Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Feb 03.
Article in English | MEDLINE | ID: mdl-38352474

ABSTRACT

Microtubule-associated protein tau is an intrinsically disordered protein (IDP) that forms characteristic fibrillar aggregates in several diseases, the most well-known of which is Alzheimer's disease (AD). Despite keen interest in disrupting or inhibiting tau aggregation to treat AD and related dementias, there are currently no FDA-approved tau-targeting drugs. This is due, in part, to the fact that tau and other IDPs do not exhibit a single well-defined conformation but instead populate a fluctuating conformational ensemble that precludes finding a stable "druggable" pocket. Despite this challenge, we previously reported the discovery of two novel families of tau ligands, including a class of aggregation inhibitors, identified through a protocol that combines molecular dynamics, structural analysis, and machine learning. Here we extend our exploration of tau druggability with the identification of tryptanthrin and its analogs as potent, substoichiometric aggregation inhibitors, with the best compounds showing potencies in the low nanomolar range even at a ~100-fold molar excess of tau4RD. Moreover, conservative changes in small molecule structure can have large impacts on inhibitory potency, demonstrating that similar structure-activity relationship (SAR) principles as used for traditional drug development also apply to tau and potentially to other IDPs.

2.
ACS Med Chem Lett ; 14(8): 1088-1094, 2023 Aug 10.
Article in English | MEDLINE | ID: mdl-37583812

ABSTRACT

Glutamate plays a key role in cognition and mood, and it has been shown that inhibiting ionotropic glutamate receptors disrupts cognition, while enhancing ionotropic receptor activity is pro-cognitive. One approach to elevating glutamatergic tone has been to antagonize presynaptic metabotropic glutamate receptor 2 (mGluR2). A desire for selectivity over the largely homologous mGluR3 motivated a strategy to achieve selectivity through the identification of mGluR2 negative allosteric modulators (NAMs). Extensive screening and optimization efforts led to the identification of a novel series of 4-arylquinoline-2-carboxamides. This series was optimized for mGluR2 NAM potency, clean off-target activity, and desirable physical properties, which resulted in the identification of improved C4 and C7 substituents. The initial lead compound from this series was Ames-positive in a single strain with metabolic activation, indicating that a reactive metabolite was likely responsible for the genetic toxicity. Metabolic profiling and Ames assessment across multiple analogs identified key structure-activity relationships associated with Ames positivity. Further optimization led to the Ames-negative mGluR2 negative allosteric modulator MK-8768.

3.
Neurobiol Aging ; 106: 12-25, 2021 10.
Article in English | MEDLINE | ID: mdl-34225000

ABSTRACT

Synucleinopathies are neurodegenerative disorders involving pathological alpha-synuclein (αSyn) protein, including dementia with Lewy bodies, multiple system atrophy and Parkinson's disease (PD). Current in vivo models of synucleinopathy include transgenic mice overexpressing αSyn variants and methods based on administration of aggregated, exogenous αSyn. Combining these techniques offers the ability to study consequences of introducing pathological αSyn into primed neuronal environments likely to develop synucleinopathy. Herein, we characterize the impacts pre-formed fibrils (PFFs) of recombinant, human αSyn have in mice overexpressing human A30P αSyn, a mutation associated with autosomal dominant PD. A30P mouse brain contains detergent insoluble αSyn biochemically similar to PD brain, and these mice develop Lewy-like synucleinopathy with age. Administration of PFFs in A30P mice resulted in regionally-specific accumulations of phosphorylated synuclein, microglial induction and a motor phenotype that differed from PFF-induced effects in wildtype mice. Surprisingly, PFF-induced losses of tyrosine hydroxylase were similar in A30P and wildtype mice. Thus, the PFF-A30P model recapitulates key aspects of synucleinopathy with induction of microglia, creating an appropriate system for evaluating neurodegenerative therapeutics.


Subject(s)
Microglia/pathology , Synucleinopathies/etiology , Synucleinopathies/pathology , alpha-Synuclein/adverse effects , Animals , Disease Models, Animal , Gene Expression , Mice, Transgenic , Parkinson Disease/etiology , Parkinson Disease/genetics , Parkinson Disease/pathology , Synucleinopathies/genetics , alpha-Synuclein/administration & dosage , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
4.
J Pharmacol Exp Ther ; 374(2): 252-263, 2020 08.
Article in English | MEDLINE | ID: mdl-32493725

ABSTRACT

Deposition of hyperphosphorylated and aggregated tau protein in the central nervous system is characteristic of Alzheimer disease and other tauopathies. Tau is subject to O-linked N-acetylglucosamine (O-GlcNAc) modification, and O-GlcNAcylation of tau has been shown to influence tau phosphorylation and aggregation. Inhibition of O-GlcNAcase (OGA), the enzyme that removes O-GlcNAc moieties, is a novel strategy to attenuate the formation of pathologic tau. Here we described the in vitro and in vivo pharmacological properties of a novel and selective OGA inhibitor, MK-8719. In vitro, this compound is a potent inhibitor of the human OGA enzyme with comparable activity against the corresponding enzymes from mouse, rat, and dog. In vivo, oral administration of MK-8719 elevates brain and peripheral blood mononuclear cell O-GlcNAc levels in a dose-dependent manner. In addition, positron emission tomography imaging studies demonstrate robust target engagement of MK-8719 in the brains of rats and rTg4510 mice. In the rTg4510 mouse model of human tauopathy, MK-8719 significantly increases brain O-GlcNAc levels and reduces pathologic tau. The reduction in tau pathology in rTg4510 mice is accompanied by attenuation of brain atrophy, including reduction of forebrain volume loss as revealed by volumetric magnetic resonance imaging analysis. These findings suggest that OGA inhibition may reduce tau pathology in tauopathies. However, since hundreds of O-GlcNAcylated proteins may be influenced by OGA inhibition, it will be critical to understand the physiologic and toxicological consequences of chronic O-GlcNAc elevation in vivo. SIGNIFICANCE STATEMENT: MK-8719 is a novel, selective, and potent O-linked N-acetylglucosamine (O-GlcNAc)-ase (OGA) inhibitor that inhibits OGA enzyme activity across multiple species with comparable in vitro potency. In vivo, MK-8719 elevates brain O-GlcNAc levels, reduces pathological tau, and ameliorates brain atrophy in the rTg4510 mouse model of tauopathy. These findings indicate that OGA inhibition may be a promising therapeutic strategy for the treatment of Alzheimer disease and other tauopathies.


Subject(s)
Enzyme Inhibitors/pharmacology , Tauopathies/drug therapy , Tauopathies/metabolism , beta-N-Acetylhexosaminidases/antagonists & inhibitors , tau Proteins/metabolism , Animals , Atrophy/drug therapy , Brain/drug effects , Brain/metabolism , Brain/pathology , Disease Models, Animal , Enzyme Inhibitors/therapeutic use , Locomotion/drug effects , Male , Mice , PC12 Cells , Rats , Tauopathies/pathology , Tauopathies/physiopathology
5.
J Med Chem ; 62(22): 10062-10097, 2019 11 27.
Article in English | MEDLINE | ID: mdl-31487175

ABSTRACT

Inhibition of O-GlcNAcase (OGA) has emerged as a promising therapeutic approach to treat tau pathology in neurodegenerative diseases such as Alzheimer's disease and progressive supranuclear palsy. Beginning with carbohydrate-based lead molecules, we pursued an optimization strategy of reducing polar surface area to align the desired drug-like properties of potency, selectivity, high central nervous system (CNS) exposure, metabolic stability, favorable pharmacokinetics, and robust in vivo pharmacodynamic response. Herein, we describe the medicinal chemistry and pharmacological studies that led to the identification of (3aR,5S,6S,7R,7aR)-5-(difluoromethyl)-2-(ethylamino)-3a,6,7,7a-tetrahydro-5H-pyrano[3,2-d]thiazole-6,7-diol 42 (MK-8719), a highly potent and selective OGA inhibitor with excellent CNS penetration that has been advanced to first-in-human phase I clinical trials.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , beta-N-Acetylhexosaminidases/antagonists & inhibitors , Administration, Oral , Animals , Biological Availability , Brain/drug effects , Dogs , Drug Discovery , Enzyme Inhibitors/blood , Enzyme Inhibitors/pharmacokinetics , Humans , Macaca mulatta , Male , PC12 Cells , Rats , Rats, Wistar , Structure-Activity Relationship , Tauopathies/drug therapy , beta-N-Acetylhexosaminidases/chemistry , beta-N-Acetylhexosaminidases/metabolism
6.
PLoS One ; 13(4): e0195486, 2018.
Article in English | MEDLINE | ID: mdl-29624602

ABSTRACT

Although tau pathology, behavioral deficits, and neuronal loss are observed in patients with tauopathies, the relationship between these endpoints has not been clearly established. Here we found that rTg4510 mice, which overexpress human mutant tau in the forebrain, develop progressive age-dependent increases in locomotor activity (LMA), which correlates with neurofibrillary tangle (NFT) pathology, hyperphosphorylated tau levels, and brain atrophy. To further clarify the relationship between these endpoints, we treated the rTg4510 mice with either doxycycline to reduce mutant tau expression or an O-GlcNAcase inhibitor Thiamet G, which has been shown to ameliorate tau pathology in animal models. We found that both doxycycline and Thiamet G treatments starting at 2 months of age prevented the progression of hyperactivity, slowed brain atrophy, and reduced brain hyperphosphorylated tau. In contrast, initiating doxycycline treatment at 4 months reduced neither brain hyperphosphorylated tau nor hyperactivity, further confirming the relationship between these measures. Collectively, our results demonstrate a unique behavioral phenotype in the rTg4510 mouse model of tauopathy that strongly correlates with disease progression, and that early interventions which reduce tau pathology ameliorate the progression of the locomotor dysfunction. These findings suggest that better understanding the relationship between locomotor deficits and tau pathology in the rTg4510 model may improve our understanding of the mechanisms underlying behavioral disturbances in patients with tauopathies.


Subject(s)
Tauopathies/drug therapy , tau Proteins/metabolism , Animals , Brain/metabolism , Brain/pathology , Disease Models, Animal , Disease Progression , Doxycycline/therapeutic use , Enzyme Inhibitors/therapeutic use , Gene Expression/drug effects , Humans , Mice , Mice, 129 Strain , Mice, Transgenic , Motor Activity/genetics , Motor Activity/physiology , Mutant Proteins/genetics , Mutant Proteins/metabolism , Neurofibrillary Tangles/pathology , Phosphorylation , Pyrans/therapeutic use , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Tauopathies/pathology , Tauopathies/physiopathology , Thiazoles/therapeutic use , beta-N-Acetylhexosaminidases/antagonists & inhibitors , tau Proteins/genetics
7.
Angew Chem Int Ed Engl ; 55(33): 9601-5, 2016 08 08.
Article in English | MEDLINE | ID: mdl-27355874

ABSTRACT

Glycogen synthase kinase-3 (GSK-3) regulates multiple cellular processes in diabetes, oncology, and neurology. N-(3-(1H-1,2,4-triazol-1-yl)propyl)-5-(3-chloro-4-methoxyphenyl)oxazole-4-carboxamide (PF-04802367 or PF-367) has been identified as a highly potent inhibitor, which is among the most selective antagonists of GSK-3 to date. Its efficacy was demonstrated in modulation of tau phosphorylation in vitro and in vivo. Whereas the kinetics of PF-367 binding in brain tissues are too fast for an effective therapeutic agent, the pharmacokinetic profile of PF-367 is ideal for discovery of radiopharmaceuticals for GSK-3 in the central nervous system. A (11) C-isotopologue of PF-367 was synthesized and preliminary PET imaging studies in non-human primates confirmed that we have overcome the two major obstacles for imaging GSK-3, namely, reasonable brain permeability and displaceable binding.


Subject(s)
Brain/drug effects , Brain/diagnostic imaging , Neuroimaging , Oxazoles/pharmacology , Positron-Emission Tomography , Protein Kinase Inhibitors/pharmacology , Triazoles/pharmacology , tau Proteins/antagonists & inhibitors , Brain/metabolism , Crystallography, X-Ray , Dose-Response Relationship, Drug , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Humans , Models, Molecular , Molecular Structure , Oxazoles/chemical synthesis , Oxazoles/chemistry , Phosphorylation/drug effects , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Triazoles/chemical synthesis , Triazoles/chemistry , tau Proteins/metabolism
8.
J Med Chem ; 59(10): 4778-89, 2016 05 26.
Article in English | MEDLINE | ID: mdl-27088900

ABSTRACT

Neurofibrillary tangles (NFTs) made up of aggregated tau protein have been identified as the pathologic hallmark of several neurodegenerative diseases including Alzheimer's disease. In vivo detection of NFTs using PET imaging represents a unique opportunity to develop a pharmacodynamic tool to accelerate the discovery of new disease modifying therapeutics targeting tau pathology. Herein, we present the discovery of 6-(fluoro-(18)F)-3-(1H-pyrrolo[2,3-c]pyridin-1-yl)isoquinolin-5-amine, 6 ([(18)F]-MK-6240), as a novel PET tracer for detecting NFTs. 6 exhibits high specificity and selectivity for binding to NFTs, with suitable physicochemical properties and in vivo pharmacokinetics.


Subject(s)
Drug Discovery , Isoquinolines/chemistry , Molecular Imaging , Neurofibrillary Tangles/pathology , Positron-Emission Tomography , Fluorine Radioisotopes/chemistry , Humans , Isoquinolines/chemical synthesis , Isoquinolines/pharmacokinetics , Molecular Structure , Neurofibrillary Tangles/metabolism
9.
Neurobiol Aging ; 37: 58-65, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26508157

ABSTRACT

In the attempt to elucidate if the "peripheral sink hypothesis" could be a potential mechanism of action for tau removal in passive immunotherapy experiments, we have examined tau levels in serum of chronically injected JNPL3 and Tg4510 transgenic animals. Measurement of tau in serum of mice treated with tau antibodies is challenging because of the antibody interference in sandwich enzyme-linked immunosorbent assays. To address this issue, we have developed a heat-treatment protocol at acidic pH to remove interfering molecules from serum, with excellent recovery of tau. The present data show that pan-tau and conformational antibodies do increase tau in mouse sera. However, these concentrations in serum do not consistently correlate with reductions of tau pathology in brain, suggesting that large elevations of tau species measured in serum are not predictive of efficacy. Here, we describe a reliable method to detect tau in serum of transgenic animals that have undergone tau immunotherapy. Levels of tau in human serum are less than the sensitivity of current assays, although artifactual signals are common. The method may be useful in similarly treated humans, a situation in which false positive signals are likely.


Subject(s)
Antibodies/therapeutic use , Immunization, Passive/methods , Tauopathies/diagnosis , Tauopathies/therapy , tau Proteins/blood , tau Proteins/immunology , Animals , Biomarkers/blood , Disease Models, Animal , Humans , Hydrogen-Ion Concentration , Mice, Transgenic , Predictive Value of Tests , Tauopathies/immunology , tau Proteins/isolation & purification
10.
J Neurosci ; 35(42): 14234-50, 2015 Oct 21.
Article in English | MEDLINE | ID: mdl-26490863

ABSTRACT

Neuronal inclusions of hyperphosphorylated and aggregated tau protein are a pathological hallmark of several neurodegenerative tauopathies, including Alzheimer's disease (AD). The hypothesis of tau transmission in AD has emerged from histopathological studies of the spatial and temporal progression of tau pathology in postmortem patient brains. Increasing evidence in cellular and animal models supports the phenomenon of intercellular spreading of tau. However, the molecular and cellular mechanisms of pathogenic tau transmission remain unknown. The studies described herein investigate tau pathology propagation using human neurons derived from induced pluripotent stem cells. Neurons were seeded with full-length human tau monomers and oligomers and chronic effects on neuronal viability and function were examined over time. Tau oligomer-treated neurons exhibited an increase in aggregated and phosphorylated pathological tau. These effects were associated with neurite retraction, loss of synapses, aberrant calcium homeostasis, and imbalanced neurotransmitter release. In contrast, tau monomer treatment did not produce any measureable changes. This work supports the hypothesis that tau oligomers are toxic species that can drive the spread of tau pathology and neurodegeneration. SIGNIFICANCE STATEMENT: Several independent studies have implicated tau protein as central to Alzheimer's disease progression and cell-to-cell pathology propagation. In this study, we investigated the ability of different tau species to propagate pathology in human neurons derived from induced pluripotent stem cells, which to date has not been shown. We demonstrated that tau oligomers, but not monomers, induce accumulation of pathological, hyperphosphorylated tau. This effect was accompanied with neurite degeneration, loss of synapses, aberrant calcium homeostasis, imbalanced neurotransmitter release, and ultimately with neuronal death. This study bridges various tau pathological phenotypes into a single and relevant induced pluripotent stem cell neuronal model of human disease that can be applied to the discovery of the mechanisms of tau-induced neurodegeneration.


Subject(s)
Induced Pluripotent Stem Cells/physiology , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Neurons/metabolism , tau Proteins/metabolism , tau Proteins/toxicity , Analysis of Variance , Calcium/metabolism , Cell Survival , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Humans , Male , Microfluidics , Microscopy, Atomic Force , Neurotransmitter Agents/metabolism , Phosphorylation , Protein Transport/physiology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , tau Proteins/chemistry
11.
Mol Neurodegener ; 9: 46, 2014 Nov 10.
Article in English | MEDLINE | ID: mdl-25384392

ABSTRACT

BACKGROUND: Tau is an axonal protein that binds to and regulates microtubule function. Hyper-phosphorylation of Tau reduces its binding to microtubules and it is associated with ß-amyloid deposition in Alzheimer's disease. Paradoxically, Tau reduction may prevent ß-amyloid pathology, raising the possibility that Tau mediates intracellular Aß clearance. The current studies investigated the role of Tau in autophagic and proteasomal intracellular Aß1-42 clearance and the subsequent effect on plaque deposition. RESULTS: Tau deletion impaired Aß clearance via autophagy, but not the proteasome, while introduction of wild type human Tau into Tau-/- mice partially restored autophagic clearance of Aß1-42, suggesting that exogenous Tau expression can support autophagic Aß1-42 clearance. Tau deletion impaired autophagic flux and resulted in Aß1-42 accumulation in pre-lysosomal autophagic vacuoles, affecting Aß1-42 deposition into the lysosome. This autophagic defect was associated with decreased intracellular Aß1-42 and increased plaque load in Tau-/- mice, which displayed less cell death. Nilotinib, an Abl tyrosine kinase inhibitor that promotes autophagic clearance mechanisms, reduced Aß1-42 only when exogenous human Tau was expressed in Tau-/- mice. CONCLUSIONS: These studies demonstrate that Tau deletion affects intracellular Aß1-42 clearance, leading to extracellular plaque.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Peptide Fragments/metabolism , tau Proteins/metabolism , Alzheimer Disease/metabolism , Animals , Autophagy , Blotting, Western , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Extracellular Space/metabolism , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Plaque, Amyloid/metabolism , Proteasome Endopeptidase Complex/metabolism
12.
J Mol Med (Berl) ; 92(4): 373-86, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24337465

ABSTRACT

UNLABELLED: Alzheimer's disease (AD) is a neurodegenerative disorder associated with amyloid accumulation and autophagic changes. Parkin is an E3 ubiquitin ligase involved in proteasomal and autophagic clearance. We previously demonstrated decreased parkin solubility and interaction with the key autophagy enzyme beclin-1 in AD, but tyrosine kinase inhibition restored parkin-beclin-1 interaction. In the current studies, we determined the mechanisms of nilotinib-induced parkin-beclin-1 interaction, which leads to amyloid clearance. Nilotinib increased endogenous parkin levels and ubiquitination, which may enhance parkin recycling via the proteasome, leading to increased activity and interaction with beclin-1. Parkin solubility was decreased and autophagy was altered in amyloid expressing mice, suggesting that amyloid stress affects parkin stability, leading to failure of protein clearance via the lysosome. Isolation of autophagic vacuoles revealed amyloid and parkin accumulation in autophagic compartments but nilotinib decreased insoluble parkin levels and facilitated amyloid deposition into lysosomes in wild type, but not parkin(-/-) mice, further underscoring an essential role for endogenous parkin in amyloid clearance. These results suggest that nilotinib boosts the autophagic machinery, leading to increased level of endogenous parkin that undergoes ubiquitination and interacts with beclin-1 to facilitate amyloid clearance. These data suggest that nilotinib-mediated autophagic changes may trigger parkin response via increased protein levels, providing a therapeutic strategy to reduce Aß and Tau in AD. KEY MESSAGE: Parkin solubility (stability) is decreased in AD and APP transgenic mice. Nilotinib-induced autophagic changes increase endogenous parkin level. Increased parkin level leads to ubiquitination and proteasomal recycling. Re-cycling decreases insoluble parkin and increases parkin-beclin-1 interaction. Beclin-1-parkin interaction enhances amyloid clearance.


Subject(s)
Amyloid/metabolism , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/drug effects , Alzheimer Disease/enzymology , Animals , Autophagy , Cell Line, Tumor , Enzyme Stability , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Rats
13.
J Neurogenet ; 25(4): 127-33, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22091726

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder that causes early memory impairment, followed by profound progressive cognitive decline, and eventually death. Neurofibrillary tangles (NFTs) are one of the histopathological hallmarks of AD. NFTs are deposits of insoluble aggregates of the microtubule-binding protein tau, left behind following neuronal loss. Intracellular aggregates of tau, either in soluble or insoluble forms, are thought to disrupt cellular machinery and synaptic function and ultimately lead to neuronal death. As the ultimate pathological endpoint in AD is neuronal loss, there is significant interest in understanding the causes of tau aggregation and deposition in the brain as a potential therapeutic avenue for AD. Post-translational modifications on tau are thought to be an important regulatory mechanism that may contribute to the propensity of tau to aggregate and form NFTs. In addition to phosphorylation, numerous other post-translational modifications have been observed on tau protein. The mechanisms that cause aggregation of tau are unknown, but it is likely that post-translational modifications other than phosphorylation also regulate this process. This review will discuss several post-translational modifications of tau and their roles in modulation of tau function and aggregation in AD.


Subject(s)
Alzheimer Disease , Protein Processing, Post-Translational/genetics , tau Proteins/genetics , tau Proteins/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Animals , Drug Design , Humans , Nerve Degeneration/drug therapy , Nerve Degeneration/metabolism , Nerve Degeneration/physiopathology , Neurofibrillary Tangles/drug effects , Neurofibrillary Tangles/genetics , Neurofibrillary Tangles/metabolism , Phosphorylation/drug effects , Phosphorylation/physiology , Protein Processing, Post-Translational/drug effects
14.
J Alzheimers Dis ; 25(4): 655-69, 2011.
Article in English | MEDLINE | ID: mdl-21483096

ABSTRACT

Accumulation of small soluble assemblies of amyloid-ß (Aß)(42) in the brain is thought to play a key role in the pathogenesis of Alzheimer's disease. As a result, there has been much interest in finding small molecules that inhibit the formation of synaptotoxic Aß(42) oligomers that necessitates sensitive methods for detecting the initial steps in the oligomerization of Aß(42). Modeling suggests that oligomerized Aß(42) adopts a conformation in which the C-terminus is embedded in the center, whereas the N-terminus is exposed at the periphery of the oligomer. Here we report that an inverse change in Aß(42) C-terminal and N-terminal epitope accessibility provides the basis of a sensitive method for assessing early steps in Aß(42) oligomerization. Using ELISA and AlphaLISA, we found that Aß(42) C-terminal immunoreactivity decreased in a time- and concentration-dependent manner under conditions favoring oligomerization. This reduction was accompanied by an increase in the N-terminal immunoreactivity, suggesting that assemblies with multiple exposed N-terminal epitopes were detected. Importantly the assay generates a robust window between monomers and oligomers at as low as 1 nM Aß(42). Using this assay, known oligomerization inhibitors produced a dose-dependent unmasking of the Aß(42) C-terminal epitope. After automation, the assay proved to be highly reproducible and effective for high throughput screening of small molecules that inhibit Aß(42) oligomerization.


Subject(s)
Amyloid beta-Peptides/analysis , Amyloid beta-Peptides/biosynthesis , Immunoassay/methods , Peptide Fragments/analysis , Peptide Fragments/biosynthesis , Alzheimer Disease/immunology , Animals , Dimerization , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Epitopes/genetics , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Light , Microscopy, Atomic Force , Neurons/metabolism , Protein Conformation , Rats , Reproducibility of Results , Scattering, Radiation
15.
Bioorg Med Chem Lett ; 21(5): 1429-33, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21295469

ABSTRACT

The synthesis and structure-activity relationships for a novel series of 6-amino-4-(pyrimidin-4-yl)pyridones derived from a high throughput screening hit are discussed. Optimization of lead matter afforded compounds with good potency, selectivity and central nervous system (CNS) exposure.


Subject(s)
Glycogen Synthase Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Pyridones/chemical synthesis , Central Nervous System/drug effects , Crystallography, X-Ray , Humans , Inhibitory Concentration 50 , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/pharmacology , Pyridones/chemistry , Pyridones/pharmacology , Structure-Activity Relationship
16.
Bioorg Med Chem Lett ; 21(9): 2631-6, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21269825

ABSTRACT

The synthesis and structure-activity relationship (SAR) of a novel series of di-substituted imidazoles, derived from modification of DAPT, are described. Subsequent optimization led to identification of a highly potent series of inhibitors that contain a ß-amine in the imidazole side-chain resulting in a robust in vivo reduction of plasma and brain Aß in guinea pigs. The therapeutic index between Aß reductions and changes in B-cell populations were studied for compound 10 h.


Subject(s)
Alzheimer Disease , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Enzyme Activation/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Imidazoles/chemical synthesis , Imidazoles/pharmacology , Amination/drug effects , Amyloid beta-Peptides/blood , Amyloid beta-Peptides/metabolism , Animals , Biological Assay , Diamide/chemical synthesis , Diamide/chemistry , Diamide/pharmacology , Enzyme Inhibitors/chemistry , Guinea Pigs , HeLa Cells , Humans , Imidazoles/chemistry , Inhibitory Concentration 50 , Molecular Structure , Structure-Activity Relationship
17.
Bioorg Med Chem Lett ; 21(9): 2637-40, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21269827

ABSTRACT

A novel series of tetralin containing amino imidazoles, derived from modification of the corresponding phenyl acetic acid derivatives is described. Replacement of the amide led to identification of a potent series of tetralin-amino imidazoles with robust central efficacy. The reduction of brain Aß in guinea pigs in the absence of changes in B-cells suggested a potential therapeutic index with respect to APP processing compared with biomarkers of notch related toxicity. Optimization of the FTOC to plasma concentrations at the brain Aß EC(50) lead to the identification of compound 14f (PF-3084014) which was selected for clinical development.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Enzyme Activation/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Tetrahydronaphthalenes/chemical synthesis , Tetrahydronaphthalenes/pharmacology , Valine/analogs & derivatives , Animals , Biological Assay , Drug Design , Enzyme Inhibitors/chemistry , Guinea Pigs , Imidazoles/chemical synthesis , Imidazoles/chemistry , Imidazoles/pharmacology , Inhibitory Concentration 50 , Molecular Structure , Structure-Activity Relationship , Tetrahydronaphthalenes/chemistry , Valine/chemical synthesis , Valine/chemistry , Valine/pharmacology
18.
Bioorg Med Chem Lett ; 19(19): 5703-7, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19700321

ABSTRACT

Utilizing structure-based drug design, a 4-aminoimidazole heterocyclic core was synthesized as a replacement for a 2-aminothiazole due to potential metabolically mediated toxicity. The synthetic route utilized allowed for ready synthesis of 1-substituted-4-aminoimidazoles. SAR exploration resulted in the identification of a novel cis-substituted cyclobutyl group that gave improved enzyme and cellular potency against cdk5/p25 with up to 30-fold selectivity over cdk2/cyclin E.


Subject(s)
Alzheimer Disease/drug therapy , Cyclin-Dependent Kinase 5/metabolism , Imidazoles/chemistry , Nerve Tissue Proteins/metabolism , Animals , Binding Sites , Caco-2 Cells , Crystallography, X-Ray , Cyclin E/antagonists & inhibitors , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Drug Design , Humans , Imidazoles/chemical synthesis , Imidazoles/pharmacology , Mice , Mice, Knockout , Nerve Tissue Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Structure-Activity Relationship
19.
J Neurosci Methods ; 169(1): 16-22, 2008 Mar 30.
Article in English | MEDLINE | ID: mdl-18160105

ABSTRACT

We have previously developed a solid-phase extraction (SPE) procedure to enable the detection of beta-amyloid (Abeta) peptides in brain tissue from non-transgenic animals. We have now adapted these methods to enrich the Abeta fraction in cerebrospinal fluid (CSF) and plasma. Human CSF and plasma and Tg2576 mouse plasma were subjected to guanidine denaturation followed by SPE in 96-well cassettes. The resulting eluates could be concentrated significantly to enhance detection of low-abundance Abeta peptides by immunoassay. The concentrated eluates diluted in a linear fashion with consistent recovery between SPE columns. This technique was therefore used to facilitate quantification of Abeta1-X, 1-40, 1-42, and 1-38 peptides in normal human CSF and plasma samples. SPE sample preparation was also applied to the plasma of mice dosed peripherally with a monoclonal antibody raised against Abeta. When such samples were assayed directly, the presence of the systemically administered antibody interfered with the subsequent immunoassay, by preventing detection of antibody-bound Abeta. After subjecting plasma from antibody-treated animals to denaturation and SPE, the antibody-antigen complex was disrupted, and the Abeta fraction could be isolated from the antibody-containing fraction. Application of this method allowed for detection of a 100-fold increase in plasma Abeta1-40 following treatment of Tg2576 mice or wild type littermate control mice with Abeta40-specific monoclonal antibody 9TL. Given the availability of a variety of SPE matrices, we hypothesize that these methods could facilitate plasma antigen retrieval using multiple therapeutic antibody approaches.


Subject(s)
Amyloid beta-Peptides/blood , Enzyme-Linked Immunosorbent Assay/methods , Immunotherapy/methods , Neurochemistry/methods , Solid Phase Extraction/methods , Alzheimer Disease/drug therapy , Alzheimer Disease/immunology , Amyloid beta-Peptides/analysis , Amyloid beta-Peptides/cerebrospinal fluid , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Brain/drug effects , Brain/immunology , Brain/metabolism , Humans , Mice , Mice, Transgenic , Peptide Fragments/analysis , Peptide Fragments/blood , Peptide Fragments/cerebrospinal fluid , Predictive Value of Tests , Treatment Outcome , Up-Regulation/drug effects , Up-Regulation/immunology
20.
Biochem Pharmacol ; 75(5): 1093-103, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18076866

ABSTRACT

Increasing beta-amyloid (Abeta) clearance may alter the course of Alzheimer's disease progression and attenuate amyloid plaque pathology. Insulin-like growth factor I (IGF-1) augmentation has been suggested to increase Abeta clearance by facilitating transport of Abeta out of the brain. The availability of safe agents that increase IGF-1 levels therefore makes IGF-1 elevation an attractive target for disease modifying therapy in AD. The present series of studies sought to replicate published paradigms in which peripheral IGF-1 administration lowered brain Abeta acutely, with reduction in plaque pathology after chronic treatment. Thus Abeta levels were measured in several animal models following treatments that elevated IGF-1. Administration of IGF-1 to young or old rats for up to 3 days had no effect on Abeta levels in brain, CSF, or plasma. In adult beagles, 4 days of dosing with the growth hormone secretagogue, CP-424391, doubled baseline plasma IGF-1 levels, yet failed to alter CSF or plasma Abeta. 5-day treatment of young Tg2576 mice with IGF-1 produced robust elevations of IGF-1 levels in plasma, but no effects on Abeta were detected in brain, CSF, or plasma. Finally, 11-month-old Tg2576 mice were implanted with subcutaneous minipumps delivering IGF-1 for 1 month. No significant changes in Abeta (by ELISA or Western blot), plaque pathology, or phospho-tau epitopes were detected. These results do not demonstrate acute or chronic actions of peripherally administered IGF-1 on Abeta levels or the phosphorylation state of tau and therefore do not suggest any disease-modifying benefits of IGF-1 restorative therapy for AD through these mechanisms.


Subject(s)
Amyloid beta-Peptides/metabolism , Brain/drug effects , Insulin-Like Growth Factor I/pharmacology , Amyloid beta-Peptides/cerebrospinal fluid , Animals , Brain/metabolism , Cell Line , Dogs , Female , Humans , Insulin-Like Growth Factor I/pharmacokinetics , Male , Mice , Mice, Transgenic , Myoblasts/cytology , Myoblasts/drug effects , Myoblasts/metabolism , Myosin Heavy Chains/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar , Recombinant Proteins/blood , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...