Network


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

Hotspot


Dive into the research topics where Chong Zhao is active.

Publication


Featured researches published by Chong Zhao.


Clinical Cancer Research | 2014

Gambogic Acid Induces Apoptosis in Imatinib-Resistant Chronic Myeloid Leukemia Cells via Inducing Proteasome Inhibition and Caspase-Dependent Bcr-Abl Downregulation

Xianping Shi; Xin Chen; Xiaofen Li; Xiaoying Lan; Chong Zhao; Shouting Liu; Hongbiao Huang; Ningning Liu; Siyan Liao; Wenbin Song; Ping Zhou; Shunqing Wang; Li Xu; Xuejun Wang; Q. Ping Dou; Jinbao Liu

Purpose: Chronic myelogenous leukemia (CML) is characterized by the constitutive activation of Bcr-Abl tyrosine kinase. Bcr-Abl-T315I is the predominant mutation that causes resistance to imatinib, cytotoxic drugs, and the second-generation tyrosine kinase inhibitors. The emergence of imatinib resistance in patients with CML leads to searching for novel approaches to the treatment of CML. Gambogic acid, a small molecule derived from Chinese herb gamboges, has been approved for phase II clinical trial for cancer therapy by the Chinese Food and Drug Administration (FDA). In this study, we investigated the effect of gambogic acid on cell survival or apoptosis in CML cells bearing Bcr-Abl-T315I or wild-type Bcr-Abl. Experimental Design: CML cell lines (KBM5, KBM5-T315I, and K562), primary cells from patients with CML with clinical resistance to imatinib, and normal monocytes from healthy volunteers were treated with gambogic acid, imatinib, or their combination, followed by measuring the effects on cell growth, apoptosis, and signal pathways. The in vivo antitumor activity of gambogic acid and its combination with imatinib was also assessed with nude xenografts. Results: Gambogic acid induced apoptosis and cell proliferation inhibition in CML cells and inhibited the growth of imatinib-resistant Bcr-Abl-T315I xenografts in nude mice. Our data suggest that GA-induced proteasome inhibition is required for caspase activation in both imatinib-resistant and -sensitive CML cells, and caspase activation is required for gambogic acid–induced Bcr-Abl downregulation and apoptotic cell death. Conclusions: These findings suggest an alternative strategy to overcome imatinib resistance by enhancing Bcr-Abl downregulation with the medicinal compound gambogic acid, which may have great clinical significance in imatinib-resistant cancer therapy. Clin Cancer Res; 20(1); 151–63. ©2013 AACR.


Cell Reports | 2013

Gambogic Acid Is a Tissue-Specific Proteasome Inhibitor In Vitro and In Vivo

Xiaofen Li; Shouting Liu; Hongbiao Huang; Ningning Liu; Chong Zhao; Siyan Liao; Changshan Yang; Yurong Liu; Canguo Zhao; Shujue Li; Xiaoyu Lu; Chunjiao Liu; Lixia Guan; Kai Zhao; Xiaoqing Shi; Wenbin Song; Ping Zhou; Xiaoxian Dong; Haiping Guo; Guanmei Wen; Change Zhang; Lili Jiang; Ningfang Ma; Bing Li; Shunqing Wang; Huo Tan; Xuejun Wang; Q. Ping Dou; Jinbao Liu

Gambogic acid (GA) is a natural compound derived from Chinese herbs that has been approved by the Chinese Food and Drug Administration for clinical trials in cancer patients; however, its molecular targets have not been thoroughly studied. Here, we report that GA inhibits tumor proteasome activity, with potency comparable to bortezomib but much less toxicity. First, GA acts as a prodrug and only gains proteasome-inhibitory function after being metabolized by intracellular CYP2E1. Second, GA-induced proteasome inhibition is a prerequisite for its cytotoxicity and anticancer effect without off-targets. Finally, because expression of the CYP2E1 gene is very high in tumor tissues but low in many normal tissues, GA could therefore produce tissue-specific proteasome inhibition and tumor-specific toxicity, with clinical significance for designing novel strategies for cancer treatment.


Scientific Reports | 2015

A novel proteasome inhibitor suppresses tumor growth via targeting both 19S proteasome deubiquitinases and 20S proteolytic peptidases

Ningning Liu; Chunjiao Liu; Xiaofen Li; Siyan Liao; Wenbin Song; Changshan Yang; Chong Zhao; Hongbiao Huang; Lixia Guan; Peiquan Zhang; Shouting Liu; Xianliang Hua; Xin-Xin Chen; Ping Zhou; Xiaoying Lan; Songgang Yi; Shunqing Wang; Xuejun Wang; Q. Ping Dou; Jinbao Liu

The successful development of bortezomib-based therapy for treatment of multiple myeloma has established proteasome inhibition as an effective therapeutic strategy, and both 20S proteasome peptidases and 19S deubiquitinases (DUBs) are becoming attractive targets of cancer therapy. It has been reported that metal complexes, such as copper complexes, inhibit tumor proteasome. However, the involved mechanism of action has not been fully characterized. Here we report that (i) copper pyrithione (CuPT), an alternative to tributyltin for antifouling paint biocides, inhibits the ubiquitin-proteasome system (UPS) via targeting both 19S proteasome-specific DUBs and 20S proteolytic peptidases with a mechanism distinct from that of the FDA-approved proteasome inhibitor bortezomib; (ii) CuPT potently inhibits proteasome-specific UCHL5 and USP14 activities; (iii) CuPT inhibits tumor growth in vivo and induces cytotoxicity in vitro and ex vivo. This study uncovers a novel class of dual inhibitors of DUBs and proteasome and suggests a potential clinical strategy for cancer therapy.


Toxicology Letters | 2014

Anacardic acid induces cell apoptosis associated with induction of ATF4-dependent endoplasmic reticulum stress.

Hongbiao Huang; Xianliang Hua; Ningning Liu; Xiaofen Li; Shouting Liu; Xin Chen; Chong Zhao; Xiaoying Lan; Changshan Yang; Q. Ping Dou; Jinbao Liu

Anacardic acid (6-pentadecylsalicylic acid, AA), a natural compound isolated from the traditional medicine Amphipterygium adstringens, has been reported to possess antitumor activities. However, its molecular targets have not been thoroughly studied. Here, we report that AA is a potent inducer of endoplasmic reticulum (ER) stress, leading to apoptosis in hepatoma HepG2 and myeloma U266 cells. Induction of ER stress by AA was supported by a dose- and time-dependent increase in expression of the ER signaling downstream molecules, such as GRP78/BiP, phosphorylated eIF2α, ATF4 and CHOP in both HepG2 and U266 cell lines. Blockage of ATF4 expression by siRNA partially inhibited, while knockdown of CHOP expression by siRNA slightly increased AA-induced cell death in these cells. In addition, AA suppressed HepG2 xenograft tumor growth, associated with increased ER stress in vivo. These results suggest that AA induces tumor cell apoptosis associated with ATF4-dependent ER stress.


Oncotarget | 2016

Two clinical drugs deubiquitinase inhibitor auranofin and aldehyde dehydrogenase inhibitor disulfiram trigger synergistic anti-tumor effects in vitro and in vivo

Hongbiao Huang; Yuning Liao; Ningning Liu; Xianliang Hua; Jianyu Cai; Changshan Yang; Huidan Long; Chong Zhao; Xin Chen; Xiaoying Lan; Dan Zang; Jinjie Wu; Xiaofen Li; Xianping Shi; Xuejun Wang; Jinbao Liu

Inhibition of proteasome-associated deubiquitinases (DUBs) is emerging as a novel strategy for cancer therapy. It was recently reported that auranofin (Aur), a gold (I)-containing compound used clinically to treat rheumatoid arthritis, is a proteasome-associated DUB inhibitor. Disulfiram (DSF), an inhibitor of aldehyde dehydrogenase, is currently in clinical use for treating alcoholism. Recent studies have indicated that DSF can also act as an antitumor agent. We investigated the effect of combining DSF and Aur on apoptosis induction and tumor growth in hepatoma cancer cells. Here we report that (i) the combined treatment of Aur and DSF results in synergistic cytotoxicity to hepatoma cells in vitro and in vivo; (ii) Aur and DSF in combination induces caspase activation, endoplasmic reticulum (ER) stress, and reactive oxygen species (ROS) production; (iii) pan-caspase inhibitor z-VAD-FMK could efficiently block apoptosis but not proteasome inhibition induced by Aur and DSF combined treatment, and ROS is not required for Aur+DSF to induce apoptosis. Collectively, we demonstrate a model of synergism between DSF and proteasome-associated DUB inhibitor Aur in the induction of apoptosis in hepatoma cancer cells, identifying a potential novel anticancer strategy for clinical use in the future.


Biochemical Pharmacology | 2016

Platinum-containing compound platinum pyrithione is stronger and safer than cisplatin in cancer therapy.

Chong Zhao; Xin Chen; Dan Zang; Xiaoying Lan; Siyan Liao; Changshan Yang; Peiquan Zhang; Jinjie Wu; Xiaofen Li; Ningning Liu; Yuning Liao; Hongbiao Huang; Xianping Shi; Lili Jiang; Xiuhua Liu; Zhimin He; Xuejun Wang; Jinbao Liu

DNA is the well-known molecular target of current platinum-based anticancer drugs; consequently, their clinical use is severely restricted by their systemic toxicities and drug resistance originating from non-selective DNA damage. Various strategies have been developed to circumvent the shortcomings of platinum-based chemotherapy but the inherent problem remains unsolved. Here we report that platinum pyrithione (PtPT), a chemically well-characterized synthetic complex of platinum, inhibits proteasome function and thereby exhibits greater and more selective cytotoxicity to multiple cancer cells than cisplatin, without showing discernible DNA damage both in vitro and in vivo. Moreover, unlike the classical proteasome inhibitor bortezomib/Velcade which inhibits the proteasome via blocking the peptidase activity of 20S proteasomes, PtPT primarily deactivates 26S proteasome-associated deubiquitinases USP14 and UCHL5. Furthermore, PtPT can selectively induce cytotoxicity and proteasome inhibition in cancer cells from leukemia patients but not peripheral blood mononuclear cells from healthy humans. In nude mice, PtPT also remarkably inhibited tumor xenograft growth, without showing the adverse effects that were induced by cisplatin. Hence, we have discovered a new platinum-based anti-tumor agent PtPT which targets 26S proteasome-associated deubiquitinases rather than DNA in the cell and thereby exerts safer and more potent anti-tumor effects, identifying a highly translatable new platinum-based anti-cancer strategy.


Toxicology Letters | 2014

The combination of proteasome inhibitors bortezomib and gambogic acid triggers synergistic cytotoxicity in vitro but not in vivo.

Ningning Liu; Hongbiao Huang; Li Xu; Xianliang Hua; Xiaofen Li; Shouting Liu; Changshan Yang; Canguo Zhao; Chong Zhao; Shujue Li; Q. Ping Dou; Jinbao Liu

The proteasome inhibitor-based combinational therapy has been reported to be an efficient cancer treatment. Our recent studies demonstrated that the natural compound gambogic acid (GA) is a tissue-specific proteasome inhibitor, comparable to bortezomib (Bor), and sensitizes malignant cells to the proteasome inhibitor MG132/MG262 both in vitro and in vivo. The aim of this study was to further extend our investigation by combining GA with the clinically used proteasome inhibitor Bor to test their combined efficacy against human hepatoma HepG2 and mouse hepatoma H22 cells. GA and Bor synergistically induced cytotoxicity and cell death in human HepG2 and mouse H22 cells, and accelerated proteasome inhibition, endoplasmic reticulum (ER) stress and caspase activation in HepG2 cancer cells. However, unexpectedly, GA did not enhance or even antagonized Bor-induced tumor growth inhibition in H22 allograft and HepG2 xenograft tumor models. These findings demonstrated that GA increased Bor activity in vitro but limited the efficacy of Bor in vivo. We suggest that the combination of GA and Bor be avoided when administering these drugs to patients.


Scientific Reports | 2015

Gambogic acid induces apoptosis in diffuse large B-cell lymphoma cells via inducing proteasome inhibition

Xianping Shi; Xiaoying Lan; Xin Chen; Chong Zhao; Xiaofen Li; Shouting Liu; Hongbiao Huang; Ningning Liu; Dan Zang; Yuning Liao; Peiquan Zhang; Xuejun Wang; Jinbao Liu

Resistance to chemotherapy is a great challenge to improving the survival of patients with diffuse large B-cell lymphoma (DLBCL), especially those with activated B-cell-like DLBCL (ABC-DLBCL). Therefore it is urgent to search for novel agents for the treatment of DLBCL. Gambogic acid (GA), a small molecule derived from Chinese herb gamboges, has been approved for Phase II clinical trial for cancer therapy by Chinese FDA. In the present study, we investigated the effect of GA on cell survival and apoptosis in DLBCL cells including both GCB- and ABC-DLBCL cells. We found that GA induced growth inhibition and apoptosis of both GCB- and ABC-DLBCL cells in vitro and in vivo, which is associated with proteasome malfunction. These findings provide significant pre-clinical evidence for potential usage of GA in DLBCL therapy particularly in ABC-DLBCL treatment.


Oncotarget | 2017

Repurposing an antidandruff agent to treating cancer: zinc pyrithione inhibits tumor growth via targeting proteasome-associated deubiquitinases

Chong Zhao; Xin Chen; Changshan Yang; Dan Zang; Xiaoying Lan; Siyan Liao; Peiquan Zhang; Jinjie Wu; Xiaofen Li; Ningning Liu; Yuning Liao; Hongbiao Huang; Xianping Shi; Lili Jiang; Xiuhua Liu; Q. Ping Dou; Xuejun Wang; Jinbao Liu

The ubiquitin-proteasome system (UPS) plays a central role in various cellular processes through selectively degrading proteins involved in critical cellular functions. Targeting UPS has been validated as a novel strategy for treating human cancer, as inhibitors of the 20S proteasome catalytic activity are currently in clinical use for treatment of multiple myeloma and other cancers, and the deubiquitinase activity associated with the proteasome is also a valid target for anticancer agents. Recent studies suggested that zinc pyrithione, an FDA-approved antidandruff agent, may have antitumor activity, but the detailed molecular mechanisms remain unclear. Here we report that zinc pyrithione (ZnPT) targets the proteasome-associated DUBs (USP14 and UCHL5) and inhibits their activities, resulting in a rapid accumulation of protein-ubiquitin conjugates, but without inhibiting the proteolytic activities of 20S proteasomes. Furthermore, ZnPT exhibits cytotoxic effects against various cancer cell lines in vitro, selectively kills bone marrow cells from leukemia patients ex vivo, and efficiently inhibits the growth of lung adenocarcinoma cancer cell xenografts in nude mice. This study has identified zinc pyrithione, an FDA-approved pharmacological agent with potential antitumor properties as a proteasomal DUB inhibitor.


Cell Death and Disease | 2017

Platinum pyrithione induces apoptosis in chronic myeloid leukemia cells resistant to imatinib via DUB inhibition-dependent caspase activation and Bcr-Abl downregulation

Xiaoying Lan; Chong Zhao; Xin Chen; Peiquan Zhang; Dan Zang; Jinjie Wu; Jinghong Chen; Huidan Long; Li Yang; Hongbiao Huang; Xuejun Wang; Xianping Shi; Jinbao Liu

Chronic myelogenous leukemia (CML) is characterized by the chimeric tyrosine kinase Bcr-Abl. T315I Bcr-Abl is the most notorious point mutation to elicit acquired resistance to imatinib (IM), leading to poor prognosis. Therefore, it is urgent to search for additional approaches and targeting strategies to overcome IM resistance. We recently reported that platinum pyrithione (PtPT) potently inhibits the ubiquitin–proteasome system (UPS) via targeting the 26 S proteasome-associated deubiquitinases (DUBs), without effecting on the 20 S proteasome. Here we further report that (i) PtPT induces apoptosis in Bcr-Abl wild-type and Bcr-Abl-T315I mutation cells including the primary mononuclear cells from CML patients clinically resistant to IM, as well as inhibits the growth of IM-resistant Bcr-Abl-T315I xenografts in vivo; (ii) PtPT downregulates Bcr-Abl level through restraining Bcr-Abl transcription, and decreasing Bcr-Abl protein mediated by DUBs inhibition-induced caspase activation; (iii) UPS inhibition is required for PtPT-induced caspase activation and cell apoptosis. These findings support that PtPT overcomes IM resistance through both Bcr-Abl-dependent and -independent mechanisms. We conclude that PtPT can be a lead compound for further drug development to overcome imatinib resistance in CML patients.

Collaboration


Dive into the Chong Zhao's collaboration.

Top Co-Authors

Avatar

Jinbao Liu

Guangzhou Medical University

View shared research outputs
Top Co-Authors

Avatar

Hongbiao Huang

Guangzhou Medical University

View shared research outputs
Top Co-Authors

Avatar

Xuejun Wang

University of South Dakota

View shared research outputs
Top Co-Authors

Avatar

Ningning Liu

Guangzhou Medical University

View shared research outputs
Top Co-Authors

Avatar

Xiaofen Li

Guangzhou Medical University

View shared research outputs
Top Co-Authors

Avatar

Xiaoying Lan

Guangzhou Medical University

View shared research outputs
Top Co-Authors

Avatar

Xin Chen

Guangzhou Medical University

View shared research outputs
Top Co-Authors

Avatar

Xianping Shi

Guangzhou Medical University

View shared research outputs
Top Co-Authors

Avatar

Changshan Yang

Guangzhou Medical University

View shared research outputs
Top Co-Authors

Avatar

Shouting Liu

Guangzhou Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge