Network


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

Hotspot


Dive into the research topics where Benyi Li is active.

Publication


Featured researches published by Benyi Li.


Molecular Cancer Therapeutics | 2005

Small-interfering RNA-induced androgen receptor silencing leads to apoptotic cell death in prostate cancer

Xinbo Liao; Siqing Tang; J. Brantley Thrasher; Tomas L. Griebling; Benyi Li

Prostate cancer is the second leading cause of cancer death in the United States and, thus far, there has been no effective therapy for the treatment of hormone-refractory disease. Recently, the androgen receptor (AR) has been shown to play a critical role in the development and progression of the disease. In this report, we showed that knocking down the AR protein level by a small interfering RNA (siRNA) approach resulted in a significant apoptotic cell death as evidenced by an increased annexin V binding, reduced mitochondrial potential, caspase-3/6 activation, and DFF45 and poly(ADP-ribose) polymerase cleavage. The apoptotic response was specifically observed in those siRNA-transfected cells that harbor a native AR gene. No cell death was found in the AR-null prostate cancer cell PC-3 or its subline that has been reconstituted with an exogenous AR gene, as well as two breast cancer cell lines that are AR positive. Moreover, in parallel with the siRNA-induced AR silencing, the antiapoptotic protein Bcl-xL was significantly reduced, which might account for the apoptotic cell death because ectopic enforced expression of Bcl-xL protein partially inhibited apoptosis after AR silencing. Taken together, our data showed that knocking down the AR protein level in prostate cancer cells leads to apoptosis by disrupting the Bcl-xL–mediated survival signal downstream of AR-dependent survival pathway.


Molecular Cancer Therapeutics | 2005

Chemical ablation of androgen receptor in prostate cancer cells by the histone deacetylase inhibitor LAQ824

Liwei Chen; Songshu Meng; Hai Wang; Purva Bali; Wenlong Bai; Benyi Li; Peter Atadja; Kapil N. Bhalla; Jie Wu

Androgen receptor plays a critical role in the development of primary as well as advanced hormone-refractory prostate cancer. Therefore, ablation of androgen receptor from prostate cancer cells is an interesting concept for developing a new therapy not only for androgen-dependent prostate cancer but also for metastatic hormone-refractory prostate cancer, for which there is no effective treatment available. We report here that LAQ824, a cinnamyl hydroxamatic acid histone deacetylase inhibitor currently in human clinical trials, effectively depleted androgen receptor in prostate cancer cells at nanomolar concentrations. LAQ824 seemed capable of depleting both the mutant and wild-type androgen receptors in either androgen-dependent and androgen-independent prostate cancer cells. Although LAQ824 may exert its effect through multiple mechanisms, several lines of evidence suggest that inactivation of the heat shock protein-90 (Hsp90) molecular chaperone is involved in LAQ824-induced androgen receptor depletion. Besides androgen receptor, LAQ824 reduced the level of Hsp90 client proteins HER-2 (ErbB2), Akt/PKB, and Raf-1 in LNCaP cells. Another Hsp90 inhibitor, 17-allyamino-17-demethoxygeldanamycin (17-AAG), also induced androgen receptor diminution. LAQ824 induced Hsp90 acetylation in LNCaP cells, which resulted in inhibition of its ATP-binding activity, dissociation of Hsp90-androgen receptor complex, and proteasome-mediated degradation of androgen receptor. Consequently, LAQ824 blocked androgen-induced prostate-specific antigen production in LNCaP cells. LAQ824 effectively inhibited cell proliferation and induced apoptosis of these prostate cancer cells. These results reveal that LAQ824 is a potent agent for depletion of androgen receptor and a potential new drug for prostate cancer.


Journal of Cell Science | 2010

GSK-3β promotes cell survival by modulating Bif-1-dependent autophagy and cell death

Jun Yang; Yoshinori Takahashi; Erdong Cheng; Jihong Liu; Paul F. Terranova; Bin Zhao; J. Brantley Thrasher; Hong-Gang Wang; Benyi Li

Glycogen synthase kinase 3 beta (GSK-3β) is constantly active in cells and its activity increases after serum deprivation, indicating that GSK-3β might play a major role in cell survival under serum starvation. In this study, we attempted to determine how GSK-3β promotes cell survival after serum depletion. Under full culture conditions (10% FBS), GSK-3β inhibition with chemical inhibitors or siRNAs failed to induce cell death in human prostate cancer cells. By contrast, under conditions of serum starvation, a profound necrotic cell death was observed as evidenced by cellular morphologic features and biochemical markers. Further analysis revealed that GSK-3β-inhibition-induced cell death was in parallel with an extensive autophagic response. Interestingly, blocking the autophagic response switched GSK-3β-inhibition-induced necrosis to apoptotic cell death. Finally, GSK-3β inhibition resulted in a remarkable elevation of Bif-1 protein levels, and silencing Bif-1 expression abrogated GSK-3β-inhibition-induced autophagic response and cell death. Taken together, our study suggests that GSK-3β promotes cell survival by modulating Bif-1-dependent autophagic response and cell death.


The Prostate | 2011

Suppression of glycogen synthase kinase 3 activity reduces tumor growth of prostate cancer in vivo.

Qing Zhu; Jun Yang; Suxia Han; Jihong Liu; Jeffery Holzbeierlein; J. Brantley Thrasher; Benyi Li

Glycogen synthase kinase 3 (GSK‐3) has been regarded as a potential therapeutic target for multiple human cancers. We previously reported that suppression of GSK‐3 activity with lithium chloride (LiCl) or small chemical inhibitors impaired cellular DNA synthesis and reduced cell proliferation in prostate cancer cells. Therefore, in this study, we extended this in vitro findings to in vivo settings in order to establish a proof of concept that inhibition of GSK‐3 activity is feasible in suppressing tumor growth of prostate cancer in vivo.


Anti-Cancer Drugs | 2012

Pharmacological ascorbate induces cytotoxicity in prostate cancer cells through ATP depletion and induction of autophagy.

Ping Chen; Jun Yu; Brain Chalmers; Jeanne Drisko; Jun Yang; Benyi Li; Qi Chen

Recent studies have revealed the scientific basis for the use of intravenous (i.v.) vitamin C or ascorbic acid (ascorbate) in treating cancers, and raised the possibility of using i.v. ascorbate as a prooxidant anticancer therapy. Through the production of H2O2, pharmacologic ascorbate can induce some cancer cell death in vitro and inhibit a number of types of tumor growth in animal models. However, the mechanism of cell death triggered by ascorbate is not well understood. In this study, we investigated the cytotoxicity of pharmacological concentrations of ascorbate to human prostate cancer cells and the mechanisms involved. The results showed that ascorbate in the millimolar range induced cytotoxicity in five of the six tested prostate cancer cell lines. The IC50 values in the sensitive prostate cancer cells ranged from 1.9 to 3.5 mmol/l, concentrations clinically achievable with i.v. ascorbate use. All tested androgen-independent cells were sensitive to ascorbate treatment. The ascorbate-insensitive cell line LaPC4 is hormonally dependent. Whereas the reasons for sensitivity/resistance to ascorbate treatment need to be investigated further, cell death in sensitive cells was dependent on H2O2. Ascorbate treatment depleted ATP and induced autophagy in sensitive prostate cancer cells, resulting in cell death. Taken together with previous studies, high-dose ascorbate has the potential to be a novel treatment option to hormone-refractory prostate cancer.


Urologic Oncology-seminars and Original Investigations | 2014

Cav1.3 channel α1D protein is overexpressed and modulates androgen receptor transactivation in prostate cancers1

Ruibao Chen; Xing Zeng; Ruitao Zhang; Jiaoti Huang; Xiangxing Kuang; Jun Yang; Jihong Liu; Ossama Tawfik; James Brantley Thrasher; Benyi Li

Widespread use of L-type calcium channel blockers for treating hypertension has led to multiple epidemiologic studies to assess the risk of prostate cancer incidence. These studies revealed a reverse correlation between the likelihood of prostate cancer risk and the use of L-type calcium channel blockers among men without family history but the mechanism was not clear. In this study, we examined the expression profiles of multiple L-type calcium channel genes in prostate cancers and determined their functional roles in androgen receptor (AR) transactivation and cell growth. By reanalyzing the ONCOMINE database, we found that L-type calcium channel CACNA1D gene expression levels in cancer tissues were significantly higher than noncancer tissues in 14 of 15 published complementary deoxyribonucleic acid microarray data sets, of which 9 data sets showed an increase of 2- to 17-folds. Quantitative polymerase chain reaction and immunostaining experiments revealed that CACNA1D gene and its coding protein α1D were highly expressed in prostate cancers, especially in castration-resistant diseases, compared with benign prostate tissues. Consistent with the notion of CACNA1D as an ERG-regulated gene, CACNA1D gene expression levels were significantly higher in prostate cancers with TMPRSS2-ERG gene fusion compared with the cases without this gene fusion. Blocking L-type channels function or knocking down CACNA1D gene expression significantly suppressed androgen-stimulated Ca(2+) influx, AR transactivation, and cell growth in prostate cancer cells. Taken together, these data suggest that CACNA1D gene overexpression is associated with prostate cancer progression and might play an important role in Ca(2+) influx, AR activation, and cell growth in prostate cancer cells.


Autophagy | 2014

Autophagy activation reduces renal tubular injury induced by urinary proteins.

Wei Jing Liu; Mian-Na Luo; Jin Tan; Wei Chen; Lei-zhao Huang; Chen Yang; Qingjun Pan; Benyi Li; Hua-feng Liu

Autophagy is shown to be beneficial for renal tubular injury caused by nephrotoxic drugs. To investigate whether autophagy could protect renal tubular epithelial cells (TECs) from injury induced by urinary proteins, we studied the activity and action of autophagy in TECs after urinary protein overload in vivo and in vitro. We found that autophagic vacuoles increased in TECs from patients with minimal change nephrotic syndrome (MCNS) and rat models with severe proteinuria induced by cationic BSA. In HK-2 cells, exposure to urinary proteins extracted from patients with MCNS led to a significant increase in autophagosome and autolysosome formation and decrease in SQSTM1/p62 protein level. Urinary protein addition also induced lysosomal turnover of LC3-II and perinuclear clustering of lysosomes. These changes were mediated by a reactive oxygen species (ROS)-dependent mechanism. Furthermore, pretreatment of HK-2 cells with rapamycin reduced the production of LCN2/NGAL and HAVCR1/KIM-1 and the level of apoptosis induced by urinary proteins. In contrast, blocking autophagy with chloroquine or BECN1 siRNAs exerted an opposite effect. Similar results were also observed in animal models with proteinuria after treatments with rapamycin and chloroquine. Taken together, our results indicated an increase in autophagic flux, which mounts an adaptive response in TECs after urinary protein overload.


Molecular Pharmaceutics | 2012

Prodrug strategy for PSMA-targeted delivery of TGX-221 to prostate cancer cells.

Yunqi Zhao; Shaofeng Duan; Xing Zeng; Chunjing Liu; Neal M. Davies; Benyi Li; M. Laird Forrest

TGX-221 is a potent, selective, and cell membrane permeable inhibitor of the PI3K p110β catalytic subunit. Recent studies showed that TGX-221 has antiproliferative activity against PTEN-deficient tumor cell lines including prostate cancers. The objective of this study was to develop an encapsulation system for parenterally delivering TGX-221 to the target tissue through a prostate-specific membrane aptamer (PSMAa10) with little or no side effects. In this study, PEG-PCL micelles were formulated to encapsulate the drug, and a prodrug strategy was pursued to improve the stability of the carrier system. Fluorescence imaging studies demonstrated that the cellular uptake of both drug and nanoparticles was significantly improved by targeted micelles in a PSMA positive cell line. The area under the plasma concentration time curve of the micelle formulation in nude mice was 2.27-fold greater than that of the naked drug, and the drug clearance rate was 6.16-fold slower. These findings suggest a novel formulation approach for improving site-specific drug delivery of a molecular-targeted prostate cancer treatment.


The Prostate | 2008

Androgen receptor-dependent regulation of Bcl-xL expression: Implication in prostate cancer progression.

Aijing Sun; Jianxi Tang; Yan Hong; Jiawu Song; Paul F. Terranova; J. Brantley Thrasher; Stan Svojanovsky; Hong-Gang Wang; Benyi Li

Recently we reported that silencing the androgen receptor (AR) gene reduced Bcl‐xL expression that was associated with a profound apoptotic cell death in prostate cancer cells. In this study we further investigated AR‐regulated Bcl‐xL expression.


The Prostate | 2016

GSK-3β controls autophagy by modulating LKB1-AMPK pathway in prostate cancer cells.

Aijing Sun; Li C; Ruibao Chen; Yiling Huang; Chen Q; Cui X; Hua-feng Liu; Thrasher Jb; Benyi Li

Glycogen synthase kinase 3β (GSK3B, GSK‐3β) is a multi‐functional protein kinase involved in various cellular processes and its activity elevates after serum deprivation. We have shown that inhibition of GSK‐3β activity triggered a profound autophagic response and subsequent necrotic cell death after serum deprivation in prostate cancer cells. In this study, we dissected the mechanisms involved in GSK‐3β inhibition‐triggered autophagy.

Collaboration


Dive into the Benyi Li's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jun Yang

University of Kansas

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jihong Liu

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge