Network


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

Hotspot


Dive into the research topics where Shengyan Xiang is active.

Publication


Featured researches published by Shengyan Xiang.


Oncogene | 2011

Inhibition of androgen receptor activity by histone deacetylase 4 through receptor SUMOylation.

Yonghua Yang; Anfernee Kai-Wing Tse; Pengfei Li; Qiuping Ma; Shengyan Xiang; Santo V. Nicosia; Edward Seto; Xiaohong Zhang; Wenlong Bai

The transcriptional activity of the androgen receptor (AR) is regulated by both ligand binding and post-translational modifications, including acetylation and small ubiquitin-like modifier (SUMO)ylation. Histone deacetylases (HDACs) are known to catalyze the removal of acetyl groups from both histones and non-histone proteins. In this study, we report that HDAC4 binds to and inhibits the activity of the AR. This inhibition was found to depend on the SUMOylation, instead of deacetylation, of the AR. Consistently, HDAC4 increases the level of AR SUMOylation in both whole-cell and cell-free assay systems, raising the possibility that the deacetylase may act as an E3 ligase for AR SUMOylation. Knock down of HDAC4 increases the activity of endogenous AR and androgen induction of prostate-specific antigen expression and prostate cancer cell growth, which is associated with decreased SUMOylation of the receptor. Overall, the studies identify HDAC4 as a positive regulator for AR SUMOylation, revealing a deacetylase-independent mechanism of HDAC action in prostate cancer cells.


Journal of Biological Chemistry | 2013

Extracellular Signal-regulated Kinase (ERK) Phosphorylates Histone Deacetylase 6 (HDAC6) at Serine 1035 to Stimulate Cell Migration

Kendra A. Williams; Mu Zhang; Shengyan Xiang; Chen Hu; Jheng Yu Wu; Shengping Zhang; Meagan Ryan; Adrienne D. Cox; Channing J. Der; Bin Fang; John M. Koomen; Eric B. Haura; Gerold Bepler; Santo V. Nicosia; Patrick Matthias; Chuangui Wang; Wenlong Bai; Xiaohong Zhang

Background: HDAC6 plays an important role in cell migration. Results: ERK interacts with and phosphorylates HDAC6 to promote cell migration. Conclusion: ERK signaling pathway promotes cell migration, in part, through phosphorylating HDAC6. Significance: Inhibition of HDAC6 activity as well as the EGFR-Ras-Raf-MEK-ERK signaling pathway may cooperatively reduce cell migration. Histone deacetylase 6 (HDAC6) is well known for its ability to promote cell migration through deacetylation of its cytoplasmic substrates such as α-tubulin. However, how HDAC6 itself is regulated to control cell motility remains elusive. Previous studies have shown that one third of extracellular signal-regulated kinase (ERK) is associated with the microtubule cytoskeleton in cells. Yet, no connection between HDAC6 and ERK has been discovered. Here, for the first time, we reveal that ERK binds to and phosphorylates HDAC6 to promote cell migration via deacetylation of α-tubulin. We have identified two novel ERK-mediated phosphorylation sites: threonine 1031 and serine 1035 in HDAC6. Both sites were phosphorylated by ERK1 in vitro, whereas Ser-1035 was phosphorylated in response to the activation of EGFR-Ras-Raf-MEK-ERK signaling pathway in vivo. HDAC6-null mouse embryonic fibroblasts rescued by the nonphosphorylation mimicking mutant displayed significantly reduced cell migration compared with those rescued by the wild type. Consistently, the nonphosphorylation mimicking mutant exerted lower tubulin deacetylase activity in vivo compared with the wild type. These data indicate that ERK/HDAC6-mediated cell motility is through deacetylation of α-tubulin. Overall, our results suggest that HDAC6-mediated cell migration could be governed by EGFR-Ras-Raf-MEK-ERK signaling.


Molecular Cell | 2014

HDAC6 Deacetylates and Ubiquitinates MSH2 to Maintain Proper Levels of MutSα

Mu Zhang; Shengyan Xiang; Heui Yun Joo; Lei Wang; Kendra A. Williams; Wei Liu; Chen Hu; Dan Tong; Joshua Haakenson; Chuangui Wang; Shengping Zhang; Ryan E. Pavlovicz; Amanda Jones; K.-H. Schmidt; Jinfu Tang; Huiqin Dong; Bin Shan; Bin Fang; Rangasudhagar Radhakrishnan; Peter M. Glazer; Patrick Matthias; John M. Koomen; Edward Seto; Gerold Bepler; Santo V. Nicosia; Jiandong Chen; Chenglong Li; Liya Gu; Guo Min Li; Wenlong Bai

MutS protein homolog 2 (MSH2) is a key DNA mismatch repair protein. It forms the MSH2-MSH6 (MutSα) and MSH2-MSH3 (MutSβ) heterodimers, which help to ensure genomic integrity. MutSα not only recognizes and repairs mismatched nucleotides but also recognizes DNA adducts induced by DNA-damaging agents, and triggers cell-cycle arrest and apoptosis. Loss or depletion of MutSα from cells leads to microsatellite instability (MSI) and resistance to DNA damage. Although the level of MutSα can be reduced by the ubiquitin-proteasome pathway, the detailed mechanisms of this regulation remain elusive. Here we report that histone deacetylase 6 (HDAC6) sequentially deacetylates and ubiquitinates MSH2, leading to MSH2 degradation. In addition, HDAC6 significantly reduces cellular sensitivity to DNA-damaging agents and decreases cellular DNA mismatch repair activities by downregulation of MSH2. Overall, these findings reveal a mechanism by which proper levels of MutSα are maintained.


PLOS ONE | 2012

Depletion of HDAC6 Enhances Cisplatin-Induced DNA Damage and Apoptosis in Non-Small Cell Lung Cancer Cells

Lei Wang; Shengyan Xiang; Kendra A. Williams; Huiqin Dong; Wenlong Bai; Santo V. Nicosia; Saadi Khochbin; Gerold Bepler; Xiaohong Zhang

Histone deacetylase inhibitors (HDACi) are promising therapeutic agents which are currently used in combination with chemotherapeutic agents in clinical trials for cancer treatment including non-small cell lung cancer (NSCLC). However, the mechanisms underlying their anti-tumor activities remain elusive. Previous studies showed that inhibition of HDAC6 induces DNA damage and sensitizes transformed cells to anti-tumor agents such as etoposide and doxorubicin. Here, we showed that depletion of HDAC6 in two NSCLC cell lines, H292 and A549, sensitized cells to cisplatin, one of the first-line chemotherapeutic agents used to treat NSCLC. We suggested that depletion of HDAC6 increased cisplatin-induced cytotoxicity was due to the enhancement of apoptosis via activating ATR/Chk1 pathway. Furthermore, we showed that HDAC6 protein levels were positively correlated with cisplatin IC50 in 15 NSCLC cell lines. Lastly, depletion of HDAC6 in H292 xenografts rendered decreased tumor weight and volume and exhibited increased basal apoptosis compared with the controls in a xenograft mouse model. In summary, our findings suggest that HDAC6 is positively associated with cisplatin resistance in NSCLC and reveal HDAC6 as a potential novel therapeutic target for platinum refractory NSCLC.


Clinical Cancer Research | 2016

Binding of released Bim to Mcl-1 is a mechanism of intrinsic resistance to ABT-199 which can be overcome by combination with daunorubicin or cytarabine in AML cells

Xiaojia Niu; Jianyun Zhao; Jun Ma; Chengzhi Xie; Holly Edwards; Guan Wang; J. Timothy Caldwell; Shengyan Xiang; Xiaohong Zhang; Roland Chu; Zhihong J. Wang; Hai Lin; Jeffrey W. Taub; Yubin Ge

Purpose: To investigate the molecular mechanism underlying intrinsic resistance to ABT-199. Experimental Design: Western blots and real-time RT-PCR were used to determine levels of Mcl-1 after ABT-199 treatment alone or in combination with cytarabine or daunorubicin. Immunoprecipitation of Bim and Mcl-1 were used to determine the effect of ABT-199 treatment on their interactions with Bcl-2 family members. Lentiviral short hairpin RNA knockdown of Bim and CRISPR knockdown of Mcl-1 were used to confirm their role in resistance to ABT-199. JC-1 assays and flow cytometry were used to determine drug-induced apoptosis. Results: Immunoprecipitation of Bim from ABT-199–treated cell lines and a primary patient sample demonstrated decreased association with Bcl-2, but increased association with Mcl-1 without corresponding change in mitochondrial outer membrane potential. ABT-199 treatment resulted in increased levels of Mcl-1 protein, unchanged or decreased Mcl-1 transcript levels, and increased Mcl-1 protein half-life, suggesting that the association with Bim plays a role in stabilizing Mcl-1 protein. Combining conventional chemotherapeutic agent cytarabine or daunorubicin with ABT-199 resulted in increased DNA damage along with decreased Mcl-1 protein levels, compared with ABT-199 alone, and synergistic induction of cell death in both AML cell lines and primary patient samples obtained from AML patients at diagnosis. Conclusions: Our results demonstrate that sequestration of Bim by Mcl-1 is a mechanism of intrinsic ABT-199 resistance and supports the clinical development of ABT-199 in combination with cytarabine or daunorubicin for the treatment of AML. Clin Cancer Res; 22(17); 4440–51. ©2016 AACR.


Journal of Biological Chemistry | 2016

Ubiquitin-Specific Peptidase 10 (USP10) Deubiquitinates and Stabilizes MutS Homolog 2 (MSH2) to Regulate Cellular Sensitivity to DNA Damage

Mu Zhang; Chen Hu; Dan Tong; Shengyan Xiang; Kendra A. Williams; Wenlong Bai; Guo Min Li; Gerold Bepler; Xiaohong Zhang

MSH2 is a key DNA mismatch repair protein, which plays an important role in genomic stability. In addition to its DNA repair function, MSH2 serves as a sensor for DNA base analogs-provoked DNA replication errors and binds to various DNA damage-induced adducts to trigger cell cycle arrest or apoptosis. Loss or depletion of MSH2 from cells renders resistance to certain DNA-damaging agents. Therefore, the level of MSH2 determines DNA damage response. Previous studies showed that the level of MSH2 protein is modulated by the ubiquitin-proteasome pathway, and histone deacetylase 6 (HDAC6) serves as an ubiquitin E3 ligase. However, the deubiquitinating enzymes, which regulate MSH2 remain unknown. Here we report that ubiquitin-specific peptidase 10 (USP10) interacts with and stabilizes MSH2. USP10 deubiquitinates MSH2 in vitro and in vivo. Moreover, the protein level of MSH2 is positively correlated with the USP10 protein level in a panel of lung cancer cell lines. Knockdown of USP10 in lung cancer cells exhibits increased cell survival and decreased apoptosis upon the treatment of DNA-methylating agent N-methyl-N′-nitro-N-nitrosoguanidine (MNNG) and antimetabolite 6-thioguanine (6-TG). The above phenotypes can be rescued by ectopic expression of MSH2. In addition, knockdown of MSH2 decreases the cellular mismatch repair activity. Overall, our results suggest a novel USP10-MSH2 pathway regulating DNA damage response and DNA mismatch repair.


Journal of Biological Chemistry | 2015

Histone Deacetylase 10 Regulates DNA Mismatch Repair and May Involve the Deacetylation of MutS Homolog 2

Rangasudhagar Radhakrishnan; Yixuan Li; Shengyan Xiang; Fenghua Yuan; Zhigang Yuan; Elphine Telles; Jia Fang; Domenico Coppola; David Shibata; William S. Lane; Yanbin Zhang; Xiaohong Zhang; Edward Seto

Background: The DNA mismatch repair protein MutS homolog 2 (MSH2) is an acetylated protein. Results: Histone deacetylase 10 (HDAC10) interacts with MSH2 and deacetylates MSH2 at Lys-73, which might stimulate MSH2 activity. Conclusion: HDAC10 promotes DNA mismatch repair activity and may involve the deacetylation of MSH2. Significance: HDAC10 has an important role in regulating DNA mismatch repair. MutS homolog 2 (MSH2) is an essential DNA mismatch repair (MMR) protein. It interacts with MSH6 or MSH3 to form the MutSα or MutSβ complex, respectively, which recognize base-base mispairs and insertions/deletions and initiate the repair process. Mutation or dysregulation of MSH2 causes genomic instability that can lead to cancer. MSH2 is acetylated at its C terminus, and histone deacetylase (HDAC6) deacetylates MSH2. However, whether other regions of MSH2 can be acetylated and whether other histone deacetylases (HDACs) and histone acetyltransferases (HATs) are involved in MSH2 deacetylation/acetylation is unknown. Here, we report that MSH2 can be acetylated at Lys-73 near the N terminus. Lys-73 is highly conserved across many species. Although several Class I and II HDACs interact with MSH2, HDAC10 is the major enzyme that deacetylates MSH2 at Lys-73. Histone acetyltransferase HBO1 might acetylate this residue. HDAC10 overexpression in HeLa cells stimulates cellular DNA MMR activity, whereas HDAC10 knockdown decreases DNA MMR activity. Thus, our study identifies an HDAC10-mediated regulatory mechanism controlling the DNA mismatch repair function of MSH2.


Conquering RAS#R##N#From Biology to Cancer Therapy | 2017

Activation of Ras by Post-Translational Modifications

Shengyan Xiang; Wenlong Bai; G. Bepler; X. Zhang

Ras proteins are molecular switches cycling between the GTP-bound state and the GDP-bound state to transduce signals from outside of the cell to the interior. They all have the CAAX motifs at the C-terminus. Post-translational modifications of the CAAX motifs determine the subcellular localization and correct biological function of Ras proteins. Here we have described how prenylation of the CAAX motifs affects membrane association of Ras and how this modification can be targeted to block Ras-transduced signaling. In addition, other post-translational modifications of Ras, such as palmitoylation, phosphorylation, acetylation, ubiquitination, and S-nitrosylation, are discussed. Because oncogenic Ras mutants are drivers for tumorigenesis, understanding how post-translational modifications of Ras mutants influence their activities will help us design therapeutic drugs to treat patients with cancer.


PLOS ONE | 2015

Murine Pancreatic Adenocarcinoma Reduces Ikaros Expression and Disrupts T Cell Homeostasis

Nadine Nelson; Shengyan Xiang; Xiaohong Zhang; Danielle L. Gilvary; Julie Y. Djeu; Kazim Husain; Mokenge P. Malafa; Nasreen A. Vohra; Shari Pilon-Thomas; Tomar Ghansah

Background Maintenance of T cell immune homeostasis is critical for adequate anti-tumor immunity. The transcription factor Ikaros is essential for lymphocyte development including T cells. Alterations in Ikaros expression occur in blood malignancies in humans and mice. In this study, we investigated the role of Ikaros in regulating T cell immune balance in pancreatic cancer mouse models. Methodology and Principal Findings Using our Panc02 tumor-bearing (TB) mouse model, western blot analysis revealed a reduction in Ikaros proteins while qRT-PCR showed no differences in Ikaros mRNA levels in TB splenocytes compared to control. Treatment of naïve splenocytes with the proteasomal inhibitor, MG132, stabilized Ikaros expression and prevented Ikaros downregulation by Panc02 cells, in vitro. Western blot analyses showed a reduction in protein phosphatase 1 (PP1) and protein kinase CK2 expression in TB splenocytes while CK2 activity was increased. Immunofluorescence microscopy revealed altered punctate staining of Ikaros in TB splenocytes. Flow cytometry revealed a significant decrease in effector CD4+ and CD8+ T cell percentages but increased CD4+CD25+ regulatory T cells in TB splenocytes. Similar alterations in T cell percentages, as well as reduced Ikaros and CK2 but not PP1 expression, were observed in a transgenic, triple mutant (TrM) pancreatic cancer model. Ikaros expression was also reduced in enriched TB CD3+ T cells. MG132 treatment of naïve CD3+ T cells stabilized Ikaros expression in the presence of Panc02 cells. Western blots showed reduced PP1 and CK2 expression in TB CD3+ T cells. Conclusions/Significance The results of this study suggest that the pancreatic tumor microenvironment may cause proteasomal degradation of Ikaros, possibly via dysregulation of PP1 and CK2 expression and activity, respectively. This loss of Ikaros expression may contribute to an imbalance in T cell percentages. Ikaros may potentially be a therapeutic target to restore T cell homeostasis in pancreatic cancer hosts, which may be critical for effective anti-tumor immunity.


Cancer immunology research | 2015

Abstract A83: The role of Ikaros in effector and regulatory T cell homeostasis in a murine pancreatic cancer model

Nadine Nelson; Shengyan Xiang; Xiaohong Zhang; Karoly Szekeres; Ghansah Tomar

The alternatively spliced transcription factor Ikaros is crucial for normal lymphocyte development. Ikaros plays a role as a tumor suppressor in murine T cells. In pancreatic cancer hosts, the balance between effector and regulatory T cells is lost, leading to immunosuppression and reduced anti-tumor immunity. In this study, we aim to identify the role of Ikaros in regulating effector CD4+ and CD8+ and regulatory T cell homeostasis in a pancreatic tumor microenvironment. Using our murine heterotopic model of pancreatic cancer, we isolated splenocytes from tumor-bearing (TB) and control mice and performed western blot and qRT-PCR analyses to evaluate Ikaros mRNA and protein expression, respectively. Flow cytometry analyses were also used to immunophenotype T cell populations in splenocytes from control and TB mice. Next, magnetic activated cell sorting (MACS) was used to sort CD3+ T cells from TB and control splenocytes for western blot analyses of Ikaros expression. We also carried out in vitro and in vivo experiments using apigenin, a plant flavonoid and protein kinase inhibitor with anti-inflammatory, anti-tumor and anti-proliferative properties, to shed insight into how Ikaros may be regulated and its involvement in T cell development. Our results showed no difference in Ikaros mRNA expression but reduced expression of all Ikaros isoforms in splenocytes from TB mice compared with controls. Ikaros protein expression was also downregulated in sorted TB T cells, compared with control. Analyses of T cells by flow cytometry revealed a significant decrease in effector CD4+ and CD8+ T cell percentages but an increase in CD4+CD25+ regulatory T cells (Treg) in TB splenocytes compared with controls. In vitro, treatment of splenocytes with apigenin stabilized Ikaros9 protein expression. In the presence of Panc02 cells, Ikaros protein expression was downregulated. However, the addition of apigenin to the co-culture blocked this downregulation. These correlated with in vivo findings that showed partial reversal of some Ikaros isoforms in TB mice treated with apigenin. Apigenin treatment also significantly increased CD4+ and CD8+ T cell percentages but decreased Treg percentages. We are currently performing experiments to determine how Ikaros may be regulated in T cells and its effect on their functions. Thus far, our findings suggest that PC progression is associated with reduced Ikaros expression, which may lead to loss of T cell homeostasis and thus, dampened anti-tumor immune responses. Citation Format: Nadine Nelson, Shengyan Xiang, Xiaohong Zhang, Karoly Szekeres, Ghansah Tomar. The role of Ikaros in effector and regulatory T cell homeostasis in a murine pancreatic cancer model. [abstract]. In: Proceedings of the AACR Special Conference: Tumor Immunology and Immunotherapy: A New Chapter; December 1-4, 2014; Orlando, FL. Philadelphia (PA): AACR; Cancer Immunol Res 2015;3(10 Suppl):Abstract nr A83.

Collaboration


Dive into the Shengyan Xiang's collaboration.

Top Co-Authors

Avatar

Xiaohong Zhang

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Wenlong Bai

University of South Florida

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kendra A. Williams

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Mu Zhang

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Santo V. Nicosia

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Bin Fang

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Edward Seto

George Washington University

View shared research outputs
Top Co-Authors

Avatar

Huiqin Dong

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Nadine Nelson

University of South Florida

View shared research outputs
Researchain Logo
Decentralizing Knowledge