Network


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

Hotspot


Dive into the research topics where Sharon Sweitzer is active.

Publication


Featured researches published by Sharon Sweitzer.


PLOS ONE | 2012

SIRT1 activators suppress inflammatory responses through promotion of p65 deacetylation and inhibition of NF-κB activity.

Hongying Yang; Wei Zhang; Heng Pan; Heidi Galonek Feldser; Elden Lainez; Christine A. Miller; Stewart Leung; Zhong Zhong; Huizhen Zhao; Sharon Sweitzer; Thomas Considine; Thomas V. Riera; Vipin Suri; Brian S. White; James L. Ellis; George P. Vlasuk; Christine Loh

Chronic inflammation is a major contributing factor in the pathogenesis of many age-associated diseases. One central protein that regulates inflammation is NF-κB, the activity of which is modulated by post-translational modifications as well as by association with co-activator and co-repressor proteins. SIRT1, an NAD+-dependent protein deacetylase, has been shown to suppress NF-κB signaling through deacetylation of the p65 subunit of NF-κB resulting in the reduction of the inflammatory responses mediated by this transcription factor. The role of SIRT1 in the regulation of NF-κB provides the necessary validation for the development of pharmacological strategies for activating SIRT1 as an approach for the development of a new class of anti-inflammatory therapeutics. We report herein the development of a quantitative assay to assess compound effects on acetylated p65 protein in the cell. We demonstrate that small molecule activators of SIRT1 (STACs) enhance deacetylation of cellular p65 protein, which results in the suppression of TNFα-induced NF-κB transcriptional activation and reduction of LPS-stimulated TNFα secretion in a SIRT1-dependent manner. In an acute mouse model of LPS-induced inflammation, the STAC SRTCX1003 decreased the production of the proinflammatory cytokines TNFα and IL-12. Our studies indicate that increasing SIRT1-mediated NF-κB deacetylation using small molecule activating compounds is a novel approach to the development of a new class of therapeutic anti-inflammatory agents.


Biochemistry | 2011

A Tale of Two Subunits: How the Neomorphic R132H IDH1 Mutation Enhances Production of αHG

Beth Pietrak; Huizhen Zhao; Hongwei Qi; Chad Quinn; Enoch Gao; Joseph G. Boyer; Nestor O. Concha; Kristin K. Brown; Chaya Duraiswami; Richard Wooster; Sharon Sweitzer; Benjamin J. Schwartz

Heterozygously expressed single-point mutations in isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2, respectively) render these dimeric enzymes capable of producing the novel metabolite α-hydroxyglutarate (αHG). Accumulation of αHG is used as a biomarker for a number of cancer types, helping to identify tumors with similar IDH mutations. With IDH1, it has been shown that one role of the mutation is to increase the rate of conversion from αKG to αHG. To improve our understanding of the function of this mutation, we have detailed the kinetics of the normal (isocitrate to αKG) and neomorphic (αKG to αHG) reactions, as well as the coupled conversion of isocitrate to αHG. We find that the mutant IDH1 is very efficient in this coupled reaction, with the ability to form αHG from isocitrate and NADP(+). The wild type/wild type IDH1 is also able to catalyze this conversion, though it is much more sensitive to concentrations of isocitrate. This difference in behavior can be attributed to the competitive binding between isocitrate and αKG, which is made more favorable for αKG by the neomorphic mutation at arginine 132. Thus, each partial reaction in the heterodimer is functionally isolated from the other. To test whether there is a cooperative effect resulting from the two subunits being in a dimer, we selectively inactivated each subunit with a secondary mutation in the NADP/H binding site. We observed that the remaining, active subunit was unaffected in its associated activity, reinforcing the notion of each subunit being functionally independent. This was further demonstrated using a monomeric form of IDH from Azotobacter vinelandii, which can be shown to gain the same neomorphic reaction when a homologous mutation is introduced into that protein.


Prostaglandins & Other Lipid Mediators | 2013

In vitro and in vivo characterization of a novel soluble epoxide hydrolase inhibitor

Patricia L. Podolin; Brian Bolognese; Joseph F. Foley; Edward Long; Brian Peck; Sandra Umbrecht; Xiaojun Zhang; Penny Zhu; Benjamin Schwartz; Wensheng Xie; Chad Quinn; Hongwei Qi; Sharon Sweitzer; Stephanie Chen; Marc Galop; Yun Ding; Svetlana L. Belyanskaya; David I. Israel; Barry Morgan; David J. Behm; Joseph P. Marino; Edit Kurali; Mary S. Barnette; Ruth J. Mayer; Catherine L. Booth-Genthe; James F. Callahan

Soluble epoxide hydrolase (sEH, EPHX2) metabolizes eicosanoid epoxides, including epoxyeicosatrienoic acids (EETs) to the corresponding dihydroxyeicosatrienoic acids (DHETs), and leukotoxin (LTX) to leukotoxin diol (LTX diol). EETs, endothelium-derived hyperpolarizing factors, exhibit potentially beneficial properties, including anti-inflammatory effects and vasodilation. A novel, potent, selective inhibitor of recombinant human, rat and mouse sEH, GSK2256294A, exhibited potent cell-based activity, a concentration-dependent inhibition of the conversion of 14,15-EET to 14,15-DHET in human, rat and mouse whole blood in vitro, and a dose-dependent increase in the LTX/LTX diol ratio in rat plasma following oral administration. Mice receiving 10 days of cigarette smoke exposure concomitant with oral administration of GSK2256294A exhibited significant, dose-dependent reductions in pulmonary leukocytes and keratinocyte chemoattractant (KC, CXCL1) levels. Mice receiving oral administration of GSK2256294A following 10 days of cigarette smoke exposure exhibited significant reductions in pulmonary leukocytes compared to vehicle-treated mice. These data indicate that GSK2256294A attenuates cigarette smoke-induced inflammation by both inhibiting its initiation and/or maintenance and promoting its resolution. Collectively, these data indicate that GSK2256294A would be an appropriate agent to evaluate the role of sEH in clinical studies, for example in diseases where cigarette smoke is a risk factor, such as chronic obstructive pulmonary disease (COPD) and cardiovascular disease.


Journal of Medicinal Chemistry | 2008

Second Generation of Hydroxyethylamine Bace-1 Inhibitors: Optimizing Potency and Oral Bioavailability.

N Charrier; B Clarke; Leanne Cutler; Emmanuel Demont; C Dingwall; R Dunsdon; P East; J Hawkins; C Howes; I Hussain; P Jeffrey; G Maile; Rosalie Matico; Julie Mosley; Alan Naylor; A O'Brien; Sally Redshaw; Paul Rowland; Soleil; Kathrine J. Smith; Sharon Sweitzer; P Theobald; David Vesey; D.S Walter; G. Wayne

BACE-1 inhibition has the potential to provide a disease-modifying therapy for the treatment of Alzheimers disease. Optimization of a first generation of BACE-1 inhibitors led to the discovery of novel hydroxyethylamines (HEAs) bearing a tricyclic nonprime side. These derivatives have nanomolar cell potency and are orally bioavailable.


Bioorganic & Medicinal Chemistry Letters | 2009

Second generation of BACE-1 inhibitors part 3: Towards non hydroxyethylamine transition state mimetics

Nicolas Charrier; B Clarke; Leanne Cutler; Emmanuel Demont; Colin Dingwall; Rachel Dunsdon; Julie Hawkins; Colin Howes; Julia A. Hubbard; Ishrut Hussain; Graham Maile; Rosalie Matico; Julie Mosley; Alan Naylor; Alistair O’Brien; Sally Redshaw; Paul Rowland; Virginie Soleil; Kathrine J. Smith; Sharon Sweitzer; Pam Theobald; David Vesey; Daryl Simon Walter; Gareth Wayne

Our first generation of hydroxyethylamine BACE-1 inhibitors proved unlikely to provide molecules that would lower amyloid in an animal model at low oral doses. This observation led us to the discovery of a second generation of inhibitors having nanomolar activity in a cell-based assay and with the potential for improved pharmacokinetic profiles. In this Letter, we describe our successful strategy for the optimization of oral bioavailability and also give insights into the design of compounds with the potential for improved brain penetration.


Bioorganic & Medicinal Chemistry Letters | 2008

Bace-1 Inhibitors Part 3: Identification of Hydroxy Ethylamines (Heas) with Nanomolar Potency in Cells.

Paul John Beswick; Nicolas Charrier; B Clarke; Emmanuel Demont; Colin Dingwall; Rachel Dunsdon; Faller A; Robert J. Gleave; Julie Hawkins; Ishrut Hussain; Christopher Norbert Johnson; David Timothy Macpherson; Graham Maile; Rosalie Matico; Peter Henry Milner; Julie Mosley; Antoinette Naylor; A O'Brien; Sally Redshaw; Riddell D; Paul Rowland; John Skidmore; Soleil; Kathrine J. Smith; Steven James Stanway; Geoffrey Stemp; A Stuart; Sharon Sweitzer; P Theobald; David Vesey

This article is focusing on further optimization of previously described hydroxy ethylamine (HEA) BACE-1 inhibitors obtained from a focused library with the support of X-ray crystallography. Optimization of the non-prime side of our inhibitors and introduction of a 6-membered sultam substituent binding to Asn-294 as well as a fluorine in the C-2 position led to derivatives with nanomolar potency in cell-based assays.


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.


Nature Communications | 2015

Crystallographic structure of a small molecule SIRT1 activator-enzyme complex.

Han Dai; April Case; Thomas V. Riera; Thomas Considine; Jessica E. Lee; Yoshitomo Hamuro; Huizhen Zhao; Yong Jiang; Sharon Sweitzer; Beth Pietrak; Benjamin J. Schwartz; Charles A. Blum; Jeremy S. Disch; Richard Caldwell; Bruce G. Szczepankiewicz; Christopher Oalmann; Pui Yee Ng; Brian H. White; Rebecca L. Casaubon; Radha Narayan; Karsten Koppetsch; Francis Bourbonais; Bo Wu; Junfeng Wang; Dongming Qian; Fan Jiang; Cheney Mao; Minghui Wang; Erding Hu; Joseph Wu

SIRT1, the founding member of the mammalian family of seven NAD+-dependent sirtuins, is composed of 747 amino acids forming a catalytic domain and extended N- and C-terminal regions. We report the design and characterization of an engineered human SIRT1 construct (mini-hSIRT1) containing the minimal structural elements required for lysine deacetylation and catalytic activation by small molecule sirtuin-activating compounds (STACs). Using this construct, we solved the crystal structure of a mini-hSIRT1-STAC complex, which revealed the STAC-binding site within the N-terminal domain of hSIRT1. Together with hydrogen-deuterium exchange mass spectrometry (HDX-MS) and site-directed mutagenesis using full-length hSIRT1, these data establish a specific STAC-binding site and identify key intermolecular interactions with hSIRT1. The determination of the interface governing the binding of STACs with human SIRT1 facilitates greater understanding of STAC activation of this enzyme, which holds significant promise as a therapeutic target for multiple human diseases.


Bioorganic & Medicinal Chemistry Letters | 2009

Second generation of BACE-1 inhibitors. Part 1: The need for improved pharmacokinetics

Nicolas Charrier; B Clarke; Leanne Cutler; Emmanuel Demont; Colin Dingwall; Rachel Dunsdon; Julie Hawkins; Colin Howes; Julia A. Hubbard; Ishrut Hussain; Graham Maile; Rosalie Matico; Julie Mosley; Antoinette Naylor; A O'Brien; Sally Redshaw; Paul Rowland; Soleil; Kathrine J. Smith; Sharon Sweitzer; P Theobald; David Vesey; Daryl Simon Walter; Gareth Wayne

Inhibition of the aspartyl protease BACE-1 has the potential to deliver a disease-modifying therapy for Alzheimers disease. We have recently disclosed a series of transition-state mimetic BACE-1 inhibitors showing nanomolar potency in cell-based assays. Amongst them, GSK188909 (compound 2) had favorable pharmacokinetics and was the first orally bioavailable inhibitor reported to demonstrate brain amyloid lowering in an animal model. In this Letter, we describe the reasons that led us to favor a second generation of inhibitors for further in vivo studies.


Bioorganic & Medicinal Chemistry Letters | 2008

BACE-1 inhibitors part 1: identification of novel hydroxy ethylamines (HEAs).

B Clarke; Emmanuel Demont; Colin Dingwall; Rachel Dunsdon; Faller A; Julie Hawkins; Ishrut Hussain; David Timothy Macpherson; Graham Maile; Rosalie Matico; Peter Henry Milner; Julie Mosley; Antoinette Naylor; A O'Brien; Sally Redshaw; Riddell D; Paul Rowland; Soleil; Kathrine J. Smith; Steven James Stanway; Geoffrey Stemp; Sharon Sweitzer; P Theobald; David Vesey; Daryl Simon Walter; Jon R. Ward; Gareth Wayne

Inhibition of the aspartyl protease BACE-1 has the potential to deliver a disease-modifying therapy for Alzheimers disease. Herein, is described the lead generation effort which resulted, with the support of X-ray crystallography, in the discovery of potent inhibitors based on a hydroxy ethylamine (HEA) transition-state mimetic. These inhibitors were capable of lowering amyloid production in a cell-based assay.

Collaboration


Dive into the Sharon Sweitzer'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