EGFR enhances MCM7 phosphorylation and DNA replication through Lyn phosphorylation in human cancer cells.

Collaboration


Dive into the Wei Chao Chang's collaboration.

Top Co-Authors

Avatar
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

Jennifer L. Hsu

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

Yan Wang

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ying Nai Wang

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

Hong Jen Lee

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
Researchain Logo
Decentralizing Knowledge