Network


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

Hotspot


Dive into the research topics where Shipra Shukla is active.

Publication


Featured researches published by Shipra Shukla.


Nature Medicine | 2013

ETS factors reprogram the androgen receptor cistrome and prime prostate tumorigenesis in response to PTEN loss

Yu Chen; Ping Chi; Shira Rockowitz; Phillip J. Iaquinta; Tambudzai Shamu; Shipra Shukla; Dong Gao; Inna Sirota; Brett S. Carver; John Wongvipat; Howard I. Scher; Deyou Zheng; Charles L. Sawyers

Studies of ETS-mediated prostate oncogenesis have been hampered by a lack of suitable experimental systems. Here we describe a new conditional mouse model that shows robust, homogenous ERG expression throughout the prostate. When combined with homozygous Pten loss, the mice developed accelerated, highly penetrant invasive prostate cancer. In mouse prostate tissue, ERG markedly increased androgen receptor (AR) binding. Robust ERG-mediated transcriptional changes, observed only in the setting of Pten loss, included the restoration of AR transcriptional output and upregulation of genes involved in cell death, migration, inflammation and angiogenesis. Similarly, ETS variant 1 (ETV1) positively regulated the AR cistrome and transcriptional output in ETV1-translocated, PTEN-deficient human prostate cancer cells. In two large clinical cohorts, expression of ERG and ETV1 correlated with higher AR transcriptional output in PTEN-deficient prostate cancer specimens. We propose that ETS factors cause prostate-specific transformation by altering the AR cistrome, priming the prostate epithelium to respond to aberrant upstream signals such as PTEN loss.


Nature | 2015

Alternative transcription initiation leads to expression of a novel ALK isoform in cancer

Thomas Wiesner; William R. Lee; Anna C. Obenauf; Leili Ran; Rajmohan Murali; Qi Fan Zhang; Elissa W.P. Wong; Wenhuo Hu; Sasinya N. Scott; Ronak Shah; Iñigo Landa; Julia Button; Nathalie Lailler; Andrea Sboner; Dong Gao; Devan Murphy; Zhen Cao; Shipra Shukla; Travis J. Hollmann; Lu Wang; Laetitia Borsu; Taha Merghoub; Gary K. Schwartz; Michael A. Postow; Charlotte E. Ariyan; James A. Fagin; Deyou Zheng; Marc Ladanyi; Michael F. Berger; Yu Chen

Activation of oncogenes by mechanisms other than genetic aberrations such as mutations, translocations, or amplifications is largely undefined. Here we report a novel isoform of the anaplastic lymphoma kinase (ALK) that is expressed in ∼11% of melanomas and sporadically in other human cancer types, but not in normal tissues. The novel ALK transcript initiates from a de novo alternative transcription initiation (ATI) site in ALK intron 19, and was termed ALKATI. In ALKATI-expressing tumours, the ATI site is enriched for H3K4me3 and RNA polymerase II, chromatin marks characteristic of active transcription initiation sites. ALKATI is expressed from both ALK alleles, and no recurrent genetic aberrations are found at the ALK locus, indicating that the transcriptional activation is independent of genetic aberrations at the ALK locus. The ALKATI transcript encodes three proteins with molecular weights of 61.1, 60.8 and 58.7 kilodaltons, consisting primarily of the intracellular tyrosine kinase domain. ALKATI stimulates multiple oncogenic signalling pathways, drives growth-factor-independent cell proliferation in vitro, and promotes tumorigenesis in vivo in mouse models. ALK inhibitors can suppress the kinase activity of ALKATI, suggesting that patients with ALKATI-expressing tumours may benefit from ALK inhibitors. Our findings suggest a novel mechanism of oncogene activation in cancer through de novo alternative transcription initiation.


Cancer Discovery | 2015

Combined Inhibition of MAP Kinase and KIT Signaling Synergistically Destabilizes ETV1 and Suppresses GIST Tumor Growth

Leili Ran; Inna Sirota; Zhen Cao; Devan Murphy; Yuedan Chen; Shipra Shukla; Yuanyuan Xie; Kaufmann Mc; Dong Gao; Zhu S; Rossi F; John Wongvipat; Taguchi T; William D. Tap; Ingo K. Mellinghoff; Peter Besmer; Cristina R. Antonescu; Ping Chi

UNLABELLED Gastrointestinal stromal tumor (GIST), originating from the interstitial cells of Cajal (ICC), is characterized by frequent activating mutations of the KIT receptor tyrosine kinase. Despite the clinical success of imatinib, which targets KIT, most patients with advanced GIST develop resistance and eventually die of the disease. The ETS family transcription factor ETV1 is a master regulator of the ICC lineage. Using mouse models of Kit activation and Etv1 ablation, we demonstrate that ETV1 is required for GIST initiation and proliferation in vivo, validating it as a therapeutic target. We further uncover a positive feedback circuit where MAP kinase activation downstream of KIT stabilizes the ETV1 protein, and ETV1 positively regulates KIT expression. Combined targeting of ETV1 stability by imatinib and MEK162 resulted in increased growth suppression in vitro and complete tumor regression in vivo. The combination strategy to target ETV1 may provide an effective therapeutic strategy in GIST clinical management. SIGNIFICANCE ETV1 is a lineage-specific oncogenic transcription factor required for the growth and survival of GIST. We describe a novel strategy of targeting ETV1 protein stability by the combination of MEK and KIT inhibitors that synergistically suppress tumor growth. This strategy has the potential to change first-line therapy in GIST clinical management.


Cancer Cell | 2017

Aberrant Activation of a Gastrointestinal Transcriptional Circuit in Prostate Cancer Mediates Castration Resistance

Shipra Shukla; Joanna Cyrta; Devan Murphy; Edward Walczak; Leili Ran; Praveen Agrawal; Yuanyuan Xie; Yuedan Chen; Shangqian Wang; Yu Zhan; Dan Li; Elissa W.P. Wong; Andrea Sboner; Himisha Beltran; Juan Miguel Mosquera; Jessica Sher; Zhen Cao; John Wongvipat; Richard Koche; Anuradha Gopalan; Deyou Zheng; Mark A. Rubin; Howard I. Scher; Ping Chi; Yu Chen

Prostate cancer exhibits a lineage-specific dependence on androgen signaling. Castration resistance involves reactivation of androgen signaling or activation of alternative lineage programs to bypass androgen requirement. We describe an aberrant gastrointestinal-lineage transcriptome expressed in ∼5% of primary prostate cancer that is characterized by abbreviated response to androgen-deprivation therapy and in ∼30% of castration-resistant prostate cancer. This program is governed by a transcriptional circuit consisting of HNF4G and HNF1A. Cistrome and chromatin analyses revealed that HNF4G is a pioneer factor that generates and maintains enhancer landscape at gastrointestinal-lineage genes, independent of androgen-receptor signaling. In HNF4G/HNF1A-double-negative prostate cancer, exogenous expression of HNF4G at physiologic levels recapitulates the gastrointestinal transcriptome, chromatin landscape, and leads to relative castration resistance.


Cancer Discovery | 2017

FOXF1 Defines the Core-Regulatory Circuitry in Gastrointestinal Stromal Tumor

Leili Ran; Yuedan Chen; Jessica Sher; Elissa W.P. Wong; Devan Murphy; Jenny Zhang; Dan Li; Kemal Deniz; Inna Sirota; Zhen Cao; Shangqian Wang; Youxin Guan; Shipra Shukla; Katie Yang Li; Alan Chramiec; Yuanyuan Xie; Deyou Zheng; Richard Koche; Cristina R. Antonescu; Yu Chen; Ping Chi

The cellular context that integrates upstream signaling and downstream nuclear response dictates the oncogenic behavior and shapes treatment responses in distinct cancer types. Here, we uncover that in gastrointestinal stromal tumor (GIST), the forkhead family member FOXF1 directly controls the transcription of two master regulators, KIT and ETV1, both required for GIST precursor-interstitial cells of Cajal lineage specification and GIST tumorigenesis. Further, FOXF1 colocalizes with ETV1 at enhancers and functions as a pioneer factor that regulates the ETV1-dependent GIST lineage-specific transcriptome through modulation of the local chromatin context, including chromatin accessibility, enhancer maintenance, and ETV1 binding. Functionally, FOXF1 is required for human GIST cell growth in vitro and murine GIST tumor growth and maintenance in vivo The simultaneous control of the upstream signaling and nuclear response sets up a unique regulatory paradigm and highlights the critical role of FOXF1 in enforcing the GIST cellular context for highly lineage-restricted clinical behavior and treatment response.Significance: We uncover that FOXF1 defines the core-regulatory circuitry in GIST through both direct transcriptional regulation and pioneer factor function. The unique and simultaneous control of signaling and transcriptional circuitry by FOXF1 sets up an enforced transcriptional addiction to FOXF1 in GIST, which can be exploited diagnostically and therapeutically. Cancer Discov; 8(2); 234-51. ©2017 AACR.See related commentary by Lee and Duensing, p. 146This article is highlighted in the In This Issue feature, p. 127.


Cancer Research | 2017

ETV1-positive cells give rise to BRAFV600E mutant gastrointestinal stromal tumors

Leili Ran; Devan Murphy; Jessica Sher; Zhen Cao; Shangqian Wang; Edward Walczak; Youxin Guan; Yuanyuan Xie; Shipra Shukla; Yu Zhan; Cristina R. Antonescu; Yu Chen; Ping Chi

Gastrointestinal stromal tumor (GIST) is the most common subtype of sarcoma. Despite clinical advances in the treatment of KIT/PDGFRA-mutant GIST, similar progress against KIT/PDGFRA wild-type GIST, including mutant BRAF-driven tumors, has been limited by a lack of model systems. ETV1 is a master regulator in the intestinal cells of Cajal (ICC), thought to be the cells of origin of GIST. Here, we present a model in which the ETV1 promoter is used to specifically and inducibly drive Cre recombinase in ICC as a strategy to study GIST pathogenesis. Using a conditional allele for BrafV600E , a mutation observed in clinical cases of GIST, we observed that BrafV600E activation was sufficient to drive ICC hyperplasia but not GIST tumorigenesis. In contrast, combining BrafV600E activation with Trp53 loss was sufficient to drive both ICC hyperplasia and formation of multifocal GIST-like tumors in the mouse gastrointestinal tract with 100% penetrance. This mouse model of sporadic GIST model was amenable to therapeutic intervention, and it recapitulated clinical responses to RAF inhibition seen in human GIST. Our work offers a useful in vivo model of human sporadic forms of BRAF-mutant GIST to help unravel its pathogenesis and therapeutic response to novel experimental agents. Cancer Res; 77(14); 3758-65. ©2017 AACR.


Cancer Research | 2014

Abstract 3396: Dual lineage inhibition of ETV1 and KIT disrupts the ETV1-KIT feed forward circuit and potentiates imatinib antitumor effect in GIST oncogenesis

Leili Ran; Inna Sirota; Zhen Cao; Devan Murphy; Shipra Shukla; Ferdinando Rossi; John Wongvipat; William D. Tap; Peter Besmer; Cristina R. Antonescu; Yu Chen; Ping Chi

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA Gastrointestinal stromal tumour (GIST) is one of the most common types of human sarcoma and is primarily defined by activating mutations in the KIT or PDGFRA receptor tyrosine kinases. Despite the initial clinical success of imatinib that specifically target mutant KIT and PDGFRA, imatinib resistance has become the biggest challenge in the management of advanced GIST patient. Novel therapeutics that can improve the efficacy of first line imatinib therapy and/or prevent and overcome imatinib resistance is imperative. The ETS family member, ETV1, has been previously identified as a lineage-specific survival factor that cooperates with mutant KIT by ETV1 protein stabilization by active MAP kinase signaling downstream of KIT signaling in GIST oncogenesis. Here we demonstrate that ETV1 is required for GIST initiation and maintenance in vivo using compound genetically engineered mouse models (GEMM). We further identified that ETV1 forms a feed forward circuit in GIST oncogenesis where the ETV1 protein is stabilized by active MAP kinase signaling downstream of KIT and stabilized ETV1enhances KIT expression through direct binding to the KIT enhancer regions in both GIST mouse models and human GIST cell lines. The dual lineage targeting of KIT by imatinib and ETV1 by MEK162 (an MEK inhibitor) disrupts the ETV1-KIT feed forward circuit and induces more apoptosis than single agent imatinib or MEK162 in human GIST cells. The combination therapy resulted in complete tumor regression whereas single agent imatinib or MEK162 resulted in stabilization of disease in human GIST xenograft studies. Moreover, the combination therapy also induced more tumor fibrosis than single agent imatinib or MEK162 in GIST GEMM. These observations demonstrate the in vivo role of ETV1 in GIST oncogenesis and the feasibility of targeting ETV1 protein stability by inhibiting MAP kinase signaling. They also suggest that the dual lineage targeting of ETV1 and KIT by the combination therapy may provide a more effective therapeutic strategy than imatinib alone in GIST management. Citation Format: Leili Ran, Inna Sirota, Zhen Cao, Devan Murphy, Shipra Shukla, Ferdinando Rossi, John Wongvipat, William D. Tap, Peter Besmer, Cristina R. Antonescu, Yu Chen, Ping Chi. Dual lineage inhibition of ETV1 and KIT disrupts the ETV1-KIT feed forward circuit and potentiates imatinib antitumor effect in GIST oncogenesis. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 3396. doi:10.1158/1538-7445.AM2014-3396


Omics A Journal of Integrative Biology | 2018

Abstract A15: Modeling sporadic gastrointestinal stromal tumor with BRAFV600E mutation

Leili Ran; Devan Murphy; Jessica Sher; Zhen Cao; Shangqian Wang; Edward Walczak; Youxin Guan; Yuanyuan Xie; Shipra Shukla; Yu Zhan; Cristina R. Antonescu; Yu Chen; Ping Chi

Gastrointestinal stromal tumor (GIST) is the most common subtype of sarcoma, characterized by activating mutations in KIT , PDGFRA , BRAF, N/H/KRA S, and loss of function genetic/epigenetic alterations in NF1 , SDH -complex. Despite clinical advances for KIT/PDGFRA-mutant GIST, the scientific and clinical advances for KIT/PDGFRA-wild type GISTs have remained extremely limited due to the lack of model systems. Due to the lack of ICC-lineage specific cre system, modeling for sporadic GIST has not been feasible. ETV1 has recently been described as an ICC and GIST-lineage specific master regulator. Here, using the cre-activated Rosa26 CAGGS-LSL-EYFP system, we demonstrate that Etv1 CREERT2 preferentially activates cre in the Etv1 -expressing ICC lineage by tamoxifen. Using the Braf CA conditional allele for cre-activated Braf V600E , we observed that Braf V600E alone, while leading to development of ICC hyperplasia, is not sufficient to drive GIST tumorigenesis. Combination of Braf V600E activation and Trp53 loss not only gives rise to ICC hyperplasia, but also leads to multifocal GIST-like tumors in mouse GI tract with 100% penetrance. We further demonstrate that the Braf V600E ; Tp53-/- sporadic GIST mouse models are amenable for therapeutic intervention and recapitulate clinical responses to RAF inhibitors in human GIST. Our observations provide the first in vivo model for understanding the pathogenesis and therapeutic responses in BRAF-mutant GISTs. Importantly, these observations provide the proof of principle for modeling sporadic GIST in adult mice using the Etv1 CREERT2 system and establish the feasibility and utility of establishing model systems for human GISTs that do not have any model systems available for mechanistic studies and therapeutic development. Citation Format: Leili Ran, Devan Murphy, Jessica Sher, Zhen Cao, Shangqian Wang, Edward Walczak, Youxin Guan, Yuanyuan Xie, Shipra Shukla, Yu Zhan, Cristina R. Antonescu, Yu Chen, Ping Chi. Modeling sporadic gastrointestinal stromal tumor with BRAFV600E mutation [abstract]. In: Proceedings of the AACR Conference on Advances in Sarcomas: From Basic Science to Clinical Translation; May 16-19, 2017; Philadelphia, PA. Philadelphia (PA): AACR; Clin Cancer Res 2018;24(2_Suppl):Abstract nr A15.


Journal of Clinical Investigation | 2018

COP1-DET1-ETS axis regulates ERK transcriptome and sensitivity to MAPK inhibitors

Yuanyuan Xie; Zhen Cao; Elissa W.P. Wong; Youxin Guan; Wenfu Ma; Jenny Zhang; Edward Walczak; Devan Murphy; Leili Ran; Inna Sirota; Shangqian Wang; Shipra Shukla; Dong Gao; Simon R.V. Knott; Kenneth Chang; Justin Leu; John Wongvipat; Cristina R. Antonescu; Gregory J. Hannon; Ping Chi; Yu Chen

Aberrant activation of MAPK signaling leads to the activation of oncogenic transcriptomes. How MAPK signaling is coupled with the transcriptional response in cancer is not fully understood. In 2 MAPK-activated tumor types, gastrointestinal stromal tumor and melanoma, we found that ETV1 and other Pea3-ETS transcription factors are critical nuclear effectors of MAPK signaling that are regulated through protein stability. Expression of stabilized Pea3-ETS factors can partially rescue the MAPK transcriptome and cell viability after MAPK inhibition. To identify the players involved in this process, we performed a pooled genome-wide RNAi screen using a fluorescence-based ETV1 protein stability sensor and identified COP1, DET1, DDB1, UBE3C, PSMD4, and COP9 signalosome members. COP1 or DET1 loss led to decoupling between MAPK signaling and the downstream transcriptional response, where MAPK inhibition failed to destabilize Pea3 factors and fully inhibit the MAPK transcriptome, thus resulting in decreased sensitivity to MAPK pathway inhibitors. We identified multiple COP1 and DET1 mutations in human tumors that were defective in the degradation of Pea3-ETS factors. Two melanoma patients had de novo DET1 mutations arising after vemurafenib treatment. These observations indicate that MAPK signaling–dependent regulation of Pea3-ETS protein stability is a key signaling node in oncogenesis and therapeutic resistance to MAPK pathway inhibition.


Cancer Research | 2012

Abstract IA15: Context-specific oncogenesis by ETS-family transcription factors

Yu Chen; Shipra Shukla; Ping Chi; Brett S. Carver; Charles L. Sawyers

Abstract Recurrent translocation involving ETS-family transcription factors including ERG and ETV1, ETV4, and ETV5 occur in over half of prostate cancers, implicating them as driver oncogenes in prostate tumorigenesis. Two observations pose important questions. 1) The same ETS factors aberrantly expressed in prostate cancer can be endogenously highly expressed in other tissues to modulate normal physiology (e.g., ERG expression in endothelial cells and ETV1 expression in neurons). 2) Forced overexpression in prostate model systems such as immortalized human primary prostate epithelial cells and mouse prostate yields minimal phenotype. These observations indicate that ETS mediated oncogenesis is highly dependent on the cellular context, which is mediated by the cell/tissue type and cooperating lesions. We asked how cellular context affects the DNA binding, transcriptional output, and phenotype of ETS proteins. We compared human ETS dependent tumor cell lines from two lineages, prostate cancer and gastrointestinal stromal tumor and show that ETS binding is largely tissue specific. Further, we have generated genetically engineered mouse models that express ETS in different tissues, within different compartments of the prostate gland, and with different cooperating lesions. We show that ERG binding to genomic DNA is largely similar in PTEN intact and PTEN null mouse prostates. Yet, ERG can only mediate a transcriptional program that mediates an aggressive phenotype in PTEN null prostates. These data suggests that cellular context is critical for ETS transcription factor function. Citation Format: Yu Chen, Shipra Shukla, Ping Chi, Brett Carver, Charles L. Sawyers. Context-specific oncogenesis by ETS-family transcription factors [abstract]. In: Proceedings of the AACR Special Conference on Advances in Prostate Cancer Research; 2012 Feb 6-9; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2012;72(4 Suppl):Abstract nr IA15.

Collaboration


Dive into the Shipra Shukla's collaboration.

Top Co-Authors

Avatar

Leili Ran

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ping Chi

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Yu Chen

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Zhen Cao

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Devan Murphy

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Cristina R. Antonescu

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Yuanyuan Xie

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Inna Sirota

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

John Wongvipat

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Shangqian Wang

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge