Network


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

Hotspot


Dive into the research topics where Peter J. Tummino is active.

Publication


Featured researches published by Peter J. Tummino.


Nature | 1999

Genomic-sequence comparison of two unrelated isolates of the human gastric pathogen Helicobacter pylori

Richard A. Alm; Lo-See L. Ling; Donald T. Moir; Benjamin L. King; Eric D. Brown; Peter Doig; Douglas R. Smith; Brian Noonan; Braydon C. Guild; Boudewijn L. deJonge; Gilles Carmel; Peter J. Tummino; Anthony Caruso; Maria Uria-Nickelsen; Debra M. Mills; Cameron Ives; Rene Gibson; David Merberg; Scott D. Mills; Qin Jiang; Diane E. Taylor; Gerald F. Vovis; Trevor J. Trust

Helicobacter pylori, one of the most common bacterial pathogens of humans, colonizes the gastric mucosa, where it appears to persist throughout the hosts life unless the patient is treated. Colonization induces chronic gastric inflammation which can progress to a variety of diseases, ranging in severity from superficial gastritis and peptic ulcer to gastric cancer and mucosal-associated lymphoma. Strain-specific genetic diversity has been proposed to be involved in the organisms ability to cause different diseases or even be beneficial to the infected host, and to participate in the lifelong chronicity of infection. Here we compare the complete genomic sequences of two unrelated H. pylori isolates. This is, to our knowledge, the first such genomic comparison. H. pylori was believed to exhibit a large degree of genomic and allelic diversity, but we find that the overall genomic organization, gene order and predicted proteomes (sets of proteins encoded by the genomes) of the two strains are quite similar. Between 6 to 7% of the genes are specific to each strain, with almost half of these genes being clustered in a single hypervariable region.


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.


Biochemistry | 2008

Residence Time of Receptor-Ligand Complexes and Its Effect on Biological Function

Peter J. Tummino; Robert A. Copeland

The formation and duration of binary receptor-ligand complexes are fundamental to many physiologic processes. Most often, the effectiveness of interaction between a receptor and its ligand is quantified in terms of closed system, equilibrium affinity measurements, such as IC50 and Kd. In the context of in vivo biology, however, the extent and duration of responses to receptor-ligand interactions depend greatly on the time period over which the ligand is in residence on its receptor. Here we define receptor-ligand complex residence time in quantitative terms and describe its significance to biological function. Examples of the importance of residence time are presented for natural ligands of different receptor types. The impact of residence time on the optimization of potential ligands as drugs for human medicine is also described.


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 | 2010

Discovery of GSK2126458, a Highly Potent Inhibitor of PI3K and the Mammalian Target of Rapamycin.

Steven David Knight; Nicholas D. Adams; Joelle L. Burgess; Amita M. Chaudhari; Michael G. Darcy; Carla A. Donatelli; Juan I. Luengo; Ken A. Newlander; Cynthia A. Parrish; Lance H. Ridgers; Martha A. Sarpong; Stanley J. Schmidt; Glenn S. Van Aller; Jeffrey D. Carson; Melody Diamond; Patricia A. Elkins; Christine M. Gardiner; Eric Garver; Seth Gilbert; Richard R. Gontarek; Jeffrey R. Jackson; Kevin L. Kershner; Lusong Luo; Kaushik Raha; Christian S. Sherk; Chiu-Mei Sung; David Sutton; Peter J. Tummino; Ronald Wegrzyn; Kurt R. Auger

Phosphoinositide 3-kinase α (PI3Kα) is a critical regulator of cell growth and transformation, and its signaling pathway is the most commonly mutated pathway in human cancers. The mammalian target of rapamycin (mTOR), a class IV PI3K protein kinase, is also a central regulator of cell growth, and mTOR inhibitors are believed to augment the antiproliferative efficacy of PI3K/AKT pathway inhibition. 2,4-Difluoro-N-{2-(methyloxy)-5-[4-(4-pyridazinyl)-6-quinolinyl]-3-pyridinyl}benzenesulfonamide (GSK2126458, 1) has been identified as a highly potent, orally bioavailable inhibitor of PI3Kα and mTOR with in vivo activity in both pharmacodynamic and tumor growth efficacy models. Compound 1 is currently being evaluated in human clinical trials for the treatment of cancer.


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.


Cancer Cell | 2015

A DNA Hypomethylation Signature Predicts Antitumor Activity of LSD1 Inhibitors in SCLC

Helai P. Mohammad; Kimberly N. Smitheman; Chandrashekhar D. Kamat; David Soong; Kelly Federowicz; Glenn S. Van Aller; Jess Schneck; Jeffrey D. Carson; Yan Liu; Michael Butticello; William G. Bonnette; Shelby A. Gorman; Yan Degenhardt; Yuchen Bai; Michael T. McCabe; Melissa B. Pappalardi; Jiri Kasparec; Xinrong Tian; Kenneth C. McNulty; Meagan B. Rouse; Patrick McDevitt; Thau Ho; Michelle Crouthamel; Timothy K. Hart; Nestor O. Concha; Charles F. McHugh; William Henry Miller; Dashyant Dhanak; Peter J. Tummino; Christopher Carpenter

Epigenetic dysregulation has emerged as an important mechanism in cancer. Alterations in epigenetic machinery have become a major focus for targeted therapies. The current report describes the discoveryxa0and biological activity of a cyclopropylamine containing inhibitor of Lysine Demethylase 1 (LSD1), GSK2879552. This small molecule is a potent, selective, orally bioavailable, mechanism-based irreversible inactivator of LSD1. A proliferation screen of cell lines representing a number of tumor types indicated that small cell lung carcinoma (SCLC) is sensitive to LSD1 inhibition. The subset of SCLC lines and primary samples that undergo growth inhibition in response to GSK2879552 exhibit DNA hypomethylation of a signature set of probes, suggesting this may be used as a predictive biomarker of activity.


Nature | 2014

Smyd3 links lysine methylation of map3k2 to ras-driven cancer

Pawel K. Mazur; Nicolas Reynoird; Purvesh Khatri; Pascal W. T. C. Jansen; Alex W. Wilkinson; Shichong Liu; Olena Barbash; Glenn S. Van Aller; Michael Huddleston; Dashyant Dhanak; Peter J. Tummino; Ryan G. Kruger; Benjamin A. Garcia; Atul J. Butte; Michiel Vermeulen; Julien Sage; Or Gozani

Deregulation of lysine methylation signalling has emerged as a common aetiological factor in cancer pathogenesis, with inhibitors of several histone lysine methyltransferases (KMTs) being developed as chemotherapeutics. The largely cytoplasmic KMT SMYD3 (SET and MYND domain containing protein 3) is overexpressed in numerous human tumours. However, the molecular mechanism by which SMYD3 regulates cancer pathways and its relationship to tumorigenesis in vivo are largely unknown. Here we show that methylation of MAP3K2 by SMYD3 increases MAP kinase signalling and promotes the formation of Ras-driven carcinomas. Using mouse models for pancreatic ductal adenocarcinoma and lung adenocarcinoma, we found that abrogating SMYD3 catalytic activity inhibits tumour development in response to oncogenic Ras. We used protein array technology to identify the MAP3K2 kinase as a target of SMYD3. In cancer cell lines, SMYD3-mediated methylation of MAP3K2 at lysine 260 potentiates activation of the Ras/Raf/MEK/ERK signalling module and SMYD3 depletion synergizes with a MEK inhibitor to block Ras-driven tumorigenesis. Finally, the PP2A phosphatase complex, a key negative regulator of the MAP kinase pathway, binds to MAP3K2 and this interaction is blocked by methylation. Together, our results elucidate a new role for lysine methylation in integrating cytoplasmic kinase-signalling cascades and establish a pivotal role for SMYD3 in the regulation of oncogenic Ras signalling.


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.


Biochemical Journal | 2008

Effects of oncogenic p110α subunit mutations on the lipid kinase activity of phosphoinositide 3-kinase

Jeffrey D. Carson; Glenn S. Van Aller; Ruth Lehr; Robert H. Sinnamon; Robert B. Kirkpatrick; Kurt R. Auger; Dashyant Dhanak; Robert A. Copeland; Richard R. Gontarek; Peter J. Tummino; Lusong Luo

The PIK3CA gene, encoding the p110α catalytic subunit of Class IA PI3Ks (phosphoinositide 3-kinases), is frequently mutated in many human tumours. The three most common tumour-derived alleles of p110α, H1047R, E542K and E545K, were shown to potently activate PI3K signalling in human epithelial cells. In the present study, we examine the biochemical activity of the recombinantly purified PI3K oncogenic mutants. The kinetic characterizations of the wt (wild-type) and the three ‘hot spot’ PI3K mutants show that the mutants all have approx. 2-fold increase in lipid kinase activities. Interestingly, the phosphorylated IRS-1 (insulin receptor substrate-1) protein shows activation of the lipid kinase activity for the wt and H1047R but not E542K and E545K PI3Kα, suggesting that these mutations represent different mechanisms of lipid kinase activation and hence transforming activity in cancer cells.

Collaboration


Dive into the Peter J. Tummino's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Olena Barbash

University of New Mexico

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge