Network


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

Hotspot


Dive into the research topics where Jean Zhao is active.

Publication


Featured researches published by Jean Zhao.


Nature Reviews Drug Discovery | 2009

Targeting the phosphoinositide 3-kinase pathway in cancer

Pixu Liu; Hailing Cheng; Thomas M. Roberts; Jean Zhao

The phosphoinositide 3-kinase (PI3K) pathway is a key signal transduction system that links oncogenes and multiple receptor classes to many essential cellular functions, and is perhaps the most commonly activated signalling pathway in human cancer. This pathway therefore presents both an opportunity and a challenge for cancer therapy. Even as inhibitors that target PI3K isoforms and other major nodes in the pathway, including AKT and mammalian target of rapamycin (mTOR), reach clinical trials, major issues remain. Here, we highlight recent progress that has been made in our understanding of the PI3K pathway and discuss the potential of and challenges for the development of therapeutic agents that target this pathway in cancer.


Nature | 2010

COT drives resistance to RAF inhibition through MAP kinase pathway reactivation

Cory M. Johannessen; Jesse S. Boehm; So Young Kim; Sapana Thomas; Leslie Wardwell; Laura A. Johnson; Caroline Emery; Nicolas Stransky; Alexandria P. Cogdill; Jordi Barretina; Giordano Caponigro; Haley Hieronymus; Ryan R. Murray; Kourosh Salehi-Ashtiani; David E. Hill; Marc Vidal; Jean Zhao; Xiaoping Yang; Ozan Alkan; Sungjoon Kim; Jennifer L. Harris; Christopher J. Wilson; Vic E. Myer; Peter Finan; David E. Root; Thomas M. Roberts; Todd R. Golub; Keith T. Flaherty; Reinhard Dummer; Barbara Weber

Oncogenic mutations in the serine/threonine kinase B-RAF (also known as BRAF) are found in 50–70% of malignant melanomas. Pre-clinical studies have demonstrated that the B-RAF(V600E) mutation predicts a dependency on the mitogen-activated protein kinase (MAPK) signalling cascade in melanoma—an observation that has been validated by the success of RAF and MEK inhibitors in clinical trials. However, clinical responses to targeted anticancer therapeutics are frequently confounded by de novo or acquired resistance. Identification of resistance mechanisms in a manner that elucidates alternative ‘druggable’ targets may inform effective long-term treatment strategies. Here we expressed ∼600 kinase and kinase-related open reading frames (ORFs) in parallel to interrogate resistance to a selective RAF kinase inhibitor. We identified MAP3K8 (the gene encoding COT/Tpl2) as a MAPK pathway agonist that drives resistance to RAF inhibition in B-RAF(V600E) cell lines. COT activates ERK primarily through MEK-dependent mechanisms that do not require RAF signalling. Moreover, COT expression is associated with de novo resistance in B-RAF(V600E) cultured cell lines and acquired resistance in melanoma cells and tissue obtained from relapsing patients following treatment with MEK or RAF inhibitors. We further identify combinatorial MAPK pathway inhibition or targeting of COT kinase activity as possible therapeutic strategies for reducing MAPK pathway activation in this setting. Together, these results provide new insights into resistance mechanisms involving the MAPK pathway and articulate an integrative approach through which high-throughput functional screens may inform the development of novel therapeutic strategies.


Cell | 2007

Integrative Genomic Approaches Identify IKBKE as a Breast Cancer Oncogene

Jesse S. Boehm; Jean Zhao; Jun Yao; So Young Kim; Ron Firestein; Ian F. Dunn; Sarah K. Sjostrom; Levi A. Garraway; Stanislawa Weremowicz; Andrea L. Richardson; Heidi Greulich; Carly J. Stewart; Laura Mulvey; Rhine R. Shen; Lauren Ambrogio; Tomoko Hirozane-Kishikawa; David E. Hill; Marc Vidal; Matthew Meyerson; Jennifer K. Grenier; Greg Hinkle; David E. Root; Thomas M. Roberts; Eric S. Lander; Kornelia Polyak; William C. Hahn

The karyotypic chaos exhibited by human epithelial cancers complicates efforts to identify mutations critical for malignant transformation. Here we integrate complementary genomic approaches to identify human oncogenes. We show that activation of the ERK and phosphatidylinositol 3-kinase (PI3K) signaling pathways cooperate to transform human cells. Using a library of activated kinases, we identify several kinases that replace PI3K signaling and render cells tumorigenic. Whole genome structural analyses reveal that one of these kinases, IKBKE (IKKepsilon), is amplified and overexpressed in breast cancer cell lines and patient-derived tumors. Suppression of IKKepsilon expression in breast cancer cell lines that harbor IKBKE amplifications induces cell death. IKKepsilon activates the nuclear factor-kappaB (NF-kappaB) pathway in both cell lines and breast cancers. These observations suggest a mechanism for NF-kappaB activation in breast cancer, implicate the NF-kappaB pathway as a downstream mediator of PI3K, and provide a framework for integrated genomic approaches in oncogene discovery.


Cell | 2005

A Genetic Screen for Candidate Tumor Suppressors Identifies REST

Thomas F. Westbrook; Eric Martin; Michael R. Schlabach; Yumei Leng; Anthony C. Liang; Bin Feng; Jean Zhao; Thomas M. Roberts; Gail Mandel; Gregory J. Hannon; Ronald A. DePinho; Lynda Chin; Stephen J. Elledge

Tumorigenesis is a multistep process characterized by a myriad of genetic and epigenetic alterations. Identifying the causal perturbations that confer malignant transformation is a central goal in cancer biology. Here we report an RNAi-based genetic screen for genes that suppress transformation of human mammary epithelial cells. We identified genes previously implicated in proliferative control and epithelial cell function including two established tumor suppressors, TGFBR2 and PTEN. In addition, we uncovered a previously unrecognized tumor suppressor role for REST/NRSF, a transcriptional repressor of neuronal gene expression. Array-CGH analysis identified REST as a frequent target of deletion in colorectal cancer. Furthermore, we detect a frameshift mutation of the REST gene in colorectal cancer cells that encodes a dominantly acting truncation capable of transforming epithelial cells. Cells lacking REST exhibit increased PI(3)K signaling and are dependent upon this pathway for their transformed phenotype. These results implicate REST as a human tumor suppressor and provide a novel approach to identifying candidate genes that suppress the development of human cancer.


Nature Reviews Cancer | 2015

PI3K in cancer: divergent roles of isoforms, modes of activation and therapeutic targeting

Lauren M. Thorpe; Haluk Yuzugullu; Jean Zhao

Phosphatidylinositol 3-kinases (PI3Ks) are crucial coordinators of intracellular signalling in response to extracellular stimuli. Hyperactivation of PI3K signalling cascades is one of the most common events in human cancers. In this Review, we discuss recent advances in our knowledge of the roles of specific PI3K isoforms in normal and oncogenic signalling, the different ways in which PI3K can be upregulated, and the current state and future potential of targeting this pathway in the clinic.


Cancer Cell | 2003

A chemical genetic screen identifies inhibitors of regulated nuclear export of a Forkhead transcription factor in PTEN-deficient tumor cells

Tweeny R. Kau; Frank C. Schroeder; Shivapriya Ramaswamy; Cheryl L Wojciechowski; Jean Zhao; Thomas M. Roberts; Jon Clardy; William R. Sellers; Pamela A. Silver

The PI3K/PTEN/Akt signal transduction pathway plays a key role in many tumors. Downstream targets of this pathway include the Forkhead family of transcription factors (FOXO1a, FOXO3a, FOXO4). In PTEN null cells, FOXO1a is inactivated by PI3K-dependent phosphorylation and mislocalization to the cytoplasm, yet still undergoes nucleocytoplasmic shuttling. Since forcible localization of FOXO1a to the nucleus can reverse tumorigenicity of PTEN null cells, a high-content, chemical genetic screen for inhibitors of FOXO1a nuclear export was performed. The compounds detected in the primary screen were retested in secondary assays, and structure-function relationships were identified. Novel general export inhibitors were found that react with CRM1 as well as a number of compounds that inhibit PI3K/Akt signaling, among which are included multiple antagonists of calmodulin signaling.


Cancer Cell | 2003

Human mammary epithelial cell transformation through the activation of phosphatidylinositol 3-kinase

Jean Zhao; Ole Gjoerup; Romesh Subramanian; Yuan Cheng; Wen Chen; Thomas M. Roberts; William C. Hahn

Recent studies have demonstrated that introduction of hTERT in combination with SV40 large T antigen (LT), small t antigen (st), and H-rasV12 suffices to transform many primary human cells. In human mammary epithelial cells (HMECs) expressing elevated c-Myc, activated H-Ras is dispensable for anchorage-independent growth. Using this system, we show that st activates the PI3K pathway and that constitutive PI3K signaling substitutes for st in transformation. Moreover, using constitutively active versions of Akt1 and Rac1, we show that these downstream pathways of PI3K synergize to achieve anchorage-independent growth. At lower levels of c-myc expression, activated PI3K also replaces st to complement H-rasV12 and LT and confers both soft agar growth and tumorigenicity. However, elevated c-myc expression cannot replace H-rasV12 for tumorigenesis. These observations begin to define the pathways perturbed during the transformation of HMECs.


Nature Medicine | 2011

Oncogenic PIK3CA-driven mammary tumors frequently recur via PI3K pathway-dependent and PI3K pathway-independent mechanisms

Pixu Liu; Hailing Cheng; Stephanie Santiago; Maria B. Ræder; Fan Zhang; Adam Isabella; Janet Yang; Derek J Semaan; Changzhong Chen; Edward A. Fox; Nathanael S. Gray; John E. Monahan; Robert Schlegel; Rameen Beroukhim; Gordon B. Mills; Jean Zhao

PIK3CA gain-of-function mutations are a common oncogenic event in human malignancy, making phosphatidylinositol 3-kinase (PI3K) a target for cancer therapy. Despite the promise of targeted therapy, resistance often develops, leading to treatment failure. To elucidate mechanisms of resistance to PI3K-targeted therapy, we constructed a mouse model of breast cancer conditionally expressing human PIK3CAH1047R. Notably, most PIK3CAH1047R-driven mammary tumors recurred after PIK3CAH1047R inactivation. Genomic analyses of recurrent tumors revealed multiple lesions, including focal amplification of Met or Myc (also known as c-Met and c-Myc, respectively). Whereas Met amplification led to tumor survival dependent on activation of endogenous PI3K, tumors with Myc amplification became independent of the PI3K pathway. Functional analyses showed that Myc contributed to oncogene independence and resistance to PI3K inhibition. Notably, PIK3CA mutations and c-MYC elevation co-occur in a substantial fraction of human breast tumors. Together, these data suggest that c-MYC elevation represents a potential mechanism by which tumors develop resistance to current PI3K-targeted therapies.


Proceedings of the National Academy of Sciences of the United States of America | 2006

The p110α isoform of PI3K is essential for proper growth factor signaling and oncogenic transformation

Jean Zhao; Hailing Cheng; Shidong Jia; Li Wang; Ole Gjoerup; Aki Mikami; Thomas M. Roberts

Growth factor signaling is mediated through Class IA phosphatidylinositol 3-kinases (PI3Ks). Among this class of enzymes, only p110α, encoded by the PIK3CA gene, has been found to be mutant in human cancers. To determine the specific functions of p110α, we generated mice carrying a conditionally targeted allele of the PIK3CA gene. Here, we report that PIK3CA-knockout mouse embryonic fibroblasts are deficient in cellular signaling in response to various growth factors, unable to differentiate into adipocytes, and resistant to oncogenic transformation induced by a variety of oncogenic receptor tyrosine kinases, indicating a fundamental role for p110α in these biological processes.


Cancer Research | 2004

Androgen-Induced Differentiation and Tumorigenicity of Human Prostate Epithelial Cells

Raanan Berger; Phillip G. Febbo; Pradip K. Majumder; Jean Zhao; Shayan Mukherjee; Sabina Signoretti; K.Thirza Campbell; William R. Sellers; Thomas M. Roberts; Massimo Loda; Todd R. Golub; William C. Hahn

Androgen ablation is the primary treatment modality for patients with metastatic prostate cancer; however, the role of androgen receptor signaling in prostate cancer development remains enigmatic. Using a series of genetically defined immortalized and tumorigenic human prostate epithelial cells, we found that introduction of the androgen receptor induced differentiation of transformed prostate epithelial cells to a luminal phenotype reminiscent of organ-confined prostate cancer when placed in the prostate microenvironment. Moreover, androgen receptor expression converted previously androgen-independent, tumorigenic prostate epithelial cells into cells dependent on testosterone for tumor formation. These observations indicate that androgen receptor expression is oncogenic and addictive for the human prostate epithelium.

Collaboration


Dive into the Jean Zhao's collaboration.

Top Co-Authors

Avatar

Craig J. Thomas

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Christopher P. Austin

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

David A. Fidock

Columbia University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge