Network


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

Hotspot


Dive into the research topics where Lorin A. Thompson is active.

Publication


Featured researches published by Lorin A. Thompson.


Proceedings of the National Academy of Sciences of the United States of America | 2001

Photochemically enhanced binding of small molecules to the tumor necrosis factor receptor-1 inhibits the binding of TNF-alpha.

Percy H. Carter; Peggy Scherle; Jodi A. Muckelbauer; Matthew E. Voss; Rui-Qin Liu; Lorin A. Thompson; Andrew J. Tebben; Kimberly A. Solomon; Yvonne C. Lo; Zhong Li; Paul Strzemienski; Nikoo Falahatpisheh; Meizhong Xu; Zhongren Wu; Neil A. Farrow; Kal Ramnarayan; Jing Wang; Darryl Rideout; Venkatachalapathi V. Yalamoori; Peter J. Domaille; Dennis J. Underwood; James M. Trzaskos; Steven M. Friedman; Robert Newton; Carl P. Decicco

The binding of tumor necrosis factor alpha (TNF-α) to the type-1 TNF receptor (TNFRc1) plays an important role in inflammation. Despite the clinical success of biologics (antibodies, soluble receptors) for treating TNF-based autoimmune conditions, no potent small molecule antagonists have been developed. Our screening of chemical libraries revealed that N-alkyl 5-arylidene-2-thioxo-1,3-thiazolidin-4-ones were antagonists of this protein–protein interaction. After chemical optimization, we discovered IW927, which potently disrupted the binding of TNF-α to TNFRc1 (IC50 = 50 nM) and also blocked TNF-stimulated phosphorylation of Iκ-B in Ramos cells (IC50 = 600 nM). This compound did not bind detectably to the related cytokine receptors TNFRc2 or CD40, and did not display any cytotoxicity at concentrations as high as 100 μM. Detailed evaluation of this and related molecules revealed that compounds in this class are “photochemically enhanced” inhibitors, in that they bind reversibly to the TNFRc1 with weak affinity (ca. 40–100 μM) and then covalently modify the receptor via a photochemical reaction. We obtained a crystal structure of IV703 (a close analog of IW927) bound to the TNFRc1. This structure clearly revealed that one of the aromatic rings of the inhibitor was covalently linked to the receptor through the main-chain nitrogen of Ala-62, a residue that has already been implicated in the binding of TNF-α to the TNFRc1. When combined with the fact that our inhibitors are reversible binders in light-excluded conditions, the results of the crystallography provide the basis for the rational design of nonphotoreactive inhibitors of the TNF-α–TNFRc1 interaction.


Journal of Pharmacology and Experimental Therapeutics | 2008

P-Glycoprotein Efflux and Other Factors Limit Brain Amyloid β Reduction by β-Site Amyloid Precursor Protein-Cleaving Enzyme 1 Inhibitors in Mice

Jere E. Meredith; Lorin A. Thompson; Jeremy H. Toyn; Donna M. Barten; Jovita Marcinkeviciene; Lisa M. Kopcho; Young Kook Kim; Alan Lin; Valerie Guss; Catherine R. Burton; Lawrence G. Iben; Craig Polson; Joe Cantone; Michael J. Ford; Dieter M. Drexler; Tracey Fiedler; Kimberley A. Lentz; James E. Grace; Janet Kolb; Jason A. Corsa; Maria Pierdomenico; Kelli M. Jones; Richard E. Olson; John E. Macor; Charles F. Albright

Alzheimers disease (AD) is a progressive neurodegenerative disease. Amyloid β (Aβ) peptides are hypothesized to cause the initiation and progression of AD based on pathologic data from AD patients, genetic analysis of mutations that cause early onset forms of AD, and preclinical studies. Based on this hypothesis, β-site amyloid precursor protein (APP)-cleaving enzyme 1 (BACE1) inhibitors are an attractive therapeutic approach for AD because cleavage of the APP by BACE1 is required to form Aβ. In this study, three potent BACE1 inhibitors are characterized. All three inhibitors decrease Aβ formation in cultured cells with IC50 values less than 10 nM. Analysis of APP C-terminal fragments by immunoblotting and Aβ peptides by mass spectrometry showed that these inhibitors decreased Aβ by inhibiting BACE1. An assay for Aβ1–40 in mice was developed and used to show that these BACE1 inhibitors decreased plasma Aβ1–40, but not brain Aβ1–40, in wild-type mice. Because these BACE1 inhibitors were substrates for P-glycoprotein (P-gp), a member of the ATP-binding cassette superfamily of efflux transporters, these inhibitors were administered to P-gp knockout (KO) mice. These studies showed that all three BACE1 inhibitors decreased brain Aβ1–40 in P-gp KO mice, demonstrating that P-gp is a major limitation for development of BACE1 inhibitors to test the amyloid hypothesis. A comparison of plasma Aβ1–40 and brain Aβ1–40 dose responses for these three compounds revealed differences in relative ED50 values, indicating that factors other than P-gp can also contribute to poor brain activity by BACE1 inhibitors.


Bioorganic & Medicinal Chemistry Letters | 2003

Both 5-arylidene-2-thioxodihydropyrimidine-4,6(1H,5H)-diones and 3-thioxo-2,3-dihydro-1H-imidazo[1,5-a]indol-1-ones are light-Dependent tumor necrosis factor-α antagonists

Matthew E. Voss; Percy H. Carter; Andrew J. Tebben; Peggy A. Scherle; Gregory D. Brown; Lorin A. Thompson; Meizhong Xu; Yvonne C. Lo; Gengjie Yang; Rui-Qin Liu; Paul Strzemienski; J.Gerry. Everlof; James M. Trzaskos; Carl P. Decicco

Based on the realization that N-alkyl 5-arylidene-2-thioxo-1,3-thiazolidin-4-ones are tumor necrosis factor-alpha antagonists, we discovered two additional classes of antagonists: 3-thioxo-2,3-dihydro-1H-imidazo[1,5-a]indol-1-ones (via rational design) and 5-arylidene-2-thioxodihydropyrimidine-4,6(1H,5H)-diones (via computer-guided screening). Chemical modification of the lead structures showed that the structure-activity relationship profiles for both of these series were dependent on the electronic properties of the molecules. Subsequent studies showed that they were light-dependent inhibitors.


Bioorganic & Medicinal Chemistry Letters | 2011

Synthesis and SAR of indole-and 7-azaindole-1,3-dicarboxamide hydroxyethylamine inhibitors of BACE-1.

Mendi A. Higgins; F. Christopher Zusi; Yunhui Zhang; Michael F. Dee; Michael F. Parker; Jodi K. Muckelbauer; Daniel M. Camac; Paul E. Morin; Vidhyashankar Ramamurthy; Andrew J. Tebben; Kimberley A. Lentz; James E. Grace; Jovita Marcinkeviciene; Lisa M. Kopcho; Catherine R. Burton; Donna M. Barten; Jeremy H. Toyn; Jere E. Meredith; Charles F. Albright; Joanne J. Bronson; John E. Macor; Lorin A. Thompson

Heterocyclic replacement of the isophthalamide phenyl ring in hydroxyethylamine (HEA) BACE-1 inhibitors was explored. A variety of indole-1,3-dicarboxamide HEAs exhibited potent BACE-1 enzyme inhibition, but displayed poor cellular activity. Improvements in cellular activity and aspartic protease selectivity were observed for 7-azaindole-1,3-dicarboxamide HEAs. A methylprolinol-bearing derivative (10n) demonstrated robust reductions in rat plasma Aβ levels, but did not lower rat brain Aβ due to poor central exposure. The same analog exhibited a high efflux ratio in a bidirectional Caco-2 assay and was likely a substrate of the efflux transporter P-glycoprotein. X-ray crystal structures are reported for two indole HEAs in complex with BACE-1.


Journal of Medicinal Chemistry | 2012

Acyl guanidine inhibitors of β-secretase (BACE-1): optimization of a micromolar hit to a nanomolar lead via iterative solid- and solution-phase library synthesis.

Samuel W. Gerritz; Weixu Zhai; Shuhao Shi; Shirong Zhu; Jeremy H. Toyn; Jere E. Meredith; Lawrence G. Iben; Catherine R. Burton; Charles F. Albright; Andrew C. Good; Andrew J. Tebben; Jodi K. Muckelbauer; Daniel M. Camac; William J. Metzler; Lynda S. Cook; Ramesh Padmanabha; Kimberley A. Lentz; Michael J. Sofia; Michael A. Poss; John E. Macor; Lorin A. Thompson

This report describes the discovery and optimization of a BACE-1 inhibitor series containing an unusual acyl guanidine chemotype that was originally synthesized as part of a 6041-membered solid-phase library. The synthesis of multiple follow-up solid- and solution-phase libraries facilitated the optimization of the original micromolar hit into a single-digit nanomolar BACE-1 inhibitor in both radioligand binding and cell-based functional assay formats. The X-ray structure of representative inhibitors bound to BACE-1 revealed a number of key ligand:protein interactions, including a hydrogen bond between the side chain amide of flap residue Gln73 and the acyl guanidine carbonyl group, and a cation-π interaction between Arg235 and the isothiazole 4-methoxyphenyl substituent. Following subcutaneous administration in rats, an acyl guanidine inhibitor with single-digit nanomolar activity in cells afforded good plasma exposures and a dose-dependent reduction in plasma Aβ levels, but poor brain exposure was observed (likely due to Pgp-mediated efflux), and significant reductions in brain Aβ levels were not obtained.


Archives of Biochemistry and Biophysics | 2003

Comparative studies of active site–ligand interactions among various recombinant constructs of human β-amyloid precursor protein cleaving enzyme

Lisa M. Kopcho; Jianhong Ma; Jovita Marcinkeviciene; Zhihong Lai; Mark R. Witmer; Janet Cheng; Joseph Yanchunas; Jeffrey Tredup; Martin J. Corbett; Deepa Calambur; Michael Wittekind; Manjula Paruchuri; Dharti Kothari; Grace Lee; Subinay Ganguly; Vidhyashankar Ramamurthy; Paul E. Morin; Daniel M. Camac; Robert W King; Amy L Lasut; O Harold Ross; Milton C Hillman; Barbara Fish; Keqiang Shen; Randine L. Dowling; Young Bun Kim; Nilsa R. Graciani; Dale Collins; Andrew P. Combs; Henry J. George

Amyloid precursor protein (APP) cleaving enzyme (BACE) is the enzyme responsible for beta-site cleavage of APP, leading to the formation of the amyloid-beta peptide that is thought to be pathogenic in Alzheimers disease (AD). Hence, BACE is an attractive pharmacological target, and numerous research groups have begun searching for potent and selective inhibitors of this enzyme as a potential mechanism for therapeutic intervention in AD. The mature enzyme is composed of a globular catalytic domain that is N-linked glycosylated in mammalian cells, a single transmembrane helix that anchors the enzyme to an intracellular membrane, and a short C-terminal domain that extends outside the phospholipid bilayer of the membrane. Here we have compared the substrate and active site-directed inhibitor binding properties of several recombinant constructs of human BACE. The constructs studied here address the importance of catalytic domain glycosylation state, inclusion of domains other than the catalytic domain, and incorporation into a membrane bilayer on the interactions of the enzyme active site with peptidic ligands. We find no significant differences in ligand binding properties among these various constructs. These data demonstrate that the nonglycosylated, soluble catalytic domain of BACE faithfully reflects the ligand binding properties of the full-length mature enzyme in its natural membrane environment. Thus, the use of the nonglycosylated, soluble catalytic domain of BACE is appropriate for studies aimed at understanding the determinants of ligand recognition by the enzyme active site.


Bioorganic & Medicinal Chemistry Letters | 2011

Monosubstituted γ-lactam and conformationally constrained 1,3-diaminopropan-2-ol transition-state isostere inhibitors of β-secretase (BACE)

Kenneth M. Boy; Jason M. Guernon; Jianliang Shi; Jeremy H. Toyn; Jere E. Meredith; Donna M. Barten; Catherine R. Burton; Charles F. Albright; Jovita Marcinkeviciene; Andrew C. Good; Andrew J. Tebben; Jodi K. Muckelbauer; Daniel M. Camac; Kimberley A. Lentz; Joanne J. Bronson; Richard E. Olson; John E. Macor; Lorin A. Thompson

The synthesis, evaluation, and structure-activity relationships of a class of γ-lactam 1,3-diaminopropan-2-ol transition-state isostere inhibitors of BACE are discussed. Two strategies for optimizing lead compound 1a are presented. Reducing the overall size of the inhibitors resulted in the identification of γ-lactam 1i, whereas the introduction of conformational constraint on the prime-side of the inhibitor generated compounds such as the 3-hydroxypyrrolidine inhibitor 28n. The full in vivo profile of 1i in rats and 28n in Tg 2576 mice is presented.


The Journal of Neuroscience | 2015

Centrally Delivered BACE1 Inhibitor Activates Microglia, and Reverses Amyloid Pathology and Cognitive Deficit in Aged Tg2576 Mice.

Deepak R. Thakker; Sethu Sankaranarayanan; Marcy R. Weatherspoon; Jonathan Harrison; Maria Pierdomenico; Jennifer M. Heisel; Lorin A. Thompson; Roy Haskell; James E. Grace; Sarah J. Taylor; Charles F. Albright; Lisa L. Shafer

Multiple small-molecule inhibitors of the β-secretase enzyme (BACE1) are under preclinical or clinical investigation for Alzheimers disease (AD). Prior work has illustrated robust lowering of central amyloid β (Aβ) after acute administration of BACE1 inhibitors. However, very few studies have assessed the overall impact of chronically administered BACE1 inhibitors on brain amyloid burden, neuropathology, and behavioral function in aged preclinical models. We investigated the effects of a potent nonbrain-penetrant BACE1 inhibitor, delivered directly to the brain using intracerebroventricular infusion in an aged transgenic mouse model. Intracerebroventricular infusion of the BACE1 inhibitor (0.3–23.5 μg/d) for 8 weeks, initiated in 17-month-old Tg2576 mice, produced dose-dependent increases in brain inhibitor concentrations (0.2–13 μm). BACE1 inhibition significantly reversed the behavioral deficit in contextual fear conditioning, and reduced brain Aβ levels, plaque burden, and associated pathology (e.g., dystrophic neurites), with maximal effects attained with ∼1 μg/d dose. Strikingly, the BACE1 inhibitor also reversed amyloid pathology below baseline levels (amyloid burden at the start of treatment), without adversely affecting cerebral amyloid angiopathy, microhemorrhages, myelination, or neuromuscular function. Inhibitor-mediated decline in brain amyloid pathology was associated with an increase in microglial ramification. This is the first demonstration of chronically administered BACE1 inhibitor to activate microglia, reverse brain amyloid pathology, and elicit functional improvement in an aged transgenic mouse model. Thus, engagement of novel glial-mediated clearance mechanisms may drive disease-modifying therapeutic benefit with BACE1 inhibition in AD.


Bioorganic & Medicinal Chemistry Letters | 2011

Synthesis and in vivo evaluation of cyclic diaminopropane BACE-1 inhibitors.

Lorin A. Thompson; Jianliang Shi; Carl P. Decicco; Andrew J. Tebben; Richard E. Olson; Kenneth M. Boy; Jason M. Guernon; Andrew Good; Ann Y. Liauw; Changsheng Zheng; Robert A. Copeland; Andrew P. Combs; George L. Trainor; Daniel M. Camac; Jodi K. Muckelbauer; Kimberley A. Lentz; James E. Grace; Catherine R. Burton; Jeremy H. Toyn; Donna M. Barten; Jovita Marcinkeviciene; Jere E. Meredith; Charles F. Albright; John E. Macor

The synthesis, evaluation, and structure-activity relationships of a set of related constrained diaminopropane inhibitors of BACE-1 are described. The full in vivo profile of an optimized inhibitor in both normal and P-gp deficient mice is compared with data generated in normal rats.


Journal of Biological Chemistry | 2002

Novel Inhibition of Porcine Pepsin by a Substituted Piperidine PREFERENCE FOR ONE OF THE ENZYME CONFORMERS

Jovita Marcinkeviciene; Lisa M. Kopcho; Tao Yang; Robert A. Copeland; Brian Glass; Andrew P. Combs; Nikoo Falahatpisheh; Lorin A. Thompson

Pepsin inhibition by 3-alkoxy-4-arylpiperidine (substituted piperidine; (3R,4R)-3-(4-bromobenzyloxy)-4-[4-(2-naphthalen-1-yl-2-oxo-ethoxy)phenyl]piperidine) has been studied using steady-state kinetic and pre-equilibrium binding methods. Data were compared with pepstatin A, a well known competitive inhibitor of pepsin. Steady-state analysis reveals that the substituted piperidine likewise behaves as a competitive inhibitor. Pre-equilibrium binding studies indicate that the substituted piperidine can displace a fluorescently labeled statine inhibitor from the enzyme active site. Simulation of the stopped-flow fluorescence transients provided estimates of the K d values of 1.4 ± 0.2 μm and 39 ± 2 nm for the piperidine and the fluorescently labeled statine, respectively. The effects of combinations of these two inhibitors resulted in a series of parallel lines when plotted by the method of Yonetani and Theorell (Yonetani, T., and Theorell, H. (1964) Arch. Biochem. Biophys. 106, 234–251), suggesting that the two inhibitors bind in a mutually exclusive fashion to pepsin. Fitting of the entire data set to the appropriate equation yielded an α factor of 8 ± 1. The magnitude of this factor (∞ > α > 1) can be explained by a conformational distinction between the enzyme species that bind each inhibitor. The effects of pH on the inhibition constants for pepstatin A and the substituted piperidine also suggest that the inhibitors bind to distinct conformational forms of the enzyme. No inhibition by the piperidine was observed at acidic pH, while pepstatin A inhibition is maximal at low pH values. Inhibition by the piperidine was maximal when a group with pK 4.8 ± 0.2 was deprotonated and another group with pK 5.9 ± 0.2 was protonated. Most likely these two groups are the catalytic aspartates with perturbed ionization properties as a result of a significant and unique conformational change. Taken together, these data suggest that the enzyme can readily interconvert between two conformers, one capable of binding substrate and pepstatin A and the other capable of binding the substituted piperidine.

Collaboration


Dive into the Lorin A. Thompson'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