Network


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

Hotspot


Dive into the research topics where Michael T. McCabe is active.

Publication


Featured researches published by Michael T. McCabe.


Nature | 2012

EZH2 inhibition as a therapeutic strategy for lymphoma with EZH2-activating mutations

Michael T. McCabe; Heidi M. Ott; Gopinath Ganji; Susan Korenchuk; Christine Thompson; Glenn S. Van Aller; Yan Liu; Alan P. Graves; Anthony Della Pietra; Elsie Diaz; Louis V. LaFrance; Mark Mellinger; Celine Duquenne; Xinrong Tian; Ryan G. Kruger; Charles F. McHugh; Martin Brandt; William Henry Miller; Dashyant Dhanak; Sharad K. Verma; Peter J. Tummino; Caretha L. Creasy

In eukaryotes, post-translational modification of histones is critical for regulation of chromatin structure and gene expression. EZH2 is the catalytic subunit of the polycomb repressive complex 2 (PRC2) and is involved in repressing gene expression through methylation of histone H3 on lysine 27 (H3K27). EZH2 overexpression is implicated in tumorigenesis and correlates with poor prognosis in several tumour types. Additionally, somatic heterozygous mutations of Y641 and A677 residues within the catalytic SET domain of EZH2 occur in diffuse large B-cell lymphoma (DLBCL) and follicular lymphoma. The Y641 residue is the most frequently mutated residue, with up to 22% of germinal centre B-cell DLBCL and follicular lymphoma harbouring mutations at this site. These lymphomas have increased H3K27 tri-methylation (H3K27me3) owing to altered substrate preferences of the mutant enzymes. However, it is unknown whether specific, direct inhibition of EZH2 methyltransferase activity will be effective in treating EZH2 mutant lymphomas. Here we demonstrate that GSK126, a potent, highly selective, S-adenosyl-methionine-competitive, small-molecule inhibitor of EZH2 methyltransferase activity, decreases global H3K27me3 levels and reactivates silenced PRC2 target genes. GSK126 effectively inhibits the proliferation of EZH2 mutant DLBCL cell lines and markedly inhibits the growth of EZH2 mutant DLBCL xenografts in mice. Together, these data demonstrate that pharmacological inhibition of EZH2 activity may provide a promising treatment for EZH2 mutant lymphoma.


Cancer Cell | 2013

EZH2 is required for germinal center formation and somatic EZH2 mutations promote lymphoid transformation

Wendy Béguelin; Relja Popovic; Matt Teater; Yanwen Jiang; Karen L. Bunting; Monica Rosen; Hao Shen; Shao Ning Yang; Ling Wang; Teresa Ezponda; Eva Martinez-Garcia; Haikuo Zhang; Sharad K. Verma; Michael T. McCabe; Heidi M. Ott; Glenn S. Van Aller; Ryan G. Kruger; Yan Liu; Charles F. McHugh; David W. Scott; Young Rock Chung; Neil L. Kelleher; Rita Shaknovich; Caretha L. Creasy; Randy D. Gascoyne; Kwok-Kin Wong; Leandro Cerchietti; Ross L. Levine; Omar Abdel-Wahab; Jonathan D. Licht

The EZH2 histone methyltransferase is highly expressed in germinal center (GC) B cells and targeted by somatic mutations in B cell lymphomas. Here, we find that EZH2 deletion or pharmacologic inhibition suppresses GC formation and functions. EZH2 represses proliferation checkpoint genes and helps establish bivalent chromatin domains at key regulatory loci to transiently suppress GC B cell differentiation. Somatic mutations reinforce these physiological effects through enhanced silencing of EZH2 targets. Conditional expression of mutant EZH2 in mice induces GC hyperplasia and accelerated lymphomagenesis in cooperation with BCL2. GC B cell (GCB)-type diffuse large B cell lymphomas (DLBCLs) are mostly addicted to EZH2 but not the more differentiated activated B cell (ABC)-type DLBCLs, thus clarifying the therapeutic scope of EZH2 targeting.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Mutation of A677 in histone methyltransferase EZH2 in human B-cell lymphoma promotes hypertrimethylation of histone H3 on lysine 27 (H3K27)

Michael T. McCabe; Alan P. Graves; Gopinath Ganji; Elsie Diaz; Wendy S. Halsey; Yong Jiang; Kimberly N. Smitheman; Heidi M. Ott; Melissa B. Pappalardi; Kimberly E. Allen; Stephanie Chen; Anthony Della Pietra; Edward Dul; Ashley M. Hughes; Seth Gilbert; Sara H. Thrall; Peter J. Tummino; Ryan G. Kruger; Martin Brandt; Benjamin J. Schwartz; Caretha L. Creasy

Trimethylation of histone H3 on lysine 27 (H3K27me3) is a repressive posttranslational modification mediated by the histone methyltransferase EZH2. EZH2 is a component of the polycomb repressive complex 2 and is overexpressed in many cancers. In B-cell lymphomas, its substrate preference is frequently altered through somatic mutation of the EZH2 Y641 residue. Herein, we identify mutation of EZH2 A677 to a glycine (A677G) among lymphoma cell lines and primary tumor specimens. Similar to Y641 mutant cell lines, an A677G mutant cell line revealed aberrantly elevated H3K27me3 and decreased monomethylated H3K27 (H3K27me1) and dimethylated H3K27 (H3K27me2). A677G EZH2 possessed catalytic activity with a substrate specificity that was distinct from those of both WT EZH2 and Y641 mutants. Whereas WT EZH2 displayed a preference for substrates with less methylation [unmethylated H3K27 (H3K27me0):me1:me2 kcat/Km ratio = 9:6:1] and Y641 mutants preferred substrates with greater methylation (H3K27me0:me1:me2 kcat/Km ratio = 1:2:13), the A677G EZH2 demonstrated nearly equal efficiency for all three substrates (H3K27me0:me1:me2 kcat/Km ratio = 1.1:0.6:1). When transiently expressed in cells, A677G EZH2, but not WT EZH2, increased global H3K27me3 and decreased H3K27me2. Structural modeling of WT and mutant EZH2 suggested that the A677G mutation acquires the ability to methylate H3K27me2 through enlargement of the lysine tunnel while preserving activity with H3K27me0/me1 substrates through retention of the Y641 residue that is crucial for orientation of these smaller substrates. This mutation highlights the interplay between Y641 and A677 residues in the substrate specificity of EZH2 and identifies another lymphoma patient population that harbors an activating mutation of EZH2.


ACS Medicinal Chemistry Letters | 2012

Identification of Potent, Selective, Cell-Active Inhibitors of the Histone Lysine Methyltransferase EZH2.

Sharad K. Verma; Xinrong Tian; Louis Vincent Lafrance; Celine Duquenne; Dominic Suarez; Kenneth A. Newlander; Stuart P. Romeril; Joelle L. Burgess; Seth W. Grant; James Brackley; Alan P. Graves; Daryl Scherzer; Art Shu; Christine Thompson; Heidi M. Ott; Glenn S. Van Aller; Carl A. Machutta; Elsie Diaz; Yong Jiang; Neil W. Johnson; Steven David Knight; Ryan G. Kruger; Michael T. McCabe; Dashyant Dhanak; Peter J. Tummino; Caretha L. Creasy; William H. Miller

The histone H3-lysine 27 (H3K27) methyltransferase EZH2 plays a critical role in regulating gene expression, and its aberrant activity is linked to the onset and progression of cancer. As part of a drug discovery program targeting EZH2, we have identified highly potent, selective, SAM-competitive, and cell-active EZH2 inhibitors, including GSK926 (3) and GSK343 (6). These compounds are small molecule chemical tools that would be useful to further explore the biology of EZH2.


Nature | 2016

Reductive carboxylation supports redox homeostasis during anchorage-independent growth

Lei Jiang; Alexander A. Shestov; Pamela Swain; Chendong Yang; Seth J. Parker; Qiong A. Wang; Lance S. Terada; Nicholas D. Adams; Michael T. McCabe; Beth Pietrak; Stan Schmidt; Christian M. Metallo; Brian P. Dranka; Benjamin Schwartz; Ralph J. DeBerardinis

Cells receive growth and survival stimuli through their attachment to an extracellular matrix (ECM). Overcoming the addiction to ECM-induced signals is required for anchorage-independent growth, a property of most malignant cells. Detachment from ECM is associated with enhanced production of reactive oxygen species (ROS) owing to altered glucose metabolism. Here we identify an unconventional pathway that supports redox homeostasis and growth during adaptation to anchorage independence. We observed that detachment from monolayer culture and growth as anchorage-independent tumour spheroids was accompanied by changes in both glucose and glutamine metabolism. Specifically, oxidation of both nutrients was suppressed in spheroids, whereas reductive formation of citrate from glutamine was enhanced. Reductive glutamine metabolism was highly dependent on cytosolic isocitrate dehydrogenase-1 (IDH1), because the activity was suppressed in cells homozygous null for IDH1 or treated with an IDH1 inhibitor. This activity occurred in absence of hypoxia, a well-known inducer of reductive metabolism. Rather, IDH1 mitigated mitochondrial ROS in spheroids, and suppressing IDH1 reduced spheroid growth through a mechanism requiring mitochondrial ROS. Isotope tracing revealed that in spheroids, isocitrate/citrate produced reductively in the cytosol could enter the mitochondria and participate in oxidative metabolism, including oxidation by IDH2. This generates NADPH in the mitochondria, enabling cells to mitigate mitochondrial ROS and maximize growth. Neither IDH1 nor IDH2 was necessary for monolayer growth, but deleting either one enhanced mitochondrial ROS and reduced spheroid size, as did deletion of the mitochondrial citrate transporter protein. Together, the data indicate that adaptation to anchorage independence requires a fundamental change in citrate metabolism, initiated by IDH1-dependent reductive carboxylation and culminating in suppression of mitochondrial ROS.


Nature Communications | 2015

The epigenetic modifier EZH2 controls melanoma growth and metastasis through silencing of distinct tumour suppressors

Daniel Zingg; Julien Debbache; Simon M. Schaefer; Eylul Tuncer; Sandra C Frommel; Phil F. Cheng; Natalia Arenas-Ramirez; Jessica Haeusel; Yudong Zhang; Mario Bonalli; Michael T. McCabe; Caretha L. Creasy; Mitchell P. Levesque; Onur Boyman; Raffaella Santoro; Olga Shakhova; Reinhard Dummer; Lukas Sommer

Increased activity of the epigenetic modifier EZH2 has been associated with different cancers. However, evidence for a functional role of EZH2 in tumorigenesis in vivo remains poor, in particular in metastasizing solid cancers. Here we reveal central roles of EZH2 in promoting growth and metastasis of cutaneous melanoma. In a melanoma mouse model, conditional Ezh2 ablation as much as treatment with the preclinical EZH2 inhibitor GSK503 stabilizes the disease through inhibition of growth and virtually abolishes metastases formation without affecting normal melanocyte biology. Comparably, in human melanoma cells, EZH2 inactivation impairs proliferation and invasiveness, accompanied by re-expression of tumour suppressors connected to increased patient survival. These EZH2 target genes suppress either melanoma growth or metastasis in vivo, revealing the dual function of EZH2 in promoting tumour progression. Thus, EZH2-mediated epigenetic repression is highly relevant especially during advanced melanoma progression, which makes EZH2 a promising target for novel melanoma therapies.


PLOS ONE | 2013

BET Inhibition Silences Expression of MYCN and BCL2 and Induces Cytotoxicity in Neuroblastoma Tumor Models

Anastasia Wyce; Gopinath Ganji; Kimberly N. Smitheman; Chun-wa Chung; Susan Korenchuk; Yuchen Bai; Olena Barbash; BaoChau Le; Peter D. Craggs; Michael T. McCabe; Karen M. Kennedy-Wilson; Lydia V. Sanchez; Romain Luc Marie Gosmini; Nigel James Parr; Charles F. McHugh; Dashyant Dhanak; Rab K. Prinjha; Kurt R. Auger; Peter J. Tummino

BET family proteins are epigenetic regulators known to control expression of genes involved in cell growth and oncogenesis. Selective inhibitors of BET proteins exhibit potent anti-proliferative activity in a number of hematologic cancer models, in part through suppression of the MYC oncogene and downstream Myc-driven pathways. However, little is currently known about the activity of BET inhibitors in solid tumor models, and whether down-regulation of MYC family genes contributes to sensitivity. Here we provide evidence for potent BET inhibitor activity in neuroblastoma, a pediatric solid tumor associated with a high frequency of MYCN amplifications. We treated a panel of neuroblastoma cell lines with a novel small molecule inhibitor of BET proteins, GSK1324726A (I-BET726), and observed potent growth inhibition and cytotoxicity in most cell lines irrespective of MYCN copy number or expression level. Gene expression analyses in neuroblastoma cell lines suggest a role of BET inhibition in apoptosis, signaling, and N-Myc-driven pathways, including the direct suppression of BCL2 and MYCN. Reversal of MYCN or BCL2 suppression reduces the potency of I-BET726-induced cytotoxicity in a cell line-specific manner; however, neither factor fully accounts for I-BET726 sensitivity. Oral administration of I-BET726 to mouse xenograft models of human neuroblastoma results in tumor growth inhibition and down-regulation MYCN and BCL2 expression, suggesting a potential role for these genes in tumor growth. Taken together, our data highlight the potential of BET inhibitors as novel therapeutics for neuroblastoma, and suggest that sensitivity is driven by pleiotropic effects on cell growth and apoptotic pathways in a context-specific manner.


Nature Chemical Biology | 2015

New IDH1 mutant inhibitors for treatment of acute myeloid leukemia

Ujunwa C. Okoye-Okafor; Boris Bartholdy; Jessy Cartier; Enoch Gao; Beth Pietrak; Alan R. Rendina; Cynthia M. Rominger; Chad Quinn; Angela Smallwood; Kenneth Wiggall; Alexander Joseph Reif; Stanley J. Schmidt; Hongwei Qi; Huizhen Zhao; Gerard Joberty; Maria Faelth-Savitski; Marcus Bantscheff; Gerard Drewes; Chaya Duraiswami; Pat Brady; Arthur Groy; Swathi Rao Narayanagari; Iléana Antony-Debré; Kelly Mitchell; Heng Rui Wang; Yun Ruei Kao; Maximilian Christopeit; Luis Carvajal; Laura Barreyro; Elisabeth Paietta

Neomorphic mutations in isocitrate dehydrogenase 1 (IDH1) are driver mutations in acute myeloid leukemia (AML) and other cancers. We report the development of new allosteric inhibitors of mutant IDH1. Crystallographic and biochemical results demonstrated that compounds of this chemical series bind to an allosteric site and lock the enzyme in a catalytically inactive conformation, thereby enabling inhibition of different clinically relevant IDH1 mutants. Treatment of IDH1 mutant primary AML cells uniformly led to a decrease in intracellular 2-HG, abrogation of the myeloid differentiation block and induction of granulocytic differentiation at the level of leukemic blasts and more immature stem-like cells, in vitro and in vivo. Molecularly, treatment with the inhibitors led to a reversal of the DNA cytosine hypermethylation patterns caused by mutant IDH1 in the cells of individuals with AML. Our study provides proof of concept for the molecular and biological activity of novel allosteric inhibitors for targeting different mutant forms of IDH1 in leukemia.


Epigenomics | 2014

EZH2 as a potential target in cancer therapy

Michael T. McCabe; Caretha L. Creasy

Over the last several years, dysregulation of epigenetic mechanisms including DNA and histone methylation has been recognized as a hallmark of cancer. Alterations of epigenetic regulators themselves, including the histone lysine methyltransferase EZH2, have been reported in numerous cancer types. With the discovery of small molecule inhibitors of EZH2, we can now begin to evaluate EZH2 as a therapeutic target in cancer. This article will provide an overview of the dysregulation of EZH2 in cancer, possible mechanisms for inhibition of EZH2 activity, and the preclinical activity of currently available EZH2 inhibitors.


Journal of Biomolecular Screening | 2012

Development and Validation of Reagents and Assays for EZH2 Peptide and Nucleosome High-Throughput Screens

Elsie Diaz; Carl A. Machutta; Stephanie Chen; Yong Jiang; Christopher J. Nixon; Glenn A. Hofmann; Danielle Key; Sharon Sweitzer; Mehul Patel; Zining Wu; Caretha L. Creasy; Ryan G. Kruger; Louis V. LaFrance; Sharad K. Verma; Melissa B. Pappalardi; BaoChau Le; Glenn S. Van Aller; Michael T. McCabe; Peter J. Tummino; Andrew J. Pope; Sara H. Thrall; Benjamin Schwartz; Martin Brandt

Histone methyltransferases (HMT) catalyze the methylation of histone tail lysines, resulting in changes in gene transcription. Misregulation of these enzymes has been associated with various forms of cancer, making this target class a potential new area for the development of novel chemotherapeutics. EZH2 is the catalytic component of the polycomb group repressive complex (PRC2), which selectively methylates histone H3 lysine 27 (H3K27). EZH2 is overexpressed in prostate, breast, bladder, brain, and other tumor types and is recognized as a molecular marker for cancer progression and aggressiveness. Several new reagents and assays were developed to aid in the identification of EZH2 inhibitors, and these were used to execute two high-throughput screening campaigns. Activity assays using either an H3K27 peptide or nucleosomes as substrates for methylation are described. The strategy to screen EZH2 with either a surrogate peptide or a natural substrate led to the identification of the same tractable series. Compounds from this series are reversible, are [3H]-S-adenosyl-L-methionine competitive, and display biochemical inhibition of H3K27 methylation.

Collaboration


Dive into the Michael T. McCabe'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