Network


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

Hotspot


Dive into the research topics where Boris Leibovitch is active.

Publication


Featured researches published by Boris Leibovitch.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Interference with Sin3 function induces epigenetic reprogramming and differentiation in breast cancer cells

Eduardo F. Farias; Kevin Petrie; Boris Leibovitch; Janice Murtagh; Manuel Boix Chornet; Tino Schenk; Arthur Zelent; Samuel Waxman

Sin3A/B is a master transcriptional scaffold and corepressor that plays an essential role in the regulation of gene transcription and maintenance of chromatin structure, and its inappropriate recruitment has been associated with aberrant gene silencing in cancer. Sin3A/B are highly related, large, multidomian proteins that interact with a wide variety of transcription factors and corepressor components, and we examined whether disruption of the function of a specific domain could lead to epigenetic reprogramming and derepression of specific subsets of genes. To this end, we selected the Sin3A/B-paired amphipathic α-helices (PAH2) domain based on its established role in mediating the effects of a relatively small number of transcription factors containing a PAH2-binding motif known as the Sin3 interaction domain (SID). Here, we show that in both human and mouse breast cancer cells, the targeted disruption of Sin3 function by introduction of a SID decoy that interferes with PAH2 binding to SID-containing partner proteins reverted the silencing of genes involved in cell growth and differentiation. In particular, the SID decoy led to epigenetic reprogramming and reexpression of the important breast cancer-associated silenced genes encoding E-cadherin, estrogen receptor α, and retinoic acid receptor β and impaired tumor growth in vivo. Interestingly, the SID decoy was effective in the triple-negative M.D. Anderson-Metastatic Breast-231 (MDA-MB-231) breast cancer cell line, restoring sensitivity to 17β-estradiol, tamoxifen, and retinoids. Therefore, the development of small molecules that can block interactions between PAH2 and SID-containing proteins offers a targeted epigenetic approach for treating this type of breast cancer that may also have wider therapeutic implications.


Oncotarget | 2015

Targeting the SIN3A-PF1 interaction inhibits epithelial to mesenchymal transition and maintenance of a stem cell phenotype in triple negative breast cancer.

Nidhi Bansal; Kevin Petrie; Rossitza Christova; Chi Yeh Chung; Boris Leibovitch; Louise Howell; Veronica Gil; Yordan Sbirkov; Eunjee Lee; Joanna Wexler; Edgardo V. Ariztia; Rajal Sharma; Jun Zhu; Emily Bernstein; Ming-Ming Zhou; Arthur Zelent; Eduardo F. Farias; Samuel Waxman

Triple negative breast cancer (TNBC) is characterized by a poorly differentiated phenotype and limited treatment options. Aberrant epigenetics in this subtype represent a potential therapeutic opportunity, but a better understanding of the mechanisms contributing to the TNBC pathogenesis is required. The SIN3 molecular scaffold performs a critical role in multiple cellular processes, including epigenetic regulation, and has been identified as a potential therapeutic target. Using a competitive peptide corresponding to the SIN3 interaction domain of MAD (Tat-SID), we investigated the functional consequences of selectively blocking the paired amphipathic α-helix (PAH2) domain of SIN3. Here, we report the identification of the SID-containing adaptor PF1 as a factor required for maintenance of the TNBC stem cell phenotype and epithelial-to-mesenchymal transition (EMT). Tat-SID peptide blocked the interaction between SIN3A and PF1, leading to epigenetic modulation and transcriptional downregulation of TNBC stem cell and EMT markers. Importantly, Tat-SID treatment also led to a reduction in primary tumor growth and disseminated metastatic disease in vivo. In support of these findings, knockdown of PF1 expression phenocopied treatment with Tat-SID both in vitro and in vivo. These results demonstrate a critical role for a complex containing SIN3A and PF1 in TNBC and provide a rational for its therapeutic targeting.


Molecular Cancer Therapeutics | 2015

Selective Inhibition of SIN3 Corepressor with Avermectins as a Novel Therapeutic Strategy in Triple-Negative Breast Cancer

Yeon Jin Kwon; Kevin Petrie; Boris Leibovitch; Lei Zeng; Mihaly Mezei; Louise Howell; Veronica Gil; Rossitza Christova; Nidhi Bansal; Shuai Yang; Rajal Sharma; Edgardo V. Ariztia; Jessica Frankum; Rachel Brough; Yordan Sbirkov; Alan Ashworth; Christopher J. Lord; Arthur Zelent; Eduardo F. Farias; Ming-Ming Zhou; Samuel Waxman

Triple-negative breast cancers (TNBC) lacking estrogen, progesterone, and HER2 receptors account for 10% to 20% of breast cancer and are indicative of poor prognosis. The development of effective treatment strategies therefore represents a pressing unmet clinical need. We previously identified a molecularly targeted approach to target aberrant epigenetics of TNBC using a peptide corresponding to the SIN3 interaction domain (SID) of MAD. SID peptide selectively blocked binding of SID-containing proteins to the paired α-helix (PAH2) domain of SIN3, resulting in epigenetic and transcriptional modulation of genes associated with epithelial–mesenchymal transition (EMT). To find small molecule inhibitor (SMI) mimetics of SID peptide, we performed an in silico screen for PAH2 domain–binding compounds. This led to the identification of the avermectin macrocyclic lactone derivatives selamectin and ivermectin (Mectizan) as candidate compounds. Both selamectin and ivermectin phenocopied the effects of SID peptide to block SIN3–PAH2 interaction with MAD, induce expression of CDH1 and ESR1, and restore tamoxifen sensitivity in MDA-MB-231 human and MMTV-Myc mouse TNBC cells in vitro. Treatment with selamectin or ivermectin led to transcriptional modulation of genes associated with EMT and maintenance of a cancer stem cell phenotype in TNBC cells. This resulted in impairment of clonogenic self-renewal in vitro and inhibition of tumor growth and metastasis in vivo. Underlining the potential of avermectins in TNBC, pathway analysis revealed that selamectin also modulated the expression of therapeutically targetable genes. Consistent with this, an unbiased drug screen in TNBC cells identified selamectin-induced sensitization to a number of drugs, including those targeting modulated genes. Mol Cancer Ther; 14(8); 1824–36. ©2015 AACR.


Developmental Biology | 2011

Adipocyte derived paracrine mediators of mammary ductal morphogenesis controlled by retinoic acid receptors.

Christine Marzan; Tara S. Kupumbati; Silvina Bertran; Tracey-Ann Samuels; Boris Leibovitch; Rafael Mira-y-Lopez; Liliana Ossowski; Eduardo F. Farias

We generated a transgenic (Tg)-mouse model expressing a dominant negative-(DN)-RARα, (RARαG303E) under adipocytes-specific promoter to explore the paracrine role of adipocyte retinoic acid receptors (RARs) in mammary morphogenesis. Transgenic adipocytes had reduced level of RARα, β and γ, which coincided with a severely underdeveloped pubertal and mature ductal tree with profoundly decreased epithelial cell proliferation. Transplantation experiments of mammary epithelium and of whole mammary glands implicated a fat-pad dependent paracrine mechanism in the stunted phenotype of the epithelial ductal tree. Co-cultures of primary adipocytes, or in vitro differentiated adipocyte cell line, with mammary epithelium showed that when activated, adipocyte-RARs contribute to generation of secreted proliferative and pro-migratory factors. Gene expression microarrays revealed a large number of genes regulated by adipocyte-RARs. Among them, pleiotrophin (PTN) was identified as the paracrine effectors of epithelial cell migration. Its expression was found to be strongly inhibited by DN-RARα, an inhibition relieved by pharmacological doses of all-trans retinoic acid (atRA) in culture and in vivo. Moreover, adipocyte-PTHR, another atRA responsive gene, was found to be an up-stream regulator of PTN. Overall, these results support the existence of a novel paracrine loop controlled by adipocyte-RAR that regulates the mammary ductal tree morphogenesis.


Oncotarget | 2016

Blocking the PAH2 domain of Sin3A inhibits tumorigenesis and confers retinoid sensitivity in triple negative breast cancer

Nidhi Bansal; Almudena Bosch; Boris Leibovitch; Lutecia Pereira; Elena Cubedo; Jianshi Yu; Keely Pierzchalski; Jace W. Jones; Melissa Fishel; Maureen A. Kane; Arthur Zelent; Samuel Waxman; Eduardo F. Farias

Triple negative breast cancer (TNBC) frequently relapses locally, regionally or as systemic metastases. Development of targeted therapy that offers significant survival benefit in TNBC is an unmet clinical need. We have previously reported that blocking interactions between PAH2 domain of chromatin regulator Sin3A and the Sin3 interaction domain (SID) containing proteins by SID decoys result in EMT reversal, and re-expression of genes associated with differentiation. Here we report a novel and therapeutically relevant combinatorial use of SID decoys. SID decoys activate RARα/β pathways that are enhanced in combination with RARα-selective agonist AM80 to induce morphogenesis and inhibit tumorsphere formation. These findings correlate with inhibition of mammary hyperplasia and a significant increase in tumor-free survival in MMTV-Myc oncomice treated with a small molecule mimetic of SID (C16). Further, in two well-established mouse TNBC models we show that treatment with C16-AM80 combination has marked anti-tumor effects, prevents lung metastases and seeding of tumor cells to bone marrow. This correlated to a remarkable 100% increase in disease-free survival with a possibility of “cure” in mice bearing a TNBC-like tumor. Targeting Sin3A by C16 alone or in combination with AM80 may thus be a promising adjuvant therapy for treating or preventing metastatic TNBC.


Oncotarget | 2017

Targeted interference of SIN3A-TGIF1 function by SID decoy treatment inhibits Wnt signaling and invasion in triple negative breast cancer cells

Yeon Jin Kwon; Boris Leibovitch; Nidhi Bansal; Lutecia Pereira; Chi Yeh Chung; Edgardo V. Ariztia; Arthur Zelent; Eduardo F. Farias; Samuel Waxman

Cancer cell invasion is an obligatory step for metastatic dissemination that contributes to rapid relapse and a poorer survival in triple negative breast cancer (TNBC) patients. Development of novel therapeutic strategies to block tumor invasion is an unmet need in the treatment of cancer. We reported that the selective inhibition of the PAH2 domain of SIN3A protein function markedly suppressed metastatic dissemination to the lungs in TNBC xenograft bearing mice. Here, we show that TNBC cell lines treated with Sin3 interaction domain (SID) decoy peptides that bind to PAH2 display a strong in vitro inhibition of transwell invasion. This is accompanied by actin cytoskeleton reorganization with increased cortical actin deposition and downregulation of known Wnt target genes that are associated with epithelial to mesenchymal transition (EMT) and cancer cell invasion. Wnt pathway inhibition by SID decoy peptide was confirmed by decreased Wnt reporter activity and altered cytoplasmic localization of nuclear β-catenin. TGIF1, a transcription factor that modulates Wnt signaling and known to interact with the PAH2 domain of SIN3A, can be dissociated from the SIN3A complex by SID decoys. TGIF1 knockdown inhibits WNT target genes and in vitro cell invasion suggesting that TGIF1 might be a key target of the SID decoys to block tumor invasion. Taken together, targeting SIN3 function using SID decoys is a novel strategy to reverse invasion and the EMT program in TNBC translating into the inhibition of metastasis dissemination and eradication of residual disease.Cancer cell invasion is an obligatory step for metastatic dissemination that contributes to rapid relapse and a poorer survival in triple negative breast cancer (TNBC) patients. Development of novel therapeutic strategies to block tumor invasion is an unmet need in the treatment of cancer. We reported that the selective inhibition of the PAH2 domain of SIN3A protein function markedly suppressed metastatic dissemination to the lungs in TNBC xenograft bearing mice. Here, we show that TNBC cell lines treated with Sin3 interaction domain (SID) decoy peptides that bind to PAH2 display a strong in vitro inhibition of transwell invasion. This is accompanied by actin cytoskeleton reorganization with increased cortical actin deposition and downregulation of known Wnt target genes that are associated with epithelial to mesenchymal transition (EMT) and cancer cell invasion. Wnt pathway inhibition by SID decoy peptide was confirmed by decreased Wnt reporter activity and altered cytoplasmic localization of nuclear β-catenin. TGIF1, a transcription factor that modulates Wnt signaling and known to interact with the PAH2 domain of SIN3A, can be dissociated from the SIN3A complex by SID decoys. TGIF1 knockdown inhibits WNT target genes and in vitro cell invasion suggesting that TGIF1 might be a key target of the SID decoys to block tumor invasion. Taken together, targeting SIN3 function using SID decoys is a novel strategy to reverse invasion and the EMT program in TNBC translating into the inhibition of metastasis dissemination and eradication of residual disease.


Cancer Research | 2014

Abstract 807: Selamectin and ivermectin are small molecule inhibitors that interfere with Sin3A-PAH2 function and exert anti-tumor activity in triple-negative breast cancer

Yeon-Jin Kwon; Boris Leibovitch; Lei Zeng; Mihaly Mezei; Rossitza Christova; Shuai Yang; Rajal Sharma; Edgardo Aritzia; Nidhi Bansal; Ming-Ming Zhou; Authur Zelent; Eduardo F. Farias; Samuel Waxman

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA INTRODUCTION: Sin3 is a complex scaffolding protein that participates in epigenetic regulation involved in a variety of biological processes, including chromatin remodeling, development, differentiation, cell cycle and survival. Paired Amphipathic domain 2 (PAH2) domain of Sin3 was shown to interact with Sin3 interacting domain (SID)-containing transcription factors, such as Mxd1, KLF10/11, and REST. Previously, we reported that SID transcripts and decoy peptides disrupt the interactions between Sin3-PAH2 domain and SID-containing transcription factors and induce epigenetic reprogramming in TNBC. RESULTS AND CONCLUSION: Here we show that fourteen SMIs mimicking SID decoys were identified by in silico computation from a chemical library composed of 115,000 compounds. We confirmed that selamectin and ivermectin are SID decoys using duo-link assays. Our NMR 15N-HSQC spectroscopy binding results further demonstrated ivermectin, which shares the same binding site as selamectin, directly interacted with PAH2. Moreover, selamectin inhibited Sin3 repression activity as measured by mammalian two-hybrid and in vitro GST pull-down assays. Notably, ivermectin was approved by Food and Drug Administration (FDA) to treat small animals and human. However, the anti-tumorigenic roles for these SMIs had not been investigated in TNBC in vitro and in vivo. Remarkably, these SMIs did not affect growth in 2-dimensional (2D) clonogenecity assay, but significantly suppressed growth in 3D matrigel and tumorsphere cultures. Moreover, selamectin significantly suppressed Myc mammary tumor growth in vivo more than 50 percent without any cytotoxicity. Selamectin and ivermectin induced re-expression of CDH1 and ESR1, restoring tamoxifen sensitivity. Treatment with these compounds inhibited in vitro invasion as measured by Boyden chamber in MCF7/ADR and MDA-MB-231 cells. Selamectin and ivermectin were shown to alleviate multidrug resistance and enhance doxorubicin and paclitaxel cytotoxicity in MCF7/ADR cells, which is unrelated to disruption of Sin3-PAH2. Based on further studies of the Sin3A complexes using selamectin and ivermectin, we seek to identify novel SMIs with better efficiency for targeting Sin3 complex and treating TNBCs in the future. Citation Format: Yeon-Jin Kwon, Boris A. Leibovitch, Lei Zeng, Mihaly Mezei, Rossitza Christova, Shuai Yang, Rajal Sharma, Edgardo Aritzia, nidhi bansal, Ming-Ming Zhou, Authur Zelent, Eduardo Farias, Samuel Waxman. Selamectin and ivermectin are small molecule inhibitors that interfere with Sin3A-PAH2 function and exert anti-tumor activity in triple-negative breast cancer. [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 807. doi:10.1158/1538-7445.AM2014-807


Cancer Research | 2016

Abstract 4115: Inhibition of triple negative breast cancer cell invasion by the targeted interference of Sin3A function affecting Wnt and TGFβ signaling

Yeon-Jin Kwon; Boris Leibovitch; Nidhi Bansal; Lutecia Pereira; Edgardo V. Ariztia; Kevin Petrie; Arthur Zelent; Ming-Ming Zhou; Eduardo F. Farias; Samuel Waxman

Cancer cell invasion is an obligatory step for metastatic dissemination that contributes to rapid relapse and a poor survival in TNBC patients. Development of novel therapeutic strategies to block tumor invasion is an unmet need for TNBC treatment and for other tumor types. We reported that decoys with the SID sequence designed to bind and inhibit the function of PAH-2 domain of Sin3A protein markedly prolong survival in the adjuvant setting due to inhibition of metastatic dissemination to the lungs and bone marrow in TNBC mouse models. Here, we show that TNBC cell lines treated with SID decoys (peptides) display a strong in vitro inhibition of migration and invasion. This is accompanied by actin cytoskeleton reorganization with increased cortical actin, and inhibition of proteolytic enzymes (MMP9; MT-MMP1 and uPA) involved in extracellular matrix degradation. DNA microarray and Ingenuity pathway analysis (IPA) showed that the SID decoys inhibit Wnt and TGFβ signaling that is associated with epithelial to mesenchymal transition (EMT). Treatment with SID decoy peptide downregulated WNT/β-catenin-driven transactivation as measured by decreased promoter H3K4me3 and decreased expression of Wnt target genes like LEF1 and TCF7L2. We also show that SID decoys induce translocation of nuclear β-catenin to the cytoplasm in TNBC at 24 hours. Wnt/β-catenin is critical for EMT, cancer stem cell self-renewal, and early invasion in TNBC. TGIF1, a transcription factor that modulates TGFβ and Wnt signaling pathways and known to to interact with the PAH2 domain of Sin3A, can be dissociated from Sin3A complex by SID decoy treatment as measured by co-immunoprecipitation (Co-IP) and proximity linked assay. DNA microarray of SID peptide treated TNBC cells shows inhibition of TGFβ signaling evidenced by downregulation of MMP9, MT1-MMP and PLAU, known target genes of this pathway. This is in line with inhibition of the EMT program predicted by the IPA analysis in SID peptide treated TNBC. Taken together, the results indicate that SID decoys have potential value as therapeutic agents to revert the EMT program in TNBC that should translate into the inhibition of metastasis dissemination and eradication of residual disease in TNBC. To test this in clinic future investigations will involve the use of our previously identified small molecule mimetic of SID peptide, selamectin that is also a FDA approved drug. Use of a recently constructed cyclic stapled peptide that inhibits PAH-2 binding and invasion at Citation Format: Yeon-Jin Kwon, Boris A. Leibovitch, Nidhi Bansal, Lutecia Pereira, Edgardo V. Ariztia, Kevin Petrie, Arthur Zelent, Ming-Ming Zhou, Eduardo F. Farias, Samuel Waxman. Inhibition of triple negative breast cancer cell invasion by the targeted interference of Sin3A function affecting Wnt and TGFβ signaling. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 4115.


Cancer Research | 2015

Abstract 3511: Targeted epigenetic reprogramming reverts tumor progression in triple-negative breast cancer models by the activation of retinoic acid receptor alpha

Nidhi Bansal; Almudena Bosch; Boris Leibovitch; Keely Pierzchalski; Zhou Ming-Ming; Maureen A. Kane; Samuel Waxman; Eduardo F. Farias

INTRODUCTION: Triple negative breast cancer (TNBC) is associated with aggressiveness, early recurrence and poor prognosis and no other therapeutic alternative besides chemotherapy. The silencing of genes associated with cell differentiation in cancer is required for tumor progression. In this process the epigenetic modifications induced by aberrant control of the chromatin remodeling machinery in transformed cells plays a key role. Therefore, finding a mechanism to restore the expression of these genes by reverting the abnormal epigenetic modifications is an important task in the fight against cancer. We showed previously that the selective interference of the Sin39s PAH2 domain with Sin3 interaction domain (SID) mimicking peptides restored expression of E-cadherin, estrogen receptor and RARa target genes such as RARb and CRBP1 leading to cell differentiation and anti-tumor effect in TNBC cells. Here we studied whether the re-expression of retinoic acid receptors (RARs) induced by the small molecule C16 (SID decoy) can make TNBC cells sensitive to selective retinoid therapies. EXPERIMENTAL APPROACH: Human and mouse cell models of TNBC as well as MMTV-Myc oncomice were used to test the effects of the SID decoys on the induction of RAR signaling, cell proliferation, induction of differentiation in 3D cultures, expansion of the cancer stem cell compartment, atypical ductal hyperplasia (ADH), ductal carcinola in situ (DCIS), tumor development and metastatic dissemination. RESULTS: Our data show that C16 induced a differential expression of RARs, ∼10 fold upregulation of RARa2, RARbeta1/2, ∼10 fold downregulation of RARg1 and ∼30% increased production of retinoic acid (RA). The combination of C16 plus RARa agonists, AM80 (tamibarotene) or AM580 induced 75% inhibition of cell proliferation in 2D cultures, induced immature acini in 3D and impaired cancer stem cell proliferation inhibiting tumorsphere formation (∼65%) in vitro. In vivo, the combination of C16 and AM580 reduced 90% tumor growth and metastasis. Interestingly, the treatment of MMTV-Myc oncomice with C16 alone or in combination with AM80 prevented mammary gland hyperplasia, DCIS and delayed tumor development (80% tumor free survival, Kaplan-Meier analysis). CONCLUSION: As a whole, the epigenetic reprogramming of the TNBC cells is promising for design of therapies based on the use of RARa agonists to induce differentiation of the tumor cells; giving these patients a chance of an alternative or complementary therapy to chemotherapy regimens. Citation Format: Nidhi Bansal, Almudena Bosch, Boris A. Leibovitch, Keely Pierzchalski, Zhou Ming-Ming, Maureen Kane, Samuel Waxman, Eduardo Farias. Targeted epigenetic reprogramming reverts tumor progression in triple-negative breast cancer models by the activation of retinoic acid receptor alpha. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 3511. doi:10.1158/1538-7445.AM2015-3511


Cancer Research | 2014

Abstract 411: Targeted PF1, JARID1B inhibition induces epigenetic reprogramming in triple negative breast cancer

Rossitza Christova; Kevin Petrie; Nidhi Bansal; Boris Leibovitch; Louise Howell; Veronica Gil; Ming-Ming Zhou; Edgardo V. Ariztia; Eduardo F. Farias; Arthur Zelent; Samuel Waxman

Triple Negative Breast cancer (TNBC) is an aggressive subtype of breast cancer associated with early recurrence and poor prognosis. The treatment options are limited due to lack of expression of common drug targets: estrogen receptor (ER), Progesterone receptor (PR) and Epidermal growth factor receptor 2 (Her2). Epigenetic programs can generate aberrant transcription contributing to TNBC progression; however the dynamic and reversible nature of epigenetic changes offers the possibility to reprogram cancer cells to re-express targets that can render TNBC sensitive to targeted therapies like tamoxifen. Envisioning such ‘epidrugs’, we previously published that targeting PAH2 domain of the master transcriptional scaffold Sin3 by stable expression of 13-mer peptide corresponding to a specific motif called SID (mSin3A interaction domain) disrupts its interaction with a small group of SID-containing transcription factors. This interference reverts the expression of important breast cancer-associated genes and impairs tumor growth in vivo. We have now extended our study towards the evaluation of a cell penetrating SID peptide (pSID) in in vitro and in vivo models to establish parameters for the design of targeted epigenetic therapy for TNBC. pSID co-localizes with Sin3A and interference with PAH2-mediated Sin3A functions by pSID is shown by disruption of Sin3A-MAD1 interactions in Co-IP and Duo-Link assays. pSID treatment in MDA-MB 231 cells results in functional re-expression of CDH1 and ER along with increased H3K4 and decreased H3K27 methylation on their promoters. We also show reduction in the tumorsphere formation by pSID-pretreated MDA-MB-231 cells indicating possible epigenetic reprogramming of tumor initiating stem cells towards a differentiated phenotype. Support to this hypothesis is added by the 50% reduction in tumor growth and re-expression of CDH1 observed in FVB mice injected with pSID-pretreated MMTV-myc cells. Moreover, microarray expression analysis indicates pSID-induced EMT reversal, increased cell adhesion and reduced cell migration. Intriguingly, upon further dissection of the mechanism of epigenetic regulation by pSID we show dissociation of two important chromatin readers/modifiers from the Sin3 complex: histone H3K4Me3/2 demethylase JARID1B and H3K4Me0 binding PHD-like domain containing protein PF1; both with significantly correlated overexpression in invasive breast carcinoma. We also observe loss of recruitment of JARID1B but not Sin3A from the CDH1 promoter. Currently studies are underway to understand the cooperative role between JARID1B and PF1 in potentiating the aberrant transcription regulation by Sin3 at important breast cancer-associated promoters that can be selectively reprogrammed by SID decoys. We believe this selectivity can limit the otherwise adverse affects that may be observed by the use of generic HDAC inhibitors and demethylating agents. Citation Format: Rossitza Christova, Kevin Petrie, Nidhi Bansal, Boris Leibovitch, Louise Howell, Veronica Gil, Ming-Ming Zhou, Edgardo Ariztia, Eduardo Farias, Arthur Zelent, Samuel Waxman. Targeted PF1, JARID1B inhibition induces epigenetic reprogramming in triple negative breast cancer. [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 411. doi:10.1158/1538-7445.AM2014-411

Collaboration


Dive into the Boris Leibovitch's collaboration.

Top Co-Authors

Avatar

Samuel Waxman

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Eduardo F. Farias

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ming-Ming Zhou

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Edgardo V. Ariztia

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Nidhi Bansal

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Kevin Petrie

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Rajal Sharma

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Yeon-Jin Kwon

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Rossitza Christova

Institute of Cancer Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge