Network


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

Hotspot


Dive into the research topics where Changshan Yang is active.

Publication


Featured researches published by Changshan Yang.


Cell Research | 2010

Physiological levels of ATP Negatively Regulate Proteasome Function

Hongbiao Huang; Xiaoyan Zhang; Shujue Li; Ningning Liu; Wen Lian; Emily J. McDowell; Ping Zhou; Canguo Zhao; Haiping Guo; Change Zhang; Changshan Yang; Guangmei Wen; Xiaoxian Dong; Li Lu; Ningfang Ma; Weihua Dong; Q. Ping Dou; Xuejun Wang; Jinbao Liu

Intracellular protein degradation by the ubiquitin-proteasome system is ATP dependent, and the optimal ATP concentration to activate proteasome function in vitro is ∼100 μM. Intracellular ATP levels are generally in the low millimolar range, but ATP at a level within this range was shown to inhibit proteasome peptidase activities in vitro. Here, we report new evidence that supports a hypothesis that intracellular ATP at the physiological levels bidirectionally regulates 26S proteasome proteolytic function in the cell. First, we confirmed that ATP exerted bidirectional regulation on the 26S proteasome in vitro, with the optimal ATP concentration (between 50 and 100 μM) stimulating proteasome chymotrypsin-like activities. Second, we found that manipulating intracellular ATP levels also led to bidirectional changes in the levels of proteasome-specific protein substrates in cultured cells. Finally, measures to increase intracellular ATP enhanced, while decreasing intracellular ATP attenuated the ability of proteasome inhibition to induce cell death. These data strongly suggest that endogenous ATP within the physiological concentration range can exert a negative impact on proteasome activities, allowing the cell to rapidly upregulate proteasome activity on ATP reduction under stress conditions.


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.


PLOS ONE | 2012

L-Carnitine Is an Endogenous HDAC Inhibitor Selectively Inhibiting Cancer Cell Growth In Vivo and In Vitro

Hongbiao Huang; Ningning Liu; Haiping Guo; Siyan Liao; Xiaofen Li; Changshan Yang; Shouting Liu; Wenbin Song; Chunjiao Liu; Lixia Guan; Bing Li; Li Xu; Change Zhang; Xuejun Wang; Q. Ping Dou; Jinbao Liu

L-carnitine (LC) is generally believed to transport long-chain acyl groups from fatty acids into the mitochondrial matrix for ATP generation via the citric acid cycle. Based on Warburgs theory that most cancer cells mainly depend on glycolysis for ATP generation, we hypothesize that, LC treatment would lead to disturbance of cellular metabolism and cytotoxicity in cancer cells. In this study, Human hepatoma HepG2, SMMC-7721 cell lines, primary cultured thymocytes and mice bearing HepG2 tumor were used. ATP content was detected by HPLC assay. Cell cycle, cell death and cell viability were assayed by flow cytometry and MTS respectively. Gene, mRNA expression and protein level were detected by gene microarray, Real-time PCR and Western blot respectively. HDAC activities and histone acetylation were detected both in test tube and in cultured cells. A molecular docking study was carried out with CDOCKER protocol of Discovery Studio 2.0 to predict the molecular interaction between L-carnitine and HDAC. Here we found that (1) LC treatment selectively inhibited cancer cell growth in vivo and in vitro; (2) LC treatment selectively induces the expression of p21cip1 gene, mRNA and protein in cancer cells but not p27kip1; (4) LC increases histone acetylation and induces accumulation of acetylated histones both in normal thymocytes and cancer cells; (5) LC directly inhibits HDAC I/II activities via binding to the active sites of HDAC and induces histone acetylation and lysine-acetylation accumulation in vitro; (6) LC treatment induces accumulation of acetylated histones in chromatin associated with the p21cip1 gene but not p27kip1 detected by ChIP assay. These data support that LC, besides transporting acyl group, works as an endogenous HDAC inhibitor in the cell, which would be of physiological and pathological importance.


Cancer Letters | 2011

Gambogic acid enhances proteasome inhibitor-induced anticancer activity

Hongbiao Huang; Di Chen; Shujue Li; Xiaofen Li; Ningning Liu; Xiaoyu Lu; Shouting Liu; Kai Zhao; Canguo Zhao; Haiping Guo; Changshan Yang; Ping Zhou; Xiaoxian Dong; Change Zhang; Guanmei; Q. Ping Dou; Jinbao Liu

Proteasome inhibition has emerged as a novel approach to anticancer therapy. Numerous natural compounds, such as gambogic acid, have been tested in vitro and in vivo as anticancer agents for cancer prevention and therapy. However, whether gambogic acid has chemosensitizing properties when combined with proteasome inhibitors in the treatment of malignant cells is still unknown. In an effort to investigate this effect, human leukemia K562 cells, mouse hepatocarcinoma H22 cells and H22 cell allografts were treated with gambogic acid, a proteasome inhibitor (MG132 or MG262) or the combination of both, followed by measurement of cellular viability, apoptosis induction and tumor growth inhibition. We report, for the first time, that: (i) the combination of natural product gambogic acid and the proteasome inhibitor MG132 or MG262 results in a synergistic inhibitory effect on growth of malignant cells and tumors in allograft animal models and (ii) there was no apparent systemic toxicity observed in the animals treated with the combination. Therefore, the findings presented in this study demonstrate that natural product gambogic acid is a valuable candidate to be used in combination with proteasome inhibitors, thus representing a compelling anticancer strategy.


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.


PLOS ONE | 2012

HDAC inhibitor L-carnitine and proteasome inhibitor bortezomib synergistically exert anti-tumor activity in vitro and in vivo.

Hongbiao Huang; Ningning Liu; Changshan Yang; Siyan Liao; Haiping Guo; Kai Zhao; Xiaofen Li; Shouting Liu; Lixia Guan; Chunjiao Liu; Li Xu; Change Zhang; Wenbin Song; Bing Li; Ping Tang; Q. Ping Dou; Jinbao Liu

Combinations of proteasome inhibitors and histone deacetylases (HDAC) inhibitors appear to be the most potent to produce synergistic cytotoxicity in preclinical trials. We have recently confirmed that L-carnitine (LC) is an endogenous HDAC inhibitor. In the current study, the anti-tumor effect of LC plus proteasome inhibitor bortezomib (velcade, Vel) was investigated both in cultured hepatoma cancer cells and in Balb/c mice bearing HepG2 tumor. Cell death and cell viability were assayed by flow cytometry and MTS, respectively. Gene, mRNA expression and protein levels were detected by gene microarray, quantitative real-time PCR and Western blot, respectively. The effect of Vel on the acetylation of histone H3 associated with the p21cip1 gene promoter was examined by using ChIP assay and proteasome peptidase activity was detected by cell-based chymotrypsin-like (CT-like) activity assay. Here we report that (i) the combination of LC and Vel synergistically induces cytotoxicity in vitro; (ii) the combination also synergistically inhibits tumor growth in vivo; (iii) two major pathways are involved in the synergistical effects of the combinational treatment: increased p21cip1 expression and histone acetylation in vitro and in vivo and enhanced Vel-induced proteasome inhibition by LC. The synergistic effect of LC and Vel in cancer therapy should have great potential in the future clinical trials.


Oncogene | 2016

A novel nickel complex works as a proteasomal deubiquitinase inhibitor for cancer therapy

Canguo 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; Q P Dou; Xuejun Wang; Jinbao Liu

Based on the central role of the ubiquitin–proteasome system (UPS) in the degradation of cellular proteins, proteasome inhibition has been considered an attractive approach for anticancer therapy. Deubiquitinases (DUBs) remove ubiquitin conjugates from diverse substrates; therefore, they are essential regulators of the UPS. DUB inhibitors, especially the inhibitors of proteasomal DUBs are becoming a research hotspot in targeted cancer therapy. Previous studies have shown that metal complexes, such as copper and zinc complexes, can induce cancer cell apoptosis through inhibiting UPS function. Moreover, we have found that copper pyrithione inhibits both 19S proteasome-associated DUBs and 20S proteasome activity with a mechanism distinct from that of the classical 20S proteasome inhibitor bortezomib. In the present study, we reveal that (i) nickel pyrithione complex (NiPT) potently inhibits the UPS via targeting the 19S proteasome-associated DUBs (UCHL5 and USP14), without effecting on the 20S proteasome; (ii) NiPT selectively induces proteasome inhibition and apoptosis in cultured tumor cells and cancer cells from acute myeloid leukemia human patients; and (iii) NiPT inhibits proteasome function and tumor growth in nude mice. This study, for the first time, uncovers a nickel complex as an effective inhibitor of the 19S proteasomal DUBs and suggests a potentially new strategy for cancer treatment.


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.

Collaboration


Dive into the Changshan Yang's collaboration.

Top Co-Authors

Avatar

Hongbiao Huang

Guangzhou Medical University

View shared research outputs
Top Co-Authors

Avatar

Jinbao Liu

Guangzhou Medical University

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

Shouting Liu

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

Q. Ping Dou

Wayne State University

View shared research outputs
Top Co-Authors

Avatar

Canguo Zhao

Guangzhou Medical University

View shared research outputs
Top Co-Authors

Avatar

Chong Zhao

Guangzhou Medical University

View shared research outputs
Top Co-Authors

Avatar

Shujue Li

Guangzhou Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge