Network


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

Hotspot


Dive into the research topics where Chunhong Yan is active.

Publication


Featured researches published by Chunhong Yan.


Biochimica et Biophysica Acta | 2013

Drugging the Undruggable: Transcription Therapy for Cancer

Chunhong Yan; Paul J. Higgins

Transcriptional regulation is often the convergence point of oncogenic signaling. It is not surprising, therefore, that aberrant gene expression is a hallmark of cancer. Transformed cells often develop a dependency on such a reprogramming highlighting the therapeutic potential of rectifying cancer-associated transcriptional abnormalities in malignant cells. Although transcription is traditionally considered as undruggable, agents have been developed that target various levels of transcriptional regulation including DNA binding by transcription factors, protein-protein interactions, and epigenetic alterations. Some of these agents have been approved for clinical use or entered clinical trials. While artificial transcription factors have been developed that can theoretically modulate expression of any given gene, the emergence of reliable reporter assays greatly facilitates the search for transcription-targeted agents. This review provides a comprehensive overview of these developments, and discusses various strategies applicable for developing transcription-targeted therapeutic agents.


Molecular Cancer Therapeutics | 2011

A Small-Molecule Inhibitor of MDMX Activates p53 and Induces Apoptosis

Hongbo Wang; Xujun Ma; Shumei Ren; John K. Buolamwini; Chunhong Yan

The p53 inactivation caused by aberrant expression of its major regulators (e.g., MDM2 and MDMX) contributes to the genesis of a large number of human cancers. Recent studies have shown that restoration of p53 activity by counteracting p53 repressors is a promising anticancer strategy. Although agents (e.g., nutlin-3a) that disrupt MDM2–p53 interaction can inhibit tumor growth, they are less effective in cancer cells that express high levels of MDMX. MDMX binds to p53 and can repress the tumor suppressor function of p53 through inhibiting its trans-activation activity and/or destabilizing the protein. Here we report the identification of a benzofuroxan derivative [7-(4-methylpiperazin-1-yl)-4-nitro-1-oxido-2,1,3-benzoxadiazol-1-ium, NSC207895] that could inhibit MDMX expression in cancer cells through a reporter-based drug screening. Treatments of MCF-7 cells with this small-molecule MDMX inhibitor activated p53, resulting in elevated expression of proapoptotic genes (e.g., PUMA, BAX, and PIG3). Importantly, this novel small-molecule p53 activator caused MCF-7 cells to undergo apoptosis and acted additively with nutlin-3a to activate p53 and decrease the viability of cancer cells. These results thus show that small molecules targeting MDMX expression would be of therapeutic benefits. Mol Cancer Ther; 10(1); 69–79. ©2010 AACR.


The FASEB Journal | 2004

A novel homologous recombination system to study 92 kDa type IV collagenase transcription demonstrates that the NF-kappaB motif drives the transition from a repressed to an activated state of gene expression.

Chunhong Yan; Heng Wang; Bharat B. Aggarwal; Douglas D. Boyd

The 92‐kDa type IV collagenase (MMP‐9) contributes to tissue remodeling in both physiology and pathology. Previous studies on the transcriptional regulation of this gene have used transiently transfected promoter–reporter constructs. However, this approach suffers from several limitations including (a) multiple copies of the plasmid and (b) the plasmid is not genomically integrated and consequently poorly chromatinized. We developed a novel system for studying MMP‐9 transcription in which a single copy of a MMP‐9 promoter–luciferase construct(s) is integrated at an identical genomic locus in HT1080 cells by homologous recombination. We report that the activity of a genomic‐integrated 2.2 kb MMP‐9 promoter sequence mirrors expression of the endogenous MMP‐9 gene in response to both physiological and pharmacological (curcumin) cues. Further, when constrained into chromatin, the integrated NF‐κB‐mutated MMP‐9 promoter is repressed by PMA, a situation not apparent using nonintegrated plasmids. Thus, we have developed a novel method for studying MMP‐9 expression that overcomes some of the limitations associated with transient transfection approaches and which may be of utility in screening for agents that repress the expression of this gene.


Journal of Biological Chemistry | 2010

Activating Transcription Factor 3 Activates p53 by Preventing E6-associated Protein from Binding to E6

Hongbo Wang; Pingli Mo; Shumei Ren; Chunhong Yan

Genomic integration of human papillomavirus (HPV) DNA accounts for more than 90% of cervical cancers. High-risk genital HPVs encode E6 proteins that can interact with a cellular ubiquitin ligase E6-associated protein (E6AP) and target the tumor suppressor p53 for ubiquitin-mediated proteolysis. Currently, how this critical event is regulated is largely unknown. Here we report that activating transcription factor 3 (ATF3), a broad DNA damage sensor whose expression is frequently downregulated in cervical cancer, interacted with E6 and prevented p53 from ubiquitination and degradation mediated by the viral protein. Consistent with its role as a potent E6 antagonist, ATF3 expressed enforcedly in HPV-positive SiHa cells activated p53, leading to expression of p53-target genes (e.g. p21 and PUMA), cell cycle arrest and apoptotic cell death. The leucine zipper domain of ATF3 appears indispensable for these effects as an ATF3 mutant lacking this domain failed to interact with E6 and activate p53 in the cervical cancer cells. The prevention of p53 degradation was unlikely caused by binding of ATF3 to the tumor suppressor, but rather was a consequence of disruption of the E6-E6AP interaction by ATF3. These results indicate that ATF3 plays a key role in a mechanism defending against HPV-induced carcinogenesis, and could serve as a novel therapeutic target for HPV-positive cancers.


Journal of Biological Chemistry | 2010

MDM2 mediates ubiquitination and degradation of activating transcription factor 3.

Pingli Mo; Hongbo Wang; Hua Lu; Douglas D. Boyd; Chunhong Yan

Activating transcription factor 3 (ATF3) is a common stress sensor, and its rapid induction by cellular stresses (e.g. DNA damage) is crucial for cells to mount appropriate responses (e.g. activating the tumor suppressor p53) and maintain homeostasis. Although emerging evidence suggests that dysregulation of ATF3 contributes to occurrences of human diseases including cancer, the mechanism(s) by which ATF3 expression is regulated is largely unknown. Here, we demonstrate that mouse double minute 2 (MDM2) is a bona fide E3 ubiquitin ligase for ATF3 and regulates ATF3 expression by promoting its degradation. MDM2 via its C-terminal RING finger can bind to the Basic region of ATF3 and mediate the addition of ubiquitin moieties to the ATF3 leucine zipper domain. As a consequence, ATF3, but not a mutant deficient in MDM2 binding (Δ80–100), is degraded by MDM2-mediated proteolysis. Consistent with these results, ablation of MDM2 in cells not only increases basal ATF3 levels, but results in stabilization of ATF3 in late stages of DNA damage responses. Because ATF3 was recently identified as a p53 activator, these results suggest that MDM2 could inactivate p53 through an additional feedback mechanism involving ATF3. Therefore, we provide the first evidence demonstrating that ATF3 is regulated by a posttranslational mechanism.


Molecular and Cellular Biology | 2012

The Stress Response Mediator ATF3 Represses Androgen Signaling by Binding the Androgen Receptor

Hongbo Wang; Ming Jiang; Hongmei Cui; Mengqian Chen; Ralph Buttyan; Simon W. Hayward; Tsonwin Hai; Zhengxin Wang; Chunhong Yan

ABSTRACT Activating transcription factor 3 (ATF3) is a common mediator of cellular stress response signaling and is often aberrantly expressed in prostate cancer. We report here that ATF3 can directly bind the androgen receptor (AR) and consequently repress AR-mediated gene expression. The ATF3-AR interaction requires the leucine zipper domain of ATF3 that independently binds the DNA-binding and ligand-binding domains of AR, and the interaction prevents AR from binding to cis-acting elements required for expression of androgen-dependent genes while inhibiting the AR N- and C-terminal interaction. The functional consequences of the loss of ATF3 expression include increased transcription of androgen-dependent genes in prostate cancer cells that correlates with increased ability to grow in low-androgen-containing medium and increased proliferative activity of the prostate epithelium in ATF3 knockout mice that is associated with prostatic hyperplasia. Our results thus demonstrate that ATF3 is a novel repressor of androgen signaling that can inhibit AR functions, allowing prostate cells to restore homeostasis and maintain integrity in the face of a broad spectrum of intrinsic and environmental insults.


Journal of Biological Chemistry | 2014

The Activating Transcription Factor 3 Protein Suppresses the Oncogenic Function of Mutant p53 Proteins

Saisai Wei; Hongbo Wang; Chunwan Lu; Sarah Malmut; Jianqiao Zhang; Shumei Ren; Guohua Yu; Wei Wang; Dale D. Tang; Chunhong Yan

Background: Mutant p53 often acquires tumor-promoting activities, including the promotion of cancer cell survival, invasion, and metastasis. Results: Activating transcription factor 3 (ATF3) bound mutant p53 proteins, sensitized cancer cells to chemotherapy, and suppressed mutant p53-mediated cell migration. Conclusion: ATF3 suppressed the tumor-promoting function of mutant p53. Significance: Targeting ATF3 could be a novel strategy for treating p53-mutated cancer. Mutant p53 proteins (mutp53) often acquire oncogenic activities, conferring drug resistance and/or promoting cancer cell migration and invasion. Although it has been well established that such a gain of function is mainly achieved through interaction with transcriptional regulators, thereby modulating cancer-associated gene expression, how the mutp53 function is regulated remains elusive. Here we report that activating transcription factor 3 (ATF3) bound common mutp53 (e.g. R175H and R273H) and, subsequently, suppressed their oncogenic activities. ATF3 repressed mutp53-induced NFKB2 expression and sensitized R175H-expressing cancer cells to cisplatin and etoposide treatments. Moreover, ATF3 appeared to suppress R175H- and R273H-mediated cancer cell migration and invasion as a consequence of preventing the transcription factor p63 from inactivation by mutp53. Accordingly, ATF3 promoted the expression of the metastasis suppressor SHARP1 in mutp53-expressing cells. An ATF3 mutant devoid of the mutp53-binding domain failed to disrupt the mutp53-p63 binding and, thus, lost the activity to suppress mutp53-mediated migration, suggesting that ATF3 binds to mutp53 to suppress its oncogenic function. In line with these results, we found that down-regulation of ATF3 expression correlated with lymph node metastasis in TP53-mutated human lung cancer. We conclude that ATF3 can suppress mutp53 oncogenic function, thereby contributing to tumor suppression in TP53-mutated cancer.


Biochemical Pharmacology | 2013

Inhibition of gastric tumor growth by a novel Hsp90 inhibitor.

Chunwan Lu; Di Liu; Jing Jin; Hemantkumar Deokar; Yi Zhang; John K. Buolamwini; Xiaoming Yu; Chunhong Yan; Xiaoguang Chen

Heat shock protein 90 (Hsp90) is a molecular chaperone engaging in multiple cellular signaling by stabilizing oncoproteins (e.g. Akt and c-Raf) in tumor cells. Whereas Hsp90 inhibitors such as 17-AAG exert promising antitumor effects in clinical trials, current efforts focus on developing agents targeting Hsp90 with improved efficacy and lower toxicity. Using a fluorescence polarization assay, we screened over a hundred of synthetic small molecules and identified a resorcinol derivative LD053 that bound the Hsp90 ATP-binding pocket. The binding of LD053 to Hsp90 dissociated the co-chaperone protein cdc37 from Hsp90, resulting in destabilization of Akt and c-Raf and subsequent inhibition of PI3K/Akt and c-Raf/Mek/Erk signaling in BGC823 gastric cancer cells. As a consequence, LD053 decreased cancer cell viability and induced apoptosis evidenced by increased subG0/G1 cell population and increased cleavage of caspase 3 and PARP. Interestingly, normal human cells appeared insensitive to LD053 treatments. Consistent with its in vitro anticancer activities, LD053 significantly inhibited growth of BGC823 xenografts in nude mice without apparent body weight loss. These results thus demonstrate that LD053 is a novel Hsp90 inhibitor and has potential to be used to treat gastric cancer.


Oncogene | 2015

Loss of ATF3 Promotes Akt Activation and Prostate Cancer Development in a Pten Knockout Mouse Model

Ziyan Wang; Dong Xu; Han Fei Ding; Jaejik Kim; Junran Zhang; Tsonwin Hai; Chunhong Yan

Activating transcription factor 3 (ATF3) responds to diverse cellular stresses, and regulates oncogenic activities (for example, proliferation, survival and migration) through direct transcriptional regulation or protein-protein interactions. Although aberrant ATF3 expression is frequently found in human cancers, the role of ATF3 in tumorigenesis is poorly understood. Here, we demonstrate that ATF3 suppresses the development of prostate cancer induced by knockout of the tumor suppressor Pten in mouse prostates. Whereas the oncogenic stress elicited by Pten loss induced ATF3 expression in prostate epithelium, we found that ATF3 deficiency increased cell proliferation and promoted cell survival, leading to early onset of mouse prostatic intraepithelial neoplasia and the progression of prostate lesions to invasive adenocarcinoma. Importantly, the loss of ATF3 promoted activation of the oncogenic AKT signaling evidenced by high levels of phosphorylated AKT and S6 proteins in ATF3-null prostate lesions. In line with these in vivo results, knockdown of ATF3 expression in human prostate cancer cells by single guided RNA-mediated targeting activated AKT and increased matrix metalloproteinase-9 expression. Our results thus link ATF3 to the AKT signaling, and suggest that ATF3 is a tumor suppressor for the major subset of prostate cancers harboring dysfunctional Pten.


Nucleic Acids Research | 2015

53BP1 promotes microhomology-mediated end-joining in G1-phase cells

Xiahui Xiong; Zhanwen Du; Ying Wang; Zhihui Feng; Pan Fan; Chunhong Yan; Henning Willers; Junran Zhang

Alternative non-homologous end joining (alt-NHEJ) was originally identified as a backup repair mechanism in the absence of classical NHEJ (c-NHEJ) factors but recent studies have demonstrated that alt-NHEJ is active even when c-NHEJ as well as homologous recombination is available. The functions of 53BP1 in NHEJ processes are not well understood. Here, we report that 53BP1 promotes DNA double-strand break (DSB) repair and genomic stability not only in c-NHEJ-proficient but also -deficient human G1-phase cells. Using an array of repair substrates we show that these effects of 53BP1 are correlated with a promotion of microhomology-mediated end-joining (MMEJ), a subtype of alt-NHEJ, in G1-phase. Consistent with a specific role in MMEJ we confirm that 53BP1 status does not affect c-NHEJ. 53BP1 supports sequence deletion during MMEJ consistent with a putative role in facilitating end-resection. Interestingly, promotion of MMEJ by 53BP1 in G1-phase cells is only observed in the presence of functional BRCA1. Depletion of both 53BP1 and BRCA1 increases repair needing microhomology usage and augments loss of DNA sequence, suggesting that MMEJ is a highly regulated DSB repair process. Together, these findings significantly expand our understanding of the cell-cycle-dependent roles of 53BP1 in DSB repair.

Collaboration


Dive into the Chunhong Yan's collaboration.

Top Co-Authors

Avatar

Junran Zhang

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Hongbo Wang

Albany Medical College

View shared research outputs
Top Co-Authors

Avatar

Han Fei Ding

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar

Ziyan Wang

Albany Medical College

View shared research outputs
Top Co-Authors

Avatar

Shumei Ren

Albany Medical College

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Zhanwen Du

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Dong Xu

Albany Medical College

View shared research outputs
Top Co-Authors

Avatar

Douglas D. Boyd

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Hongmei Cui

Albany Medical College

View shared research outputs
Researchain Logo
Decentralizing Knowledge