Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Int J Neuropsychopharmacol ; 17(1): 117-26, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24103729

ABSTRACT

Prefibrillar assembly of amyloid-ß (Aß) is a major event underlying the development of neuropathology and dementia in Alzheimer's disease (AD). This study determined the neuroprotective properties of an orally bioavailable Aß synaptotoxicity inhibitor, SEN1576. Binding of SEN1576 to monomeric Aß 1-42 was measured using surface plasmon resonance. Thioflavin-T and MTT assays determined the ability of SEN1576 to block Aß 1-42-induced aggregation and reduction in cell viability, respectively. In vivo long-term potentiation (LTP) determined effects on synaptic toxicity induced by intracerebroventricular (i.c.v.) injection of cell-derived Aß oligomers. An operant behavioural schedule measured effects of oral administration following i.c.v. injection of Aß oligomers in normal rats. SEN1576 bound to monomeric Aß 1-42, protected neuronal cells exposed to Aß 1-42, reduced deficits in in vivo LTP and behaviour. SEN1576 exhibits the necessary features of a drug candidate for further development as a disease modifying treatment for the early stages of AD-like dementia.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Peptide Fragments/antagonists & inhibitors , Pyrimidines/pharmacology , Amyloid beta-Peptides/administration & dosage , Amyloid beta-Peptides/metabolism , Animals , Cell Survival/drug effects , Cells, Cultured , Conditioning, Operant/drug effects , Dose-Response Relationship, Drug , Guinea Pigs , Infusions, Intraventricular , Long-Term Potentiation/drug effects , Male , Neurons/drug effects , Neuroprotective Agents/adverse effects , Neuroprotective Agents/therapeutic use , Peptide Fragments/administration & dosage , Peptide Fragments/metabolism , Pyrimidines/adverse effects , Pyrimidines/therapeutic use , Rats
2.
Neuropharmacology ; 75: 458-66, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24035915

ABSTRACT

In the Alzheimer's disease (AD) brain, accumulation of Aß1-42 peptides is suggested to initiate a cascade of pathological events. To date, no treatments are available that can reverse or delay AD-related symptoms in patients. In the current study, we introduce a new Aß toxicity inhibitor, SEN1500, which in addition to its block effect on Aß1-42 toxicity in synaptophysin assays, can be administered orally and cross the blood-brain barrier without adverse effects in mice. In a different set of animals, APPPS1-21 mice were fed with three different doses of SEN1500 (1 mg/kg, 5 mg/kg and 20 mg/kg) for a period of 5 months. Cognition was assessed in a variety of behavioral tests (Morris water maze, social recognition, conditioned taste aversion and passive avoidance). Results suggest a positive effect on cognition with 20 mg/kg SEN1500 compared to control APPPS1-21 mice. However, no changes in soluble or insoluble Aß1-40 and Aß1-42 were detected in the brains of SEN1500-fed mice. SEN1500 also attenuated the effect of Aß1-42 on synaptophysin levels in mouse cortical neurons, which indicated that the compound blocked the synaptic toxicity of Aß1-42. In vitro and in vivo effects presented here suggest that SEN1500 could be an interesting AD therapeutic.


Subject(s)
Alzheimer Disease/complications , Amyloid beta-Peptides/antagonists & inhibitors , Learning Disabilities/drug therapy , Learning Disabilities/etiology , Memory Disorders/etiology , Nitriles/administration & dosage , Peptide Fragments/antagonists & inhibitors , Administration, Oral , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Aniline Compounds/chemistry , Aniline Compounds/pharmacology , Aniline Compounds/therapeutic use , Animals , Avoidance Learning/drug effects , Disease Models, Animal , Gene Expression Regulation/drug effects , Humans , Maze Learning/drug effects , Mice , Mice, Transgenic , Mutation/genetics , Nitriles/chemistry , Presenilin-1/genetics , Pyrimidines/administration & dosage , Pyrimidines/chemistry , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Synaptophysin/metabolism , Taste/drug effects
3.
J Alzheimers Dis ; 37(1): 77-88, 2013.
Article in English | MEDLINE | ID: mdl-23780661

ABSTRACT

The current study examined behavioral and histological effects of amyloid-ß (Aß) protein precursor (AßPP) overexpression in transgenic (Tg) rats created using the same gene, mutation, and promoter as the Tg2576 mouse model of Alzheimer's disease (AD). Male Tg+ rats were bred with female wild-type rats to generate litters of hemizygous Tg+ and Tg- offspring. Tg+ rats and Tg- littermates were tested for memory deficits at 4, 8, and 12 months old using a water-maze procedure. There were no significant behavioral differences between Tg+ rats and Tg- littermates at 4 months old but there were significant differences at 8 and 12 months old, and in probe trials at 8 and 12 months old, the Tg+ rats spent significantly less time and covered less distance in the platform zone. Under acquisition of a fixed-consecutive number schedule at 3 months old, Tg- littermates demonstrated a longer latency to learning the response rule than Tg+ rats; while this might seem paradoxical, it is consistent with the role of overexpression of AßPP in learning. Histological analyses revealed activated astrocytes in brains of Tg+ rats but not Tg- littermates at 6 months old, and thioflavin-S positive staining in the hippocampus and cortex of 17-month old Tg+ rats but not Tg- littermates. Quantification of Aß load in the brain at 22 months indicated high levels of Aß38, Aß40, and Aß42 in the Tg+ rats. These data suggest this model might provide a valuable resource for AD research.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/biosynthesis , Brain/metabolism , Disease Models, Animal , Gene Expression Regulation , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Animals , Brain/pathology , Cricetinae , Female , Humans , Male , Maze Learning/physiology , Mice , Mice, Transgenic , Rats , Rats, Transgenic
4.
Neurobiol Aging ; 34(4): 1116-25, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23164690

ABSTRACT

Oligomers of beta-amyloid (Aß) are implicated in the early memory impairment seen in Alzheimer's disease before to the onset of discernable neurodegeneration. Here, the capacity of a novel orally bioavailable, central nervous system-penetrating small molecule 5-aryloxypyrimidine, SEN1500, to prevent cell-derived (7PA2 [conditioned medium] CM) Aß-induced deficits in synaptic plasticity and learned behavior was assessed. Biochemically, SEN1500 bound to Aß monomer and oligomers, produced a reduction in thioflavin-T fluorescence, and protected a neuronal cell line and primary cortical neurons exposed to synthetic soluble oligomeric Aß(1-42). Electrophysiologically, SEN1500 alleviated the in vitro depression of long-term potentiation induced by both synthetic Aß(1-42) and 7PA2 CM, and alleviated the in vivo depression of long-term potentiation induced by 7PA2 CM, after systemic administration. Behaviorally, oral administration of SEN1500 significantly reduced memory-related deficits in operant responding induced after intracerebroventricular injection of 7PA2 CM. SEN1500 reduced cytotoxicity, acute synaptotoxicity, and behavioral deterioration after in vitro and in vivo exposure to synthetic Aß and 7PA2 CM, and shows promise for development as a clinically viable disease-modifying Alzheimer's disease treatment.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/physiopathology , Memory Disorders/drug therapy , Memory Disorders/physiopathology , Memory/drug effects , Pyrimidines/administration & dosage , Synaptic Transmission/drug effects , Administration, Oral , Alzheimer Disease/complications , Animals , Male , Memory Disorders/complications , Pyrimidines/pharmacokinetics , Rats , Rats, Sprague-Dawley
5.
Biochemistry ; 51(42): 8338-52, 2012 Oct 23.
Article in English | MEDLINE | ID: mdl-23025847

ABSTRACT

Oligomeric forms of ß-amyloid (Aß) have potent neurotoxic activity and are the primary cause of neuronal injury and cell death in Alzheimer's disease (AD). Compounds that perturb oligomer formation or structure may therefore be therapeutic for AD. We previously reported that d-[(chGly)-(Tyr)-(chGly)-(chGly)-(mLeu)]-NH(2) (SEN304) is able to inhibit Aß aggregation and toxicity, shown primarily by thioflavin T fluorescence and MTT (Kokkoni, N. et al. (2006) N-Methylated peptide inhibitors of ß-amyloid aggregation and toxicity. Optimisation of inhibitor structure. Biochemistry 45, 9906-9918). Here we extensively characterize how SEN304 affects Aß(1-42) aggregation and toxicity, using biophysical assays (thioflavin T, circular dichroism, SDS-PAGE, size exclusion chromatography, surface plasmon resonance, traveling wave ion mobility mass spectrometry, electron microscopy, ELISA), toxicity assays in cell culture (MTT and lactate dehydrogenase in human SH-SHY5Y cells, mouse neuronal cell death and synaptophysin) and long-term potentiation in a rat hippocampal brain slice. These data, with dose response curves, show that SEN304 is a powerful inhibitor of Aß(1-42) toxicity, particularly effective at preventing Aß inhibition of long-term potentiation. It can bind directly to Aß(1-42), delay ß-sheet formation and promote aggregation of toxic oligomers into a nontoxic form, with a different morphology that cannot bind thioflavin T. SEN304 appears to work by inducing aggregation, and hence removal, of Aß oligomers. It is therefore a promising lead compound for Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Oligopeptides/pharmacology , Peptide Fragments/antagonists & inhibitors , Protein Multimerization/drug effects , Alzheimer Disease , Animals , Benzothiazoles , Cell Survival , Circular Dichroism , Humans , Long-Term Potentiation/drug effects , Male , Mice , Neurons/drug effects , Protein Structure, Quaternary , Rats , Surface Plasmon Resonance , Thiazoles , Tumor Cells, Cultured
6.
J Alzheimers Dis ; 25(2): 219-29, 2011.
Article in English | MEDLINE | ID: mdl-21654060

ABSTRACT

Behavioral effects of a novel anti-inflammatory SEN1176 were investigated. This pyrrolo[3,2-e][1,2,4]triazolo[1,5-a]pyrimidine suppresses amyloid-ß (Aß)1-42-induced macrophage production of nitric oxide, TNF-α, IL-1ß, and IL-6 in a dose-dependent fashion, an activity profile consistent with SEN1176 being a neuroinflammation inhibitor. Using male Sprague-Dawley rats, SEN1176 was examined relative to detrimental behavioral effects induced following bilateral intrahippocampal (IH) injections of aggregated Aß1-42. The rats were trained to respond under an alternating-lever cyclic-ratio (ALCR) schedule of food reinforcement, enabling measurement of parameters of operant performance that reflect aspects of learning and memory. Under the ALCR schedule, orally administered SEN1176 at 5, 20, or 30 mg/kg was effective in reducing the behavioral deficit caused by bilateral IH aggregated Aß1-42 injections in a dose-related manner over a 90-day treatment period. SEN1176 at 20 and 30 mg/kg significantly reduced lever switching errors and, at doses of 5, 10, and 30 mg/kg, significantly reduced incorrect lever perseverations, indicating a reduction of the behavioral deficit induced as a result of inflammation following IH Aß1-42 injections. When treatment with SEN1176 was instigated 30 days after IH Aß1-42 injections, it resulted in progressive protection, and withdrawal of SEN1176 treatment 60 days after IH Aß1-42 injections revealed partial retention of the protective effect. SEN1176 also significantly reduced numbers of activated astrocytes adjacent to the aggregated Aß1-42 injection sites. These results indicate the potential of SEN1176 for alleviating chronic neuroinflammatory processes related to brain Aß deposition that affect learning and memory in Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/toxicity , Anti-Inflammatory Agents/therapeutic use , Behavioral Symptoms/chemically induced , Behavioral Symptoms/drug therapy , Hippocampus/drug effects , Peptide Fragments/toxicity , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacokinetics , Behavior, Animal/drug effects , Cells, Cultured , Cytokines/metabolism , Dose-Response Relationship, Drug , Macrophages/chemistry , Macrophages/drug effects , Male , Mice , Mice, Inbred BALB C , Nitric Oxide/metabolism , Pyrimidines/chemistry , Pyrimidines/pharmacokinetics , Pyrimidines/therapeutic use , Pyrroles/pharmacokinetics , Pyrroles/therapeutic use , Rats , Rats, Sprague-Dawley , Time Factors , Triazoles/pharmacokinetics , Triazoles/therapeutic use
7.
Behav Brain Res ; 210(1): 32-7, 2010 Jun 26.
Article in English | MEDLINE | ID: mdl-20138088

ABSTRACT

Clinically accessible compounds that arrest or reverse the effects of amyloid-beta (Abeta) on progressively developing behavioural symptomatology and neuropathology in Alzheimer's disease (AD) have yet to become available. However, a viable strategy may be to target and neutralise soluble Abeta oligomers, which have been shown to mediate synaptic dysfunction and to produce cognitive deficits in the intact organism. Inhibiting the aggregation of Abeta is therapeutically attractive, as Abeta aggregation is a pathological event and pharmacological interventions targeting this are likely to have a non-toxic profile. A behavioural assay, the alternating-lever cyclic-ratio schedule, was used to assess the effect of Abeta oligomers and the non-peptide small molecule RS-0406 in male Sprague-Dawley rats. RS-0406 has been shown to inhibit Abeta(1-42) fibrillogenesis and protect against Abeta(1-42)-induced cytotoxicity in primary hippocampal neurons. In the current study, RS-0406 ameliorated the adverse effects of secreted oligomers of human Abeta on behaviour and dose dependently reduced the behavioural effects of Abeta oligomers, with the highest dose, 10microM, maintaining behaviour approximately at control levels. This effect appeared to be central; peripheral confounds having been extensively investigated. This is the first published report on the effects of RS-0406 in vivo and indicates that RS-0406 has potential as a pharmacotherapeutic intervention for behavioural deficits seen in the early stages of AD, and possibly as an intervention in the development of AD neuropathology. Indeed, an analogue of RS-0406 that could be administered peripherally might be a realistic candidate for the clinical treatment of AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Cognition Disorders/drug therapy , Cognition Disorders/metabolism , Diamines/pharmacology , Neuroprotective Agents/pharmacology , Pyridazines/pharmacology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Cell Line , Cognition Disorders/chemically induced , Diamines/administration & dosage , Diamines/chemistry , Dose-Response Relationship, Drug , Humans , Male , Motor Activity/drug effects , Motor Activity/physiology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/chemistry , Neuropsychological Tests , Pyridazines/administration & dosage , Pyridazines/chemistry , Random Allocation , Rats , Rats, Sprague-Dawley , Reinforcement Schedule
8.
J Alzheimers Dis ; 17(1): 33-47, 2009.
Article in English | MEDLINE | ID: mdl-19221409

ABSTRACT

Amyloid-beta (Abeta) peptide is one of the most promising targets for the development of new therapies for Alzheimer's disease (AD). A growing body of evidence suggests a key pathogenic role for soluble oligomers of Abeta, and therapeutics which block the generation of toxic Abeta assemblies may provide successful new treatments for AD. This is therapeutically attractive because the aggregation process is believed to be an exclusively pathological event and therefore compounds targeting this mechanism are more likely to have an acceptable safety profile. A number of studies have shown that AD severity correlates more closely with soluble oligomeric forms of Abeta than with fibrillar forms of the peptide. Thus, blocking the initial stages of Abeta aggregation with small molecules could hold considerable promise as an entry to new therapies for AD. The rapid development in our understanding of toxic amyloid assemblies now provides fresh impetus for this interesting approach, and this review assesses the status of drug development in this area. Recent progress with clinical studies and highlights of new structural series that are showing promise in the discovery/pre-clinical phase are discussed.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid , Peptide Fragments/therapeutic use , Alzheimer Disease/metabolism , Amyloid/antagonists & inhibitors , Amyloid/chemistry , Amyloid/metabolism , Animals , Humans , Peptide Fragments/chemistry
9.
Bioorg Med Chem Lett ; 15(9): 2295-9, 2005 May 02.
Article in English | MEDLINE | ID: mdl-15837312

ABSTRACT

Using a furanylthiazole acetic acid as a starting point, a novel series of benzoxazol-5-yl acetic acid derivatives have been identified as heparanase inhibitors. Several compounds possess an IC50 of approximately 200 nM against heparanase, for example, trans 2-[4-[3-(3,4-dichlorophenylamino)-3-oxo-1-propenyl]-2-fluorophenyl]benzoxazol-5-yl acetic acid (16e). Several of the compounds show anti-angiogenic properties. Improvement to the DMPK profile of compounds has provided compounds of potential use in in vivo models.


Subject(s)
Acetates/pharmacology , Benzoxazoles/pharmacology , Enzyme Inhibitors/pharmacology , Glucuronidase/antagonists & inhibitors , Thiazoles/pharmacology , Acetates/chemical synthesis , Acetates/chemistry , Animals , Benzoxazoles/chemical synthesis , Benzoxazoles/chemistry , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Glucuronidase/blood , Kinetics , Mice , Models, Molecular , Molecular Structure , Structure-Activity Relationship , Thiazoles/chemical synthesis , Thiazoles/chemistry
10.
J Am Chem Soc ; 127(2): 506-7, 2005 Jan 19.
Article in English | MEDLINE | ID: mdl-15643858

ABSTRACT

Chlorination-elimination chemistry coupled with three-component Joullié-Ugi reaction and facile deprotection allowed efficient access to an array of polyhydroxylated pyrrolidines through parallel synthesis that may be considered to be a library of imino (aza) sugars (glycomimetics) and/or dihydroxyprolyl peptides (peptidomimetics). The utility of generating such a library was illustrated by screening against 15 different targets that revealed potent and selective inhibition of the Gaucher's disease glycosyltransferase enzyme glucosylceramide synthase and of primary pathogen model for human hepatitis C virus (HCV) and bovine diarrhoeal virus (BVDV). An observed selectivity for this HCV model over hepatitis B virus and remarkably low toxicity suggest a novel mode of action.


Subject(s)
Antiviral Agents/chemistry , Biomimetic Materials/chemistry , Glycopeptides/chemistry , Pyrrolidines/chemistry , Antiviral Agents/pharmacology , Aza Compounds/chemistry , Aza Compounds/pharmacology , Biomimetic Materials/pharmacology , Carbohydrates/chemistry , Carbohydrates/pharmacology , Diarrhea Viruses, Bovine Viral/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Erythritol/chemistry , Erythritol/pharmacology , Glycopeptides/pharmacology , Glycoside Hydrolases/antagonists & inhibitors , Hepatitis B virus/drug effects , Hydroxyproline/analogs & derivatives , Hydroxyproline/pharmacology , Pyrrolidines/pharmacology , Sugar Alcohols/chemistry , Sugar Alcohols/pharmacology
11.
Bioorg Med Chem Lett ; 14(15): 3975-8, 2004 Aug 02.
Article in English | MEDLINE | ID: mdl-15225710

ABSTRACT

The first inhibitors of fungal protein: mannosyl transferase 1 (PMT1) are described. They are based upon rhodanine-3-acetic acid and several compounds have been identified, for example, 5-[[3-(1-phenylethoxy)-4-(2-phenylethoxy)phenyl]methylene]-4-oxo-2-thioxo-3-thiazolidineacetic acid (5a), which inhibit Candida albicans PMT1 with IC(50)s in the range 0.2-0.5 microM. Members of the series are effective in inducing changes in morphology of C. albicans in vitro that have previously been associated with loss of the transferase activity. These compounds could serve as useful tools for studying the effects of protein O-mannosylation and its relevance in the search for novel antifungal agents.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Mannosyltransferases/antagonists & inhibitors , Rhodanine/analogs & derivatives , Rhodanine/pharmacology , Candida albicans/drug effects , Candida albicans/enzymology , Enzyme Inhibitors/pharmacology , Fungi/drug effects , Fungi/enzymology , Microbial Sensitivity Tests , Rhodanine/chemical synthesis , Structure-Activity Relationship
12.
Bioorg Med Chem Lett ; 14(12): 3269-73, 2004 Jun 21.
Article in English | MEDLINE | ID: mdl-15149688

ABSTRACT

A novel class of 2,3-dihydro-1,3-dioxo-1H-isoindole-5-carboxylic acids are described as inhibitors of the endo-beta-glucuronidase heparanase. Several of the compounds, for example, 2-[4-propylamino-5-[5-(4-chloro)phenyl-benzoxazol-2-yl]phenyl]-2,3-dihydro-1,3-dioxo-1H-isoindole-5-carboxylic acid (9c), display potent heparanase inhibitory activity (IC(50) 200-500 nM) and have high selectivity (>100-fold) over human beta-glucuronidase. They also show anti-angiogenic effects. Such compounds should serve as useful biological tools and may provide a basis for the design of novel therapeutic agents.


Subject(s)
Carboxylic Acids/chemistry , Enzyme Inhibitors/chemistry , Glucuronidase/antagonists & inhibitors , Carboxylic Acids/pharmacology , Enzyme Inhibitors/pharmacology , Glucuronidase/metabolism , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...