35 results on '"Patrick McDevitt"'
Search Results
2. How Strongly Do Physical Examination Estimates and Ultrasonographic Measurements of Liver Size Correlate? A Prospective Study
- Author
-
Michael A. Bruno, Patrick McDevitt, Ankit Patel, Justin Loloi, and Thomas Riley
- Subjects
medicine.diagnostic_test ,business.industry ,Liver span ,Ultrasound ,Physical examination ,General Medicine ,030204 cardiovascular system & hematology ,medicine.disease ,03 medical and health sciences ,Liver disease ,0302 clinical medicine ,MIdclavicular line ,Liver biopsy ,Medicine ,030212 general & internal medicine ,business ,Nuclear medicine ,Prospective cohort study ,Liver size - Abstract
Background Liver size assessed by physical examination and ultrasound has long been used to gain useful clinical information. The size measurements obtained by these modalities have been difficult to compare as they are measured in 2 different axes (transaxial vs midclavicular). Our objective was to identify a measurement correlation between ultrasound and physical examination liver size findings. We aimed to develop a correction factor whereby the liver size could be translated between the measured transaxial size obtained by ultrasound and physical examination size when measured in the midclavicular line. Methods We conducted a prospective study including 101 adult patients with liver disease undergoing liver biopsy between April 2008 and November 2008 at Penn State Health Milton S Hershey Medical Center. Liver measurements were obtained by physical examination and ultrasound, which were performed by a single hepatologist. Results The average physical examination size using the midclavicular approach was 8.9 cm ± 1.13. On ultrasound, the average transaxial measurement was 14.3 cm ± 1.6. A ratio was made between measurements from the midclavicular line physical examination size and transaxial ultrasound size, and found to have a mean correction factor of 1.6 ± 0.14. The correction factor was applied to the physical examination-determined liver size and compared with ultrasound findings, with 76% of values (77/101) falling within 10% of the ultrasound-determined liver size. Conclusion This study proves that a strong correlation exists between physical examination estimates of liver size and the measured size on ultrasonography. Multiplying the percussed liver span by a correction factor consistently yields accurate predictions of the transaxial liver span.
- Published
- 2019
- Full Text
- View/download PDF
3. Anchoring Cultural Change and Organizational Change: Case Study Research Evaluation Project All Hallows College Dublin 1995-2015
- Author
-
Patrick McDevitt and Patrick McDevitt
- Subjects
- Organizational change--Ireland--Dublin, Universities and colleges--Ireland--Dublin--Administration
- Abstract
This book describes the organizational processes and changes coupled with leadership changes over three distinct eras from 1995-2015. It illustrates the challenges the college faced, and the actions taken to resolve issues and make changes. The successes, and the barriers encountered as the organization worked toward solutions to the many interrelated and confounding social and financial issues with which the college was facing, are also described. In the book, John Kotter's Steps of Organizational change and culture is the theoretical context in the analysis of data. Kotter stresses the point that in Organizational change the “Culture” must be anchored in order for change to take place successfully. Kotter understands “Culture” as the Organization's Identity and the Organization's attitude for “Change”. The concept of “Culture” also includes how “Identity” and “Change” interrelate to one another. Unfortunately, this “anchoring of culture” does not often happen in many organizations which leads to failure and the dying of Organizations. In general, Kotter's theory is typically used in for-profit organizations, whereas the All Hallows'study applies Kotter's theory to a faith-based and non-profit organization. Although All Hallows enjoyed 172 years of educational contributions, the book will illustrate how legacy challenges, sense of complacency, lack of vision and mission identity at critical times of change failed to inculcate and anchor an Organizational Culture and Identity for Change.
- Published
- 2020
4. Nucleosome Binding Alters the Substrate Bonding Environment of Histone H3 Lysine 36 Methyltransferase NSD2
- Author
-
Patrick McDevitt, Rosalie Matico, Vern L. Schramm, Wangfang Hou, Myles B. Poulin, Marc A. Holbert, and Jessica L. Schneck
- Subjects
Models, Molecular ,0301 basic medicine ,S-Adenosylmethionine ,010402 general chemistry ,Methylation ,01 natural sciences ,Biochemistry ,Article ,Catalysis ,03 medical and health sciences ,Histone H3 ,Colloid and Surface Chemistry ,Histone methylation ,Humans ,Nucleosome ,Histone octamer ,Nucleosome binding ,Binding Sites ,Chemistry ,Computational Biology ,Histone-Lysine N-Methyltransferase ,General Chemistry ,Models, Theoretical ,Linker DNA ,Nucleosomes ,0104 chemical sciences ,Repressor Proteins ,030104 developmental biology ,Histone methyltransferase ,Chromatosome ,Biophysics ,Protein Binding - Abstract
Nuclear receptor-binding SET domain protein 2 (NSD2) is a histone H3 lysine 36 (H3K36)-specific methyltransferase enzyme that is overexpressed in a number of cancers, including multiple myeloma. NSD2 binds to S-adenosyl-L-methionine (SAM) and nucleosome substrates to catalyze the transfer of a methyl group from SAM to the ε-amino group of histone H3K36. Equilibrium binding isotope effects and density functional theory calculations indicate that the SAM methyl group is sterically constrained in complex with NSD2, and that this steric constraint is released upon nucleosome binding. Together, these results show that nucleosome binding to NSD2 induces a significant change in the chemical environment of enzyme-bound SAM.
- Published
- 2016
- Full Text
- View/download PDF
5. Discovery of a Novel 2,6-Disubstituted Glucosamine Series of Potent and Selective Hexokinase 2 Inhibitors
- Author
-
Mary Ann Hardwicke, Hong Lin, Karl F. Erhard, John D. Martin, Ren Xie, Kaushik Raha, Rosanna Tedesco, Jin Zeng, Yanqiu Qian, Patricia M. Kratz, Yong Jiang, Ted Cecconie, Stan F. Martens, Angela Smallwood, Patrick McDevitt, James F. Mack, Baoguang Zhao, Stephenie B. Chen, Anthony J. Jurewicz, Juan I. Luengo, Mark J. Schulz, Nino Campobasso, Catherine A. Oleykowski, Lawrence M. Szewczuk, Alan R. Rendina, Cynthia M. Rominger, Benjamin J. Schwartz, Leng Nickels, Junya Qu, and Jacques Briand
- Subjects
0301 basic medicine ,chemistry.chemical_classification ,Stereochemistry ,Organic Chemistry ,Biology ,Biochemistry ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,Hexokinase-2 ,Enzyme ,chemistry ,Cell culture ,Glucosamine ,030220 oncology & carcinogenesis ,Drug Discovery ,Bladder tumor ,Glycolysis ,Selectivity - Abstract
A novel series of potent and selective hexokinase 2 (HK2) inhibitors, 2,6-disubstituted glucosamines, has been identified based on HTS hits, exemplified by compound 1. Inhibitor-bound crystal structures revealed that the HK2 enzyme could adopt an "induced-fit" conformation. The SAR study led to the identification of potent HK2 inhibitors, such as compound 34 with greater than 100-fold selectivity over HK1. Compound 25 inhibits in situ glycolysis in a UM-UC-3 bladder tumor cell line via (13)CNMR measurement of [3-(13)C]lactate produced from [1,6-(13)C2]glucose added to the cell culture.
- Published
- 2016
- Full Text
- View/download PDF
6. Transition state for the NSD2-catalyzed methylation of histone H3 lysine 36
- Author
-
Rosalie Matico, Wangfang Hou, Thomas D. Meek, Sara H. Thrall, Patrick McDevitt, Jessica L. Schneck, Johnson Neil W, Myles B. Poulin, Michael J. Huddleston, and Vern L. Schramm
- Subjects
Models, Molecular ,0301 basic medicine ,Reaction mechanism ,Stereochemistry ,Methylation ,Catalysis ,Histones ,03 medical and health sciences ,Histone H3 ,Nucleophile ,Kinetic isotope effect ,Humans ,Nucleosome ,Multidisciplinary ,030102 biochemistry & molecular biology ,Chemistry ,Lysine ,digestive, oral, and skin physiology ,Leaving group ,Histone-Lysine N-Methyltransferase ,Biological Sciences ,Repressor Proteins ,030104 developmental biology ,Biochemistry ,SN2 reaction ,HeLa Cells - Abstract
Nuclear receptor SET domain containing protein 2 (NSD2) catalyzes the methylation of histone H3 lysine 36 (H3K36). It is a determinant in Wolf-Hirschhorn syndrome and is overexpressed in human multiple myeloma. Despite the relevance of NSD2 to cancer, there are no potent, selective inhibitors of this enzyme reported. Here, a combination of kinetic isotope effect measurements and quantum chemical modeling was used to provide subangstrom details of the transition state structure for NSD2 enzymatic activity. Kinetic isotope effects were measured for the methylation of isolated HeLa cell nucleosomes by NSD2. NSD2 preferentially catalyzes the dimethylation of H3K36 along with a reduced preference for H3K36 monomethylation. Primary Me-(14)C and (36)S and secondary Me-(3)H3, Me-(2)H3, 5'-(14)C, and 5'-(3)H2 kinetic isotope effects were measured for the methylation of H3K36 using specifically labeled S-adenosyl-l-methionine. The intrinsic kinetic isotope effects were used as boundary constraints for quantum mechanical calculations for the NSD2 transition state. The experimental and calculated kinetic isotope effects are consistent with an SN2 chemical mechanism with methyl transfer as the first irreversible chemical step in the reaction mechanism. The transition state is a late, asymmetric nucleophilic displacement with bond separation from the leaving group at (2.53 Å) and bond making to the attacking nucleophile (2.10 Å) advanced at the transition state. The transition state structure can be represented in a molecular electrostatic potential map to guide the design of inhibitors that mimic the transition state geometry and charge.
- Published
- 2016
- Full Text
- View/download PDF
7. Abstract NS6: Born to Run: Advanced Practice Provider-Led Mobile Stroke Unit Care Measures Up to Vascular Neurologists’ Diagnosis and Management
- Author
-
Joshua Fletcher, Andrew Bouche, Joseph Rike, Christopher Crockett, Ian Barber, James Harrold, Keri M Rogers, James Orr, Patrick McDevitt, Andrei V Alexandrov, Adam S Arthur, Stanley Bey, Ted Novak, Wendy Dusenbury, Jordan S Carlow, Chris McKendry, Charles Bullock, William Stacks, Michael Jestice, Stephen McDaniel, Tyrone Ware, Marc D. Malkoff, Anne W. Alexandrov, Mathew Crockett, Thomas Mathenia, Victoria Swatzell, Ira Crisp, and Sarah McCormick
- Subjects
Advanced and Specialized Nursing ,Telemedicine ,business.industry ,Nurse practitioners ,Staffing ,Stroke units ,Stroke unit care ,medicine.disease ,Medicine ,Neurology (clinical) ,Medical emergency ,Cardiology and Cardiovascular Medicine ,business ,psychological phenomena and processes - Abstract
Background: Mobile Stroke Units (MSU) are growing in numbers throughout the U.S. and abroad, with numerous staffing configurations, telemedicine, and differing imaging capabilities. We aimed to test the diagnostic accuracy and treatment safety, alongside time to diagnosis and treatment delivery of a novel advanced practice provider (APP) led MSU team. Methods: We launched an MSU housing a hospital-grade Siemens Somatom CT with CTA capabilities, and hired APPs with advanced neurovascular practitioner board certification to lead field medical diagnosis and order/initiate treatment for encountered stroke patients. Consecutive MSU patients were evaluated for differences between APPs and Vascular Neurologists (VNs) diagnosis and management, and scene diagnosis and treatment times were collected. Results: Agreement between APP field medical diagnosis and MD hospital diagnosis was 100%; stroke mimic diagnosis agreement was 98%. Overall agreement for field interpretation of CT/CTA was 97%, with discrepancies not associated with stroke treatment decisions. MDs’ agreement with APPs’ identification/treatment of ICH was 100%, and IVtPA treatment decisions 98% (APPs more conservative). Scene arrival to medical diagnosis (including clinical exam and imaging completion/interpretation) ranged from 7-10 minutes, of which 4 minutes were CT/CTA start to finish times. Scene arrival to IVtPA bolus ranged from 16 minutes to 33 minutes and was driven primarily by need for control of excessive hypertension, with scene arrival to start of nicardipine premix infusion ranging from 10-14 minutes. Conclusions: Use of an APP-led MSU is safe and non-inferior to VN diagnosis/management, and may be faster than telemedicine guided MSU treatment.
- Published
- 2017
- Full Text
- View/download PDF
8. A Scalable Platform for Producing Recombinant Nucleosomes with Codified Histone Methyltransferase Substrate Preferences
- Author
-
Patricia M. McCormick, Jessica L. Schneck, Wangfang Hou, Patrick McDevitt, Robert B. Kirkpatrick, Elsie Diaz, Rosalie E. Matico, and Michael J. Huddleston
- Subjects
0106 biological sciences ,Methyltransferase ,Gene Expression ,Computational biology ,01 natural sciences ,Substrate Specificity ,law.invention ,Histones ,03 medical and health sciences ,chemistry.chemical_compound ,law ,010608 biotechnology ,Escherichia coli ,Humans ,Nucleosome ,030304 developmental biology ,0303 health sciences ,biology ,Chemistry ,Drug discovery ,Histone-Lysine N-Methyltransferase ,Methylation ,Recombinant Proteins ,Nucleosomes ,Repressor Proteins ,Histone ,Histone methyltransferase ,Recombinant DNA ,biology.protein ,DNA ,HeLa Cells ,Biotechnology - Abstract
Wolf-Hirschhorn Syndrome Candidate 1 (WHSC1; also known as NSD2) is a SET domain-containing histone lysine methyltransferase. A chromosomal translocation occurs in 15-20% of multiple myeloma patients and is associated with increased production of WHSC1 and poor clinical prognosis. To define the substrate requirements of NSD2, we established a platform for the large-scale production of recombinant polynucleosomes, based on authentic human histone proteins, expressed in E. coli, and complexed with linearized DNA. A brief survey of methyltransferases whose substrate requirements are recorded in the literature yielded expected results, lending credence to the fitness of our approach. This platform was readily 'codified' with respect to both position and extent of methylation at histone 3 lysines 18 and 36 and led to the conclusion that the most readily discernible activity of NSD2 in contact with a nucleosome substrate is dimethylation of histone 3 lysine 36. We further explored reaction mechanism, and conclude a processive, rather than distributive mechanism best describes the interaction of NSD2 with intact nucleosome substrates. The methods developed feature scale and flexibility and are suited to thorough pharmaceutical-scale drug discovery campaigns.
- Published
- 2019
- Full Text
- View/download PDF
9. Discovery of Novel Cyanamide-Based Inhibitors of Cathepsin C
- Author
-
Roderick S. Davis, Emilie Veronique Petitjean, Jessica L. Schneck, Alicia Bacon Davis, Palovich Michael R, Haibo Xie, Deng Jianghe, Jim Villa, David Bettoun, Baoguang Zhao, Brian Peck, Robert Midgett, Patrick McDevitt, Casey Kmett, Dramane I. Laine, Neysa Nevins, Brent W. Mccleland, Lin Guoliang, Anais Jolit, Sandra Umbrecht, and Isabelle Delhom
- Subjects
Cathepsin ,Organic Chemistry ,Pharmacology ,Biochemistry ,Cysteine protease ,Cathepsin C ,chemistry.chemical_compound ,Cathepsin O ,chemistry ,In vivo ,Drug Discovery ,Cigarette smoke ,Cyanamide ,Selectivity - Abstract
The discovery of potent and selective cyanamide-based inhibitors of the cysteine protease cathepsin C is detailed. Optimization of the template with regard to plasma stability led to the identification of compound 17, a potent cathepsin C inhibitor with excellent selectivity over other cathepsins and potent in vivo activity in a cigarette smoke mouse model.
- Published
- 2010
- Full Text
- View/download PDF
10. Baculovirus production of fully-active phosphoinositide 3-kinase alpha as a p85α–p110α fusion for X-ray crystallographic analysis with ATP competitive enzyme inhibitors
- Author
-
Matthew C. Burns, Kyung O. Johanson, Don T. Fisher, Patrick McDevitt, Vaughan R. Leydon, Patricia A. Elkins, Hongwei Qi, Kurt R. Auger, Paris Ward, Maggie Grimes, Thomas D. Sweitzer, Martin Brandt, Benjamin Schwartz, Yu Xue, Robert H. Sinnamon, Ruth Lehr, Thau F. Ho, Christopher S. Jones, Beth Pietrak, Robert B. Kirkpatrick, and Giorgia Vincentini
- Subjects
Models, Molecular ,Cell signaling ,Recombinant Fusion Proteins ,Context (language use) ,Spodoptera ,P110α ,Crystallography, X-Ray ,Inhibitory Concentration 50 ,Adenosine Triphosphate ,X-Ray Diffraction ,Animals ,Enzyme Inhibitors ,Cells, Cultured ,PI3K/AKT/mTOR pathway ,Class II Phosphatidylinositol 3-Kinases ,chemistry.chemical_classification ,Binding Sites ,Phosphoinositide 3-kinase ,biology ,Drug discovery ,Active site ,Artificial Gene Fusion ,Class Ia Phosphatidylinositol 3-Kinase ,Crystallography ,Enzyme ,chemistry ,Biochemistry ,Drug Design ,biology.protein ,Baculoviridae ,Biotechnology - Abstract
Phosphoinositide 3-kinases have been targeted for therapeutic research because they are key components of a cell signaling cascade controlling proliferation, growth, and survival. Direct activation of the PI3Kalpha pathway contributes to the development and progression of solid tumors in breast, endometrial, colon, ovarian, and gastric cancers. In the context of a drug discovery effort, the availability of a robust crystallographic system is a means to understand the subtle differences between ATP competitive inhibitor interactions with the active site and their selectivity against other PI3Kinase enzymes. To generate a suitable recombinant design for this purpose, a p85alpha-p110alpha fusion system was developed which enabled the expression and purification of a stoichiometrically homogeneous, constitutively active enzyme for structure determination with potent ATP competitive inhibitors (Raha et al., in preparation) [56]. This approach has yielded preparations with activity and inhibition characteristics comparable to those of the full-length PI3Kalpha from which X-ray diffracting crystals were grown with inhibitors bound in the active site.
- Published
- 2010
- Full Text
- View/download PDF
11. Optimized procedures for producing biologically active chemokines
- Author
-
John J. Kerrigan, Dulcie B. Schmidt, James J. Foley, Abby J. Sukman, Tia Lewis, Patrick McDevitt, Kathleen T. Gallagher, Kyung O. Johanson, Robert S. Ames, James A. Fornwald, Taylor L. Graham, Matthew C. Burns, Quinn Lu, Xiaoyan Tang, and Gerald E. Hunsberger
- Subjects
Protein Folding ,Chemokine ,biology ,Chemistry ,Recombinant Fusion Proteins ,Biological activity ,Protein Engineering ,medicine.disease_cause ,Polymerase Chain Reaction ,Fusion protein ,Redox ,Inclusion bodies ,Biochemistry ,Affinity chromatography ,Cytoplasm ,Escherichia coli ,biology.protein ,medicine ,Chemokines ,Cloning, Molecular ,Oxidation-Reduction ,Biotechnology - Abstract
We describe here two strategies to produce biologically active chemokines with authentic N-terminal amino acid residues. The first involves producing the target chemokine with an N-terminal 6×His-SUMO tag in Escherichia coli as inclusion bodies. The fusion protein is solubilized and purified with Ni–NTA–agarose in denaturing reagents. This is further followed by tag removal and refolding in a redox refolding buffer. The second approach involves expressing the target chemokine with an N-terminal 6×His-Trx-SUMO tag in an engineered E. coli strain that facilitates formation of disulfide bonds in the cytoplasm. Following purification of the fusion protein via Ni–NTA and tag removal, the target chemokine is refolded without redox buffer and purified by reverse phase chromatography. Using the procedures, we have produced more than 15 biologically active chemokines, with a yield of up to 15 mg/L.
- Published
- 2009
- Full Text
- View/download PDF
12. Regulatory RNAs in Planarians
- Author
-
Pawlicka, Kamila, primary, Perrigue, Patrick McDevitt, primary, and Barciszewski, Jan, primary
- Published
- 2017
- Full Text
- View/download PDF
13. Structure-Based Design of a Novel SMYD3 Inhibitor that Bridges the SAM-and MEKK2-Binding Pockets
- Author
-
Ryan G. Kruger, Roland S. Annan, Patricia A. Elkins, Tony W. Dean, Patrick McDevitt, Helai P. Mohammad, Francesca Zappacosta, Alan P. Graves, William G. Bonnette, Dai-Shi Su, Glenn S. Van Aller, and Christopher L. Carpenter
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,S-Adenosylmethionine ,Methyltransferase ,Binding Sites ,030102 biochemistry & molecular biology ,Lysine ,Methylation ,Plasma protein binding ,Histone-Lysine N-Methyltransferase ,Biology ,MAP Kinase Kinase Kinase 2 ,Molecular Docking Simulation ,03 medical and health sciences ,030104 developmental biology ,Structural Biology ,Mutation ,Cancer research ,Transferase ,Humans ,Binding site ,Enzyme Inhibitors ,Molecular Biology ,Protein Binding - Abstract
SMYD3 is a lysine methyltransferase overexpressed in colorectal, breast, prostate, and hepatocellular tumors, and has been implicated as an oncogene in human malignancies. Methylation of MEKK2 by SMYD3 is important for regulation of the MEK/ERK pathway, suggesting the possibility of selectively targeting SMYD3 in RAS-driven cancers. Structural and kinetic characterization of SMYD3 was undertaken leading to a co-crystal structure of SMYD3 with a MEKK2-peptide substrate bound, and the observation that SMYD3 follows a partially processive mechanism. These insights allowed for the design of GSK2807, a potent and selective, SAM-competitive inhibitor of SMYD3 (Ki = 14 nM). A high-resolution crystal structure reveals that GSK2807 bridges the gap between the SAM-binding pocket and the substrate lysine tunnel of SMYD3. Taken together, our data demonstrate that small-molecule inhibitors of SMYD3 can be designed to prevent methylation of MEKK2 and these could have potential use as anticancer therapeutics.
- Published
- 2015
14. A DNA Hypomethylation Signature Predicts Antitumor Activity of LSD1 Inhibitors in SCLC
- Author
-
Thau F. Ho, Yan Liu, Xinrong Tian, Patrick McDevitt, Michael Butticello, Timothy K. Hart, Kimberly N. Smitheman, David Soong, Jessica L. Schneck, Peter J. Tummino, Christine L. Hann, Glenn S. Van Aller, Yuchen Bai, Nestor O. Concha, Kasparec Jiri, Melissa B. Pappalardi, Christopher L. Carpenter, Mcnulty Kenneth C, Kelly Federowicz, Jeffrey D. Carson, William H. Miller, Johnson Neil W, Chandrashekhar D. Kamat, Rouse Meagan B, Charles F. McHugh, Michael T. McCabe, Ryan G. Kruger, Dashyant Dhanak, Helai P. Mohammad, Michelle Crouthamel, Yan Degenhardt, William G. Bonnette, and Shelby A. Gorman
- Subjects
Cyclopropanes ,Cancer Research ,animal structures ,Lung Neoplasms ,Molecular Sequence Data ,Administration, Oral ,Antineoplastic Agents ,Benzoates ,Article ,Epigenesis, Genetic ,chemistry.chemical_compound ,Mice ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Epigenetics ,Enzyme Inhibitors ,Cell Proliferation ,Histone Demethylases ,biology ,Cancer ,KDM1A ,Biological activity ,Cell Biology ,DNA Methylation ,medicine.disease ,Small Cell Lung Carcinoma ,Xenograft Model Antitumor Assays ,Gene Expression Regulation, Neoplastic ,chemistry ,Biochemistry ,Oncology ,Cell culture ,biology.protein ,Cancer research ,Demethylase ,Growth inhibition ,DNA hypomethylation - Abstract
SummaryEpigenetic 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 discovery and 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.
- Published
- 2015
15. The King of Sports: Polo in late Victorian and Edwardian India
- Author
-
Patrick McDevitt
- Subjects
History ,Social Sciences (miscellaneous) - Published
- 2003
- Full Text
- View/download PDF
16. Smyd3 regulates cancer cell phenotypes and catalyzes histone H4 lysine 5 methylation
- Author
-
Glenn S. Van Aller, Peter J. Tummino, BaoChau Le, Gloria Mas, Roland S. Annan, Ryan G. Kruger, Patrick McDevitt, Nicolas Reynoird, Olena Barbash, Michael J. Huddleston, Or Gozani, Julien Sage, Shichong Liu, Benjamin A. Garcia, Robert H. Sinnamon, Anne-Flore Zmoos, Institute for Advanced Biosciences / Institut pour l'Avancée des Biosciences (Grenoble) (IAB), and Etablissement français du sang - Auvergne-Rhône-Alpes (EFS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre Hospitalier Universitaire [Grenoble] (CHU)-Centre National de la Recherche Scientifique (CNRS)-Université Grenoble Alpes (UGA)
- Subjects
Cancer Research ,[SDV]Life Sciences [q-bio] ,Blotting, Western ,Biology ,Methylation ,Chromatin remodeling ,Substrate Specificity ,Histones ,Histone H4 ,Mice ,Histone H1 ,Peptide Library ,oncogene ,Smyd3 ,Histone methylation ,Histone H2A ,Animals ,Humans ,cancer ,Histone code ,RNA, Small Interfering ,Molecular Biology ,ComputingMilieux_MISCELLANEOUS ,Mice, Knockout ,lysine ,epigenetics ,Brief Report ,Genetic Complementation Test ,EZH2 ,Histone-Lysine N-Methyltransferase ,Fibroblasts ,Chromatin ,Recombinant Proteins ,3. Good health ,Enzyme Activation ,Gene Expression Regulation, Neoplastic ,Mice, Inbred C57BL ,Phenotype ,Biochemistry ,Histone methyltransferase ,oncology ,Mutagenesis, Site-Directed ,HeLa Cells ,Plasmids - Abstract
Smyd3 is a lysine methyltransferase implicated in chromatin and cancer regulation. Here we show that Smyd3 catalyzes histone H4 methylation at lysine 5 (H4K5me). This novel histone methylation mark is detected in diverse cell types and its formation is attenuated by depletion of Smyd3 protein. Further, Smyd3-driven cancer cell phenotypes require its enzymatic activity. Thus, Smyd3, via H4K5 methylation, provides a potential new link between chromatin dynamics and neoplastic disease.
- Published
- 2014
- Full Text
- View/download PDF
17. Potent and Selective Nonpeptide Inhibitors of Caspases 3 and 7 Inhibit Apoptosis and Maintain Cell Functionality
- Author
-
Michael W. Lark, Christine Debouck, Kalindi Vaidya, Mark E. Nuttall, Kyung O. Johanson, James D. Winkler, Alison M. Badger, Martha S. Head, Paul M. Keller, Mark Alan Levy, R. Curtis Haltiwanger, Larry J. Suva, Cheryl A. Janson, Sherin S. Abdel-Meguid, Po-Huang Liang, Patrick McDevitt, George K. Chan, Nestor O. Concha, Daniel P. Nadeau, Kristine Kikly, Dennis Lee, Scott A. Long, Winnie Chan, Walter E. DeWolf, Ryan M Dominic, Susan B. Richardson, Maxine Gowen, Thaddeus A. Tomaszek, Terry A. Francis, Chiu-Mei Sung, and Jerry L. Adams
- Subjects
Isatin ,Models, Molecular ,Programmed cell death ,Neutrophils ,Apoptosis ,Caspase 3 ,Crystallography, X-Ray ,Biochemistry ,Caspase 7 ,Amino Acid Chloromethyl Ketones ,Mice ,Chondrocytes ,Osteoarthritis ,Animals ,Humans ,Enzyme Inhibitors ,Promoter Regions, Genetic ,Molecular Biology ,Caspase ,Sulfonamides ,Binding Sites ,Molecular Structure ,biology ,NLRP1 ,Intrinsic apoptosis ,Cell Biology ,Caspase Inhibitors ,Recombinant Proteins ,Cell biology ,XIAP ,biology.protein ,Camptothecin ,Collagen - Abstract
Caspases have been strongly implicated to play an essential role in apoptosis. A critical question regarding the role(s) of these proteases is whether selective inhibition of an effector caspase(s) will prevent cell death. We have identified potent and selective non-peptide inhibitors of the effector caspases 3 and 7. The inhibition of apoptosis and maintenance of cell functionality with a caspase 3/7-selective inhibitor is demonstrated for the first time, and suggests that targeting these two caspases alone is sufficient for blocking apoptosis. Furthermore, an x-ray co-crystal structure of the complex between recombinant human caspase 3 and an isatin sulfonamide inhibitor has been solved to 2.8-A resolution. In contrast to previously reported peptide-based caspase inhibitors, the isatin sulfonamides derive their selectivity for caspases 3 and 7 by interacting primarily with the S(2) subsite, and do not bind in the caspase primary aspartic acid binding pocket (S(1)). These inhibitors blocked apoptosis in murine bone marrow neutrophils and human chondrocytes. Furthermore, in camptothecin-induced chondrocyte apoptosis, cell functionality as measured by type II collagen promoter activity is maintained, an activity considered essential for cartilage homeostasis. These data suggest that inhibiting chondrocyte cell death with a caspase 3/7-selective inhibitor may provide a novel therapeutic approach for the prevention and treatment of osteoarthritis, or other disease states characterized by excessive apoptosis.
- Published
- 2000
- Full Text
- View/download PDF
18. Identification of Unique Truncated KC/GROβ Chemokines with Potent Hematopoietic and Anti-Infective Activities
- Author
-
Kyung O. Johanson, Joanna Maria Balcarek, Zdenka Ludmila Jonak, John R. White, Andrew G. King, Pradip K. Bhatnagar, Louis M. Pelus, Carrie L. Frey, Patrick McDevitt, Dean E. McNulty, and Peter Lawrence Demarsh
- Subjects
Chemokine ,Antifungal Agents ,Stromal cell ,Chemokine CXCL1 ,Molecular Sequence Data ,Immunology ,Cell ,Bone Marrow Cells ,Neutrophil Activation ,Cell Line ,Microbiology ,Mice ,Adjuvants, Immunologic ,In vivo ,medicine ,Animals ,Humans ,Immunology and Allergy ,Amino Acid Sequence ,Growth Substances ,Chemotactic Factors ,biology ,Immune Sera ,Candidiasis ,Drug Synergism ,Molecular biology ,Peptide Fragments ,Recombinant Proteins ,In vitro ,Respiratory burst ,Mice, Inbred C57BL ,Haematopoiesis ,medicine.anatomical_structure ,Cell culture ,Macrophages, Peritoneal ,biology.protein ,Intercellular Signaling Peptides and Proteins ,Female ,Stromal Cells ,Chemokines, CXC ,Oligopeptides ,Injections, Intraperitoneal - Abstract
SK&F 107647, a previously described synthetic immunomodulatory peptide, indirectly stimulates bone marrow progenitor cells and phagocytic cells, and enhances host defense effector mechanisms in bacterial and fungal infection models in vivo. In vitro, SK&F 107647 induces the production of a soluble mediator that augments colony forming cell (CFU-GM) formation in the presence of CSFs. In this paper we purified and sequenced the stromal cell-derived hematopoietic synergistic factors (HSF) secreted from both murine and human cell lines stimulated with SK&F 107647. Murine HSF is an N-terminal 4-aa truncated form of the CXC chemokine, KC, while human HSF was identified as an N-terminal 4-aa truncated form of the CXC chemokine, GROβ. In comparison to their full-length forms, truncated KC and truncated GROβ were 10 million times more potent as synergistic growth stimulants for CFU-GM. Enhanced potency of these novel truncated chemokines relative to their full-length forms was also demonstrated in respiratory burst assays, CD11b Ag expression, and intracellular killing of the opportunistic pathogen, Candida albicans. Administration of truncated KC significantly enhanced survival of mice lethally infected with C. albicans. The results reported herein delineate the biological mechanism of action of SK&F 107647, which functions via the induction of unique specific truncated forms of the chemokines KC and GROβ. To our knowledge, this represents the first example where any form of KC or GROβ were purified from marrow stromal cells. Additionally, this is the first demonstration of in vivo efficacy of a CXC chemokine in an animal infectious fungal disease model.
- Published
- 2000
- Full Text
- View/download PDF
19. Nuclear magnetic resonance solution structure of truncated human GROβ [5–73] and its structural comparison with CXC chemokine family members GROα and IL-8
- Author
-
Yan Qiu Qian, Patrick McDevitt, and Kyung O. Johanson
- Subjects
Models, Molecular ,Stereochemistry ,Chemokine CXCL1 ,Dimer ,Amino Acid Motifs ,Chemokine CXCL2 ,Static Electricity ,Dihedral angle ,Crystallography, X-Ray ,Antiparallel (biochemistry) ,Protein Structure, Secondary ,Cell Line ,Structure-Activity Relationship ,chemistry.chemical_compound ,Nuclear magnetic resonance ,Bone Marrow ,Structural Biology ,Humans ,Disulfides ,Growth Substances ,Nuclear Magnetic Resonance, Biomolecular ,Molecular Biology ,Conformational isomerism ,Conserved Sequence ,Sequence Deletion ,chemistry.chemical_classification ,Chemotactic Factors ,C-terminus ,Interleukin-8 ,Biological activity ,Peptide Fragments ,Amino acid ,Solutions ,Crystallography ,chemistry ,Intercellular Signaling Peptides and Proteins ,Stromal Cells ,Chemokines, CXC ,Dimerization ,Two-dimensional nuclear magnetic resonance spectroscopy - Abstract
The three-dimensional structure of a novel four amino acid truncated form of the CXC chemokine GRObeta [5-73] isolated from bone marrow stromal cells with potent hematopoietic and anti-infective activities has been determined by two-dimensional (1)H nuclear magnetic resonance (NMR) spectroscopy in solution. On the basis of 1878 upper distance constraints derived from nuclear Overhauser effects (NOE) and 314 dihedral angle constraints, a group of 20 conformers representing the solution structure of the human GRObeta [5-73] was computed with the program DYANA. At the concentrations used for NMR study, GRObeta [5-73] forms a dimer in solution that is architectured by a six-stranded antiparallel beta-sheet (residues 25 to 29, 39 to 44, 49 to 52) and a pair of helices (residues 58 to 68) with 2-fold symmetry, while the C terminus of the protein is disordered. The average of the pairwise root-mean-square deviations of individual NMR conformers relative to the mean coordinates for the backbone atoms N, C(alpha) and C' of residues 5 to 68 is 0.47 A. Overall, the global fold of GRObeta [5-73] is similar to that of the previously reported NMR structure of GROalpha and the NMR and X-ray structures of interleukin-8. Among these three CXC chemokines, GRObeta [5-73] is most similar in structure to GROalpha. Significant differences between GRObeta [5-73], GROalpha and interleukin-8 are in the N-terminal loop comprising residues 12 to 19. The N-terminal arm containing the conserved ELR motif and the loop of residues 30 to 38 containing the GPH motif are different among these three CXC chemokines. The structural differences in these two regions may be responsible for the specificity of the receptor binding and biological activity of different chemokines.
- Published
- 1999
- Full Text
- View/download PDF
20. Structures of Human DPP7 Reveal the Molecular Basis of Specific Inhibition and the Architectural Diversity of Proline-Specific Peptidases
- Author
-
Annie M. Hassell, Lisa M. Shewchuk, Patrick McDevitt, Elena Dobrovetsky, Lissete Crombett, Thomas D. Sweitzer, Doug Cossar, Aiping Dong, Karen M. Kennedy-Wilson, Sirano Dhe-Paganon, Almagul Seitova, Alexey Bochkarev, Sharon Sweitzer, Karl Gruber, Gustavo Arruda Bezerra, and Kyung O. Johanson
- Subjects
Insecta ,Hydrolases ,lcsh:Medicine ,Plasma protein binding ,Biochemistry ,Substrate Specificity ,Catalytic Domain ,Cricetinae ,Drug Discovery ,Macromolecular Structure Analysis ,Biomacromolecule-Ligand Interactions ,Amino Acids ,lcsh:Science ,chemistry.chemical_classification ,0303 health sciences ,Multidisciplinary ,biology ,Enzyme Classes ,030302 biochemistry & molecular biology ,Enzyme structure ,Amino acid ,Enzymes ,Dimerization ,Research Article ,Protein Binding ,Protein Structure ,Proline ,Dipeptidyl Peptidase 4 ,Molecular Sequence Data ,Biophysics ,CHO Cells ,Catalysis ,Evolution, Molecular ,03 medical and health sciences ,Hydrolase ,Catalytic triad ,Animals ,Humans ,Protein Interactions ,Dipeptidyl-Peptidases and Tripeptidyl-Peptidases ,Biology ,Dipeptidyl peptidase-4 ,030304 developmental biology ,Base Sequence ,lcsh:R ,Active site ,Proteins ,Computational Biology ,Hormones ,Protein Structure, Tertiary ,Enzyme ,chemistry ,Enzyme Structure ,biology.protein ,lcsh:Q ,Globular Proteins - Abstract
Proline-specific dipeptidyl peptidases (DPPs) are emerging targets for drug development. DPP4 inhibitors are approved in many countries, and other dipeptidyl peptidases are often referred to as DPP4 activity- and/or structure-homologues (DASH). Members of the DASH family have overlapping substrate specificities, and, even though they share low sequence identity, therapeutic or clinical cross-reactivity is a concern. Here, we report the structure of human DPP7 and its complex with a selective inhibitor Dab-Pip (L-2,4-diaminobutyryl-piperidinamide) and compare it with that of DPP4. Both enzymes share a common catalytic domain (α/β-hydrolase). The catalytic pocket is located in the interior of DPP7, deep inside the cleft between the two domains. Substrates might access the active site via a narrow tunnel. The DPP7 catalytic triad is completely conserved and comprises Ser162, Asp418 and His443 (corresponding to Ser630, Asp708 and His740 in DPP4), while other residues lining the catalytic pockets differ considerably. The "specificity domains" are structurally also completely different exhibiting a β-propeller fold in DPP4 compared to a rare, completely helical fold in DPP7. Comparing the structures of DPP7 and DPP4 allows the design of specific inhibitors and thus the development of less cross-reactive drugs. Furthermore, the reported DPP7 structures shed some light onto the evolutionary relationship of prolyl-specific peptidases through the analysis of the architectural organization of their domains.
- Published
- 2012
21. The soluble form of E-selectin is an asymmetric monomer. Expression, purification, and characterization of the recombinant protein
- Author
-
Ian Brooks, Patrick McDevitt, K D Kopple, Don E. Griswold, John A. Feild, Janice R. Connor, Preston Hensley, John J. Trill, Dean E. McNulty, and N V Kumar
- Subjects
Chromatography ,Chinese hamster ovary cell ,Size-exclusion chromatography ,Cell Biology ,Biochemistry ,law.invention ,Sedimentation coefficient ,chemistry.chemical_compound ,Monomer ,chemistry ,law ,Complementary DNA ,Recombinant DNA ,Ultracentrifuge ,Molecular Biology ,Two-dimensional nuclear magnetic resonance spectroscopy - Abstract
The gene coding for a soluble form of human E-selectin (sE-selectin) has been expressed in Chinese hamster ovary (CHO) cells. Cells seeded into a hollow fiber reactor secreted protein at a level of 160 mg/liter. The protein was purified to > 95% pure and low endotoxin (< 2 ng/mg), using physiological pH and buffers. The amino acid composition and N-terminal sequence were as predicted from the cDNA sequence. HL-60 cells bound to sE-selectin-coated plates in a dose-dependent manner, and this binding could be blocked up to 100% by pretreatment of HL60 cells with sE-selectin. The concentration of sE-selectin required for 50% inhibition was 1 microM. This value puts an upper limit for the affinity of E-selectin for its natural receptor. sE-selectin also inhibited inflammatory migration of neutrophils in a selective fashion. Purified sE-selectin exhibited a broad band of M(r) approximately 75,000 on nonreducing SDS-PAGE. sE-selectin eluted with M(r) approximately 310,000 from size exclusion chromatography at physiological pH and buffers, suggesting an oligomeric state. Matrix-assisted laser-desorption MS gave a molecular weight of 80,000, while the minimum monomer molecular weight from the gene sequence should be 58,571, demonstrating that the monomeric molecule thus expressed had 27% carbohydrate. Equilibrium analytical ultracentrifugation gave an average solution molecular weight of 81,600 (+/- 4,500). Velocity ultracentrifugation gave a sedimentation coefficient of 4.3 S and, from this, an apparent axial ratio of 10.5:1, assuming a prolate ellipsoid of revolution. An analysis of the NMR NOESY spectra of sE-selectin, sialyl-Lewis X, and sE-selectin with sialyl-Lewis X demonstrates that the recombinant protein binds sialyl-Lewis X productively. Hence, in solution, sE-selectin is a functional elongated monomer.
- Published
- 1994
- Full Text
- View/download PDF
22. Kinetic mechanism and rate-limiting steps of focal adhesion kinase-1
- Author
-
James P. Villa, Kang Yan, Angela Smallwood, Khyati Oza, Robert B. Kirkpatrick, Jessica L. Schneck, Sara H. Thrall, Baoguang Zhao, Patrick McDevitt, Jacques Briand, Stephanie Chen, Ruth Lehr, Nestor O. Concha, and Thomas D. Meek
- Subjects
Models, Molecular ,Stereochemistry ,Kinetics ,Adenylyl Imidodiphosphate ,Molecular Sequence Data ,chemistry.chemical_element ,Kinetic energy ,Crystallography, X-Ray ,Biochemistry ,Oxygen ,Models, Biological ,Catalysis ,chemistry.chemical_compound ,Adenosine Triphosphate ,Humans ,Amino Acid Sequence ,Phosphorylation ,Autophosphorylation ,Solvent ,Crystallography ,chemistry ,Focal Adhesion Kinase 1 ,Peptides ,Adenosine triphosphate ,Protein Binding - Abstract
Steady-state kinetic analysis of focal adhesion kinase-1 (FAK1) was performed using radiometric measurement of phosphorylation of a synthetic peptide substrate (Ac-RRRRRRSETDDYAEIID-NH(2), FAK-tide) which corresponds to the sequence of an autophosphorylation site in FAK1. Initial velocity studies were consistent with a sequential kinetic mechanism, for which apparent kinetic values k(cat) (0.052 +/- 0.001 s(-1)), K(MgATP) (1.2 +/- 0.1 microM), K(iMgATP) (1.3 +/- 0.2 microM), K(FAK-tide) (5.6 +/- 0.4 microM), and K(iFAK-tide) (6.1 +/- 1.1 microM) were obtained. Product and dead-end inhibition data indicated that enzymatic phosphorylation of FAK-tide by FAK1 was best described by a random bi bi kinetic mechanism, for which both E-MgADP-FAK-tide and E-MgATP-P-FAK-tide dead-end complexes form. FAK1 catalyzed the betagamma-bridge:beta-nonbridge positional oxygen exchange of [gamma-(18)O(4)]ATP in the presence of 1 mM [gamma-(18)O(4)]ATP and 1.5 mM FAK-tide with a progressive time course which was commensurate with catalysis, resulting in a rate of exchange to catalysis of k(x)/k(cat) = 0.14 +/- 0.01. These results indicate that phosphoryl transfer is reversible and that a slow kinetic step follows formation of the E-MgADP-P-FAK-tide complex. Further kinetic studies performed in the presence of the microscopic viscosogen sucrose revealed that solvent viscosity had no effect on k(cat)/K(FAK-tide), while k(cat) and k(cat)/K(MgATP) were both decreased linearly at increasing solvent viscosity. Crystallographic characterization of inactive versus AMP-PNP-liganded structures of FAK1 showed that a large conformational motion of the activation loop upon ATP binding may be an essential step during catalysis and would explain the viscosity effect observed on k(cat)/K(m) for MgATP but not on k(cat)/K(m) for FAK-tide. From the positional isotope exchange, viscosity, and structural data it may be concluded that enzyme turnover (k(cat)) is rate-limited by both reversible phosphoryl group transfer (k(forward) approximately 0.2 s(-1) and k(reverse) approximately 0.04 s(-1)) and a slow step (k(conf) approximately 0.1 s(-1)) which is probably the opening of the activation loop after phosphoryl group transfer but preceding product release.
- Published
- 2010
23. Chemical mechanism of a cysteine protease, cathepsin C, as revealed by integration of both steady-state and pre-steady-state solvent kinetic isotope effects
- Author
-
Michael S. McQueney, Thomas D. Meek, James P. Villa, Patrick McDevitt, Jessica L. Schneck, and Sara H. Thrall
- Subjects
Binding Sites ,Chemistry ,Stereochemistry ,Protein Conformation ,Kinetics ,Water ,Hydrogen-Ion Concentration ,Deuterium ,Biochemistry ,Cathepsin C ,Acylation ,Burst kinetics ,Reaction rate constant ,Tetrahedral carbonyl addition compound ,Kinetic isotope effect ,Solvents ,Humans ,Sodium Hydroxide ,Steady state (chemistry) ,Cloning, Molecular - Abstract
Cathepsin C, or dipeptidyl peptidase I, is a lysosomal cysteine protease of the papain family that catalyzes the sequential removal of dipeptides from the free N-termini of proteins and peptides. Using the dipeptide substrate Ser-Tyr-AMC, cathepsin C was characterized in both steady-state and pre-steady-state kinetic modes. The pH(D) rate profiles for both log k cat/ K m and log k cat conformed to bell-shaped curves for which an inverse solvent kinetic isotope effect (sKIE) of 0.71 +/- 0.14 for (D)( k cat/ K a) and a normal sKIE of 2.76 +/- 0.03 for (D) k cat were obtained. Pre-steady-state kinetics exhibited a single-exponential burst of AMC formation in which the maximal acylation rate ( k ac = 397 +/- 5 s (-1)) was found to be nearly 30-fold greater than the rate-limiting deacylation rate ( k dac = 13.95 +/- 0.013 s (-1)) and turnover number ( k cat = 13.92 +/- 0.001 s (-1)). Analysis of pre-steady-state burst kinetics in D 2O allowed abstraction of a normal sKIE for the acylation half-reaction that was not observed in steady-state kinetics. Since normal sKIEs were obtained for all measurable acylation steps in the presteady state [ (D) k ac = 1.31 +/- 0.04, and the transient kinetic isotope effect at time zero (tKIE (0)) = 2.3 +/- 0.2], the kinetic step(s) contributing to the inverse sKIE of (D)( k cat/ K a) must occur more rapidly than the experimental time frame of the transient kinetics. Results are consistent with a chemical mechanism in which acylation occurs via a two-step process: the thiolate form of Cys-234, which is enriched in D 2O and gives rise to the inverse value of (D)( k cat/ K a), attacks the substrate to form a tetrahedral intermediate that proceeds to form an acyl-enzyme intermediate during a proton transfer step expressing a normal sKIE. The subsequent deacylation half-reaction is rate-limiting, with proton transfers exhibiting normal sKIEs. Through derivation of 12 equations describing all kinetic parameters and sKIEs for the proposed cathepsin C mechanism, integration of both steady-state and pre-steady-state kinetics with sKIEs allowed the provision of at least one self-consistent set of values for all 13 rate constants in this cysteine protease's chemical mechanism. Simulation of the resulting kinetic profile showed that at steady state approximately 80% of the enzyme exists in an active-site cysteine-acylated form in the mechanistic pathway. The chemical and kinetic details deduced from this work provide a potential roadmap to help steer drug discovery efforts for this and other disease-relevant cysteine proteases.
- Published
- 2008
24. Abstract 3513: Inhibition of LSD1 for the treatment of cancer
- Author
-
Dominic Suarez, Glenn S. Van Aller, Jess Schneck, Kasparec Jiri, Kimberly N. Smitheman, Xinrong Tian, Yan Liu, Patrick McDevitt, Ryan G. Kruger, Helai P. Mohammad, William H. Miller, Christine L. Hann, Melissa B. Pappalardi, Johnson Neil W, Kelly Federowicz, James J. Foley, Jeffrey D. Carson, Monica Cusan, Thau F. Ho, Shekhar Kamat, Scott A. Armstrong, Michael Butticello, and Charles F. McHugh
- Subjects
CD86 ,Cancer Research ,Cell ,Myeloid leukemia ,Biology ,chemistry.chemical_compound ,Haematopoiesis ,medicine.anatomical_structure ,Immunophenotyping ,Oncology ,chemistry ,Cell culture ,Immunology ,medicine ,Cancer research ,Bone marrow ,Growth inhibition - Abstract
Lysine specific demethylase 1 (LSD1) is a histone H3K4me1/2 demethylase found in various transcriptional co-repressor complexes. LSD1 mediated H3K4 demethylation can result in a repressive chromatin environment that silences gene expression and has been shown to play a role in hematopoietic differentiation. LSD1 is also overexpressed in multiple tumor types. These studies implicate LSD1 as a key regulator of the epigenome that modulates gene expression through post-translational modification of histones and its presence in transcriptional complexes. The current study describes the anti-tumor effects of a novel, irreversible, GSK LSD1 inhibitor (GSK2879552) in acute myeloid leukemia (AML) and small cell lung cancer (SCLC). GSK2879552 is a potent, selective, mechanism-based inhibitor of LSD1. Screening of over 150 cancer cell lines revealed that AML and SCLC cells have a unique requirement for LSD1. While GSK2879552 treatment did not affect the global levels of H3K4me1 or H3K4me2, local changes in these histone marks were observed near transcriptional start sites of genes whose expression increased with LSD1 inhibition. Treatment of AML cell lines with GSK2879552 increased cell surface expression of CD11b and CD86, markers associated with a differentiated immunophenotype. Six days of GSK2879552 treatment resulted in potent anti-proliferative growth effects in 19 of 25 AML cell lines representing a range of AML subtypes. Treating for longer time periods revealed sensitivity in all AML cell lines. AML blast colony forming ability was also inhibited in 4 of 5 bone marrow samples derived from primary AML patient samples. The effects of LSD1 inhibition were further characterized in vivo using a mouse model of AML induced by transduction of mouse hematopoietic progenitor cells with a retrovirus encoding MLL-AF9 and GFP. Primary AML cells were transplanted into a cohort of secondary recipient mice and were treated upon engraftment. After 17 days of treatment, control mice had 80% GFP+ cells in the bone marrow whereas treated mice had only 2.8% GFP positive cells (p Growth inhibition was also observed in a subset of SCLC cell lines. GSK2879552 treatment of mice engrafted with SCLC lines resulted in greater than 80% tumor growth inhibition. Studies using patient derived primary SCLC showed similar efficacy demonstrating the growth inhibition of SCLC with an LSD1 inhibitor extended beyond cell lines. Together, these data demonstrate that pharmacological inhibition of LSD1 may provide a promising treatment for AML and SCLC. A Phase I clinical trial using GSK2879552 was initiated in March, 2014. All studies were conducted in accordance with the GSK Policy on the Care, Welfare and Treatment of Laboratory Animals and were reviewed by the Institutional Animal Care and Use Committee either at GSK or by the ethical review process at the institution where the work was performed. Citation Format: Kimberly Smitheman, Monica Cusan, Yan Liu, Michael Butticello, Melissa Pappalardi, James Foley, Kelly Federowicz, Glenn Van Aller, Jiri Kasparec, Xinrong Tian, Dominic Suarez, Jess Schneck, Jeff Carson, Patrick McDevitt, Thau Ho, Charles McHugh, William Miller, Scott Armstrong, Christine Hann, Neil Johnson, Ryan G. Kruger, Helai P. Mohammad, Shekhar Kamat. Inhibition of LSD1 for the treatment of cancer. [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 3513. doi:10.1158/1538-7445.AM2015-3513
- Published
- 2015
- Full Text
- View/download PDF
25. Abstract 4357: Key differences revealed in NSD2 kinetics using truncated versus full-length protein
- Author
-
Michael J. Huddleston, Jessica L. Schneck, Melissa B. Pappalardi, Roland S. Annan, Ryan G. Kruger, Robert B. Kirkpatrick, Patrick McDevitt, Rosalie Matico, and Wangfang Hou
- Subjects
Cancer Research ,Sinefungin ,Histone H3 ,Histone ,Methyltransferase ,Oncology ,biology ,Biochemistry ,Lysine ,biology.protein ,Nucleosome ,Methylation ,Methylation Site - Abstract
Histone lysine methyltransferases (HKMTs) are a class of enzymes that transfer a methyl group from S-adenosyl-L-methionine (SAM) to lysine residues on histone tails. The nuclear receptor SET domain-containing (NSD) family of proteins is known to methylate lysine 36 of histone H3 (H3K36). Abnormal methylation at H3K36 has been widely implicated in a variety of cancers and diseases, therefore, the enzymes responsible for this posttranslational modification are of interest from a drug-discovery standpoint. NSD2 (MMSET/WHSC1), a representative of this family, was found to be highly expressed in a multitude of human tumors and has been directly linked to multiple myeloma and Wolf-Hirschhorn syndrome. The chromosomal translocation t(4:14)(p16;q32) that occurs in 15% of myeloma patients is associated with both increased production of NSD2 and poor prognosis while loss of the NSD2 gene at the 4p16.3 region results in Wolf-Hirschhorn syndrome. In order to better understand the multi-dimensional nature of NSD2, full-length and truncated versions of the protein were generated to evaluate NSD2 kinetics. The biochemical activity of each NSD2 construct was assessed using (1) a radioactive assay measuring 3H transfer from SAM to the histone substrate or (2) LC-MS/MS analysis of the NSD2-dependent product. Full-length NSD2 prefers a nucleosomal substrate; whereas, C-terminal truncation of a highly charged region (AA 12-14-1240) resulted in a loss of nucleosomal activity and a gain of activity using a peptide derived from Histone H3. Additionally, LC-MS/MS mapping revealed a shift in the methylation site from H3K36 to H3K18 when using the truncated system. More detailed kinetic analysis revealed that the FL/nucleosome reaction catalyzes processive methylation while truncated NSD2 methylates the peptide distributively. Lastly, key changes in inhibitor specificity were observed. An alternative C terminal region, residues 1341-1365, was required to maintain potency of the product inhibitor SAH but not the close analog sinefungin. This may be indicative of the ping-pong kinetics proposed for FL NSD2 implying that SAH targets the NSD2-nucleosome bound form of the enzyme (UC vs. nucleosome) while sinefungin does not (NC vs. nucleosome). While a truncated NSD2/peptide system would be much easier to screen, profile and characterize in the search for inhibitors, the results found herein indicate that screening a full-length NSD2/nucleosome system may be more physiologically relevant. Citation Format: Melissa B. Pappalardi, Jessica L. Schneck, Rosalie Matico, Michael Huddleston, Wangfang Hou, Patrick McDevitt, Roland Annan, Robert Kirkpatrick, Ryan Kruger. Key differences revealed in NSD2 kinetics using truncated versus full-length protein. [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 4357. doi:10.1158/1538-7445.AM2015-4357
- Published
- 2015
- Full Text
- View/download PDF
26. A Novel Polypectomy Evaluation Platform for the Objective Bench-top Testing of Submucosal Injectables to Improve the Safety and Efficacy of Polypectomy and Endoscopic Mucosal Resection
- Author
-
Brooke Ancrile, Travis Holtzman, Ryan Gaffney, Patrick McDevitt, and Matthew T. Moyer
- Subjects
medicine.medical_specialty ,Hepatology ,business.industry ,medicine.medical_treatment ,Gastroenterology ,medicine ,Endoscopic mucosal resection ,business ,Polypectomy ,Surgery - Published
- 2013
- Full Text
- View/download PDF
27. A bicistronic expression system for bacterial production of authentic human interleukin-18
- Author
-
Kyung O. Johanson, Jim A. Fornwald, Amy Calamari, Joyce Mao, Dwight D Moore, Kristin A. Knecht, Allan R. Shatzman, Stephen H. Trulli, Peter M. Tapley, Rosalie Matico, Robert B. Kirkpatrick, Louis Elefante, John J. Trill, Silas Nwagwu, Pramathesh S Patel, James F. Kane, Patrick McDevitt, Megan M. McLaughlin, Ganesh M. Sathe, Zdenka Ludmila Jonak, and Adam F Yorke
- Subjects
Signal peptide ,DNA, Complementary ,Time Factors ,Transcription, Genetic ,Molecular Sequence Data ,Dithionitrobenzoic Acid ,law.invention ,Methionine ,Ubiquitin ,law ,Transcription (biology) ,Escherichia coli ,Humans ,Amino Acid Sequence ,Cysteine ,Codon ,Caspase ,Gene Library ,Innate immune system ,biology ,Base Sequence ,Dose-Response Relationship, Drug ,Caspase 1 ,Sulfhydryl Reagents ,Interleukin-18 ,Temperature ,Biological activity ,In vitro ,Caspases, Initiator ,Recombinant Proteins ,Protein Structure, Tertiary ,Enzyme Activation ,Biochemistry ,Caspases ,Protein Biosynthesis ,Recombinant DNA ,biology.protein ,Biological Assay ,Biotechnology ,Plasmids - Abstract
Interleukin-18 (IL-18) is activated and released from immune effector cells to stimulate acquired and innate immune responses involving T and natural killer (NK) cells. The release of IL-18 from mammalian cells is linked to its proteolytic activation by caspases including interleukin 1 converting enzyme (ICE). The absence of a signal peptide sequence and the requirement for coupled activation and cellular release have presented challenges for the large-scale recombinant production of IL-18. In this study, we have explored methods for the direct production of authentic human IL-18 toward the development of a large-scale production system. Expression of mature IL-18 directly in Escherichia coli with a methionine initiating codon leads to the production of MetIL-18 that is dramatically less potent in bioassays than IL-18 produced as a pro-peptide and activated in vitro. To produce an authentic IL-18, we have devised a bicistronic expression system for the coupled transcription and translation of ProIL-18 with caspase-1 (ICE) or caspase-4 (ICE-rel II, TX, ICH-2). Mature IL-18 with an authentic N-terminus was produced and has a biological activity and potency comparable to that of in vitro processed mature IL-18. Optimization of this system for the maximal production yields can be accomplished by modulating the temperature, to affect the rate of caspase activation and to favor the accumulation of ProIL-18, prior to its proteolytic processing by activated caspase. The effect of temperature is particularly profound for the caspase-4 co-expression process, enabling optimized production levels of over 150 mg/L in shake flasks at 25 ° C. An alternative bicistronic expression design utilizing a precise ubiquitin IL-18 fusion, processed by co-expressed ubiquitinase, was also successfully used to generate fully active IL-18, thereby demonstrating that the pro-sequence of IL-18 is not required for recombinant IL-18 production.
- Published
- 2003
28. Structural basis for Chk1 inhibition by UCN-01
- Author
-
Kyung O. Johanson, Stephen T. Davis, Baoguang Zhao, Michael J. Bower, Bin-Bing S. Zhou, Nestor O. Concha, Huizhen Zhao, Patrick McDevitt, and Susan M. Green
- Subjects
Models, Molecular ,animal structures ,Stereochemistry ,DNA damage ,Protein Conformation ,Molecular Sequence Data ,environment and public health ,Biochemistry ,Hydrophobic effect ,chemistry.chemical_compound ,Structure-Activity Relationship ,Protein structure ,Alkaloids ,otorhinolaryngologic diseases ,medicine ,Moiety ,Staurosporine ,Transferase ,Amino Acid Sequence ,Molecular Biology ,Protein Kinase Inhibitors ,Kinase ,Cell Biology ,enzymes and coenzymes (carbohydrates) ,chemistry ,Checkpoint Kinase 1 ,Lactam ,biological phenomena, cell phenomena, and immunity ,Protein Kinases ,medicine.drug - Abstract
Chk1 is a serine-threonine kinase that plays an important role in the DNA damage response, including G(2)/M cell cycle control. UCN-01 (7-hydroxystaurosporine), currently in clinical trials, has recently been shown to be a potent Chk1 inhibitor that abrogates the G(2)/M checkpoint induced by DNA-damaging agents. To understand the structural basis of Chk1 inhibition by UCN-01, we determined the crystal structure of the Chk1 kinase domain in complex with UCN-01. Chk1 structures with staurosporine and its analog SB-218078 were also determined. All three compounds bind in the ATP-binding pocket of Chk1, producing only slight changes in the protein conformation. Selectivity of UCN-01 toward Chk1 over cyclin-dependent kinases can be explained by the presence of a hydroxyl group in the lactam moiety interacting with the ATP-binding pocket. Hydrophobic interactions and hydrogen-bonding interactions were observed in the structures between UCN-01 and the Chk1 kinase domain. The high structural complementarity of these interactions is consistent with the potency and selectivity of UCN-01.
- Published
- 2002
29. Crystal structure of Staphylococcus aureus tyrosyl-tRNA synthetase in complex with a class of potent and specific inhibitors
- Author
-
Kyung O. Johanson, Ward W. Smith, Andrew P. Fosberry, Judith LaLonde, Patrick McDevitt, Pamela Brown, Xiayang Qiu, Susan M. Green, Martin Hibbs, Richard L. Jarvest, Catherine S. V. Houge-Frydrych, Cheryl A. Janson, Ceri J. Lewis, Alison F Chalker, Paul S. Carter, and John M. Berge
- Subjects
Models, Molecular ,Staphylococcus aureus ,Stereochemistry ,Protein Conformation ,Mutant ,Molecular Sequence Data ,Biology ,medicine.disease_cause ,Crystallography, X-Ray ,Biochemistry ,Article ,Piperidines ,Tyrosine-tRNA Ligase ,medicine ,Amino Acid Sequence ,Binding site ,Enzyme Inhibitors ,Furans ,Molecular Biology ,chemistry.chemical_classification ,DNA ligase ,Sequence Homology, Amino Acid ,Substrate (chemistry) ,Dipeptides ,Antimicrobial ,Bridged Bicyclo Compounds, Heterocyclic ,Enzyme ,Tyrosine—tRNA ligase ,chemistry ,Crystallization - Abstract
SB-219383 and its analogues are a class of potent and specific inhibitors of bacterial tyrosyl-tRNA synthetases. Crystal structures of these inhibitors have been solved in complex with the tyrosyl-tRNA synthetase from Staphylococcus aureus, the bacterium that is largely responsible for hospital-acquired infections. The full-length enzyme yielded crystals that diffracted to 2.8 A resolution, but a truncated version of the enzyme allowed the resolution to be extended to 2.2 A. These inhibitors not only occupy the known substrate binding sites in unique ways, but also reveal a butyl binding pocket. It was reported that the Bacillus stearothermophilus TyrRS T51P mutant has much increased catalytic activity. The S. aureus enzyme happens to have a proline at position 51. Therefore, our structures may contribute to the understanding of the catalytic mechanism and provide the structural basis for designing novel antimicrobial agents.
- Published
- 2001
30. Massive abdominal venous cavernous transformation diagnosed by use of EUS
- Author
-
Abraham Mathew, Patrick McDevitt, and Matthew T. Moyer
- Subjects
Adult ,Male ,medicine.medical_specialty ,Biopsy ,Endoscopic ultrasonography ,Sensitivity and Specificity ,Endosonography ,Varicose Veins ,Postoperative Complications ,Varicose veins ,medicine ,Humans ,Radiology, Nuclear Medicine and imaging ,Splanchnic Circulation ,Vein ,Ultrasonography, Interventional ,medicine.diagnostic_test ,Portal Vein ,business.industry ,Angiography ,Gastroenterology ,Hematemesis ,Thrombosis ,Ultrasonography, Doppler ,medicine.disease ,Magnetic Resonance Imaging ,Surgery ,medicine.anatomical_structure ,Pancreatitis ,Abdomen ,Radiology ,medicine.symptom ,Tomography, X-Ray Computed ,business ,Splenorenal Shunt, Surgical - Published
- 2010
- Full Text
- View/download PDF
31. Translating the Reported Ultrasound Liver Size to the Physical Exam Liver Size
- Author
-
Michael A. Bruno, Thomas Riley, and Patrick McDevitt
- Subjects
medicine.medical_specialty ,Hepatology ,business.industry ,Gastroenterology ,medicine ,Physical exam ,Radiology ,Ultrasound liver ,business ,Liver size - Published
- 2009
- Full Text
- View/download PDF
32. Inhibition Of LSD1 As a Therapeutic Strategy For The Treatment Of Acute Myeloid Leukemia
- Author
-
Jessica L. Schneck, Dominic Suarez, Peter J. Tummino, Scott A. Armstrong, Yan Liu, Patrick McDevitt, Helai P. Mohammad, Glenn S. Van Aller, Xinrong Tian, William H. Miller, Kimberly N. Smitheman, Kelly Federowicz, Thau F. Ho, Jeffrey D. Carson, Melissa B. Pappalardi, Johnson Neil W, Monica Cusan, Kasparec Jiri, Rouse Meagan B, Charles F. McHugh, and Ryan G. Kruger
- Subjects
CD86 ,business.industry ,Cellular differentiation ,Immunology ,Myeloid leukemia ,Cell Biology ,Hematology ,Biochemistry ,chemistry.chemical_compound ,Haematopoiesis ,Immunophenotyping ,medicine.anatomical_structure ,chemistry ,Cancer research ,medicine ,Bone marrow ,Growth inhibition ,Stem cell ,business - Abstract
Lysine specific demethylase 1 (LSD1) is a histone H3K4me1/2 demethylase found in various transcriptional co-repressor complexes. These complexes include Histone Deacetylases (HDAC1/2) and Co-Repressor for Element-1-Silencing Transcription factor (CoREST). LSD1 mediated H3K4 demethylation can result in a repressive chromatin environment that silences gene expression. LSD1 has been shown to play a role in development in various contexts. LSD1 can interact with pluripotency factors in human embryonic stem cells and is important for decommissioning enhancers in stem cell differentiation. Beyond embryonic settings, LSD1 is also critical for hematopoietic differentiation. LSD1 is overexpressed in multiple cancer types and recent studies suggest inhibition of LSD1 reactivates the all-trans retinoic acid receptor pathway in acute myeloid leukemia (AML). These studies implicate LSD1 as a key regulator of the epigenome that modulates gene expression through post-translational modification of histones and through its presence in transcriptional complexes. The current study describes the anti-tumor effects of a novel LSD1 inhibitor (GSK2879552) in AML. GSK2879552 is a potent, selective, mechanism-based, irreversible inhibitor of LSD1. Screening of over 150 cancer cell lines revealed that AML cells have a unique requirement for LSD1. While LSD1 inhibition did not affect the global levels of H3K4me1 or H3K4me2, local changes in these histone marks were observed near transcriptional start sites of putative LSD1 target genes. This increase in the transcriptionally activating histone modification correlated with a dose dependent increase in gene expression. Treatment with GSK2879552 promoted the expression of cell surface markers, including CD11b and CD86, associated with a differentiated immunophenotype in 12 of 13 AML cell lines. For example, in SKM-1 cells, increases in cell surface expression of CD86 and CD11b occurred after as early as one day of treatment with EC50 values of 13 and 7 nM respectively. In a separate study using an MV-4-11 engraftment model, increases in CD86 and CD11b were observed as early as 8 hours post dosing. GSK2879552 treatment resulted in a potent anti-proliferative growth effect in 19 of 25 AML cell lines (average EC50 = 38 nM), representing a range of AML subtypes. Potent growth inhibition was also observed on AML blast colony forming ability in 4 out of 5 bone marrow samples derived from primary AML patient samples (average EC50 = 205 nM). The effects of LSD1 inhibition were further characterized in an in vivo mouse model of AML induced by transduction of mouse hematopoietic progenitor cells with a retrovirus encoding MLL-AF9 and GFP. Primary AML cells were transplanted into a cohort of secondary recipient mice and upon engraftment, the mice were treated for 17 days. After 17 days of treatment, control treated mice had 80% GFP+ cells in the bone marrow whereas treated mice possessed 2.8% GFP positive cells (p Together, these data demonstrate that pharmacological inhibition of LSD1 may provide a promising treatment for AML by promoting differentiation and subsequent growth inhibition of AML blasts. GSK2879552 is currently in late preclinical development and clinical trials are anticipated to start in 2014. All studies were conducted in accordance with the GSK Policy on the Care, Welfare and Treatment of Laboratory Animals and were reviewed the Institutional Animal Care and Use Committee either at GSK or by the ethical review process at the institution where the work was performed. Disclosures: Kruger: GlaxoSmithKline Pharmaceuticals: Employment. Mohammad:GlaxoSmithKline Pharmaceuticals: Employment. Smitheman:GlaxoSmithKline Pharmaceuticals: Employment. Liu:GlaxoSmithKline Pharmaceuticals: Employment. Pappalardi:GlaxoSmithKline Pharmaceuticals: Employment. Federowicz:GlaxoSmithKline Pharmaceuticals: Employment. Van Aller:GlaxoSmithKline Pharmaceuticals: Employment. Kasparec:GlaxoSmithKline Pharmaceuticals: Employment. Tian:GlaxoSmithKline Pharmaceuticals: Employment. Suarez:GlaxoSmithKline Pharmaceuticals: Employment. Rouse:GlaxoSmithKline Pharmaceuticals: Employment. Schneck:GlaxoSmithKline Pharmaceuticals: Employment. Carson:GlaxoSmithKline Pharmaceuticals: Employment. McDevitt:GlaxoSmithKline Pharmaceuticals: Employment. Ho:GlaxoSmithKline Pharmaceuticals: Employment. McHugh:GlaxoSmithKline Pharmaceuticals: Employment. Miller:GlaxoSmithKline Pharmaceuticals: Employment. Johnson:GlaxoSmithKline Pharmaceuticals: Employment. Armstrong:Epizyme Inc.: Has consulted for Epizyme Inc. Other. Tummino:GlaxoSmithKline Pharmaceuticals: Employment.
- Published
- 2013
- Full Text
- View/download PDF
33. Osteopontin-stimulated vascular smooth muscle cell migration is mediated by beta 3 integrin
- Author
-
Mary C. Farach-Carson, Patrick J. McKenna, Jeffrey M. Stadel, Eliot H. Ohlstein, William T. Butler, Kyung O. Johanson, Tian Li Yue, Patrick McDevitt, and Giora Z. Feuerstein
- Subjects
Neointima ,Male ,medicine.medical_specialty ,Integrins ,Smooth muscle cell migration ,Sialoglycoproteins ,Cell ,Integrin ,Muscle, Smooth, Vascular ,Rats, Sprague-Dawley ,stomatognathic system ,Cell Movement ,Internal medicine ,medicine ,Extracellular ,Cell Adhesion ,Animals ,Osteopontin ,Aorta ,Cells, Cultured ,Platelet-Derived Growth Factor ,biology ,Dose-Response Relationship, Drug ,Integrin beta3 ,Cell Biology ,Cell biology ,Rats ,medicine.anatomical_structure ,Endocrinology ,biology.protein ,Phosphorylation ,Vitronectin ,Oligopeptides - Abstract
Osteopontin (OPN), a 41-kDa phosphorylated glycoprotein, has been detected in rat aorta and carotid arteries, and expression of its mRNA in blood vessels is strongly increased in response to vascular injury. To investigate the potential role of OPN in vascular pathophysiology, we studied the effect of rat OPN on aortic smooth muscle cell migration and proliferation in vitro . OPN enhanced the migration of rat smooth muscle cells in a time- and concentration-dependent manner with an EC 50 value of 46 ± 11 nmol/liter ( n = 5). The maximal increase in cell migration by OPN was 29-fold over basal levels. OPN-induced smooth muscle cell migration was inhibited in a concentration-dependent manner by the monoclonal antibody F11, which recognizes the rat integrin subunit β 3 . In contrast, polyclonal antiserum recognizing the rat integrin β 1 subunit did not inhibit smooth muscle cell migration in response to OPN, but did block fibronectin-promoted migration. Moreover, OPN-induced smooth muscle cell migration was dependent on the presence of extracellular divalent cations and was significantly inhibited by anti-OPN antibodies. OPN did not stimulate [ 3 H]thymidine incorporation into cultured smooth muscle cells, indicating that it selectively enhanced migration. In view of the pathological significance of arterial smooth muscle cell migration in the formation of intimal thickening, our results suggest that smooth muscle cell recognition of OPN, probably through the vitronectin receptor, α v β 3 , could play a role in the cells' response to vascular injury and especially neointima formation.
- Published
- 1994
34. Mo1357 A Novel Polypectomy Evaluation Platform (PEP) for Objective Benchtop Evaluation of Submucosal Injectables to Improve Safetey and Efficacy of Polypectomy and Endoscopic Mucosal Resection
- Author
-
Travis Holtzman, Matthew T. Moyer, and Patrick McDevitt
- Subjects
medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Gastroenterology ,medicine ,Radiology, Nuclear Medicine and imaging ,Endoscopic mucosal resection ,business ,Polypectomy ,Surgery - Published
- 2011
- Full Text
- View/download PDF
35. Dissection of an Unusual Gastric Mass Using Rat-Tooth Forceps
- Author
-
Patrick McDevitt, Abraham Mathew, and Arnab Biswas
- Subjects
medicine.medical_specialty ,Hepatology ,business.industry ,Forceps ,Gastroenterology ,medicine ,Dissection (medical) ,Gastric mass ,business ,medicine.disease ,Surgery - Published
- 2007
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.