Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Glia ; 66(3): 492-504, 2018 03.
Article in English | MEDLINE | ID: mdl-29134678

ABSTRACT

Chronic inflammation represents a central component in the pathogenesis of Alzheimer's disease (AD). Recent work suggests that breaking immune tolerance by Programmed cell Death-1 (PD1) checkpoint inhibition produces an IFN-γ-dependent systemic immune response, with infiltration of the brain by peripheral myeloid cells and neuropathological as well as functional improvements even in mice with advanced amyloid pathology (Baruch et al., (): Nature Medicine, 22:135-137). Immune checkpoint inhibition was therefore suggested as potential treatment for neurodegenerative disorders when activation of the immune system is appropriate. Because a xenogeneic rat antibody (mAb) was used in the study, whether the effect was specific to PD1 target engagement was uncertain. In the present study we examined whether PD1 immunotherapy can lower amyloid-ß pathology in a range of different amyloid transgenic models performed at three pharmaceutical companies with the exact same anti-PD1 isotype and two mouse chimeric variants. Although PD1 immunotherapy stimulated systemic activation of the peripheral immune system, monocyte-derived macrophage infiltration into the brain was not detected, and progression of brain amyloid pathology was not altered. Similar negative results of the effect of PD1 immunotherapy on amyloid brain pathology were obtained in two additional models in two separate institutions. These results show that inhibition of PD1 checkpoint signaling by itself is not sufficient to reduce amyloid pathology and that additional factors might have contributed to previously published results (Baruch et al., (): Nature Medicine, 22:135-137). Until such factors are elucidated, animal model data do not support further evaluation of PD1 checkpoint inhibition as a therapeutic modality for Alzheimer's disease.


Subject(s)
Alzheimer Disease/therapy , Amyloid beta-Peptides/metabolism , Antibodies/administration & dosage , Brain/immunology , Immunotherapy , Programmed Cell Death 1 Receptor/immunology , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Animals , Antibodies/metabolism , Brain/pathology , Disease Models, Animal , Female , Humans , Immunohistochemistry , Interferon-gamma/blood , Male , Mice, Inbred C57BL , Mice, Transgenic , Presenilin-1/genetics , Presenilin-1/metabolism , RNA, Messenger/metabolism , Random Allocation , Spleen/immunology
2.
J Neurosci ; 28(45): 11445-53, 2008 Nov 05.
Article in English | MEDLINE | ID: mdl-18987181

ABSTRACT

Inheritance of the apoE4 allele (epsilon4) increases the risk of developing Alzheimer's disease; however, the mechanisms underlying this association remain elusive. Recent data suggest that inheritance of epsilon4 may lead to reduced apoE protein levels in the CNS. We therefore examined apoE protein levels in the brains, CSF and plasma of epsilon2/2, epsilon3/3, and epsilon4/4 targeted replacement mice. These apoE mice showed a genotype-dependent decrease in apoE levels; epsilon2/2 >epsilon3/3 >epsilon4/4. Next, we sought to examine the relative contributions of apoE4 and apoE3 in the epsilon3/4 mouse brains. ApoE4 represented 30-40% of the total apoE. Moreover, the absolute amount of apoE3 per allele was similar between epsilon3/3 and epsilon3/4 mice, implying that the reduced levels of total apoE in epsilon3/4 mice can be explained by the reduction in apoE4 levels. In culture medium from epsilon3/4 human astrocytoma or epsilon3/3, epsilon4/4 and epsilon3/4 primary astrocytes, apoE4 levels were consistently lower than apoE3. Secreted cholesterol levels were also lower from epsilon4/4 astrocytes. Pulse-chase experiments showed an enhanced degradation and reduced half-life of newly synthesized apoE4 compared with apoE3. Together, these data suggest that astrocytes preferentially degrade apoE4, leading to reduced apoE4 secretion and ultimately to reduced brain apoE levels. Moreover, the genotype-dependent decrease in CNS apoE levels, mirror the relative risk of developing AD, and suggest that low levels of total apoE exhibited by epsilon4 carriers may directly contribute to the disease progression, perhaps by reducing the capacity of apoE to promote synaptic repair and/or Abeta clearance.


Subject(s)
Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Brain/metabolism , Polymorphism, Genetic , Analysis of Variance , Animals , Animals, Newborn , Cells, Cultured , Culture Media, Conditioned/pharmacology , DNA Mutational Analysis , Dose-Response Relationship, Drug , Humans , Hydrocarbons, Fluorinated/pharmacology , Male , Mice , Mice, Transgenic , Neuroglia/drug effects , Neuroglia/metabolism , RNA, Messenger/metabolism , Sulfonamides/pharmacology , Time Factors
3.
J Pharmacol Exp Ther ; 331(2): 598-608, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19671883

ABSTRACT

The presenilin containing gamma-secretase complex is responsible for the regulated intramembraneous proteolysis of the amyloid precursor protein (APP), the Notch receptor, and a multitude of other substrates. gamma-Secretase catalyzes the final step in the generation of Abeta(40) and Abeta(42) peptides from APP. Amyloid beta-peptides (Abeta peptides) aggregate to form neurotoxic oligomers, senile plaques, and congophilic angiopathy, some of the cardinal pathologies associated with Alzheimer's disease. Although inhibition of this protease acting on APP may result in potentially therapeutic reductions of neurotoxic Abeta peptides, nonselective inhibition of the enzyme may cause severe adverse events as a result of impaired Notch receptor processing. Here, we report the preclinical pharmacological profile of GSI-953 (begacestat), a novel thiophene sulfonamide gamma-secretase inhibitor (GSI) that selectively inhibits cleavage of APP over Notch. This GSI inhibits Abeta production with low nanomolar potency in cellular and cell-free assays of gamma-secretase function, and displaces a tritiated analog of GSI-953 from enriched gamma-secretase enzyme complexes with similar potency. Cellular assays of Notch cleavage reveal that this compound is approximately 16-fold selective for the inhibition of APP cleavage. In the human APP-overexpressing Tg2576 transgenic mouse, treatment with this orally active compound results in a robust reduction in brain, plasma, and cerebral spinal fluid Abeta levels, and a reversal of contextual fear-conditioning deficits that are correlated with Abeta load. In healthy human volunteers, oral administration of a single dose of GSI-953 produces dose-dependent changes in plasma Abeta levels, confirming pharmacodynamic activity of GSI-953 in humans.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Sulfonamides/pharmacology , Thiophenes/pharmacology , Adolescent , Adult , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Binding, Competitive , CHO Cells , Cell Line , Cricetinae , Cricetulus , Dogs , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/toxicity , Fear/psychology , Female , Humans , Male , Mice , Mice, Transgenic , Middle Aged , Rats , Rats, Sprague-Dawley , Receptors, Notch/physiology , Signal Transduction/drug effects , Sulfonamides/pharmacokinetics , Sulfonamides/toxicity , Thiophenes/pharmacokinetics , Thiophenes/toxicity , Young Adult
4.
Bioorg Med Chem Lett ; 19(3): 926-9, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-19097890

ABSTRACT

Accumulation of beta-amyloid (Abeta), produced by the proteolytic cleavage of amyloid precursor protein (APP) by beta- and gamma-secretase, is widely believed to be associated with Alzheimer's disease (AD). Research around the high-throughput screening hit (S)-4-chlorophenylsulfonyl isoleucinol led to the identification of the Notch-1-sparing (9.5-fold) gamma-secretase inhibitor (S)-N-(5-chlorothiophene-2-sulfonyl)-beta,beta-diethylalaninol 7.b.2 (Abeta(40/42) EC(50)=28 nM), which is efficacious in reduction of Abeta production in vivo.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Isoleucine/analogs & derivatives , Receptor, Notch1/metabolism , Alcohols , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/chemistry , Animals , Drug Design , Humans , Isoleucine/chemistry , Models, Chemical , Propanolamines/chemistry , Sulfonamides/chemistry
5.
Nat Commun ; 7: 13042, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27727204

ABSTRACT

Inhibition of ß-secretase BACE1 is considered one of the most promising approaches for treating Alzheimer's disease. Several structurally distinct BACE1 inhibitors have been withdrawn from development after inducing ocular toxicity in animal models, but the target mediating this toxicity has not been identified. Here we use a clickable photoaffinity probe to identify cathepsin D (CatD) as a principal off-target of BACE1 inhibitors in human cells. We find that several BACE1 inhibitors blocked CatD activity in cells with much greater potency than that displayed in cell-free assays with purified protein. Through a series of exploratory toxicology studies, we show that quantifying CatD target engagement in cells with the probe is predictive of ocular toxicity in vivo. Taken together, our findings designate off-target inhibition of CatD as a principal driver of ocular toxicity for BACE1 inhibitors and more generally underscore the power of chemical proteomics for discerning mechanisms of drug action.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Cathepsin D/metabolism , Enzyme Inhibitors/toxicity , Eye/pathology , Proteomics/methods , Toxicity Tests , Amyloid Precursor Protein Secretases/metabolism , Animals , Cell Line , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Eye/drug effects , Humans , Inhibitory Concentration 50 , Mass Spectrometry , Mice, Knockout , Molecular Probes/chemical synthesis , Molecular Probes/chemistry , Peptides/metabolism , Protein Binding , Rats, Wistar , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/pathology , Staining and Labeling
6.
J Med Chem ; 58(6): 2678-702, 2015 Mar 26.
Article in English | MEDLINE | ID: mdl-25695670

ABSTRACT

The identification of centrally efficacious ß-secretase (BACE1) inhibitors for the treatment of Alzheimer's disease (AD) has historically been thwarted by an inability to maintain alignment of potency, brain availability, and desired absorption, distribution, metabolism, and excretion (ADME) properties. In this paper, we describe a series of truncated, fused thioamidines that are efficiently selective in garnering BACE1 activity without simultaneously inhibiting the closely related cathepsin D or negatively impacting brain penetration and ADME alignment, as exemplified by 36. Upon oral administration, these inhibitors exhibit robust brain availability and are efficacious in lowering central Amyloid ß (Aß) levels in mouse and dog. In addition, chronic treatment in aged PS1/APP mice effects a decrease in the number and size of Aß-derived plaques. Most importantly, evaluation of 36 in a 2-week exploratory toxicology study revealed no accumulation of autofluorescent material in retinal pigment epithelium or histology findings in the eye, issues observed with earlier BACE1 inhibitors.


Subject(s)
Amidines/chemistry , Amidines/therapeutic use , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Brain/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/therapeutic use , Plaque, Amyloid/drug therapy , Alzheimer Disease/drug therapy , Amidines/pharmacokinetics , Amidines/pharmacology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Animals , Aspartic Acid Endopeptidases/metabolism , Brain/metabolism , Brain/pathology , Dogs , Drug Design , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Humans , Male , Mice , Models, Molecular , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Rats , Rats, Wistar , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/pharmacokinetics , Sulfhydryl Compounds/pharmacology , Sulfhydryl Compounds/therapeutic use
7.
J Med Chem ; 55(7): 3414-24, 2012 Apr 12.
Article in English | MEDLINE | ID: mdl-22420884

ABSTRACT

Replacement of the central, para-substituted fluorophenyl ring in the γ-secretase inhibitor 1 (BMS-708,163) with the bicyclo[1.1.1]pentane motif led to the discovery of compound 3, an equipotent enzyme inhibitor with significant improvements in passive permeability and aqueous solubility. The modified biopharmaceutical properties of 3 translated into excellent oral absorption characteristics (~4-fold ↑ C(max) and AUC values relative to 1) in a mouse model of γ-secretase inhibition. In addition, SAR studies into other fluorophenyl replacements indicate the intrinsic advantages of the bicyclo[1.1.1]pentane moiety over conventional phenyl ring replacements with respect to achieving an optimal balance of properties (e.g., γ-secretase inhibition, aqueous solubility/permeability, in vitro metabolic stability). Overall, this work enhances the scope of the [1.1.1]-bicycle beyond that of a mere "spacer" unit and presents a compelling case for its broader application as a phenyl group replacement in scenarios where the aromatic ring count impacts physicochemical parameters and overall drug-likeness.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Bridged Bicyclo Compounds/chemical synthesis , Oxadiazoles/chemical synthesis , Pentanes/chemical synthesis , Sulfonamides/chemical synthesis , Administration, Oral , Animals , Biological Availability , Brain/metabolism , Bridged Bicyclo Compounds/pharmacokinetics , Bridged Bicyclo Compounds/pharmacology , Cell Line , Dogs , Female , Humans , Mice , Microsomes, Liver/metabolism , Oxadiazoles/pharmacokinetics , Oxadiazoles/pharmacology , Pentanes/pharmacokinetics , Pentanes/pharmacology , Rats , Stereoisomerism , Structure-Activity Relationship , Sulfonamides/pharmacokinetics , Sulfonamides/pharmacology , Tissue Distribution
8.
Alzheimers Res Ther ; 3(5): 28, 2011 Sep 28.
Article in English | MEDLINE | ID: mdl-21943025

ABSTRACT

Animal models have contributed significantly to our understanding of the underlying biological mechanisms of Alzheimer's disease (AD). As a result, over 300 interventions have been investigated and reported to mitigate pathological phenotypes or improve behavior in AD animal models or both. To date, however, very few of these findings have resulted in target validation in humans or successful translation to disease-modifying therapies. Challenges in translating preclinical studies to clinical trials include the inability of animal models to recapitulate the human disease, variations in breeding and colony maintenance, lack of standards in design, conduct and analysis of animal trials, and publication bias due to under-reporting of negative results in the scientific literature. The quality of animal model research on novel therapeutics can be improved by bringing the rigor of human clinical trials to animal studies. Research communities in several disease areas have developed recommendations for the conduct and reporting of preclinical studies in order to increase their validity, reproducibility, and predictive value. To address these issues in the AD community, the Alzheimer's Drug Discovery Foundation partnered with Charles River Discovery Services (Morrisville, NC, USA) and Cerebricon Ltd. (Kuopio, Finland) to convene an expert advisory panel of academic, industry, and government scientists to make recommendations on best practices for animal studies testing investigational AD therapies. The panel produced recommendations regarding the measurement, analysis, and reporting of relevant AD targets, th choice of animal model, quality control measures for breeding and colony maintenance, and preclinical animal study design. Major considerations to incorporate into preclinical study design include a priori hypotheses, pharmacokinetics-pharmacodynamics studies prior to proof-of-concept testing, biomarker measurements, sample size determination, and power analysis. The panel also recommended distinguishing between pilot 'exploratory' animal studies and more extensive 'therapeutic' studies to guide interpretation. Finally, the panel proposed infrastructure and resource development, such as the establishment of a public data repository in which both positive animal studies and negative ones could be reported. By promoting best practices, these recommendations can improve the methodological quality and predictive value of AD animal studies and make the translation to human clinical trials more efficient and reliable.

9.
J Med Chem ; 51(23): 7348-51, 2008 Dec 11.
Article in English | MEDLINE | ID: mdl-19012391

ABSTRACT

SAR on HTS hits 1 and 2 led to the potent, Notch-1-sparing GSI 9, which lowered brain Abeta in Tg2576 mice at 100 mg/kg po. Converting the metabolically labile methyl groups in 9 to trifluoromethyl groups afforded the more stable analogue 10, which had improved in vivo potency. Further side chain modification afforded the potent Notch-1-sparing GSI begacestat (5), which was selected for development for the treatment of Alzheimer's disease.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Drug Discovery , Enzyme Inhibitors/pharmacology , Receptor, Notch1/metabolism , Sulfonamides/pharmacology , Thiophenes/pharmacology , Alzheimer Disease/enzymology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Animals , Crystallography, X-Ray , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Mice , Mice, Transgenic , Models, Molecular , Molecular Conformation , Stereoisomerism , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/chemistry , Thiophenes/chemical synthesis , Thiophenes/chemistry
10.
Mol Cell Neurosci ; 34(4): 621-8, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17336088

ABSTRACT

Recent studies show that intracellular cholesterol levels can modulate the processing of amyloid precursor protein to Abeta peptide. Moreover, cholesterol-rich apoE-containing lipoproteins may also promote Abeta clearance. Agonists of the liver X receptor (LXR) transcriptionally induce genes involved in intracellular lipid efflux and transport, including apoE. Thus, LXR agonists have the potential to both inhibit APP processing and promote Abeta clearance. Here we show that LXR agonist, TO901317, increased hippocampal ABCA1 and apoE and decreased Abeta42 levels in APP transgenic mice. TO901317 had no significant effects on levels of Abeta40, full length APP, or the APP processing products. Next, we examined the effects of TO901317 in the contextual fear conditioning paradigm; TO901317 completely reversed the contextual memory deficit in these mice. These data demonstrate that LXR agonists do not directly inhibit APP processing but rather facilitate the clearance of Abeta42 and may represent a novel therapeutic approach to Alzheimer's disease.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , DNA-Binding Proteins/agonists , Hippocampus/metabolism , Memory/drug effects , Peptide Fragments/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , Sulfonamides/pharmacology , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/metabolism , Alzheimer Disease/metabolism , Animals , Apolipoproteins E/metabolism , Hippocampus/drug effects , Humans , Hydrocarbons, Fluorinated , Liver X Receptors , Male , Memory/physiology , Mice , Mice, Transgenic , Orphan Nuclear Receptors , Reverse Transcriptase Polymerase Chain Reaction
11.
J Biol Chem ; 278(38): 36264-8, 2003 Sep 19.
Article in English | MEDLINE | ID: mdl-12857759

ABSTRACT

Generation of the amyloid peptide through proteolytic processing of the amyloid precursor protein by beta- and gamma-secretases is central to the etiology of Alzheimer's disease. beta-secretase, known more widely as the beta-site amyloid precursor protein cleaving enzyme 1 (BACE1), has been identified as a transmembrane aspartic proteinase, and its ectodomain has been reported to be cleaved and secreted from cells in a soluble form. The extracellular domains of many diverse proteins are known to be cleaved and secreted from cells by a process known as ectodomain shedding. Here we confirm that the ectodomain of BACE1 is secreted from cells and that this processing is up-regulated by agents that activate protein kinase C. A metalloproteinase is involved in the cleavage of BACE1 as hydroxamic acid-based metalloproteinase inhibitors abolish the release of shed BACE1. Using potent and selective inhibitors, we demonstrate that ADAM10 is a strong candidate for the BACE1 sheddase. In addition, we show that the BACE1 sheddase is distinct from alpha-secretase and, importantly, that inhibition of BACE1 shedding does not influence amyloid precursor protein processing at the beta-site.


Subject(s)
Aspartic Acid Endopeptidases/chemistry , Amyloid Precursor Protein Secretases , Binding Sites , Blotting, Western , Cell Line , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Endopeptidases/chemistry , Enzyme Activation , Humans , Hydroxamic Acids/chemistry , Inhibitory Concentration 50 , Models, Chemical , Mutagenesis, Site-Directed , Protein Binding , Protein Kinase C/metabolism , Protein Structure, Tertiary , Up-Regulation
12.
Mol Cell Neurosci ; 22(3): 344-52, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12691736

ABSTRACT

The interaction between myelin-associated glycoprotein (MAG), expressed at the periaxonal membrane of myelin, and receptors on neurons initiates a bidirectional signalling system that results in inhibition of neurite outgrowth and maintenance of myelin integrity. We show that this involves a lipid-raft to lipid-raft interaction on opposing cell membranes. MAG is exclusively located in low buoyancy Lubrol WX-insoluble membrane fractions isolated from whole brain, primary oligodendrocytes, or MAG-expressing CHO cells. Localisation within these domains is dependent on cellular cholesterol and occurs following terminal glycosylation in the trans-Golgi network, characteristics of association with lipid rafts. Furthermore, a recombinant form of MAG interacts specifically with lipid-raft fractions from whole brain and cultured cerebellar granule cells, containing functional MAG receptors GT1b and Nogo-66 receptor and molecules required for transduction of signal from MAG into neurons. The localisation of both MAG and MAG receptors within lipid rafts on the surface of opposing cells may create discrete areas of high avidity multivalent interaction, known to be critical for signalling into both cell types. Localisation within lipid rafts may provide a molecular environment that facilitates the interaction between MAG and multiple receptors and also between MAG ligands and molecules involved in signal transduction.


Subject(s)
Membrane Microdomains/metabolism , Myelin Sheath/metabolism , Myelin-Associated Glycoprotein/metabolism , Neurons/metabolism , Receptors, Cell Surface/metabolism , Animals , CHO Cells , Cricetinae , Detergents/pharmacology , Mice , Neurons/cytology , Polyethylene Glycols/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL