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










Publication year range
1.
Behav Brain Res ; 345: 21-29, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29476896

ABSTRACT

Impulsive choice behavior, which can be assessed using the delay discounting task, is a characteristic of various psychiatric disorders, including attention-deficit/hyperactivity disorder (ADHD). Guanfacine is a selective α2A-adrenergic receptor agonist that is clinically effective in treating ADHD. However, there is no clear evidence that systemic guanfacine administration reduces impulsive choice behavior in the delay discounting task in rats. In the present study, we examined the effect of systemic guanfacine administration on food-motivated impulsive choice behavior in rats and the neuronal mechanism underlying this effect. Repeated administration of either guanfacine, methylphenidate, or atomoxetine significantly enhanced impulse control, increasing the number of times the rats chose a large but delayed reward in a dose-dependent manner. The effect of guanfacine was significantly blocked by pretreatment with an α2A-adrenergic receptor antagonist. Furthermore, the effect of guanfacine remained unaffected in rats pretreated with a selective noradrenergic neurotoxin, consistent with a post-synaptic action. In contrast, the effect of atomoxetine on impulsive choice behavior was attenuated by pretreatment with the noradrenergic neurotoxin. These results provide the first evidence that systemically administered guanfacine reduces impulsive choice behavior in rats and that direct stimulation of postsynaptic, rather than presynaptic, α2A-adrenergic receptors is involved in this effect.


Subject(s)
Adrenergic alpha-2 Receptor Agonists/pharmacology , Choice Behavior/drug effects , Feeding Behavior/drug effects , Guanfacine/pharmacology , Impulsive Behavior/drug effects , Motivation/drug effects , Adrenergic alpha-2 Receptor Antagonists/pharmacology , Animals , Atomoxetine Hydrochloride/pharmacology , Choice Behavior/physiology , Dose-Response Relationship, Drug , Feeding Behavior/physiology , Feeding Behavior/psychology , Food , Impulsive Behavior/physiology , Male , Methylphenidate/pharmacology , Motivation/physiology , Neurons/drug effects , Neurons/metabolism , Random Allocation , Rats, Wistar , Receptors, Adrenergic, alpha-2/metabolism
2.
Cell Rep ; 21(1): 259-273, 2017 Oct 03.
Article in English | MEDLINE | ID: mdl-28978478

ABSTRACT

γ-secretase inhibitors (GSI) are drugs developed to decrease amyloid-ß peptide (Aß) production by inhibiting intramembranous cleavage of ß-amyloid protein precursor (ßAPP). However, a large phase 3 trial of semagacestat, a potential non-transition state analog (non-TSA) GSI, in patients with Alzheimer's disease (AD) was terminated due to unexpected aggravation of cognitive deficits and side effects. Here, we show that some semagacestat effects are clearly different from a phenotype caused by a loss of function of presenilins, core proteins in the γ-secretase complex. Semagacestat increases intracellular byproduct peptides, produced along with Aß through serial γ-cleavage of ßAPP, as well as intracellular long Aß species, in cell-based and in vivo studies of AD model mice. Other potential non-TSA GSIs, but not L685,458, a TSA GSI, have similar effects. Furthermore, semagacestat inhibits release of de novo intramembranous γ-byproducts to the soluble space. Thus, semagacestat is a pseudo-GSI, and therefore, the semagacestat clinical trial did not truly test the Aß hypothesis.


Subject(s)
Alanine/analogs & derivatives , Amyloid Precursor Protein Secretases/genetics , Azepines/pharmacology , Enzyme Inhibitors/pharmacology , Neurons/drug effects , Alanine/pharmacology , Alzheimer Disease , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Carbamates/pharmacology , Cell Differentiation , Clinical Trials as Topic , Dipeptides/pharmacology , Disease Models, Animal , Drug Administration Schedule , Gene Expression Regulation , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/enzymology , Mice , Neurons/enzymology , Neurons/pathology
3.
Proc Natl Acad Sci U S A ; 111(7): 2638-43, 2014 Feb 18.
Article in English | MEDLINE | ID: mdl-24497505

ABSTRACT

Alzheimer's disease (AD) is characterized by the accumulation of amyloid-ß (Aß). The genes that govern this process, however, have remained elusive. To this end, we combined distinct mouse strains with transcriptomics to directly identify disease-relevant genes. We show that AD model mice (APP-Tg) with DBA/2 genetic backgrounds have significantly lower levels of Aß accumulation compared with SJL and C57BL/6 mice. We then applied brain transcriptomics to reveal the genes in DBA/2 that suppress Aß accumulation. To avoid detecting secondarily affected genes by Aß, we used non-Tg mice in the absence of Aß pathology and selected candidate genes differently expressed in DBA/2 mice. Additional transcriptome analysis of APP-Tg mice with mixed genetic backgrounds revealed kinesin light chain-1 (Klc1) as an Aß modifier, indicating a role for intracellular trafficking in Aß accumulation. Aß levels correlated with the expression levels of Klc1 splice variant E and the genotype of Klc1 in these APP-Tg mice. In humans, the expression levels of KLC1 variant E in brain and lymphocyte were significantly higher in AD patients compared with unaffected individuals. Finally, functional analysis using neuroblastoma cells showed that overexpression or knockdown of KLC1 variant E increases or decreases the production of Aß, respectively. The identification of KLC1 variant E suggests that the dysfunction of intracellular trafficking is a causative factor of Aß pathology. This unique combination of distinct mouse strains and model mice with transcriptomics is expected to be useful for the study of genetic mechanisms of other complex diseases.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Microtubule-Associated Proteins/metabolism , Protein Isoforms/metabolism , Alzheimer Disease/genetics , Animals , Brain/metabolism , Crosses, Genetic , Gene Expression Profiling , Humans , Kinesins , Mice , Microtubule-Associated Proteins/genetics , Protein Isoforms/genetics , Species Specificity
4.
Proc Natl Acad Sci U S A ; 108(39): 16410-5, 2011 Sep 27.
Article in English | MEDLINE | ID: mdl-21930904

ABSTRACT

Gram-positive bacteria of the genus Streptomyces are industrially important microorganisms, producing >70% of commercially important antibiotics. The production of these compounds is often regulated by low-molecular-weight bacterial hormones called autoregulators. Although 60% of Streptomyces strains may use γ-butyrolactone-type molecules as autoregulators and some use furan-type molecules, little is known about the signaling molecules used to regulate antibiotic production in many other members of this genus. Here, we purified a signaling molecule (avenolide) from Streptomyces avermitilis--the producer of the important anthelmintic agent avermectin with annual world sales of $850 million--and determined its structure, including stereochemistry, by spectroscopic analysis and chemical synthesis as (4S,10R)-10-hydroxy-10-methyl-9-oxo-dodec-2-en-1,4-olide, a class of Streptomyces autoregulator. Avenolide is essential for eliciting avermectin production and is effective at nanomolar concentrations with a minimum effective concentration of 4 nM. The aco gene of S. avermitilis, which encodes an acyl-CoA oxidase, is required for avenolide biosynthesis, and homologs are also present in Streptomyces fradiae, Streptomyces ghanaensis, and Streptomyces griseoauranticus, suggesting that butenolide-type autoregulators may represent a widespread and another class of Streptomyces autoregulator involved in regulating antibiotic production.


Subject(s)
Anthelmintics/metabolism , Hormones/metabolism , Ivermectin/analogs & derivatives , Streptomyces/metabolism , Culture Media , Hormones/chemistry , Ivermectin/metabolism , Ligands , Mass Spectrometry , Nuclear Magnetic Resonance, Biomolecular , Solvents
5.
Neurodegener Dis ; 6(5-6): 230-9, 2009.
Article in English | MEDLINE | ID: mdl-19738368

ABSTRACT

Alzheimer-disease-associated beta-amyloid (Abeta) is produced by sequential endoproteolysis of beta-amyloid protein precursor (betaAPP): the extracellular portion is shed by cleavage in the juxtamembrane region by beta-amyloid-cleaving enzyme (BACE)/beta-secretase, after which it is cleaved by presenilin (PS)/gamma-secretase near the middle of the transmembrane domain. Thus, inhibition of either of the secretases reduces Abeta generation and is a fundamental strategy for the development of drugs to prevent Alzheimer disease. However, it is not clear how small compounds reduce Abeta production without inhibition of the secretases. Such compounds are expected to avoid some of the side effects of secretase inhibitors. Here, we report that destruxin E (Dx-E), a natural cyclic hexadepsipeptide, reduces Abeta generation without affecting BACE or PS/gamma-secretase activity. In agreement with this, Dx-E did not inhibit Notch signaling. We found that Dx-E decreases colocalization of BACE1 and betaAPP, which reduces beta-cleavage of betaAPP. Therefore, the data demonstrate that Dx-E represents a novel Abeta-reducing process which could have fewer side effects than secretase inhibitors.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Depsipeptides/pharmacology , Fungal Proteins/pharmacology , Gene Expression Regulation/drug effects , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Protein Precursor/genetics , Cell Line, Transformed , Dose-Response Relationship, Drug , Humans , Presenilin-1/genetics , Receptors, Notch/genetics , Time Factors , Transfection/methods
6.
EMBO Mol Med ; 1(4): 223-35, 2009 Jul.
Article in English | MEDLINE | ID: mdl-20049724

ABSTRACT

Surrogate markers for the Alzheimer disease (AD)-associated 42-amino acid form of amyloid-beta (Abeta42) have been sought because they may aid in the diagnosis of AD and for clarification of disease pathogenesis. Here, we demonstrate that human cerebrospinal fluid (CSF) contains three APLP1-derived Abeta-like peptides (APL1beta) that are generated by beta- and gamma-cleavages at a concentration of approximately 4.5 nM. These novel peptides, APL1beta25, APL1beta27 and APL1beta28, were not deposited in AD brains. Interestingly, most gamma-secretase modulators (GSMs) and familial AD-associated presenilin1 mutants that up-regulate the relative production of Abeta42 cause a parallel increase in the production of APL1beta28 in cultured cells. Moreover, in CSF from patients with pathological mutations in presenilin1 gene, the relative APL1beta28 levels are higher than in non-AD controls, while the relative Abeta42 levels are unchanged or lower. Most strikingly, the relative APL1beta28 levels are higher in CSF from sporadic AD patients (regardless of whether they are at mild cognitive impairment or AD stage), than those of non-AD controls. Based on these results, we propose the relative level of APL1beta28 in the CSF as a candidate surrogate marker for the relative level of Abeta42 production in the brain.


Subject(s)
Alzheimer Disease/diagnosis , Amyloid beta-Protein Precursor/cerebrospinal fluid , Biomarkers/cerebrospinal fluid , Peptide Fragments/cerebrospinal fluid , Adult , Alzheimer Disease/cerebrospinal fluid , Amino Acid Sequence , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/analysis , Amyloid beta-Protein Precursor/metabolism , Aspartic Acid Endopeptidases/antagonists & inhibitors , Aspartic Acid Endopeptidases/metabolism , Biomarkers/analysis , Biomarkers/metabolism , Cell Line , Cell Line, Tumor , Humans , Middle Aged , Molecular Sequence Data , Mutation , Peptide Fragments/analysis , Peptide Fragments/metabolism , Presenilin-1/genetics
7.
Mol Cell Biol ; 28(1): 165-76, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17967888

ABSTRACT

Intramembrane proteolysis by presenilin-dependent gamma-secretase produces the Notch intracellular cytoplasmic domain (NCID) and Alzheimer disease-associated amyloid-beta. Here, we show that upon Notch signaling the intracellular domain of Notch-1 is cleaved into two distinct types of NICD species due to diversity in the site of S3 cleavage. Consistent with the N-end rule, the S3-V cleavage produces stable NICD with Val at the N terminus, whereas the S3-S/S3-L cleavage generates unstable NICD with Ser/Leu at the N terminus. Moreover, intracellular Notch signal transmission with unstable NICDs is much weaker than that with stable NICD. Importantly, the extent of endocytosis in target cells affects the relative production ratio of the two types of NICD, which changes in parallel with Notch signaling. Surprisingly, substantial amounts of unstable NICD species are generated from the Val-->Gly and the Lys-->Arg mutants, which have been reported to decrease S3 cleavage efficiency in cultured cells. Thus, we suggest that the existence of two distinct types of NICD points to a novel aspect of the intracellular signaling and that changes in the precision of S3 cleavage play an important role in the process of conversion from extracellular to intracellular Notch signaling.


Subject(s)
Peptide Hydrolases/metabolism , Receptors, Notch/chemistry , Receptors, Notch/metabolism , Signal Transduction , Amino Acid Sequence , Amyloid Precursor Protein Secretases/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line , Cell Survival , Cell-Free System , Cricetinae , Cytoplasm/metabolism , Endocytosis , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Molecular Sequence Data , Mutation/genetics , Promoter Regions, Genetic/genetics , Protein Structure, Tertiary , Receptors, Notch/genetics , Transcriptional Activation/genetics
8.
Eur J Neurosci ; 26(9): 2458-68, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17970733

ABSTRACT

Accumulation of amyloid beta (Abeta) is a pathological hallmark of Alzheimer's disease, and lowering Abeta is a promising therapeutic approach. Intact anti-Abeta antibodies reduce brain Abeta through two pathways: enhanced microglial phagocytosis and Abeta transfer from the brain to the periphery (Abeta sequestration). While activation of microglia, which is essential for microglial phagocytosis, is necessarily accompanied by undesired neuroinflammatory events, the capacity for sequestration does not seem to be linked to such effects. We and other groups have found that simple Abeta binding agents are sufficient to reduce brain Abeta through the sequestration pathway. In this study, we aimed to eliminate potentially deleterious immune activation from antibodies without affecting the ability to induce sequestration. The glycan portion of immunoglobulin is critically involved in interactions with immune effectors including the Fc receptor and complement c1q; deglycosylation eliminates these interactions, while antigen (Abeta)-binding affinity is maintained. In this study, we investigated whether deglycosylated anti-Abeta antibodies reduce microglial phagocytosis and neuroinflammation without altering the capacity to induce Abeta sequestration. Deglycosylated antibodies maintained Abeta binding affinity. Deglycosylated antibodies did not enhance Abeta phagocytosis or cytokine release in primary cultured microglia, whereas intact antibodies did so significantly. Intravenous injection of deglycosylated antibodies elevated plasma Abeta levels and induced Abeta sequestration to a similar or greater degree compared with intact antibodies in an Alzheimer's transgenic mouse model without or with Abeta plaque pathology. We conclude that deglycosylated antibodies effectively induced Abeta sequestration without provoking neuroinflammation; thus, these deglycosylated antibodies may be optimal for sequestration therapy for Alzheimer's disease.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/antagonists & inhibitors , Antibodies/pharmacology , Cytokines/drug effects , Microglia/drug effects , Phagocytosis/drug effects , Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/chemical synthesis , Amyloid beta-Peptides/immunology , Animals , Animals, Newborn , Antibodies/immunology , Antibodies/therapeutic use , Brain/drug effects , Brain/immunology , Brain/metabolism , Cells, Cultured , Coculture Techniques , Cytokines/metabolism , Down-Regulation/drug effects , Down-Regulation/immunology , Encephalitis/chemically induced , Encephalitis/immunology , Encephalitis/prevention & control , Gliosis/chemically induced , Gliosis/immunology , Gliosis/prevention & control , Glycosylation , Humans , Injections, Intravenous , Mice , Mice, Transgenic , Microglia/metabolism , Phagocytosis/immunology , Polysaccharides/chemistry , Polysaccharides/immunology , Rats , Rats, Wistar , Treatment Outcome
9.
Hum Mol Genet ; 16(1): 15-23, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-17135279

ABSTRACT

We scanned throughout chromosome 21 to assess genetic associations with late-onset Alzheimer disease (AD) using 374 Japanese patients and 375 population-based controls, because trisomy 21 is known to be associated with early deposition of beta-amyloid (Abeta) in the brain. Among 417 markers spanning 33 Mb, 22 markers showed associations with either the allele or the genotype frequency (P < 0.05). Logistic regression analysis with age, sex and apolipoprotein E (APOE)-epsilon4 dose supported genetic risk of 17 markers, of which eight markers were linked to the SAMSN1, PRSS7, NCAM2, RUNX1, DYRK1A and KCNJ6 genes. In logistic regression, the DYRK1A (dual-specificity tyrosine-regulated kinase 1A) gene, located in the Down syndrome critical region, showed the highest significance [OR = 2.99 (95% CI: 1.72-5.19), P = 0.001], whereas the RUNX1 gene showed a high odds ratio [OR = 23.3 (95% CI: 2.76-196.5), P = 0.038]. DYRK1A mRNA level in the hippocampus was significantly elevated in patients with AD when compared with pathological controls (P < 0.01). DYRK1A mRNA level was upregulated along with an increase in the Abeta-level in the brain of transgenic mice, overproducing Abeta at 9 months of age. In neuroblastoma cells, Abeta induced an increase in the DYRK1A transcript, which also led to tau phosphorylation at Thr212 under the overexpression of tau. Therefore, the upregulation of DYRK1A transcription results from Abeta loading, further leading to tau phosphorylation. Our result indicates that DYRK1A could be a key molecule bridging between beta-amyloid production and tau phosphorylation in AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/biosynthesis , Chromosomes, Human, Pair 21/genetics , Down Syndrome/genetics , Membrane Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Animals , Brain/metabolism , Case-Control Studies , Cell Line, Tumor , Female , Haplotypes , Humans , Male , Mice , Mice, Transgenic , Middle Aged , Phosphorylation , Polymorphism, Single Nucleotide , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , RNA, Messenger/metabolism , Dyrk Kinases
10.
J Neurochem ; 99(6): 1555-63, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17083447

ABSTRACT

Accumulation of amyloid beta peptide (Abeta) in brain is a hallmark of Alzheimer's disease (AD). Inhibition of beta-site amyloid precursor protein (APP)-cleaving enzyme-1 (BACE1), the enzyme that initiates Abeta production, and other Abeta-lowering strategies are commonly tested in transgenic mice overexpressing mutant APP. However, sporadic AD cases, which represent the majority of AD patients, are free from the mutation and do not necessarily have overproduction of APP. In addition, the commonly used Swedish mutant APP alters APP cleavage. Therefore, testing Abeta-lowering strategies in transgenic mice may not be optimal. In this study, we investigated the impact of BACE1 inhibition in non-transgenic mice with physiologically relevant APP expression. Existing Abeta ELISAs are either relatively insensitive to mouse Abeta or not specific to full-length Abeta. A newly developed ELISA detected a significant reduction of full-length soluble Abeta 1-40 in mice with the BACE1 homozygous gene deletion or BACE1 inhibitor treatment, while the level of x-40 Abeta was moderately reduced due to detection of non-full-length Abeta and compensatory activation of alpha-secretase. These results confirmed the feasibility of Abeta reduction through BACE1 inhibition under physiological conditions. Studies using our new ELISA in non-transgenic mice provide more accurate evaluation of Abeta-reducing strategies than was previously feasible.


Subject(s)
Amyloid Precursor Protein Secretases/physiology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Aspartic Acid Endopeptidases/physiology , Neural Inhibition/drug effects , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/deficiency , Animals , Aspartic Acid Endopeptidases/antagonists & inhibitors , Aspartic Acid Endopeptidases/deficiency , Blotting, Western/methods , Cells, Cultured , Cerebral Cortex/cytology , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay/methods , Humans , In Vitro Techniques , Mice , Mice, Knockout , Neurons/metabolism , Triglycerides , Up-Regulation/drug effects , gamma-Aminobutyric Acid/analogs & derivatives , gamma-Aminobutyric Acid/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...