Network


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

Hotspot


Dive into the research topics where Sharon Shacham is active.

Publication


Featured researches published by Sharon Shacham.


Nature Genetics | 2014

Genomic and molecular characterization of esophageal squamous cell carcinoma

De-Chen Lin; Jia-Jie Hao; Yasunobu Nagata; Liang Xu; Li Shang; Xuan Meng; Yusuke Sato; Yusuke Okuno; Ana Maria Varela; Ling-Wen Ding; Manoj Garg; Li-Zhen Liu; Henry Yang; Dong Yin; Zhi-Zhou Shi; Yan-Yi Jiang; Wen-Yue Gu; Ting Gong; Yu Zhang; Xin Xu; Ori Kalid; Sharon Shacham; Seishi Ogawa; Ming-Rong Wang; H. Phillip Koeffler

Esophageal squamous cell carcinoma (ESCC) is prevalent worldwide and particularly common in certain regions of Asia. Here we report the whole-exome or targeted deep sequencing of 139 paired ESCC cases, and analysis of somatic copy number variations (SCNV) of over 180 ESCCs. We identified previously uncharacterized mutated genes such as FAT1, FAT2, ZNF750 and KMT2D, in addition to those already known (TP53, PIK3CA and NOTCH1). Further SCNV evaluation, immunohistochemistry and biological analysis suggested their functional relevance in ESCC. Notably, RTK-MAPK-PI3K pathways, cell cycle and epigenetic regulation are frequently dysregulated by multiple molecular mechanisms in this cancer. Our approaches also uncovered many druggable candidates, and XPO1 was further explored as a therapeutic target because it showed both gene mutation and protein overexpression. Our integrated study unmasks a number of novel genetic lesions in ESCC and provides an important molecular foundation for understanding esophageal tumors and developing therapeutic targets.


Blood | 2012

Selective inhibitors of nuclear export show that CRM1/XPO1 is a target in chronic lymphocytic leukemia

Rosa Lapalombella; Qingxiang Sun; Katie Williams; Larissa Tangeman; Shruti Jha; Yiming Zhong; Virginia M. Goettl; Emilia Mahoney; Caroline Berglund; Sneha V. Gupta; Alicia Farmer; Rajeswaran Mani; Amy J. Johnson; David M. Lucas; Xiaokui Mo; Dirk Daelemans; Vincent Sandanayaka; Sharon Shechter; Dilara McCauley; Sharon Shacham; Michael Kauffman; Yuh Min Chook; John C. Byrd

The nuclear export protein XPO1 is overexpressed in cancer, leading to the cytoplasmic mislocalization of multiple tumor suppressor proteins. Existing XPO1-targeting agents lack selectivity and have been associated with significant toxicity. Small molecule selective inhibitors of nuclear export (SINEs) were designed that specifically inhibit XPO1. Genetic experiments and X-ray structures demonstrate that SINE covalently bind to a cysteine residue in the cargo-binding groove of XPO1, thereby inhibiting nuclear export of cargo proteins. The clinical relevance of SINEs was explored in chronic lymphocytic leukemia (CLL), a disease associated with recurrent XPO1 mutations. Evidence is presented that SINEs can restore normal regulation to the majority of the dysregulated pathways in CLL both in vitro and in vivo and induce apoptosis of CLL cells with a favorable therapeutic index, with enhanced killing of genomically high-risk CLL cells that are typically unresponsive to traditional therapies. More importantly, SINE slows disease progression, and improves overall survival in the Eμ-TCL1-SCID mouse model of CLL with minimal weight loss or other toxicities. Together, these findings demonstrate that XPO1 is a valid target in CLL with minimal effects on normal cells and provide a basis for the development of SINEs in CLL and related hematologic malignancies.


Endocrinology | 1997

Mechanism of Mitogen-Activated Protein Kinase Activation by Gonadotropin-Releasing Hormone in the Pituitary αT3–1 Cell Line: Differential Roles of Calcium and Protein Kinase C1

Nachum Reiss; Linet N. Llevi; Sharon Shacham; Dagan Harris; Rony Seger; Zvi Naor

The mechanism of mitogen-activated protein kinase (MAPK, ERK) stimulation by the GnRH analog [d-Trp6]GnRH (GnRH-a) was investigated in the gonadotroph-derived αT3–1 cell line. GnRH-a as well as the protein kinase C (PKC) activator 12-O-tetradecanoyl phorbol-13-acetate (TPA) stimulated a sustained response of MAPK activity, whereas epidermal growth factor (EGF) stimulated a transient response. MAPK kinase (MEK) is also activated by GnRH-a, but in a transient manner. GnRH-a and TPA apparently activated mainly the MAPK isoform ERK1, as revealed by Mono-Q fast protein liquid chromatography followed by Western blotting as well as by gel kinase assay. GnRH-a and TPA stimulated the tyrosine phosphorylation of several proteins, and this effect as well as the stimulation of MAPK activity were inhibited by the PKC inhibitor GF 109203X. Similarly, down-regulation of TPA-sensitive PKC subspecies nearly abolished the effect of GnRH-a and TPA on MAPK activity. Furthermore, the protein tyrosine kinase (PTK) inhibitor ge...


Blood | 2012

Preclinical activity of a novel CRM1 inhibitor in acute myeloid leukemia.

Parvathi Ranganathan; Xueyan Yu; Caroline Na; Ramasamy Santhanam; Sharon Shacham; Michael Kauffman; Alison Walker; Rebecca B. Klisovic; William Blum; Michael A. Caligiuri; Carlo M. Croce; Guido Marcucci; Ramiro Garzon

Chromosome maintenance protein 1 (CRM1) is a nuclear export receptor involved in the active transport of tumor suppressors (e.g., p53 and nucleophosmin) whose function is altered in cancer because of increased expression and overactive transport. Blocking CRM1-mediated nuclear export of such proteins is a novel therapeutic strategy to restore tumor suppressor function. Orally bioavailable selective inhibitors of nuclear export (SINE) that irreversibly bind to CRM1 and block the function of this protein have been recently developed. Here we investigated the antileukemic activity of KPT-SINE (KPT-185 and KPT-276) in vitro and in vivo in acute myeloid leukemia (AML). KPT-185 displayed potent antiproliferative properties at submicromolar concentrations (IC50 values; 100-500 nM), induced apoptosis (average 5-fold increase), cell-cycle arrest, and myeloid differentiation in AML cell lines and patient blasts. A strong down-regulation of the oncogene FLT3 after KPT treatment in both FLT3-ITD and wild-type cell lines was observed. Finally, using the FLT3-ITD-positive MV4-11 xenograft murine model, we show that treatment of mice with oral KPT-276 (analog of KPT-185 for in vivo studies) significantly prolongs survival of leukemic mice (P < .01). In summary, KPT-SINE are highly potent in vitro and in vivo in AML. The preclinical results reported here support clinical trials of KPT-SINE in AML.


Frontiers in Neuroendocrinology | 1998

Mechanism of GnRH Receptor Signaling: Combinatorial Cross-Talk of Ca2+and Protein Kinase C☆☆☆

Zvi Naor; Dagan Harris; Sharon Shacham

Gonadotropin-releasing hormone (GnRH), the first key hormone of reproduction, is synthesized in the hypothalamus and is released in a pulsatile manner to stimulate pituitary gonadotrope-luteinizing hormone (LH) and follicle-stimulating hormone (FSH) synthesis and release. Gonadotropes represent only about 10% of pituitary cells and are divided into monohormonal cells (18% LH and 22% FSH cells) and 60% multihormonal (LH + FSH) cells. GnRH binds to a specific seven transmembrane domain receptor which is coupled to Gq and activates sequentially different phospholipases to provide Ca2+ and lipid-derived messenger molecules. Initially, phospholipase C is activated, followed by activation of both phospholipase A2 (PLA2) and phospholipase D (PLD). Generation of the second messengers inositol 1,4,5-trisphosphate and diacylglycerol (DAG) lead to mobilization of intracellular pools of Ca2+ and activation of protein kinase C (PKC). Early DAG and Ca2+, derived via enhanced phosphoinositide turnover, might be involved in rapid activation of selective Ca(2+)-dependent, conventional PKC isoforms (cPKC). On the other hand, late DAG, derived from phosphatidic acid (PA) via PLD, may activate Ca(2+)-independent novel PKC isoforms (nPKC). In addition, arachidonic acid (AA) which is liberated by activated PLA2, might also support selective activation of PKC isoforms (PKCs) with or without other cofactors. Differential cross-talk of Ca2+, AA, and selective PKCs might generate a compartmentalized signal transduction cascade to downstream elements which are activated during the neurohormone action. Among those elements is the mitogen-activated protein kinase (MAPK) cascade which is activated by GnRH in a PKC-, Ca(2+)-, and protein tyrosine kinase (PTK)-dependent fashion. Transcriptional regulation can be mediated by the activation of transcription factors such as c-fos by MAPK. Indeed, GnRH activates the expression of both c-jun and c-fos which might participate in gene regulation via the formation of AP-1. The signaling cascade leading to gonadotropin (LH and FSH) gene regulation by GnRH is still not known and might involve the above-mentioned cascades. AA and selective lipoxygenase products such as leukotriene C4 also participate in GnRH action, possibly by cross-talk with PKCs, or by an autocrine/paracrine amplification cycle. A complex combinatorial, spatial and temporal cross-talk of the above messenger molecules seems to mediate the diverse effects elicited by GnRH, the first key hormone of the reproductive cycle.


Leukemia | 2014

CRM1 inhibition induces tumor cell cytotoxicity and impairs osteoclastogenesis in multiple myeloma: molecular mechanisms and therapeutic implications

Yu-Tzu Tai; Yosef Landesman; Chirag Acharya; Yolanda Calle; Mike Zhong; Michele Cea; Daniel Tannenbaum; Antonia Cagnetta; Michaela R. Reagan; Aditya Munshi; William Senapedis; J. R. Saint-Martin; T. Kashyap; Sharon Shacham; Michael Kauffman; Yumei Gu; Lizi Wu; Irene M. Ghobrial; Fenghuang Zhan; Andrew L. Kung; S. A. Schey; Paul G. Richardson; Nikhil C. Munshi; Kenneth C. Anderson

The key nuclear export protein CRM1/XPO1 may represent a promising novel therapeutic target in human multiple myeloma (MM). Here we showed that chromosome region maintenance 1 (CRM1) is highly expressed in patients with MM, plasma cell leukemia cells and increased in patient cells resistant to bortezomib treatment. CRM1 expression also correlates with increased lytic bone and shorter survival. Importantly, CRM1 knockdown inhibits MM cell viability. Novel, oral, irreversible selective inhibitors of nuclear export (SINEs) targeting CRM1 (KPT-185, KPT-330) induce cytotoxicity against MM cells (ED50<200 nM), alone and cocultured with bone marrow stromal cells (BMSCs) or osteoclasts (OC). SINEs trigger nuclear accumulation of multiple CRM1 cargo tumor suppressor proteins followed by growth arrest and apoptosis in MM cells. They further block c-myc, Mcl-1, and nuclear factor κB (NF-κB) activity. SINEs induce proteasome-dependent CRM1 protein degradation; concurrently, they upregulate CRM1, p53-targeted, apoptosis-related, anti-inflammatory and stress-related gene transcripts in MM cells. In SCID mice with diffuse human MM bone lesions, SINEs show strong anti-MM activity, inhibit MM-induced bone lysis and prolong survival. Moreover, SINEs directly impair osteoclastogenesis and bone resorption via blockade of RANKL-induced NF-κB and NFATc1, with minimal impact on osteoblasts and BMSCs. These results support clinical development of SINE CRM1 antagonists to improve patient outcome in MM.


Leukemia | 2013

Antileukemic activity of nuclear export inhibitors that spare normal hematopoietic cells

Julia Etchin; Qi Sun; Alex Kentsis; Alicia Farmer; Zi Chao Zhang; Takaomi Sanda; Marc R. Mansour; C Barcelo; Dilara McCauley; Michael Kauffman; Sharon Shacham; Amanda L. Christie; Andrew L. Kung; Scott J. Rodig; Yuh Min Chook; A T Look

Drugs that target the chief mediator of nuclear export, chromosome region maintenance 1 protein (CRM1) have potential as therapeutics for leukemia, but existing CRM1 inhibitors show variable potencies and a broad range of cytotoxic effects. Here, we report the structural analysis and antileukemic activity of a new generation of small-molecule inhibitors of CRM1. Designated selective inhibitors of nuclear export (SINE), these compounds were developed using molecular modeling to screen a small virtual library of compounds against the nuclear export signal (NES) groove of CRM1. The 2.2-Å crystal structure of the CRM1-Ran-RanBP1 complex bound to KPT-251, a representative molecule of this class of inhibitors, shows that the drug occupies part of the groove in CRM1 that is usually occupied by the NES, but penetrates much deeper into the groove and blocks CRM1-directed protein export. SINE inhibitors exhibit potent antileukemic activity, inducing apoptosis at nanomolar concentrations in a panel of 14 human acute myeloid leukemia (AML) cell lines representing different molecular subtypes of the disease. When administered orally to immunodeficient mice engrafted with human AML cells, KPT-251 had potent antileukemic activity with negligible toxicity to normal hematopoietic cells. Thus, KPT-SINE CRM1 antagonists represent a novel class of drugs that warrant further testing in AML patients.


Proteins | 2004

PREDICT modeling and in‐silico screening for G‐protein coupled receptors

Sharon Shacham; Yael Marantz; Shay Bar-Haim; Ori Kalid; Dora Warshaviak; Noa Avisar; Boaz Inbal; Alexander Heifetz; Merav Fichman; Maya Topf; Zvi Naor; Silvia Noiman; Oren M. Becker

G‐protein coupled receptors (GPCRs) are a major group of drug targets for which only one x‐ray structure is known (the nondrugable rhodopsin), limiting the application of structure‐based drug discovery to GPCRs. In this paper we present the details of PREDICT, a new algorithmic approach for modeling the 3D structure of GPCRs without relying on homology to rhodopsin. PREDICT, which focuses on the transmembrane domain of GPCRs, starts from the primary sequence of the receptor, simultaneously optimizing multiple ‘decoy’ conformations of the protein in order to find its most stable structure, culminating in a virtual receptor‐ligand complex. In this paper we present a comprehensive analysis of three PREDICT models for the dopamine D2, neurokinin NK1, and neuropeptide Y Y1 receptors. A shorter discussion of the CCR3 receptor model is also included. All models were found to be in good agreement with a large body of experimental data. The quality of the PREDICT models, at least for drug discovery purposes, was evaluated by their successful utilization in in‐silico screening. Virtual screening using all three PREDICT models yielded enrichment factors 9‐fold to 44‐fold better than random screening. Namely, the PREDICT models can be used to identify active small‐molecule ligands embedded in large compound libraries with an efficiency comparable to that obtained using crystal structures for non‐GPCR targets. Proteins 2004.


British Journal of Haematology | 2013

KPT-330 inhibitor of CRM1 (XPO1)-mediated nuclear export has selective anti-leukaemic activity in preclinical models of T-cell acute lymphoblastic leukaemia and acute myeloid leukaemia

Julia Etchin; Takaomi Sanda; Marc R. Mansour; Alex Kentsis; Joan Montero; Bonnie Thi Le; Amanda L. Christie; Dilara McCauley; Scott J. Rodig; Michael Kauffman; Sharon Shacham; Richard Stone; Anthony Letai; Andrew L. Kung; A. Thomas Look

This study explored the anti‐leukaemic efficacy of novel irreversible inhibitors of the major nuclear export receptor, chromosome region maintenance 1 (CRM1, also termed XPO1). We found that these novel CRM1 antagonists, termed SINE (Selective Inhibitors of Nuclear Export), induced rapid apoptosis at low nanomolar concentrations in a panel of 14 human T‐cell acute lymphoblastic leukaemia (T‐ALL) cell lines representing different molecular subtypes of the disease. To assess in vivo anti‐leukaemia cell activity, we engrafted immunodeficient mice intravenously with the human T‐ALL MOLT‐4 cells, which harbour activating mutations of NOTCH1 and NRAS as well as loss of function of the CDKN2A, PTEN and TP53 tumour suppressors and express a high level of oncogenic transcription factor TAL1. Importantly, we examined the in vivo anti‐leukaemic efficacy of the clinical SINE compound KPT‐330 against T‐ALL and acute myeloid leukaemia (AML) cells. These studies demonstrated striking in vivo activity of KPT‐330 against T‐ALL and AML cells, with little toxicity to normal murine haematopoietic cells. Taken together, our results show that SINE CRM1 antagonists represent promising ‘first‐in‐class’ drugs with a novel mechanism of action and wide therapeutic index, and imply that drugs of this class show promise for the targeted therapy of T‐ALL and AML.


Blood | 2013

Prognostic impact and targeting of CRM1 in acute myeloid leukemia

Kensuke Kojima; Steven M. Kornblau; Vivian Ruvolo; Archana Dilip; Seshagiri Duvvuri; R. Eric Davis; Min Zhang; Zhiqiang Wang; Kevin R. Coombes; Nianxiang Zhang; Yi Hua Qiu; Jared K. Burks; Hagop M. Kantarjian; Sharon Shacham; Michael Kauffman; Michael Andreeff

Chromosomal region maintenance 1 (CRM1) is a nuclear export receptor recognizing proteins bearing a leucine-rich nuclear export signal. CRM1 is involved in nuclear export of tumor suppressors such as p53. We investigated the prognostic significance of CRM1 in acute myeloid leukemia (AML) and effects of a novel small-molecule selective inhibitor of CRM1. CRM1 protein expression was determined in 511 newly diagnosed AML patients and was correlated with mouse double minute 2 (MDM2) and p53 levels. High CRM1 expression was associated with short survival of patients and remained an adverse prognostic factor in multivariate analysis. CRM1 inhibitor KPT-185 induced mainly full-length p53 and apoptosis in a p53-dependent manner, whereas inhibition of proliferation was p53 independent. Patient samples with p53 mutations showed low sensitivity to KPT-185. Nuclear retention of p53 induced by CRM1 inhibition synergized with increased levels of p53 induced by MDM2 inhibition in apoptosis induction. KPT-185 and Nutlin-3a, alone and in combination, induced synergistic apoptosis in patient-derived CD34(+)/CD38(-) AML, but not in normal progenitor cells. Data suggest that CRM1 exerts an antiapoptotic function and is highly prognostic in AML. We propose a novel combinatorial approach for the therapy of AML, aimed at maximal activation of p53-mediated apoptosis by concomitant MDM2 and CRM1 inhibition.

Collaboration


Dive into the Sharon Shacham's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dilara McCauley

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

William Senapedis

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Erkan Baloglu

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Trinayan Kashyap

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Boris Klebanov

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Sharon Shechter

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge