Network


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

Hotspot


Dive into the research topics where Huiyuan Zhang is active.

Publication


Featured researches published by Huiyuan Zhang.


Journal of Experimental Medicine | 2014

USP21 negatively regulates antiviral response by acting as a RIG-I deubiquitinase

Yihui Fan; Renfang Mao; Yang Yu; Shangfeng Liu; Zhongcheng Shi; Jin Cheng; Huiyuan Zhang; Lei An; Yanling Zhao; Xin Xu; Zhenghu Chen; Mari Kogiso; Dekai Zhang; Hong Zhang; Pumin Zhang; Jae U. Jung; Xiao-Nan Li; Guo-Tong Xu; Jianhua Yang

The deubiquitinase USP21 targets RIG-I for deubiquitination, thus dampening interferon production and activation of IFN-responsive genes in response to RNA viruses.


Apoptosis | 2015

MTOR ATP-competitive inhibitor INK128 inhibits neuroblastoma growth via blocking mTORC signaling

Huiyuan Zhang; Jun Dou; Yang Yu; Yanling Zhao; Yihui Fan; Jin Cheng; Xin Xu; Wei Liu; Shan Guan; Zhenghu Chen; Yan Shi; Roma Patel; Sanjeev A. Vasudevan; Peter E. Zage; Hong Zhang; Jed G. Nuchtern; Eugene S. Kim; Songbin Fu; Jianhua Yang

High-risk neuroblastoma often develops resistance to high-dose chemotherapy. The mTOR signaling cascade is frequently deregulated in human cancers and targeting mTOR signaling sensitizes many cancer types to chemotherapy. Here, using a panel of neuroblastoma cell lines, we found that the mTOR inhibitor INK128 showed inhibitory effects on both anchorage-dependent and independent growth of neuroblastoma cells and significantly enhanced the cytotoxic effects of doxorubicin (Dox) on these cell lines. Treatment of neuroblastoma cells with INK128 blocked the activation of downstream mTOR signaling and enhanced Dox-induced apoptosis. Moreover, INK128 was able to overcome the established chemoresistance in the LA-N-6 cell line. Using an orthotopic neuroblastoma mouse model, we found that INK128 significantly inhibited tumor growth in vivo. In conclusion, we have shown that INK128-mediated mTOR inhibition possessed substantial antitumor activity and could significantly increase the sensitivity of neuroblastoma cells to Dox therapy. Taken together, our results indicate that using INK128 can provide additional efficacy to current chemotherapeutic regimens and represent a new paradigm in restoring drug sensitivity in neuroblastoma.


Scientific Reports | 2016

Novel ALK inhibitor AZD3463 inhibits neuroblastoma growth by overcoming crizotinib resistance and inducing apoptosis

Yongfeng Wang; Long Wang; Shan Guan; Wenming Cao; Hao Wang; Zhenghu Chen; Yanling Zhao; Yang Yu; Huiyuan Zhang; Jonathan C. Pang; Sophia L. Huang; Yo Akiyama; Yifan Yang; Wenjing Sun; Xin Xu; Yan Shi; Hong Zhang; Eugene S. Kim; Jodi A. Muscal; Fengmin Lu; Jianhua Yang

ALK receptor tyrosine kinase has been shown to be a therapeutic target in neuroblastoma. Germline ALK activating mutations are responsible for the majority of hereditary neuroblastoma and somatic ALK activating mutations are also frequently observed in sporadic cases of advanced NB. Crizotinib, a first-line therapy in the treatment of advanced non-small cell lung cancer (NSCLC) harboring ALK rearrangements, demonstrates striking efficacy against ALK-rearranged NB. However, crizotinib fails to effectively inhibit the activity of ALK when activating mutations are present within its kinase domain, as with the F1174L mutation. Here we show that a new ALK inhibitor AZD3463 effectively suppressed the proliferation of NB cell lines with wild type ALK (WT) as well as ALK activating mutations (F1174L and D1091N) by blocking the ALK-mediated PI3K/AKT/mTOR pathway and ultimately induced apoptosis and autophagy. In addition, AZD3463 enhanced the cytotoxic effects of doxorubicin on NB cells. AZD3463 also exhibited significant therapeutic efficacy on the growth of the NB tumors with WT and F1174L activating mutation ALK in orthotopic xenograft mouse models. These results indicate that AZD3463 is a promising therapeutic agent in the treatment of NB.


Scientific Reports | 2016

TAK1 inhibitor NG25 enhances doxorubicin-mediated apoptosis in breast cancer cells

Zhenyu Wang; Huiyuan Zhang; Minghao Shi; Yang Yu; Hao Wang; Wenming Cao; Yanling Zhao; Hong Zhang

Doxorubicin (Dox, Adriamycin) has been widely used in breast cancer treatment. But its severe cardio-toxic side effects limited the clinical use. Dox treatment can induce DNA damage and other accompanying effects in cancer cells, and subsequently activates nuclear factor κB (NF-κB) pathway which has a strong pro-survival role in different types of malignancy. We hypothesize that blocking NF-κB pathway may sensitize breast cancer cells to Dox chemotherapy. TGFβ-activated kinase-1 (TAK1) is a key intracellular molecule participating in genotoxic stresses-induced NF-κB activation. Targeting TAK1 as a strategy to enhance cancer treatment efficacy has been studied in several malignancies. We showed that NG25, a synthesized TAK1 inhibitor, greatly enhanced Dox treatment efficacy in a panel of breast cancer cell lines. In this pre-clinical study, we found that NG25 partially blocked Dox-induced p38 phosphorylation and IκBα degradation and enhanced Dox-induced cytotoxic effects and apoptosis in all breast cancer cell lines tested. Taken together, we provided clear evidence that NG25 sensitizes the breast cancer cells to Dox treatment in vitro. This combination may be an effective and feasible therapeutic option maximizing Dox efficacy and meanwhile minimizing Dox side effects in treating breast cancer.


The Journal of Pathology | 2014

Amplification and over-expression of MAP3K3 gene in human breast cancer promotes formation and survival of breast cancer cells

Yihui Fan; Ningling Ge; Xiaosong Wang; Wenjing Sun; Renfang Mao; Wen Bu; Chad J. Creighton; Pingju Zheng; Sanjeev A. Vasudevan; Lei An; Jinshu Yang; Yi-Jue Zhao; Huiyuan Zhang; Xiao-Nan Li; Pulivarthi H. Rao; Eastwood Leung; Yong-Jie Lu; Joe W. Gray; Rachel Schiff; Susan G. Hilsenbeck; C. Kent Osborne; Jianhua Yang; Hong Zhang

Gene amplifications in the 17q chromosomal region are observed frequently in breast cancers. An integrative bioinformatics analysis of this region nominated the MAP3K3 gene as a potential therapeutic target in breast cancer. This gene encodes mitogen‐activated protein kinase kinase kinase 3 (MAP3K3/MEKK3), which has not yet been reported to be associated with cancer‐causing genetic aberrations. We found that MAP3K3 was amplified in approximately 8–20% of breast cancers. Knockdown of MAP3K3 expression significantly inhibited cell proliferation and colony formation in MAP3K3‐amplified breast cancer cell lines MCF‐7 and MDA‐MB‐361 but not in MAP3K3 non‐amplified breast cancer cells. Knockdown of MAP3K3 expression in MAP3K3‐amplified breast cancer cells sensitized breast cancer cells to apoptotic induction by TNFα and TRAIL, as well as doxorubicin, VP‐16 and fluorouracil, three commonly used chemotherapeutic drugs for treating breast cancer. In addition, ectopic expression of MAP3K3, in collaboration with Ras, induced colony formation in both primary mouse embryonic fibroblasts and immortalized human breast epithelial cells (MCF‐10A). Combined, these results suggest that MAP3K3 contributes to breast carcinogenesis and may endow resistance of breast cancer cells to cytotoxic chemotherapy. Therefore, MAP3K3 may be a valuable therapeutic target in patients with MAP3K3‐amplified breast cancers, and blocking MAP3K3 kinase activity with a small molecule inhibitor may sensitize MAP3K3‐amplified breast cancer cells to chemotherapy. Copyright


Cancer Letters | 2017

The second-generation ALK inhibitor alectinib effectively induces apoptosis in human neuroblastoma cells and inhibits tumor growth in a TH-MYCN transgenic neuroblastoma mouse model

Jiaxiong Lu; Shan Guan; Yanling Zhao; Yang Yu; Sarah E. Woodfield; Huiyuan Zhang; Kristine L. Yang; Shayahati Bieerkehazhi; Lin Qi; Xiao-Nan Li; Jerry Gu; Xin Xu; Jingling Jin; Jodi A. Muscal; Tianshu Yang; Guo-Tong Xu; Jianhua Yang

Activating germline mutations of anaplastic lymphoma kinase (ALK) occur in most cases of hereditary neuroblastoma (NB) and the constitutively active kinase activity of ALK promotes cell proliferation and survival in NB. Therefore, ALK kinase is a potential therapeutic target for NB. In this study, we show that the novel ALK inhibitor alectinib effectively suppressed cell proliferation and induces apoptosis in NB cell lines with either wild-type ALK or mutated ALK (F1174L and D1091N) by blocking ALK-mediated PI3K/Akt/mTOR signaling. In addition, alectinib enhanced doxorubicin-induced cytotoxicity and apoptosis in NB cells. Furthermore, alectinib induced apoptosis in an orthotopic xenograft NB mouse model. Also, in the TH-MYCN transgenic mouse model, alectinib resulted in decreased tumor growth and prolonged survival time. These results indicate that alectinib may be a promising therapeutic agent for the treatment of NB.


Scientific Reports | 2016

Wip1 inhibitor GSK2830371 inhibits neuroblastoma growth by inducing Chk2/p53-mediated apoptosis

Zhenghu Chen; Long Wang; Dayong Yao; Tianshu Yang; Wen Ming Cao; Jun Dou; Jonathan C. Pang; Shan Guan; Huiyuan Zhang; Yang Yu; Yanling Zhao; Yongfeng Wang; Xin Xu; Yan Shi; Roma Patel; Hong Zhang; Sanjeev A. Vasudevan; Shangfeng Liu; Jianhua Yang; Jed G. Nuchtern

Neuroblastoma (NB) is the most common extracranial tumor in children. Unlike in most adult tumors, tumor suppressor protein 53 (p53) mutations occur with a relatively low frequency in NB and the downstream function of p53 is intact in NB cell lines. Wip1 is a negative regulator of p53 and hindrance of Wip1 activity by novel inhibitor GSK2830371 is a potential strategy to activate p53’s tumor suppressing function in NB. Yet, the in vivo efficacy and the possible mechanisms of GSK2830371 in NB have not yet been elucidated. Here we report that novel Wip1 inhibitor GSK2830371 induced Chk2/p53-mediated apoptosis in NB cells in a p53-dependent manner. In addition, GSK2830371 suppressed the colony-formation potential of p53 wild-type NB cell lines. Furthermore, GSK2830371 enhanced doxorubicin- (Dox) and etoposide- (VP-16) induced cytotoxicity in a subset of NB cell lines, including the chemoresistant LA-N-6 cell line. More importantly, GSK2830371 significantly inhibited tumor growth in an orthotopic xenograft NB mouse model by inducing Chk2/p53-mediated apoptosis in vivo. Taken together, this study suggests that GSK2830371 induces Chk2/p53-mediated apoptosis both in vitro and in vivo in a p53 dependent manner.


Oncotarget | 2017

Novel multi-targeted ErbB family inhibitor afatinib blocks EGF-induced signaling and induces apoptosis in neuroblastoma

Xinfang Mao; Zhenghu Chen; Yanling Zhao; Yang Yu; Shan Guan; Sarah E. Woodfield; Sanjeev A. Vasudevan; Ling Tao; Jonathan C. Pang; Jiaxiong Lu; Huiyuan Zhang; Fuchun Zhang; Jianhua Yang

Neuroblastoma is the most common extracranial solid tumor in children. The ErbB family of proteins is a group of receptor tyrosine kinases that promote the progression of various malignant cancers including neuroblastoma. Thus, targeting them with small molecule inhibitors is a promising strategy for neuroblastoma therapy. In this study, we investigated the anti-tumor effect of afatinib, an irreversible inhibitor of members of the ErbB family, on neuroblastoma. We found that afatinib suppressed the proliferation and colony formation ability of neuroblastoma cell lines in a dose-dependent manner. Afatinib also induced apoptosis and blocked EGF-induced activation of PI3K/AKT/mTOR signaling in all neuroblastoma cell lines tested. In addition, afatinib enhanced doxorubicin-induced cytotoxicity in neuroblastoma cells, including the chemoresistant LA-N-6 cell line. Finally, afatinib exhibited antitumor efficacy in vivo by inducing apoptosis in an orthotopic xenograft neuroblastoma mouse model. Taken together, these results show that afatinib inhibits neuroblastoma growth both in vitro and in vivo by suppressing EGFR-mediated PI3K/AKT/mTOR signaling. Our study supports the idea that EGFR is a potential therapeutic target in neuroblastoma. And targeting ErbB family protein kinases with small molecule inhibitors like afatinib alone or in combination with doxorubicin is a viable option for treating neuroblastoma.


International Journal of Oncology | 2016

The novel mTORC1/2 dual inhibitor INK128 enhances radiosensitivity of breast cancer cell line MCF-7.

Zhi-gang Liu; Jiao Tang; Zhenghu Chen; Huiyuan Zhang; Hui Wang; Jianhua Yang; Hong Zhang

mTOR, a member of the PIKK family, is crucial for cell growth, survival, motility, proliferation, protein synthesis and DNA transcription. Many studies have demonstrated that mTOR inhibitor could enhance radiosensitivity. However, the effect of the novel mTORC1/2 dual inhibitor, INK128, on the radiosensitivity of breast cancer and the underlying mechanisms are still vague. In the present study, the cell viability was estimated using CCK-8 assay, and the dose-survival relationship was analyzed using a clonogenic survival assay. Cell cycle was evaluated by flow cytometry. The staining of γH2AX foci was assessed by immunofluorescence. In addition, we used western blots to verify the downregulating signal protein and to detect the potential related pathway. We found that the exposure of MCF-7 cells to INK128 decreased the cell viability. Exposure of MCF-7 cells to INK128 and combined ionizing radiation greatly reduced the survival rate. INK128 combined radiotherapy significantly induced G2/M arrest, double strand breaks and inhibited its repair. Furthermore, INK128 plus radiation downregulated p-Chk2, p21 and upregulated cleaved PARP, LC3B expression. These findings suggest that mTOR inhibitor could be used as a novel radiosensitizing target for breast cancer patients.


Oncotarget | 2017

Novel multiple tyrosine kinase inhibitor ponatinib inhibits bFGF- activated signaling in neuroblastoma cells and suppresses neuroblastoma growth in vivo

Haoyu Li; Yongfeng Wang; Zhenghu Chen; Jiaxiong Lu; Jessie Pan; Yang Yu; Yanling Zhao; Huiyuan Zhang; Ting Hu; Qing Liu; Jianhua Yang

Neuroblastoma (NB) is one of the most common pediatric malignancies in children. Abnormal activation of receptor tyrosine kinases contributes to the pathological development of NB. Therefore, targeting tyrosine kinase receptors to cure NB is a promising strategy. Here, we report that a multi-targeted tyrosine kinase inhibitor ponatinib inhibited NB cell proliferation and induced NB cell apoptosis in a dose-dependent manner. In addition, ponatinib suppressed the colony formation ability of NB cells. Mechanistically, ponatinib effectively inhibited the FGFR1-activated signaling pathway. Ponatinib also enhanced the cytotoxic effects of doxorubicin on NB cells. Furthermore, ponatinib demonstrated anti-tumor efficacy in vivo by inhibiting tumor growth in an orthotopic xenograft NB mouse model. In summary, our results showed that ponatinib inhibited NB growth both in vitro and in vivo.

Collaboration


Dive into the Huiyuan Zhang's collaboration.

Top Co-Authors

Avatar

Xiao-Nan Li

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Frank Braun

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Holly Lindsay

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Lin Qi

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Mari Kogiso

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Sibo Zhao

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Yuchen Du

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Sarah Injac

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Patricia Baxter

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Jack Su

Baylor College of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge