Network


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

Hotspot


Dive into the research topics where Gregory David is active.

Publication


Featured researches published by Gregory David.


Molecular and Cellular Biology | 2011

A Novel Mammalian Complex Containing Sin3B Mitigates Histone Acetylation and RNA Polymerase II Progression within Transcribed Loci

Petar Jelinic; Jessica Pellegrino; Gregory David

ABSTRACT Transcription requires the progression of RNA polymerase II (RNAP II) through a permissive chromatin structure. Recent studies of Saccharomyces cerevisiae have demonstrated that the yeast Sin3 protein contributes to the restoration of the repressed chromatin structure at actively transcribed loci. Yet, the mechanisms underlying the restoration of the repressive chromatin structure at transcribed loci and its significance in gene expression have not been investigated in mammals. We report here the identification of a mammalian complex containing the corepressor Sin3B, the histone deacetylase HDAC1, Mrg15, and the PHD finger-containing Pf1 and show that this complex plays important roles in regulation of transcription. We demonstrate that this complex localizes at discrete loci approximately 1 kb downstream of the transcription start site of transcribed genes, and this localization requires both Pf1s and Mrg15s interaction with chromatin. Inactivation of this mammalian complex promotes increased RNAP II progression within transcribed regions and subsequent increased transcription. Our results define a novel mammalian complex that contributes to the regulation of transcription and point to divergent uses of the Sin3 protein homologues throughout evolution in the modulation of transcription.


Nature Cell Biology | 2007

The F-box protein Fbl10 is a novel transcriptional repressor of c-Jun.

Ryo Koyama-Nasu; Gregory David; Naoko Tanese

c-Jun is a component of the heterodimeric transcription factor AP-1 that is rapidly activated in response to ultraviolet light (UV). In unstressed cells, c-Jun activity is negatively regulated by transcriptional repressor complexes. Here we show that the F-box protein Fbl10/JHDM1B interacts with c-Jun and represses c-Jun-mediated transcription. Chromatin-immunoprecipitation assays demonstrate that Fbl10 is present at the c-jun promoter, and that c-Jun is required for the recruitment of Fbl10. Fbl10 binds to the unmethylated CpG sequences in the c-jun promoter through the CxxC zinc finger and tethers transcriptional repressor complexes. Suppression of Fbl10 expression by RNA interference (RNAi) induces transcription of c-jun and other c-Jun-target genes, and causes an aberrant cell-cycle progression and increased UV-induced cell death. Furthermore, Fbl10 protein and messenger RNA are downregulated in response to UV in an inverse correlation with c-Jun. Taken together, our results demonstrate that Fbl10 is a key regulator of c-Jun function.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Inhibition of androgen receptor and β-catenin activity in prostate cancer

Eugine Lee; Aviv Madar; Gregory David; Michael J. Garabedian; Ramanuj DasGupta; Susan K. Logan

Androgen receptor (AR) is the major therapeutic target in aggressive prostate cancer. However, targeting AR alone can result in drug resistance and disease recurrence. Therefore, simultaneous targeting of multiple pathways could in principle be an effective approach to treating prostate cancer. Here we provide proof-of-concept that a small-molecule inhibitor of nuclear β-catenin activity (called C3) can inhibit both the AR and β-catenin–signaling pathways that are often misregulated in prostate cancer. Treatment with C3 ablated prostate cancer cell growth by disruption of both β-catenin/T-cell factor and β-catenin/AR protein interaction, reflecting the fact that T-cell factor and AR have overlapping binding sites on β-catenin. Given that AR interacts with, and is transcriptionally regulated by β-catenin, C3 treatment also resulted in decreased occupancy of β-catenin on the AR promoter and diminished AR and AR/β-catenin target gene expression. Interestingly, C3 treatment resulted in decreased AR binding to target genes accompanied by decreased recruitment of an AR and β-catenin cofactor, coactivator-associated arginine methyltransferase 1 (CARM1), providing insight into the unrecognized function of β-catenin in prostate cancer. Importantly, C3 inhibited tumor growth in an in vivo xenograft model and blocked renewal of bicalutamide-resistant sphere-forming cells, indicating the therapeutic potential of this approach.


Current Cancer Drug Targets | 2010

Ras-Induced Senescence and its Physiological Relevance in Cancer

Teresa DiMauro; Gregory David

Activated oncogenes like Ras have traditionally been thought as promoting unrestrained proliferation; therefore, the concept of oncogene-induced senescence has been, and still is, controversial. The counter-intuitive notion that activation of oncogenes leads to the prevention of cellular proliferation has initially been fueled by in vitro studies using ectopic expression of activated Ras in primary fibroblasts. While these initial studies demonstrated unambiguously the existence of a new type of cellular senescence, induced by oncogenes in an ex-vivo system, questions were raised about the physiological relevance of this process. Indeed, recent technical advances in mouse modeling for cancer have suggested that the occurrence of Ras-induced senescence is highly dependent on the cellular context, as well as the level of expression of activated Ras, and may not be pertinent to the study of human cancer initiation and/or progression. However, our increased knowledge of the molecular basis for cellular senescence has led to a better understanding of the molecular events modulating cancer progression in vivo. Recent studies have not only clearly established the incidence of cellular senescence in pre-neoplasic lesions, but also its role as a potential tumor-suppressor mechanism in vivo. Here, we review the recent and exciting new findings regarding the physiological relevance of Ras-induced senescence, and discuss their implications in terms of cancer therapy.


Cancer Research | 2009

Sin3B Expression Is Required for Cellular Senescence and Is Up-regulated upon Oncogenic Stress

Kathryn B. Grandinetti; Petar Jelinic; Teresa DiMauro; Jessica Pellegrino; Ruben Rodriguez; Patricia M. Finnerty; Rachel Ruoff; Nabeel Bardeesy; Susan K. Logan; Gregory David

Serial passage of primary mammalian cells or strong mitogenic signals induce a permanent exit from the cell cycle called senescence. A characteristic of senescent cells is the heterochromatinization of loci encoding pro-proliferative genes, leading to their transcriptional silencing. Senescence is thought to represent a defense mechanism against uncontrolled proliferation and cancer. Consequently, genetic alterations that allow senescence bypass are associated with susceptibility to oncogenic transformation. We show that fibroblasts genetically inactivated for the chromatin-associated Sin3B protein are refractory to replicative and oncogene-induced senescence. Conversely, overexpression of Sin3B triggers senescence and the formation of senescence-associated heterochromatic foci. Although Sin3B is strongly up-regulated upon oncogenic stress, decrease in expression of Sin3B is associated with tumor progression in vivo, suggesting that expression of Sin3B may represent a barrier against transformation. Together, these results underscore the contribution of senescence in tumor suppression and suggest that expression of chromatin modifiers is modulated at specific stages of cellular transformation. Consequently, these findings suggest that modulation of Sin3B-associated activities may represent new therapeutic opportunities for treatment of cancers.


Cell Cycle | 2008

Sin3B: an essential regulator of chromatin modifications at E2F target promoters during cell cycle withdrawal.

Kathryn B. Grandinetti; Gregory David

Efficient and accurate cell cycle exit is intimately linked to cellular differentiation, and by inference, to the prevention of tumorigenesis. Perhaps the most important axis of control for this process involves the interactions of the E2F family of DNA binding proteins with the retinoblastoma (Rb) and Rb-related “pocket protein” (p107 and p130) family of tumor suppressors. Not surprisingly, alterations in this pathway are present in a large number of human malignancies. The molecular basis for the controls exercised by the Rb family of proteins has been widely investigated, but is still not completely understood. Elegant in vitro studies had previously suggested the participation of histone deacetylase (HDAC)-associated Sin3B in E2F-mediated repression. Using genetically modified mice, we have recently uncovered a role for the Sin3B protein as a specific and essential actor in promoting cell cycle exit via the E2F-Rb pathway. We demonstrated its absolute requirement not only for cell cycle exit in vitro and in vivo, but also for biological processes linked to cellular differentiation. These observations strongly suggest that Sin3B plays an essential role in coordinating the chromatin modifying activities required for the transient repression of pro-proliferation genes in quiescence, as well as stable silencing of these genes upon terminal differentiation.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Structural insights into the assembly of the histone deacetylase-associated Sin3L/Rpd3L corepressor complex

Michael David Clark; Ryan Marcum; Richard Graveline; Clarence W. Chan; Tao Xie; Zhonglei Chen; Yujia Ding; Yongbo Zhang; Alfonso Mondragón; Gregory David; Ishwar Radhakrishnan

Significance Gene transcription in eukaryotes is regulated by enzymes that posttranslationally add or remove acetyl groups from histones and render the underlying DNA more or less accessible to the transcription machinery. How histone deacetylases (HDACs), the enzymes responsible for deacetylation that are commonly found in multiprotein complexes, are assembled and targeted to their sites of action to affect transcription repression is largely unknown. We show biochemically and structurally how two key subunits of a conserved HDAC complex recruit multiple copies of HDACs into the complex in a manner that allows the enzymes to explore a large conformational space when the complex is targeted to specific genomic loci. This complex seems to be tailored for efficient deacetylation of nucleosomes that are situated far apart. Acetylation is correlated with chromatin decondensation and transcriptional activation, but its regulation by histone deacetylase (HDAC)-bearing corepressor complexes is poorly understood. Here, we describe the mechanism of assembly of the mammalian Sin3L/Rpd3L complex facilitated by Sds3, a conserved subunit deemed critical for proper assembly. Sds3 engages a globular, helical region of the HDAC interaction domain (HID) of the scaffolding protein Sin3A through a bipartite motif comprising a helix and an adjacent extended segment. Sds3 dimerizes through not only one of the predicted coiled-coil motifs but also, the segment preceding it, forming an ∼150-Å-long antiparallel dimer. Contrary to previous findings in yeast, Sin3A rather than Sds3 functions in recruiting HDAC1 into the complex by engaging the latter through a highly conserved segment adjacent to the helical HID subdomain. In the resulting model for the ternary complex, the two copies of the HDACs are situated distally and dynamically because of a natively unstructured linker connecting the dimerization domain and the Sin3A interaction domain of Sds3; these features contrast with the static organization described previously for the NuRD (nucleosome remodeling and deacetylase) complex. The Sds3 linker features several conserved basic residues that could potentially maintain the complex on chromatin by nonspecific interactions with DNA after initial recruitment by sequence-specific DNA-binding repressors.


Oncogene | 2015

Transcriptional repression of Sin3B by Bmi-1 prevents cellular senescence and is relieved by oncogene activation

Teresa DiMauro; David J. Cantor; Anthony J. Bainor; Gregory David

The Polycomb group protein Bmi-1 is an essential regulator of cellular senescence and is believed to function largely through the direct repression of the Ink4a/Arf locus. However, concurrent deletion of Ink4a/Arf does not fully rescue the defects detected in Bmi-1−/− mice, indicating that additional Bmi-1 targets remain to be identified. The expression of the chromatin-associated Sin3B protein is stimulated by oncogenic stress, and is required for oncogene-induced senescence. Here we demonstrate that oncogenic stress leads to the dissociation of Bmi-1 from the Sin3B locus, resulting in increased Sin3B expression and subsequent entry into cellular senescence. Furthermore, Sin3B is required for the senescent phenotype and elevated levels of reactive oxygen species elicited upon Bmi-1 depletion. Altogether, these results identify Sin3B as a novel direct target of Bmi-1, and establish Bmi-1-driven repression of Sin3B as an essential regulator of cellular senescence.


Blood | 2017

The chromatin-associated Sin3B protein is required for hematopoietic stem cell functions in mice

David J. Cantor; Gregory David

Hematopoietic stem cells (HSCs) reside at the top of the hematopoietic hierarchy and are the origin of all blood cells produced throughout an individuals life. The balance between HSC self-renewal and differentiation is maintained by various intrinsic and extrinsic mechanisms. Among these, the molecular pathways that restrict cell cycle progression are critical to the maintenance of functional HSCs. Alterations in the regulation of cell cycle progression in HSCs invariably lead to the development of hematologic malignancies or bone marrow failure syndromes. Here we report that hematopoietic-specific genetic inactivation of Sin3B, an essential component of the mammalian Sin3-histone deacetylase corepressor complex, severely impairs the competitive repopulation capacity of HSCs. Sin3B-deleted HSCs accumulate and fail to properly differentiate following transplantation. Moreover, Sin3B inactivation impairs HSC quiescence and sensitizes mice to myelosuppressive therapy. Together, these results identify Sin3B as a novel and critical regulator of HSC functions.


Methods of Molecular Biology | 2017

Senescence Phenotypes Induced by Ras in Primary Cells

Lena Lau; Gregory David

Cellular senescence is defined as a state of stable cell-cycle arrest that is distinct from quiescence and terminal differentiation. Many stimuli can induce senescence, including telomere shortening and oncogene activation. The phenotypes elicited by pro-senescent signals can be heterogeneous depending on the stimulus and the cell type affected. To date, there is not a definitive marker that can ubiquitously and specifically mark all senescent cells. Therefore, several independent markers must be utilized to ascertain the senescent state of a cell or group of cells. Here, we describe common assays used to assess oncogenic Ras-induced senescence.

Collaboration


Dive into the Gregory David's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge