Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Bioorg Med Chem Lett ; 28(23-24): 3793-3797, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30327146

ABSTRACT

Neuronal voltage-gated potassium channels, KV7s, are the molecular mediators of the M current and regulate membrane excitability in the central and peripheral neuronal systems. Herein, we report novel small molecule KV7 openers that demonstrate anti-seizure activities in electroshock and pentylenetetrazol-induced seizure models without influencing Rotarod readouts in mice. The anti-seizure activity was determined to be proportional to the unbound concentration in the brain. KV7 channels are also expressed in the bladder smooth muscle (detrusor) and activation of these channels may cause localized undesired effects. Therefore, the impact of individual KV7 isoforms was investigated in human detrusor tissue using a panel of KV7 openers with distinct activity profiles among KV7 isoforms. KCNQ4 and KCNQ5 mRNA were highly expressed in detrusor tissue, yet a compound that has significantly reduced activity on homomeric KV7.4 did not reduce detrusor contraction. This may suggest that the homomeric KV7.4 channel plays a less significant role in bladder contraction and further investigation is needed.


Subject(s)
Anticonvulsants/chemistry , Anticonvulsants/pharmacology , Epilepsy/drug therapy , KCNQ Potassium Channels/metabolism , Seizures/drug therapy , Animals , Anticonvulsants/therapeutic use , Epilepsy/metabolism , Humans , Mice , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Protein Isoforms/metabolism , Seizures/metabolism , Urinary Bladder/drug effects , Urinary Bladder/metabolism
2.
Antioxid Redox Signal ; 23(5): 358-74, 2015 Aug 10.
Article in English | MEDLINE | ID: mdl-26135714

ABSTRACT

AIMS: The NADPH oxidase (NOX) family of enzymes catalyzes the formation of reactive oxygen species (ROS). NOX enzymes not only have a key role in a variety of physiological processes but also contribute to oxidative stress in certain disease states. To date, while numerous small molecule inhibitors have been reported (in particular for NOX2), none have demonstrated inhibitory activity in vivo. As such, there is a need for the identification of improved NOX inhibitors to enable further evaluation of the biological functions of NOX enzymes in vivo as well as the therapeutic potential of NOX inhibition. In this study, both the in vitro and in vivo pharmacological profiles of GSK2795039, a novel NOX2 inhibitor, were characterized in comparison with other published NOX inhibitors. RESULTS: GSK2795039 inhibited both the formation of ROS and the utilization of the enzyme substrates, NADPH and oxygen, in a variety of semirecombinant cell-free and cell-based NOX2 assays. It inhibited NOX2 in an NADPH competitive manner and was selective over other NOX isoforms, xanthine oxidase, and endothelial nitric oxide synthase enzymes. Following systemic administration in mice, GSK2795039 abolished the production of ROS by activated NOX2 enzyme in a paw inflammation model. Furthermore, GSK2795039 showed activity in a murine model of acute pancreatitis, reducing the levels of serum amylase triggered by systemic injection of cerulein. INNOVATION AND CONCLUSIONS: GSK2795039 is a novel NOX2 inhibitor that is the first small molecule to demonstrate inhibition of the NOX2 enzyme in vivo.


Subject(s)
Aminopyridines/pharmacology , Drug Discovery , Enzyme Inhibitors/pharmacology , Membrane Glycoproteins/metabolism , NADPH Oxidases/metabolism , Sulfonamides/pharmacology , Aminopyridines/chemistry , Animals , Cells, Cultured , Enzyme Inhibitors/therapeutic use , Male , Membrane Glycoproteins/antagonists & inhibitors , Mice, Inbred C57BL , NADPH Oxidase 2 , NADPH Oxidases/antagonists & inhibitors , Pancreatitis/drug therapy , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Sulfonamides/chemistry
4.
Neuropharmacology ; 70: 278-86, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23485401

ABSTRACT

Accumulation of amyloid ß (Aß) in brain is a pathological hallmark of Alzheimer's disease (AD). Aß is generated after sequential cleavage of its parental molecule, amyloid precursor protein (APP), by ß- and γ-secretases. Inhibition of γ-secretase activity is an effective approach for the reduction of Aß levels. Since γ-secretase targets many different substrates, selective inhibition of its cleavage of APP is believed to be critical in order to avoid undesirable side effects. γ-Secretase modulator (GSM) shifts the cleavage site on APP and production of amyloidogenic to non-amyloidogenic Aß fragments. Since GSMs only modulate and do not block cleavage of γ-secretase substrates, they are believed less likely to produce untoward adverse reactions. Here, we report in vivo Aß-lowering profiles of a pyridazine and a pyridine-derived GSM: GSM-C (Wan et al., 2011a) and GSM-D (Wan et al., 2011b). Both compounds reduced Aß40 and Aß42 productions, increased shorter Aß fragments, and had little effect on Notch signaling (∼100-fold selective). They had excellent oral bioavailability (97.8% for GSM-C, ∼100% for GSM-D) and good brain permeability (free brain to free blood AUC ratio of 0.41 and 1.10 for GSM-C and GSM-D, respectively). Oral administration of these compounds in both acute and sub-chronic conditions reduced Aß levels in plasma and brain in rats in a dose- and time-dependent manner. Therefore, GSM-C and GSM-D represent two GSMs that are orally bioavailable and brain-permeable. They could serve as excellent tools in the investigation of the role of Aß peptides in AD pathogenesis.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Pyridazines/pharmacology , Pyridazines/pharmacokinetics , Pyridines/pharmacology , Pyridines/pharmacokinetics , Administration, Oral , Amyloid beta-Peptides/blood , Animals , Biological Availability , Brain/drug effects , Brain/metabolism , Dose-Response Relationship, Drug , Male , Neurons/enzymology , Neurons/metabolism , Primary Cell Culture , Pyridazines/administration & dosage , Pyridines/administration & dosage , Rats , Receptors, Notch/metabolism , Thymocytes/drug effects
5.
J Neurochem ; 124(1): 59-68, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23083210

ABSTRACT

Several epidemiological and preclinical studies suggest that non-steroidal anti-inflammatory drugs (NSAIDs), which inhibit cyclooxygenase (COX), reduce the risk of Alzheimer's disease (AD) and can lower ß-amyloid (Aß) production and inhibit neuroinflammation. However, follow-up clinical trials, mostly using selective cyclooxygenase (COX)-2 inhibitors, failed to show any beneficial effect in AD patients with mild to severe cognitive deficits. Recent data indicated that COX-1, classically viewed as the homeostatic isoform, is localized in microglia and is actively involved in brain injury induced by pro-inflammatory stimuli including Aß, lipopolysaccharide, and interleukins. We hypothesized that neuroinflammation is critical for disease progression and selective COX-1 inhibition, rather than COX-2 inhibition, can reduce neuroinflammation and AD pathology. Here, we show that treatment of 20-month-old triple transgenic AD (3 × Tg-AD) mice with the COX-1 selective inhibitor SC-560 improved spatial learning and memory, and reduced amyloid deposits and tau hyperphosphorylation. SC-560 also reduced glial activation and brain expression of inflammatory markers in 3 × Tg-AD mice, and switched the activated microglia phenotype promoting their phagocytic ability. The present findings are the first to demonstrate that selective COX-1 inhibition reduces neuroinflammation, neuropathology, and improves cognitive function in 3 × Tg-AD mice. Thus, selective COX-1 inhibition should be further investigated as a potential therapeutic approach for AD.


Subject(s)
Alzheimer Disease/complications , Amyloidogenic Proteins/metabolism , Cyclooxygenase Inhibitors/therapeutic use , Memory Disorders/drug therapy , Memory Disorders/etiology , Pyrazoles/therapeutic use , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Animals , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Glial Fibrillary Acidic Protein/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , Mutation/genetics , Phagocytes/drug effects , Phosphorylation/drug effects , Presenilin-1/genetics , tau Proteins/genetics
6.
Neurobiol Aging ; 33(4): 825.e15-24, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21741124

ABSTRACT

Fyn kinase phosphorylates tau and exacerbates amyloid beta (Aß)-mediated synaptic dysfunction. However, Fyn also increases the nonpathological cleavage of amyloid precursor protein (APP), suggesting opposing roles for Fyn in the pathogenesis of Alzheimer's disease (AD). To determine the effect of Fyn on both Aß and tau pathologies, we crossed homozygous Alzheimer's disease triple transgenic (3×Tg) mice harboring mutations in amyloid precursor protein, presenilin-1, and tau with wild-type or Fyn knockout mice to generate Fyn(+/+)3×Tg(+/-) or Fyn(+/-)3×Tg(+/-) mice. We found that Fyn(+/-)3×Tg(+/-) mice had increased soluble and intracellular Aß, and these changes were accompanied by impaired performance on the Morris water maze at 18 months. Fyn(+/-)3×Tg(+/-) mice had decreased phosphorylated tau at 15-18 months (as did Fyn knockout mice), but Fyn(+/-)3×Tg(+/-) mice had increased phosphorylated tau by 24 months. In addition, we observed that Fyn(+/-)3×Tg(+/-) males were delayed in developing Aß pathology compared with females, and displayed better spatial learning performance at 18 months. Overall, these findings suggest that loss of Fyn at early stages of disease increases soluble Aß accumulation and worsens spatial learning in the absence of changes in tau phosphorylation.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides/metabolism , Brain/metabolism , Learning Disabilities/etiology , Proto-Oncogene Proteins c-fyn/deficiency , Space Perception/physiology , tau Proteins/metabolism , Age Factors , Alzheimer Disease/complications , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Animals , Brain/pathology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression Regulation/genetics , Humans , Learning Disabilities/genetics , Male , Maze Learning/physiology , Mice , Mice, Transgenic , Presenilin-1/genetics , Sex Factors , tau Proteins/genetics
7.
J Neurochem ; 121(2): 277-86, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22035227

ABSTRACT

γ-secretase inhibitors (GSIs) have been developed to reduce amyloid-ß (Aß) production for the treatment of Alzheimer's disease by inhibiting the cleavage of amyloid precursor protein (APP). However, cross-inhibitory activity on the processing of Notch can cause adverse reactions. To avoid these undesirable effects, γ-secretase modulators (GSMs) are being developed to selectively reduce toxic Aß production without perturbing Notch signaling. As it is also known that GSIs can cause a paradoxical increase of plasma Aß over the baseline after a transient reduction (known as Aß-rebound), we asked if GSMs would cause a similar rebound and what the potential mechanism might be. Our studies were performed with one GSI (LY-450139) and two chemically distinct GSMs. Although LY-450139 caused Aß-rebound as expected in rat plasma, the two GSMs did not. Inhibition of APP processing by LY-450139 induced an accumulation of γ-secretase substrates, α- and ß-C-terminal fragments of APP, but neither GSM caused such an accumulation. In conclusion, we discover that GSMs, unlike GSIs, do not cause Aß-rebound, possibly because of the lack of accumulation of ß-C-terminal fragments. GSMs may be superior to GSIs in the treatment of Alzheimer's disease not only by sparing Notch signaling but also by avoiding Aß-rebound.


Subject(s)
Alanine/analogs & derivatives , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/drug effects , Amyloid beta-Peptides/biosynthesis , Azepines/pharmacology , Alanine/pharmacokinetics , Alanine/pharmacology , Amyloid beta-Peptides/genetics , Animals , Area Under Curve , Azepines/pharmacokinetics , Cell Line , Cells, Cultured , Chromatography, High Pressure Liquid , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Humans , Male , Mice , Mice, Transgenic , Neurons/pathology , Peptide Fragments/metabolism , Plaque, Amyloid/pathology , Rats , Rats, Sprague-Dawley , Receptors, Notch/drug effects , Signal Transduction/drug effects
8.
Bioorg Med Chem Lett ; 21(16): 4832-5, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21742495

ABSTRACT

SAR of a novel series of pyridine-derived γ-secretase modulators is described. Compound 5 was found to be a potent modulator in vitro, which on further profiling, was found to decrease Aß42 and Aß40, and maintain (or increase) the levels of total Aß. Furthermore, representative compounds 1 and 5 demonstrated in vivo efficacy to lower Aß42 in the brain without altering Notch processing in the peripheral.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Pyridines/pharmacology , Animals , Biological Availability , Cytochrome P-450 Enzyme Inhibitors , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Molecular Structure , Pyridines/chemical synthesis , Pyridines/chemistry , Rats , Stereoisomerism , Structure-Activity Relationship
9.
Bioorg Med Chem Lett ; 21(13): 4016-9, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21636276

ABSTRACT

SAR of a novel series of pyridazine-derived γ-secretase modulators is described. Compound 25 was found to be a potent modulator in vitro, which on further profiling, was found to decrease Aß42 and Aß40, and maintain the levels of total Aß. Furthermore, 25 demonstrated excellent pharmacokinetic parameters as well as good CNS penetration in the rat.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Pyridazines/chemical synthesis , Amyloid beta-Peptides/metabolism , Animals , Cell Survival , Cells, Cultured , Enzyme Activation/drug effects , Humans , Inhibitory Concentration 50 , Pyridazines/chemistry , Pyridazines/pharmacology , Rats , Structure-Activity Relationship
10.
Bioorg Med Chem Lett ; 21(13): 3992-6, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21621412

ABSTRACT

In the course of a ß-site APP-cleaving enzyme 1 (BACE1) inhibitor discovery project an in situ synthesis/screening protocol was employed to prepare 120 triazole-linked reduced amide isostere inhibitors. Among these compounds, four showed modest (single digit micromolar) BACE1 inhibition. Our ligand design was based on a potent reduced amide isostere 1, wherein the P(2) amide moiety was replaced with an anti-1,2,3-triazole unit. Unfortunately, this replacement resulted in a 1000-fold decrease in potency. Docking studies of triazole-linked reduced amide isostere A3Z10 and potent oxadiazole-linked tertiary carbinamine 2a with BACE1 suggests that the docking poses of A3Z10 and 2a in the active sites are quite similar, with one exception. In the docked structures the placement of the protonated amine that engages D228 differs considerably between 2a and A3Z10. This difference could account for the lower BACE1 inhibition potency of A3Z10 and related compounds relative to 2a.


Subject(s)
Alzheimer Disease/drug therapy , Amides/chemical synthesis , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Drug Discovery , Enzyme Inhibitors/chemical synthesis , Triazoles/chemistry , Amides/chemistry , Amides/therapeutic use , Catalytic Domain , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/therapeutic use , Humans , Inhibitory Concentration 50 , Models, Molecular , Molecular Structure , Oxidation-Reduction , Small Molecule Libraries
11.
PLoS One ; 6(1): e16259, 2011 Jan 17.
Article in English | MEDLINE | ID: mdl-21264226

ABSTRACT

Humanin (HN), a 24-residue peptide, was identified as a novel neuroprotective factor and shows anti-cell death activity against a wide spectrum of Alzheimer's disease (AD)-related cytotoxicities, including exposure to amyloid beta (Abeta), in vitro. We previously demonstrated that the injection of S14G-HN, a highly potent HN derivative, into brain ameliorated memory loss in an Abeta-injection mouse model. To fully understand HN's functions under AD-associated pathological conditions, we examined the effect of S14G-HN on triple transgenic mice harboring APP(swe), tau(P310L), and PS-1(M146V) that show the age-dependent development of multiple pathologies relating to AD. After 3 months of intranasal treatment, behavioral analyses showed that S14G-HN ameliorated cognitive impairment in male mice. Moreover, ELISA and immunohistochemical analyses showed that Abeta levels in brains were markedly lower in S14G-HN-treated male and female mice than in vehicle control mice. We also found the expression level of neprilysin, an Abeta degrading enzyme, in the outer molecular layer of hippocampal formation was increased in S14G-HN-treated mouse brains. NEP activity was also elevated by S14G-HN treatment in vitro. These findings suggest that decreased Abeta level in these mice is at least partly attributed to S14G-HN-induced increase of neprilysin level. Although HN was identified as an anti-neuronal death factor, these results indicate that HN may also have a therapeutic effect on amyloid accumulation in AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Intracellular Signaling Peptides and Proteins/pharmacology , Memory Disorders/drug therapy , Age Factors , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/drug effects , Animals , Brain/metabolism , Female , Hippocampus , Intracellular Signaling Peptides and Proteins/administration & dosage , Intracellular Signaling Peptides and Proteins/therapeutic use , Male , Mice , Mice, Transgenic , Neprilysin/biosynthesis
12.
J Neuroinflammation ; 7: 57, 2010 Sep 28.
Article in English | MEDLINE | ID: mdl-20920207

ABSTRACT

BACKGROUND: Passive immunization with antibodies directed to Aß decreases brain Aß/amyloid burden and preserves memory in transgenic mouse models of Alzheimer's disease (AD). This therapeutic strategy is under intense scrutiny in clinical studies, but its application is limited by neuroinflammatory side effects (autoimmune encephalitis and vasogenic edema). METHODS: We intravenously administered the monoclonal Aß protofibril antibody PFA1 to aged (22 month) male and female 3 × tg AD mice with intermediate or advanced AD-like neuropathologies, respectively, and measured brain and serum Aß and CNS cytokine levels. We also examined 17 month old 3 × tg AD female mice with intermediate pathology to determine the effect of amyloid burden on responses to passive immunization. RESULTS: The 22 month old male mice immunized with PFA1 had decreased brain Aß, increased serum Aß, and no change in CNS cytokine levels. In contrast, 22 month old immunized female mice revealed no change in brain Aß, decreased serum Aß, and increased CNS cytokine levels. Identical experiments in younger (17 month old) female 3 × tg AD mice with intermediate AD-like neuropathologies revealed a trend towards decreased brain Aß and increased serum Aß accompanied by a decrease in CNS MCP-1. CONCLUSIONS: These data suggest that passive immunization with PFA1 in 3 × tg AD mice with intermediate disease burden, regardless of sex, is effective in mediating potentially therapeutic effects such as lowering brain Aß. In contrast, passive immunization of mice with a more advanced amyloid burden may result in potentially adverse effects (encephalitis and vasogenic edema) mediated by certain proinflammatory cytokines.


Subject(s)
Alzheimer Disease/immunology , Alzheimer Vaccines/therapeutic use , Amyloid beta-Peptides/metabolism , Brain/pathology , Immunization, Passive , Alzheimer Disease/pathology , Alzheimer Disease/prevention & control , Amyloid beta-Peptides/immunology , Animals , Blotting, Western , Brain/immunology , Brain/metabolism , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Male , Mice , Mice, Transgenic , tau Proteins/immunology , tau Proteins/metabolism
13.
PLoS One ; 4(9): e7134, 2009 Sep 22.
Article in English | MEDLINE | ID: mdl-19771166

ABSTRACT

The metabolism of the amyloid precursor protein (APP) and tau are central to the pathobiology of Alzheimer's disease (AD). We have examined the in vivo turnover of APP, secreted APP (sAPP), Abeta and tau in the wild-type and Tg2576 mouse brain using cycloheximide to block protein synthesis. In spite of overexpression of APP in the Tg2576 mouse, APP is rapidly degraded, similar to the rapid turnover of the endogenous protein in the wild-type mouse. sAPP is cleared from the brain more slowly, particularly in the Tg2576 model where the half-life of both the endogenous murine and transgene-derived human sAPP is nearly doubled compared to wild-type mice. The important Abeta degrading enzymes neprilysin and IDE were found to be highly stable in the brain, and soluble Abeta40 and Abeta42 levels in both wild-type and Tg2576 mice rapidly declined following the depletion of APP. The cytoskeletal-associated protein tau was found to be highly stable in both wild-type and Tg2576 mice. Our findings unexpectedly show that of these various AD-relevant protein metabolites, sAPP turnover in the brain is the most different when comparing a wild-type mouse and a beta-amyloid depositing, APP overexpressing transgenic model. Given the neurotrophic roles attributed to sAPP, the enhanced stability of sAPP in the beta-amyloid depositing Tg2576 mice may represent a neuroprotective response.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid beta-Protein Precursor/metabolism , tau Proteins/metabolism , Animals , Brain/metabolism , Cells, Cultured , Cycloheximide/pharmacology , Cytoskeleton/metabolism , Humans , Mice , Mice, Transgenic , Protein Synthesis Inhibitors/pharmacology , Rats , Time Factors
14.
Alzheimer Dis Assoc Disord ; 23(4): 315-8, 2009.
Article in English | MEDLINE | ID: mdl-19571732

ABSTRACT

To study the relationship between plasma levels of amyloid beta (Abeta) peptides and dementia in aging individuals with Down syndrome, we investigated the relationship among plasma Abeta, apolipoprotein E genotype and cognitive and clinical factors using baseline specimens form participants in an ongoing clinical trial in individuals with Down syndrome 50 years of age and older. Because of substantial skew in the distribution of peptide levels, analyses used log transformations of the data. The ratio of Abeta42 to Abeta40 was associated with the presence of dementia (P=0.003, df=196, F=9.37); this association persisted after adjustment for age, sex level of mental retardation, and apolipoprotein E genotype. Consistent with recent reports regarding the effect of presenilin mutations on peptide generation, our finding supports the theory that the ratio of Abeta42 to Abeta40 rather than absolute levels of the peptides is important to the pathophysiology of Alzheimer's disease in genetically susceptible populations.


Subject(s)
Aging/blood , Amyloid beta-Peptides/blood , Dementia/blood , Down Syndrome/blood , Aged , Aging/genetics , Amyloid beta-Peptides/genetics , Dementia/complications , Dementia/genetics , Down Syndrome/complications , Down Syndrome/genetics , Female , Humans , Internationality , Male , Middle Aged
15.
Am J Pathol ; 175(1): 17-24, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19497998

ABSTRACT

Synaptic deficits are closely correlated with cognitive dysfunction in Alzheimer's disease (AD), and synaptic integrity is regulated by the actin cytoskeleton. We demonstrated here that the Wiskott-Aldrich syndrome protein family verprolin-homologous protein (WAVE), a key molecule for actin assembly, co-aggregated with both hyperphosphorylated tau and phosphorylated collapsin response mediator protein 2 (CRMP2) in neurofibrillary tangles and abnormal neurites of the AD brain. Although phosphorylated CRMP2 accumulation was induced in the brains of JNPL3 mice, WAVE accumulation was not detected in the brains of either JNPL3 or Tg2576 mice that developed neurofibrillary tangles and amyloid-beta (Abeta) plaques, respectively. Interestingly, both phosphorylated CRMP2 accumulation and WAVE accumulation were recapitulated in the brains of 3xTg-AD mice that developed neurofibrillary tangles and Abeta plaques. In addition, we found an interaction between WAVE, CRMP2, and hyperphosphorylated tau in the cytosolic fraction of the AD brain. Taken together, WAVE accumulation may require both Abeta/amyloid precursor protein and tau pathologies, and an interaction between WAVE, CRMP2, and hyperphosphorylated tau may be involved in this process. Thus, WAVE accumulation may be involved in Abeta/amyloid precursor protein mediated-tangle modification, suggesting a possible correlation between WAVE accumulation and synaptic deficits induced by disturbances in actin assembly in AD brains.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Brain/pathology , Wiskott-Aldrich Syndrome Protein Family/metabolism , Aged , Alzheimer Disease/metabolism , Animals , Blotting, Western , Brain/metabolism , Humans , Image Processing, Computer-Assisted , Immunohistochemistry , Immunoprecipitation , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Transgenic , Microscopy, Confocal , Nerve Tissue Proteins/metabolism , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Phosphorylation , tau Proteins/metabolism
16.
Biochem Biophys Res Commun ; 385(2): 247-50, 2009 Jul 24.
Article in English | MEDLINE | ID: mdl-19450545

ABSTRACT

The overproduction and extracellular buildup of amyloid-beta peptide (Abeta) is a critical step in the etiology of Alzheimer's disease. Recent data suggest that intracellular trafficking is of central importance in the production of Abeta. Here we use a neuronal cell line to examine two structurally similar clathrin assembly proteins, AP180 and CALM. We show that RNA interference-mediated knockdown of AP180 reduces the generation of Abeta1-40 and Abeta1-42, whereas CALM knockdown has no effect on Abeta generation. Thus AP180 is among the traffic controllers that oversee and regulate amyloid precursor protein processing pathways. Our results also suggest that AP180 and CALM, while similar in their domain structures and biochemical properties, are in fact dedicated to separate trafficking pathways in neurons.


Subject(s)
Amyloid beta-Peptides/biosynthesis , Clathrin/metabolism , Monomeric Clathrin Assembly Proteins/metabolism , Neurons/metabolism , Peptide Fragments/biosynthesis , Cell Line, Tumor , Clathrin/chemistry , Clathrin/genetics , Gene Knockdown Techniques , Humans , Monomeric Clathrin Assembly Proteins/chemistry , Monomeric Clathrin Assembly Proteins/genetics , Protein Structure, Tertiary , RNA Interference
17.
Nat Med ; 15(4): 377-9, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19287391

ABSTRACT

Amyloid-beta (Abeta) peptides, found in Alzheimer's disease brain, accumulate rapidly after traumatic brain injury (TBI) in both humans and animals. Here we show that blocking either beta- or gamma-secretase, enzymes required for production of Abeta from amyloid precursor protein (APP), can ameliorate motor and cognitive deficits and reduce cell loss after experimental TBI in mice. Thus, APP secretases are promising targets for treatment of TBI.


Subject(s)
Amyloid Precursor Protein Secretases/therapeutic use , Brain Injuries/drug therapy , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/deficiency , Animals , Aspartic Acid Endopeptidases/deficiency , Brain Injuries/prevention & control , Cognition/drug effects , Disease Models, Animal , Humans , Mice , Motor Activity/drug effects
18.
J Biol Chem ; 284(13): 8495-506, 2009 Mar 27.
Article in English | MEDLINE | ID: mdl-19164281

ABSTRACT

The amyloid precursor protein (APP) is cleaved to produce the Alzheimer disease-associated peptide Abeta, but the normal functions of uncleaved APP in the brain are unknown. We found that APP was present in the postsynaptic density of central excitatory synapses and coimmunoprecipitated with N-methyl-d-aspartate receptors (NMDARs). The presence of APP in the postsynaptic density was supported by the observation that NMDARs regulated trafficking and processing of APP; overexpression of the NR1 subunit increased surface levels of APP, whereas activation of NMDARs decreased surface APP and promoted production of Abeta. We transfected APP or APP RNA interference into primary neurons and used electrophysiological techniques to explore the effects of APP on postsynaptic function. Reduction of APP decreased (and overexpression of APP increased) NMDAR whole cell current density and peak amplitude of spontaneous miniature excitatory postsynaptic currents. The increase in NMDAR current by APP was due to specific recruitment of additional NR2B-containing receptors. Consistent with these findings, immunohistochemical experiments demonstrated that APP increased the surface levels and decreased internalization of NR2B subunits. These results demonstrate a novel physiological role of postsynaptic APP in enhancing NMDAR function.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Serum Amyloid A Protein/metabolism , Synaptic Membranes/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Brain/pathology , COS Cells , Chlorocebus aethiops , Gene Expression , Mice , Mice, Knockout , Protein Transport/genetics , RNA Interference , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/genetics , Serum Amyloid A Protein/genetics , Synaptic Membranes/genetics , Synaptic Membranes/pathology
19.
Neurosci Lett ; 450(1): 51-5, 2009 Jan 23.
Article in English | MEDLINE | ID: mdl-19022346

ABSTRACT

The clinical progression of Alzheimer's disease is closely related to tau pathology. Hyperphosphorylation of tau precedes histopathological evidence of tangle formation, and modulation of tau phosphorylation is a promising therapeutic target. Although some phosphorylation sites are more critical in pathological processes, the importance of each phosphorylation site is unclear. In this study, we found that levels of phosphorylated tau drastically increased in crude and insoluble tau fractions with aging in a transgenic mouse model of Alzheimer-type tauopathy. However, changes in the soluble tau fraction were minor and phosphorylation at some sites was even reduced with aging. Total soluble (presumably functional) tau was reduced, while insoluble tau increased with aging. Synaptic proteins were reduced as insoluble tau increased. Taken together, these findings suggest that levels of soluble and insoluble tau are indicative of overall levels of tau phosphorylation, and may be useful markers to evaluate the effects of anti-tau therapeutic strategies in vivo.


Subject(s)
Aging/physiology , tau Proteins/metabolism , Alzheimer Disease/physiopathology , Analysis of Variance , Animals , Brain/physiopathology , Disease Models, Animal , Disks Large Homolog 4 Protein , Guanylate Kinases , Intracellular Signaling Peptides and Proteins/metabolism , Male , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Neurons/metabolism , Phosphorylation , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Solubility , Synaptophysin/metabolism , tau Proteins/chemistry
20.
Neurobiol Aging ; 30(11): 1777-91, 2009 Nov.
Article in English | MEDLINE | ID: mdl-18258340

ABSTRACT

Gangliosides have been shown to be necessary for beta-amyloid (Abeta) binding and aggregation. GD3 synthase (GD3S) is responsible for biosynthesis of the b- and c-series gangliosides, including two of the four major brain gangliosides. We examined Abeta-ganglioside interactions in neural tissue from mice lacking the gene coding for GD3S (St8sia1), and in a double-transgenic (APP/PSEN1) mouse model of Alzheimer's disease cross-bred with GD3S-/- mice. In primary neurons and astrocytes lacking GD3S, Abeta-induced cell death and Abeta aggregation were inhibited. Like GD3S-/- and APP/PSEN1 double-transgenic mice, APP/PSEN1/GD3S-/- "triple-mutant" mice are indistinguishable from wild-type mice on casual examination. APP/PSEN1 double-transgenics exhibit robust impairments on a number of reference-memory tasks. In contrast, APP/PSEN1/GD3S-/- triple-mutant mice performed as well as wild-type control and GD3S-/- mice. Consistent with the behavioral improvements, both aggregated and unaggregated Abeta and associated neuropathology were almost completely eliminated in triple-mutant mice. These results suggest that GD3 synthase may be a novel therapeutic target to combat the cognitive deficits, amyloid plaque formation, and neurodegeneration that afflict Alzheimer's patients.


Subject(s)
Amyloid/metabolism , Memory/physiology , Plaque, Amyloid/pathology , Sialyltransferases/deficiency , Alzheimer Disease , Amyloid beta-Protein Precursor/genetics , Animals , CD11b Antigen/metabolism , Cells, Cultured , Cerebral Cortex/pathology , Disease Models, Animal , Gas Chromatography-Mass Spectrometry/methods , Humans , Lipid Peroxidation/genetics , Maze Learning/physiology , Memory Disorders/genetics , Mice , Mice, Transgenic , Mutation/genetics , Neurons/drug effects , Neurons/metabolism , Plaque, Amyloid/genetics , Presenilin-1/genetics , Protein Binding/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...