Network


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

Hotspot


Dive into the research topics where Hua Xie is active.

Publication


Featured researches published by Hua Xie.


Cancer Prevention Research | 2013

Direct Targeting of MEK1/2 and RSK2 by Silybin Induces Cell-Cycle Arrest and Inhibits Melanoma Cell Growth

Mee Hyun Lee; Zunnan Huang; Dong Joon Kim; Sung Hyun Kim; Myoung Ok Kim; Sung Young Lee; Hua Xie; Si Jun Park; Jae Young Kim; Joydeb Kumar Kundu; Ann M. Bode; Young-Joon Surh; Zigang Dong

Abnormal functioning of multiple gene products underlies the neoplastic transformation of cells. Thus, chemopreventive and/or chemotherapeutic agents with multigene targets hold promise in the development of effective anticancer drugs. Silybin, a component of milk thistle, is a natural anticancer agent. In the present study, we investigated the effect of silybin on melanoma cell growth and elucidated its molecular targets. Our study revealed that silybin attenuated the growth of melanoma xenograft tumors in nude mice. Silybin inhibited the kinase activity of mitogen-activated protein kinase (MEK)-1/2 and ribosomal S6 kinase (RSK)-2 in melanoma cells. The direct binding of silybin with MEK1/2 and RSK2 was explored using a computational docking model. Treatment of melanoma cells with silybin attenuated the phosphorylation of extracellular signal-regulated kinase (ERK)-1/2 and RSK2, which are regulated by the upstream kinases MEK1/2. The blockade of MEK1/2-ERK1/2-RSK2 signaling by silybin resulted in a reduced activation of NF-κB, activator protein-1, and STAT3, which are transcriptional regulators of a variety of proliferative genes in melanomas. Silybin, by blocking the activation of these transcription factors, induced cell-cycle arrest at the G1 phase and inhibited melanoma cell growth in vitro and in vivo. Taken together, silybin suppresses melanoma growth by directly targeting MEK- and RSK-mediated signaling pathways. Cancer Prev Res; 6(5); 455–65. ©2013 AACR.


Carcinogenesis | 2014

[6]-Shogaol inhibits growth and induces apoptosis of non-small cell lung cancer cells by directly regulating Akt1/2

Myoung Ok Kim; Mee Hyun Lee; Naomi Oi; Sung Hyun Kim; Ki Beom Bae; Zunnan Huang; Dong Joon Kim; Kanamata Reddy; Sung Young Lee; Si Jun Park; Jae Young Kim; Hua Xie; Joydeb Kumar Kundu; Zae Young Ryoo; Ann M. Bode; Young-Joon Surh; Zigang Dong

Non-small cell lung cancer (NSCLC) is the leading cause of cancer mortality worldwide. Despite progress in developing chemotherapeutics for the treatment of NSCLC, primary and secondary resistance limits therapeutic success. NSCLC cells exhibit multiple mutations in the epidermal growth factor receptor (EGFR), which cause aberrant activation of diverse cell signaling pathways. Therefore, suppression of the inappropriate amplification of EGFR downstream signaling cascades is considered to be a rational therapeutic and preventive strategy for the management of NSCLC. Our initial molecular target-oriented virtual screening revealed that the ginger components, including [6]-shogaol, [6]-paradol and [6]-gingerol, seem to be potential candidates for the prevention and treatment of NSCLC. Among the compounds, [6]-shogaol showed the greatest inhibitory effects on the NSCLC cell proliferation and anchorage-independent growth. [6]-Shogaol induced cell cycle arrest (G1 or G2/M) and apoptosis. Furthermore, [6]-shogaol inhibited Akt kinase activity, a downstream mediator of EGFR signaling, by binding with an allosteric site of Akt. In NCI-H1650 lung cancer cells, [6]-shogaol reduced the constitutive phosphorylation of signal transducer and activator of transcription-3 (STAT3) and decreased the expression of cyclin D1/3, which are target proteins in the Akt signaling pathway. The induction of apoptosis in NCI-H1650 cells by [6]-shogaol corresponded with the cleavage of caspase-3 and caspase-7. Moreover, intraperitoneal administration of [6]-shogaol inhibited the growth of NCI-H1650 cells as tumor xenografts in nude mice. [6]-Shogaol suppressed the expression of Ki-67, cyclin D1 and phosphorylated Akt and STAT3 and increased terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling-positivity in xenograft tumors. The current study clearly indicates that [6]-shogaol can be exploited for the prevention and/or treatment of NSCLC.


Cancer Research | 2013

Identification of an Aurora Kinase Inhibitor Specific for the Aurora B Isoform

Hua Xie; Mee Hyun Lee; Feng Zhu; Kanamata Reddy; Cong Peng; Yan Li; Do Young Lim; Dong Joon Kim; Xiang Li; Soouk Kang; Haitao Li; Wei Ya Ma; Ronald A. Lubet; Jian Ding; Ann M. Bode; Zigang Dong

Aurora kinases play an important role in chromosome alignment, segregation, and cytokinesis during mitosis. In the present study, we used a ligand docking method to explore the novel scaffold of potential Aurora B inhibitors. One thousand compounds from our in-house compound library were screened against the Aurora B structure and one compound, (E)-3-((E)-4-(benzo[d][1,3]dioxol-5-yl)-2-oxobut-3-en-1-ylidene)indolin-2-one (designated herein as HOI-07) was selected for further study. HOI-07 potently inhibited in vitro Aurora B kinase activity in a dose-dependent manner, without obvious inhibition of another 49 kinases, including Aurora A. This compound suppressed Aurora B kinase activity in lung cancer cells, evidenced by the inhibition of the phosphorylation of histone H3 on Ser10 in a dose- and time-dependent manner. This inhibition resulted in apoptosis induction, G(2)-M arrest, polyploidy cells, and attenuation of cancer cell anchorage-independent growth. Moreover, knocking down the expression of Aurora B effectively reduced the sensitivity of cancer cells to HOI-07. Results of an in vivo xenograft mouse study showed that HOI-07 treatment effectively suppressed the growth of A549 xenografts, without affecting the body weight of mice. The expression of phospho-histone H3, phospho-Aurora B, and Ki-67 was also suppressed in the HOI-07 treatment group. Taken together, we identified HOI-07 as a specific Aurora B inhibitor, which deserves further investigation.


Molecular Cancer Therapeutics | 2013

Discovery of the novel mTOR inhibitor and its antitumor activities in vitro and in vivo

Hua Xie; Mee Hyun Lee; Feng Zhu; Kanamata Reddy; Zunnan Huang; Dong Joon Kim; Yan Li; Cong Peng; Do Young Lim; Soouk Kang; Sung Keun Jung; Xiang Li; Haitao Li; Wei Ya Ma; Ronald A. Lubet; Jian Ding; Ann M. Bode; Zigang Dong

The phosphoinositide 3-kinase (PI3-K)/Akt and mTOR signaling pathway plays a critical role in cell survival and proliferation and is often aberrantly activated in many types of cancer. The mTOR kinase protein, one of the key molecules in this pathway, has been shown to be an important target for cancer therapy. In the present study, a ligand docking method was used to screen for novel scaffold mTOR inhibitors. Sixty thousand compounds in the Natural Product Database were screened against the mTOR homologous structure, and 13 commercially available compounds listed in the top-ranked 100 compounds were selected for further examination. Compound [(E)-3-(4-(benzo[d][1,3]dioxol-5-yl)-2-oxobut-3-en-1-yl)- 3-hydroxyindolin-2-one; designated herein as 3HOI-BA-01] was then selected for further study of its antitumor activity. An in vitro study has shown that 3HOI-BA-01 inhibited mTOR kinase activity in a dose-dependent manner by directly binding with mTOR. In a panel of non–small cell lung cancer cells, the compound also attenuated mTOR downstream signaling, including the phosphorylation of p70S6K, S6, and Akt, resulting in G1 cell-cycle arrest and growth inhibition. Results of an in vivo study have shown that intraperitoneal injection of 3HOI-BA-01 in A549 lung tumor–bearing mice effectively suppressed cancer growth without affecting the body weight of the mice. The expression of downstream signaling molecules in the mTOR pathway in tumor tissues was also reduced after 3HOI-BA-01 treatment. Taken together, we identified 3HOI-BA-01 as a novel and effective mTOR inhibitor. Mol Cancer Ther; 12(6); 950–8. ©2013 AACR.


Carcinogenesis | 2012

Identification of mammalian target of rapamycin as a direct target of fenretinide both in vitro and in vivo

Hua Xie; Feng Zhu; Zunnan Huang; Mee Hyun Lee; Dong Joon Kim; Xiang Li; Do Young Lim; Sung Keun Jung; Soouk Kang; Haitao Li; Kanamata Reddy; Lei Wang; Wei Ya Ma; Ronald A. Lubet; Ann M. Bode; Zigang Dong

N-(4-hydroxyphenyl) retinamide (4HPR, fenretinide) is a synthetic retinoid that has been tested in clinical trials as a cancer therapeutic and chemopreventive agent. Although 4HPR has been shown to be cytotoxic to many kinds of cancer cells, the underlying molecular mechanisms are only partially understood. Until now, no direct cancer-related molecular target has been reported to be involved in the antitumor activities of 4HPR. Herein, we found that 4HPR inhibited mammalian target of rapamycin (mTOR) kinase activity by directly binding with mTOR, which suppressed the activities of both the mTORC1 and the mTORC2 complexes. The predicted binding mode of 4HPR with mTOR was based on a homology computer model, which showed that 4HPR could bind in the ATP-binding pocket of the mTOR protein through hydrogen bonds and hydrophobic interactions. In vitro studies also showed that 4HPR attenuated mTOR downstream signaling in a panel of non-small-cell lung cancer cells, resulting in growth inhibition. Moreover, knockdown of mTOR in cancer cells decreased their sensitivity to 4HPR. Results of an in vivo study demonstrated that i.p. injection of 4HPR in A549 lung tumor-bearing mice effectively suppressed cancer growth. The expression of mTOR downstream signaling molecules in tumor tissues was also decreased after 4HPR treatment. Taken together, our results are the first to identify mTOR as a direct antitumor target of 4HPR both in vitro and in vivo, providing a valuable rationale for guiding the clinical uses of 4HPR.


Carcinogenesis | 2012

Ceftriaxone, an FDA-approved cephalosporin antibiotic, suppresses lung cancer growth by targeting Aurora B

Xiang Li; Haitao Li; Shengqing Li; Feng Zhu; Dong Joon Kim; Hua Xie; Yan Li; Janos Nadas; Naomi Oi; Tatyana A. Zykova; Dong Hoon Yu; Mee Hyun Lee; Myoung Ok Kim; Lei Wang; Wei Ya Ma; Ronald A. Lubet; Ann M. Bode; Ziming Dong; Zigang Dong

Ceftriaxone, an FDA-approved third-generation cephalosporin antibiotic, has antimicrobial activity against both gram-positive and gram-negative organisms. Generally, ceftriaxone is used for a variety of infections such as community-acquired pneumonia, meningitis and gonorrhea. Its primary molecular targets are the penicillin-binding proteins. However, other activities of ceftriaxone remain unknown. Herein, we report for the first time that ceftriaxone has antitumor activity in vitro and in vivo. Kinase profiling results predicted that Aurora B might be a potential off target of ceftriaxone. Pull-down assay data confirmed that ceftriaxone could bind with Aurora B in vitro and in A549 cells. Furthermore, ceftriaxone (500 µM) suppressed anchorage-independent cell growth by targeting Aurora B in A549, H520 and H1650 lung cancer cells. Importantly, in vivo xenograft animal model results showed that ceftriaxone effectively suppressed A549 and H520 lung tumor growth by inhibiting Aurora B. These data suggest the anticancer efficacy of ceftriaxone for the treatment of lung cancers through its inhibition of Aurora B.


Carcinogenesis | 2013

CinQ-03, a novel allosteric MEK inhibitor, suppresses cancer growth in vitro and in vivo

Dong Joon Kim; Mee Hyun Lee; Kanamata Reddy; Yani Li; Do Young Lim; Hua Xie; Sung Young Lee; Young Il Yeom; Ann M. Bode; Zigang Dong

The mitogen-activated protein kinase kinase 1 and 2 signaling pathway is a major component of the RAS (Rat sarcoma)/RAF (Radpidly accelerated fibrosarcoma)/MEK (mitogen-activated protein kinase kinase)/ERKs (Extracellular signal-regulated kinases) signaling axis that regulates tumorigenesis and cancer cell growth. MEK is frequently activated in various cancers that have mutations in the KRAS and BRAF oncogenes. Therefore, MEK has been suggested as a therapeutic target for inhibitor development against tumors that are dependent on the activating mutations in mitogen-activated protein kinase signaling. Herein, we report the discovery of three novel MEK inhibitors, herein referred to as CInQ-01, CInQ-03 and CInQ-06. All three inhibitors were highly effective in suppressing MEK1 and MEK2 in vitro kinase activity as well as anchorage-dependent and anchorage-independent cell growth. The inhibitory activity was associated with markedly reduced phosphorylation of ERKs and ribosomal S6 kinases. Furthermore, administration of CInQ-03 inhibited colon cancer cell growth in an in vivo xenograft mouse model and showed no skin toxicity. Overall, these results suggest that these novel MEK inhibitors might be used for chemotherapy or prevention.


npj Precision Oncology | 2017

Inhibiting breast cancer by targeting the thromboxane A 2 pathway

Haitao Li; Mee-Hyun Lee; Kangdong Liu; Ting Wang; Mengqiu Song; Yaping Han; Ke Yao; Hua Xie; Feng Zhu; Michael E. Grossmann; Margot P. Cleary; Wei Chen; Ann M. Bode; Zigang Dong

Targeting the estrogen receptor as a strategy has been the gold standard for breast cancer chemoprevention or breast cancer recurrence, but its benefit is limited to estrogen receptor-positive tumors. Cyclooxygenases have been implicated in mammary tumorigenesis. We sought to identify the key prostaglandin responsible for the pro-neoplastic effect of cyclooxygenases and develop prostaglandin-targeted strategies for breast cancer chemoprevention or therapy. Immunohistochemical analysis revealed that either thromboxane A2 synthase 1 or the thromboxane A2 receptor is highly expressed in human breast tumors as well as premalignant lesions, but not in normal mammary tissues. Clinically, the thromboxane A2 pathway might be associated with HER2-positive and axillary lymph node metastasis in human breast cancer. We found that the thromboxane A2 pathway was required for breast cancer cell growth, anchorage-independent growth and invasion capabilities. Importantly, we discovered that switching off thromboxane A2 biosynthesis effectively suppressed either MMTV-HER2-driven mammary tumorigenesis or breast cancer metastasis in preclinical animal models. Taken together, this study established a critical pathophysiological role of the thromboxane A2 pathway in breast cancer, and provided a rationale for introducing a strategy targeting thromboxane A2 for breast cancer chemoprevention and therapy.Breast cancer: Pathway target identifiedThe identification of a signaling pathway connected to the progression of breast cancer could prove a valuable therapeutic target. The breast cancer treatment tamoxifen, although successful, is limited to certain tumor types, and so the search is on to pinpoint molecular targets that are ubiquitous across breast cancers. Zigang Dong at the University of Minnesota, US, and co-workers identified that the thromboxane A2 (TXA2) pathway is highly-expressed in human breast tumors and premalignant lesions. Further examination showed that TXA2 is crucial for tumor cell growth and metastasis. When the researchers knocked-out TXA2 production in mouse models, this suppressed both the formation and spread of tumors. The team advise caution because TXA2 is also required for wound healing and other key processes, although short-term bursts of pathway suppression may be an option for breast cancer therapy.


Cancer Research | 2012

Abstract 154: [6]-Shogaol inhibits cell growth and induces apoptosis in non-small-cell lung cancer (NSCLC) cells by regulating multiple targets

Mee-Hyun Lee; Myoung Ok Kim; Naomi Oi; Ki Beom Bae; Zunnan Huang; Soouk Kang; Hua Xie; Tae-Gyu Lim; Ann M. Bode; Zigang Dong

Proceedings: AACR 103rd Annual Meeting 2012‐‐ Mar 31‐Apr 4, 2012; Chicago, ILnnNon-small-cell lung cancer (NSCLC) is the leading cause of cancer mortality worldwide. Despite substantial progress in developing chemotherapeutics for the treatment of NSCLC, primary and secondary resistance limits the success of these therapies. NSCLC cells exhibit different EGFR mutations and EGFR has extensive cross-talk with other cell signaling pathways. Therefore, suppression of the inappropriate expression of multiple targets is considered to be a rational therapeutic and preventive strategy for the management of NSCLC. Our initial molecular-target oriented virtual screening revealed that the ingredients of ginger, including [6]-gingerol, [6]-shogaol and [6]-paradol appeared to be potential candidates for the prevention and therapy of NSCLC cells. Among the three compounds, [6]-shogaol showed the greatest inhibitory effect on growth and soft agar colony formation of NSCLC cells. [6]-Shogaol induced cell cycle arrest (G1 or G2/M) and apoptosis. Furthermore, [6]-shogaol interacted with PI3-K and AKT and inhibited AKT kinase activity. Finally, [6]-shogaol increased the expression of p27, which is one of the target proteins in the AKT signaling pathway. The present results clearly indicate that [6]-shogaol exerted a strong anti-cancer effect and can be regarded as useful tool for the treatment of NSCLC.nnCitation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 154. doi:1538-7445.AM2012-154


Cancer Research | 2012

Abstract 4968: Phosphorylation of caspase-8 (Thr263) by ribosomal S6 kinase 2 (RSK2) mediates caspase-8 ubiquitination and stability

Cong Peng; Yong-Yeon Cho; Feng Zhu; Haitao Li; Xiang Li; Hua Xie; Ann M. Bode; Zigang Dong

The ribosomal S6 kinase 2 (RSK2) is a member of the p90 ribosomal S6 kinase (p90RSK) family of proteins and plays a critical role in proliferation, cell cycle and transformation. Here, we report that RSK2 phosphorylates caspase-8 and Thr263 was identified as a novel caspase-8 phosphorylation site. Phosphorylation of Thr263 is associated with the ubiquitination and degradation of caspase through the proteasome pathway. Results suggested that RSK2 regulates caspase-8 stability through its phosphorylation of this caspase. These data provide a direct link between RSK2 and caspase-8 and identify a novel molecular mechanism for caspase-8 modulation by RSK2. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 4968. doi:1538-7445.AM2012-4968

Collaboration


Dive into the Hua Xie's collaboration.

Top Co-Authors

Avatar

Ann M. Bode

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar

Zigang Dong

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Feng Zhu

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar

Haitao Li

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar

Mee Hyun Lee

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar

Xiang Li

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ronald A. Lubet

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Wei Ya Ma

University of Minnesota

View shared research outputs
Researchain Logo
Decentralizing Knowledge