Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Bioorg Med Chem Lett ; 25(4): 841-6, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25575659

ABSTRACT

We present the design, synthesis and biological evaluation of compounds containing a 2-(benzylidene)hexanoic acid scaffold as multi-target directed γ-secretase-modulators. Broad structural variations were undertaken to elucidate the structure-activity-relationships at the 5-position of the aromatic core. Compound 13 showed the most potent activity profile with IC50 values of 0.79µM (Aß42), 0.3µM (5-lipoxygenase) and an EC50 value of 4.64µM for PPARγ-activation. This derivative is the first compound exhibiting low micromolar to nanomolar activities for these three targets. Combining γ-secretase-modulation, PPARγ-agonism and inhibition of 5-lipoxygenase in one compound could be a novel disease-modifying multi-target-strategy for Alzheimer's disease to concurrently address the causative amyloid pathology and secondary pathologies like chronic brain inflammation.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid Precursor Protein Secretases/drug effects , Arachidonate 5-Lipoxygenase/drug effects , Caproates/therapeutic use , Lipoxygenase Inhibitors/pharmacology , PPAR gamma/agonists , Caproates/chemistry , Caproates/pharmacology , Humans , Lipoxygenase Inhibitors/therapeutic use , Structure-Activity Relationship
2.
Bioorg Med Chem ; 20(21): 6523-32, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23000293

ABSTRACT

Supramolecular self-assembly of amyloidogenic peptides is closely associated with numerous pathological conditions. For instance, Alzheimer´s disease (AD) is characterized by abundant amyloid plaques originating from the proteolytic cleavage of the amyloid precursor protein (APP) by ß- and γ-secretases. Compounds named γ-secretase modulators (GSMs) can shift the substrate cleavage specificity of γ-secretase toward the production of non-amyloidogenic, shorter Aß fragments. Herein, we describe the synthesis of highly potent acidic GSMs, equipped with a photoreactive diazirine moiety for photoaffinity labeling. The probes labeled the N-terminal fragment of presenilin (the catalytic subunit of γ-secretase), supporting a mode of action involving binding to γ-secretase. This fundamental step toward the elucidation of the molecular mechanism governing the GSM-induced shift in γ-secretase proteolytic specificity should pave the way for the development of improved drugs against AD.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Azirines/chemistry , Azirines/pharmacology , Animals , Azirines/chemical synthesis , Azirines/radiation effects , CHO Cells , Cricetinae , Dose-Response Relationship, Drug , Models, Molecular , Molecular Structure , Photochemical Processes/radiation effects , Structure-Activity Relationship
3.
ACS Chem Biol ; 7(9): 1488-95, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22725102

ABSTRACT

We present an integrated approach to identify and optimize a novel class of γ-secretase modulators (GSMs) with a unique pharmacological profile. Our strategy included (i) virtual screening through application of a recently developed protocol (PhAST), (ii) synthetic chemistry to discover structure-activity relationships, and (iii) detailed in vitro pharmacological characterization. GSMs are promising agents for treatment or prevention of Alzheimer's disease. They modulate the γ-secretase product spectrum (i.e., amyloid-ß (Aß) peptides of different length) and induce a shift from toxic Aß42 to shorter Aß species such as Aß38 with no or minimal effect on the overall rate of γ-secretase cleavage. We describe the identification of a series of 4-hydroxypyridin-2-one derivatives, which display a novel type of γ-secretase modulation with equipotent inhibition of Aß42 and Aß38 peptide species.


Subject(s)
Alzheimer Disease/enzymology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Pyridines/chemistry , Pyridines/pharmacology , Alzheimer Disease/drug therapy , Amino Acid Sequence , Amyloid Precursor Protein Secretases/chemistry , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Animals , CHO Cells , Cricetinae , Drug Design , Humans , Molecular Sequence Data , Pyridones , Structure-Activity Relationship
4.
PLoS One ; 7(1): e30484, 2012.
Article in English | MEDLINE | ID: mdl-22238696

ABSTRACT

The intramembrane-cleaving protease γ-secretase catalyzes the last step in the generation of toxic amyloid-ß (Aß) peptides and is a principal therapeutic target in Alzheimer's disease. Both preclinical and clinical studies have demonstrated that inhibition of γ-secretase is associated with prohibitive side effects due to suppression of Notch processing and signaling. Potentially safer are γ-secretase modulators (GSMs), which are small molecules that selectively lower generation of the highly amyloidogenic Aß42 peptides but spare Notch processing. GSMs with nanomolar potency and favorable pharmacological properties have been described, but the molecular mechanism of GSMs remains uncertain and both the substrate amyloid precursor protein (APP) and subunits of the γ-secretase complex have been proposed as the molecular target of GSMs. We have generated a potent photo-probe based on an acidic GSM that lowers Aß42 generation with an IC(50) of 290 nM in cellular assays. By combining in vivo photo-crosslinking with affinity purification, we demonstrated that this probe binds the N-terminal fragment of presenilin (PSEN), the catalytic subunit of the γ-secretase complex, in living cells. Labeling was not observed for APP or any of the other γ-secretase subunits. Binding was readily competed by structurally divergent acidic and non-acidic GSMs suggesting a shared mode of action. These findings indicate that potent acidic GSMs target presenilin to modulate the enzymatic activity of the γ-secretase complex.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Cells/drug effects , Enzyme Inhibitors/pharmacology , Presenilins/antagonists & inhibitors , Presenilins/physiology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , CHO Cells , Cells/metabolism , Cells, Cultured , Cricetinae , Cricetulus , Drug Evaluation, Preclinical , HEK293 Cells , Humans , Inhibitory Concentration 50 , Models, Biological , Molecular Targeted Therapy
5.
Bioorg Med Chem ; 19(18): 5372-82, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21873070

ABSTRACT

A novel set of dual γ-secretase/PPARγ modulators characterized by a 2-benzyl hexanoic acid scaffold is presented. Synthetic efforts were focused on the variation of the substitution pattern of the central benzene. Finally, we obtained a new class of 2,5-disubstituted 2-benzylidene hexanoic acid derivatives, which act as dual γ-secretase/PPARγ modulators in the low micromolar range. We have explored broad SAR and successfully improved the dual pharmacological activity and the selectivity profile against potential off-targets such as NOTCH and COX. Compound 17 showed an IC(50) Aß42=2.4 µM and an EC(50) PPARγ=7.2 µM and could be a valuable tool to further evaluate the concept of dual γ-secretase/PPARγ modulators in animal models of Alzheimer's disease.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Caproates/pharmacology , Cyclooxygenase Inhibitors/pharmacology , PPAR gamma/antagonists & inhibitors , Animals , CHO Cells , COS Cells , Caproates/chemical synthesis , Caproates/chemistry , Cell Survival/drug effects , Chlorocebus aethiops , Cricetinae , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Cyclooxygenase Inhibitors/chemical synthesis , Cyclooxygenase Inhibitors/chemistry , Dose-Response Relationship, Drug , Humans , Molecular Conformation , Recombinant Proteins/antagonists & inhibitors , Sheep , Stereoisomerism , Structure-Activity Relationship
6.
Curr Neuropharmacol ; 9(4): 598-622, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22798753

ABSTRACT

Comprehensive evidence supports that oligomerization and accumulation of amyloidogenic Aß42 peptides in brain is crucial in the pathogenesis of both familial and sporadic forms of Alzheimer's disease. Imaging studies indicate that the buildup of Aß begins many years before the onset of clinical symptoms, and that subsequent neurodegeneration and cognitive decline may proceed independently of Aß. This implies the necessity for early intervention in cognitively normal individuals with therapeutic strategies that prioritize safety. The aspartyl protease γ-secretase catalyses the last step in the cellular generation of Aß42 peptides, and is a principal target for anti-amyloidogenic intervention strategies. Due to the essential role of γ-secretase in the NOTCH signaling pathway, overt mechanism-based toxicity has been observed with the first generation of γ-secretase inhibitors, and safety of this approach has been questioned. However, two new classes of small molecules, γ-secretase modulators (GSMs) and NOTCH-sparing γ-secretase inhibitors, have revitalized γ-secretase as a drug target in AD. GSMs are small molecules that cause a product shift from Aß42 towards shorter and less toxic Ab peptides. Importantly, GSMs spare other physiologically important substrates of the γ-secretase complex like NOTCH. Recently, GSMs with nanomolar potency and favorable in vivo properties have been described. In this review, we summarize the knowledge about the unusual proteolytic activity of γ-secretase, and the chemical biology, molecular mechanisms and clinical perspective of compounds that target the γ-secretase complex, with a particular focus on GSMs.

7.
J Neurochem ; 116(3): 385-95, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21091478

ABSTRACT

γ-Secretase modulators (GSMs) inhibit the generation of amyloidogenic Aß42 peptides and are promising agents for treatment or prevention of Alzheimer's disease (AD). Recently, a second generation of GSMs with favorable pharmacological properties has emerged, but preclinical studies to assess their efficacy in vivo are lacking. Such studies rely on transgenic mouse models that express amyloid precursor protein (APP) and presenilin (PSEN) mutations associated with early-onset familial AD. Previously, we have shown that certain PSEN1 mutations attenuated the response of cultured cells to GSMs and potentially confound in vivo studies in AD mouse models. However, different combinations of familial AD mutations might have synergistic or opposing effects, and we have now systematically determined the response of APP and PSEN1 mutations present in current AD models. Using a potent acidic GSM, we found that APP mutations, either single mutations or in combination, did not affect the potency of GSMs. In contrast, all PSEN1 mutations that have been used to accelerate pathological changes in AD models strongly attenuated the Aß42-lowering activity of GSMs with two exceptions (M146L, A246E). Similar results were obtained with potent non-acidic GSMs indicating that the attenuating effect of PSEN1 mutations cannot simply be overcome by increased potency or structural changes. Notably, two non-acidic compounds fully compensated the attenuating effect of the PSEN1-G384A mutation. Taken together, our findings indicate that most AD models with rapid pathology and advanced phenotypes are unsuitable for preclinical GSM studies. However, we also provide evidence that additional compound screens could discover GSMs that are able to break the attenuating effects of PSEN mutations.


Subject(s)
Alzheimer Disease/enzymology , Alzheimer Disease/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/genetics , Mutation/genetics , Presenilin-1/genetics , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/physiology , Animals , CHO Cells , Cricetinae , Cricetulus , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Presenilin-1/physiology , Structure-Activity Relationship
8.
Proc Natl Acad Sci U S A ; 107(33): 14597-602, 2010 Aug 17.
Article in English | MEDLINE | ID: mdl-20679249

ABSTRACT

Following ectodomain shedding by beta-secretase, successive proteolytic cleavages within the transmembrane sequence (TMS) of the amyloid precursor protein (APP) catalyzed by gamma-secretase result in the release of amyloid-beta (Abeta) peptides of variable length. Abeta peptides with 42 amino acids appear to be the key pathogenic species in Alzheimer's disease, as they are believed to initiate neuronal degeneration. Sulindac sulfide, which is known as a potent gamma-secretase modulator (GSM), selectively reduces Abeta42 production in favor of shorter Abeta species, such as Abeta38. By studying APP-TMS dimerization we previously showed that an attenuated interaction similarly decreased Abeta42 levels and concomitantly increased Abeta38 levels. However, the precise molecular mechanism by which GSMs modulate Abeta production is still unclear. In this study, using a reporter gene-based dimerization assay, we found that APP-TMS dimers are destabilized by sulindac sulfide and related Abeta42-lowering compounds in a concentration-dependent manner. By surface plasmon resonance analysis and NMR spectroscopy, we show that sulindac sulfide and novel sulindac-derived compounds directly bind to the Abeta sequence. Strikingly, the attenuated APP-TMS interaction by GSMs correlated strongly with Abeta42-lowering activity and binding strength to the Abeta sequence. Molecular docking analyses suggest that certain GSMs bind to the GxxxG dimerization motif in the APP-TMS. We conclude that these GSMs decrease Abeta42 levels by modulating APP-TMS interactions. This effect specifically emphasizes the importance of the dimeric APP-TMS as a promising drug target in Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Protein Precursor/metabolism , Peptide Fragments/antagonists & inhibitors , Sulindac/analogs & derivatives , Amino Acid Sequence , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/genetics , Animals , CHO Cells , Cricetinae , Cricetulus , Humans , Models, Molecular , Molecular Structure , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Binding/drug effects , Protein Multimerization/drug effects , Sulindac/chemistry , Sulindac/pharmacology , Surface Plasmon Resonance
9.
J Med Chem ; 53(12): 4691-700, 2010 Jun 24.
Article in English | MEDLINE | ID: mdl-20503989

ABSTRACT

We present a novel class of dual modulators of gamma-secretase and peroxisome proliferator-activated receptor gamma (PPARgamma) based on the structure of 2-(bis(phenethoxy)pyrimidine-2-ylthio)hexanoic acid 8 (IC(50)(Abeta42) = 22.8 microM, EC(50)(PPARgamma) = 8.3 microM). The modulation of both targets with approved drugs (i.e., amyloid-beta 42 (Abeta42)-lowering NSAIDs for gamma-secretase and glitazones for PPARgamma) has demonstrated beneficial effects in in vitro and in vivo models of Alzheimer's disease (AD). However, although NSAIDs and PPARgamma agonists share similar structural features, no druglike compounds with dual activities as gamma-secretase modulators (GSMs) and PPARgamma agonists have been designed so far. On the basis of our initial lead structure 8, we present the structure-activity relationships (SARs) of broad structural variations. A significant improvement was reached by the introduction of p-trifluoromethyl substituents at the phenyl residues yielding compound 16 (IC(50)(Abeta42) = 6.0 microM, EC(50)(PPARgamma) = 11.0 microM) and the replacement of the two phenyl residues of 8 by cyclohexyl yielding compound 22 (IC(50)(Abeta42) = 5.1 microM, EC(50)(PPARgamma) = 6.6 microM).


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Caproates/chemical synthesis , PPAR gamma/agonists , Pyrimidines/chemical synthesis , Amyloid beta-Peptides/metabolism , Animals , CHO Cells , COS Cells , Caproates/chemistry , Caproates/pharmacology , Chlorocebus aethiops , Cricetinae , Cricetulus , Cyclooxygenase Inhibitors/chemical synthesis , Cyclooxygenase Inhibitors/chemistry , Cyclooxygenase Inhibitors/pharmacology , Drug Design , Humans , PPAR gamma/genetics , PPAR gamma/metabolism , Peptide Fragments/metabolism , Pyrimidines/chemistry , Pyrimidines/pharmacology , Receptors, Notch/genetics , Receptors, Notch/metabolism , Sheep , Structure-Activity Relationship , Transcriptional Activation/drug effects
10.
Neurodegener Dis ; 6(1-2): 1-8, 2009.
Article in English | MEDLINE | ID: mdl-18349521

ABSTRACT

BACKGROUND: Epidemiological studies have suggested that long-term use of nonsteroidal anti-inflammatory drugs (NSAIDs) is associated with a reduced incidence of Alzheimer's disease (AD). Several mechanisms have been proposed to explain these findings including increased shedding of the soluble ectodomain of the amyloid precursor protein (sAPP), which functions as a neurotrophic and neuroprotective factor in vitroand in vivo. OBJECTIVE: To clarify whether NSAIDs consistently stimulate sAPP secretion. METHODS: 293-EBNA cells with stable overexpression of an APP-alkaline phosphatase fusion protein (APP-AP), SH-SY5Y and PC12 cells or primary telencephalic chicken neurons were treated with ibuprofen or indomethacin. APP shedding was then determined by measuring AP activity in conditioned media, Western blot analysis with antibodies against total sAPP or specific for sAPP-alpha, or in a pulse-chase paradigm. RESULTS: AP activity in conditioned media was not increased after NSAID treatment of 293-EBNA cells whereas it was elevated by phorbol ester. Surprisingly, ibuprofen or indomethacin treatment of SH-SY5Y and PC12 cells expressing endogenous APP did not cause changes in sAPP or sAPP-alpha secretion or downregulation of cellular APP. These findings were further corroborated in primary chicken neuronal cultures. CONCLUSIONS: Using various experimental settings, we were unable to confirm sAPP or sAPP-alpha stimulation with the NSAIDs ibuprofen and indomethacin in transfected and nontransfected cells of neuronal and nonneuronal origin. Importantly, these findings seem to rule out chronic sAPP stimulation as an alternative mechanism of NSAID action in AD.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Alkaline Phosphatase/adverse effects , Amyloid beta-Protein Precursor/drug effects , Animals , CHO Cells , Cell Line , Cell Line, Tumor , Cricetinae , Cricetulus , Ibuprofen/pharmacology , Indomethacin/pharmacology , Kinetics , Neuroblastoma , PC12 Cells/drug effects , Rats , Tetradecanoylphorbol Acetate/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...