Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
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.
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
3.
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
4.
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
5.
Biochem Soc Trans ; 37(Pt 4): 692-6, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19614577

ABSTRACT

The aggregation of numerous peptides or proteins has been linked to the onset of disease, including Abeta (amyloid beta-peptide) in AD (Alzheimer's disease), asyn (alpha-synuclein) in Parkinson's disease and amylin in Type 2 diabetes. Diverse amyloidogenic proteins can often be cut down to an SRE (self-recognition element) of as few as five residues that retains the ability to aggregate. SREs can be used as a starting point for aggregation inhibitors. In particular, N-methylated SREs can bind to a target on one side, but have hydrogen-bonding blocked on their methylated face, interfering with further assembly. We applied this strategy to develop Abeta toxicity inhibitors. Our compounds, and a range of compounds from the literature, were compared under the same conditions, using biophysical and toxicity assays. Two N-methylated D-peptide inhibitors with unnatural side chains were the most effective and can reverse Abeta-induced inhibition of LTP (long-term potentiation) at concentrations as low as 10 nM. An SRE in asyn (VAQKTV) was identified using solid-state NMR. When VAQKTV was N-methylated, it was able to disrupt asyn aggregation. N-methylated derivatives of the SRE of amylin are also able to inhibit amylin aggregation.


Subject(s)
Peptide Fragments/metabolism , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/chemistry , Animals , Drug Design , Humans , Magnetic Resonance Spectroscopy , Molecular Structure , Parkinsonian Disorders/drug therapy , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Peptides/chemical synthesis , Peptides/chemistry , Peptides/pharmacology , Protein Folding/drug effects , alpha-Synuclein/antagonists & inhibitors , alpha-Synuclein/chemistry
6.
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
7.
Biochemistry ; 45(32): 9906-18, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16893191

ABSTRACT

The key pathogenic event in the onset of Alzheimer's disease (AD) is believed to be the aggregation of the beta-amyloid (Abeta) peptide into toxic oligomers. Molecules that interfere with this process may therefore act as therapeutic agents for the treatment of AD. N-Methylated peptides (meptides) are a general class of peptide aggregation inhibitors that act by binding to one face of the aggregating peptide but are unable to hydrogen bond on the other face, because of the N-methyl group replacing a backbone NH group. Here, we optimize the structure of meptide inhibitors of Abeta aggregation, starting with the KLVFF sequence that is known to bind to Abeta. We varied the meptide length, N-methylation sites, acetylation, and amidation of the N and C termini, side-chain identity, and chirality, via five compound libraries. Inhibitor activity was tested by thioflavin T binding, affinity chromatography, electron microscopy, and an 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide toxicity assay. We found that inhibitors should have all d chirality, have a free N terminus but an amidated C terminus, and have large, branched hydrophobic side chains at positions 1-4, while the side chain at position 5 was less important. A single N-methyl group was necessary and sufficient. The most active compound, d-[(chGly)-(Tyr)-(chGly)-(chGly)-(mLeu)]-NH(2), was more active than all previously reported peptide inhibitors. Its related non-N-methylated analogues were insoluble and toxic.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/toxicity , Oligopeptides/pharmacology , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/toxicity , Peptides/pharmacology , Protein Structure, Quaternary/drug effects , Amino Acid Sequence , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/ultrastructure , Animals , Chromatography, Affinity , Molecular Sequence Data , PC12 Cells , Peptide Fragments/chemistry , Peptide Fragments/ultrastructure , Peptide Library , Rats , Structure-Activity Relationship , Tetrazolium Salts/metabolism , Thiazoles/metabolism , Toxicity Tests
SELECTION OF CITATIONS
SEARCH DETAIL
...