Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
JCI Insight ; 4(22)2019 11 14.
Article in English | MEDLINE | ID: mdl-31593554

ABSTRACT

Anemia of ß-thalassemia is caused by ineffective erythropoiesis and reduced red cell survival. Several lines of evidence indicate that iron/heme restriction is a potential therapeutic strategy for the disease. Glycine is a key initial substrate for heme and globin synthesis. We provide evidence that bitopertin, a glycine transport inhibitor administered orally, improves anemia, reduces hemolysis, diminishes ineffective erythropoiesis, and increases red cell survival in a mouse model of ß-thalassemia (Hbbth3/+ mice). Bitopertin ameliorates erythroid oxidant damage, as indicated by a reduction in membrane-associated free α-globin chain aggregates, in reactive oxygen species cellular content, in membrane-bound hemichromes, and in heme-regulated inhibitor activation and eIF2α phosphorylation. The improvement of ß-thalassemic ineffective erythropoiesis is associated with diminished mTOR activation and Rab5, Lamp1, and p62 accumulation, indicating an improved autophagy. Bitopertin also upregulates liver hepcidin and diminishes liver iron overload. The hematologic improvements achieved by bitopertin are blunted by the concomitant administration of the iron chelator deferiprone, suggesting that an excessive restriction of iron availability might negate the beneficial effects of bitopertin. These data provide important and clinically relevant insights into glycine restriction and reduced heme synthesis strategies for the treatment of ß-thalassemia.


Subject(s)
Erythrocytes/drug effects , Glycine Plasma Membrane Transport Proteins/antagonists & inhibitors , Piperazines/pharmacology , Sulfones/pharmacology , beta-Thalassemia/metabolism , Animals , Cell Survival/drug effects , Disease Models, Animal , Erythrocytes/metabolism , Female , Hemolysis/drug effects , Iron/metabolism , Iron Overload/metabolism , Liver/drug effects , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic
2.
ACS Chem Biol ; 14(1): 37-49, 2019 01 18.
Article in English | MEDLINE | ID: mdl-30452219

ABSTRACT

The importance of Discoidin Domain Receptor 1 (DDR1) in renal fibrosis has been shown via gene knockout and use of antisense oligonucleotides; however, these techniques act via a reduction of DDR1 protein, while we prove the therapeutic potential of inhibiting DDR1 phosphorylation with a small molecule. To date, efforts to generate a selective small-molecule to specifically modulate the activity of DDR1 in an in vivo model have been unsuccessful. We performed parallel DNA encoded library screens against DDR1 and DDR2, and discovered a chemical series that is highly selective for DDR1 over DDR2. Structure-guided optimization efforts yielded the potent DDR1 inhibitor 2.45, which possesses excellent kinome selectivity (including 64-fold selectivity over DDR2 in a biochemical assay), a clean in vitro safety profile, and favorable pharmacokinetic and physicochemical properties. As desired, compound 2.45 modulates DDR1 phosphorylation in vitro as well as prevents collagen-induced activation of renal epithelial cells expressing DDR1. Compound 2.45 preserves renal function and reduces tissue damage in Col4a3-/- mice (the preclinical mouse model of Alport syndrome) when employing a therapeutic dosing regime, indicating the real therapeutic value of selectively inhibiting DDR1 phosphorylation in vivo. Our results may have wider significance as Col4a3-/- mice also represent a model for chronic kidney disease, a disease which affects 10% of the global population.


Subject(s)
DNA/genetics , Discoidin Domain Receptor 1/antagonists & inhibitors , Kidney/physiopathology , Nephritis, Hereditary/genetics , Animals , Autoantigens/genetics , Autoantigens/metabolism , Collagen Type IV/genetics , Collagen Type IV/metabolism , Discoidin Domain Receptor 1/metabolism , Disease Models, Animal , Epithelial Cells/metabolism , Kidney Function Tests , Mice , Mice, Knockout , Nephritis, Hereditary/physiopathology , Phosphorylation , Src Homology 2 Domain-Containing, Transforming Protein 1/metabolism
3.
Front Pharmacol ; 9: 924, 2018.
Article in English | MEDLINE | ID: mdl-30158871

ABSTRACT

Individuals of many species rely on odors to communicate, find breeding partners, locate resources and sense dangers. In vertebrates, odorants are detected by chemosensory receptors of the olfactory system. One class of these receptors, the trace amine-associated receptors (TAARs), was recently suggested to mediate male sexual interest and mate choice. Here we tested this hypothesis in mice by generating a cluster deletion mouse (Taar2-9-/-) lacking all TAARs expressed in the olfactory epithelium, and evaluating transduction pathways from odorants to TAARs, neural activity and behaviors reflecting sexual interest. We found that a urinary volatile amine, isobutylamine (IBA), was a potent ligand for TAAR3 (but not TAAR1, 4, 5, and 6). When males were exposed to IBA, brain regions associated with sexual behaviors were less active in Taar2-9-/- than in wild type males. Accordingly, Taar2-9-/- males spent less time sniffing both the urine of females and pure IBA than wild type males. This is the first demonstration of a comprehensive transduction pathway linking odorants to TAARs and male sexual interest. Interestingly, the concentration of IBA in female urine varied across the estrus cycle with a peak during estrus. This variation in IBA concentration may represent a simple olfactory cue for males to recognize receptive females. Our results are consistent with the hypothesis that IBA and TAARs play an important role in the recognition of breeding partners and mate choice.

4.
Sci Rep ; 5: 15410, 2015 Oct 29.
Article in English | MEDLINE | ID: mdl-26510576

ABSTRACT

The amyloid-ß42 (Aß42) peptide is believed to be the main culprit in the pathogenesis of Alzheimer disease (AD), impairing synaptic function and initiating neuronal degeneration. Soluble Aß42 oligomers are highly toxic and contribute to progressive neuronal dysfunction, loss of synaptic spine density, and affect long-term potentiation (LTP). We have characterized a short, L-amino acid Aß-oligomer Interacting Peptide (AIP) that targets a relatively well-defined population of low-n Aß42 oligomers, rather than simply inhibiting the aggregation of Aß monomers into oligomers. Our data show that AIP diminishes the loss of Aß42-induced synaptic spine density and rescues LTP in organotypic hippocampal slice cultures. Notably, the AIP enantiomer (comprised of D-amino acids) attenuated the rough-eye phenotype in a transgenic Aß42 fly model and significantly improved the function of photoreceptors of these flies in electroretinography tests. Overall, our results indicate that specifically "trapping" low-n oligomers provides a novel strategy for toxic Aß42-oligomer recognition and removal.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/antagonists & inhibitors , Oligopeptides/pharmacology , Peptide Fragments/antagonists & inhibitors , Protein Aggregation, Pathological/drug therapy , Synapses/metabolism , Synaptic Transmission/drug effects , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Peptide Fragments/metabolism , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , Rats , Rats, Wistar , Synapses/pathology
5.
Eur Neuropsychopharmacol ; 25(11): 2049-61, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26372541

ABSTRACT

Trace amine-associated receptor 1 (TAAR1) activation by selective endogenous agonists modulates dopaminergic neurotransmission. This results in antipsychotic-like behavior in vivo which might be initiated by an interaction of TAAR1 and dopamine D2L receptor (D2R). Here we analyzed the functional link between TAAR1 and D2R using highly potent and selective TAAR1 agonists, and newly generated tools such as TAAR1 knock-out and TAAR1 overexpressing rats as well as specific anti-rat TAAR1 antibodies. We provide data from co-immunoprecipitation experiments supporting a functional interaction of the two receptors in heterologous cells and in brain tissue. Interaction of TAAR1 with D2R altered the subcellular localization of TAAR1 and increased D2R agonist binding affinity. Using specific ß-arrestin 2 (ßArr2) complementation assays we show that the interaction of TAAR1 with D2R reduced ßArr2 recruitment to D2R. In addition, we report that besides Gαs-protein signaling TAAR1 also signals via ßArr2. In the presence of D2R, cAMP signaling of TAAR1 was reduced while its ßArr2 signaling was enhanced, resulting in reduced GSK3ß activation. These results demonstrate that ßArr2 signaling may be an important pathway for TAAR1 function and that the activation of the TAAR1-D2R complex negatively modulates GSK3ß signaling. Given that patients with schizophrenia or bipolar disorder show increased GSK3ß signaling, such a reduction of GSK3ß signaling triggered by the interaction of D2R with activated TAAR1 further supports TAAR1 as a target for the treatment of psychiatric disorders.


Subject(s)
Arrestins/metabolism , Brain/metabolism , Glycogen Synthase Kinase 3/metabolism , Receptors, Dopamine D2/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , CHO Cells , Cell Membrane/drug effects , Cell Membrane/metabolism , Cricetulus , Cyclic AMP/metabolism , Gene Knockout Techniques , Glycogen Synthase Kinase 3 beta , HEK293 Cells , Humans , Rats, Transgenic , Rats, Wistar , Receptors, Dopamine D2/agonists , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/genetics , beta-Arrestin 2 , beta-Arrestins
6.
Front Neurosci ; 9: 39, 2015.
Article in English | MEDLINE | ID: mdl-25762894

ABSTRACT

The newly discovered trace amine-associated receptor 1 (TAAR1) has the ability to regulate both dopamine function and psychostimulant action. Here, we tested in rats the ability of RO5203648, a selective TAAR1 partial agonist, to modulate the physiological and behavioral effects of methamphetamine (METH). In experiment 1, RO5203468 dose- and time-dependently altered METH-induced locomotor activity, manifested as an early attenuation followed by a late potentiation of METH's stimulating effects. In experiment 2, rats received a 14-day treatment regimen during which RO5203648 was co-administered with METH. RO5203648 dose-dependently attenuated METH-stimulated hyperactivity, with the effects becoming more apparent as the treatments progressed. After chronic exposure and 3-day withdrawal, rats were tested for locomotor sensitization. RO5203648 administration during the sensitizing phase prevented the development of METH sensitization. However, RO5203648, at the high dose, cross-sensitized with METH. In experiment 3, RO5203648 dose-dependently blocked METH self-administration without affecting operant responding maintained by sucrose, and exhibited lack of reinforcing efficacy when tested as a METH's substitute. Neurochemical data showed that RO5203648 did not affect METH-mediated DA efflux and uptake inhibition in striatal synaptosomes. In vivo, however, RO5203648 was able to transiently inhibit METH-induced accumulation of extracellular DA levels in the nucleus accumbens. Taken together, these data highlight the significant potential of TAAR1 to modulate METH's neurochemical and behavioral effects.

7.
Neuropsychopharmacology ; 40(9): 2217-27, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25749299

ABSTRACT

Trace Amine-Associated Receptor 1 (TAAR1) is a G protein-coupled receptor expressed in the mammalian brain and known to influence subcortical monoaminergic transmission. Monoamines, such as dopamine, also play an important role within the prefrontal cortex (PFC) circuitry, which is critically involved in high-o5rder cognitive processes. TAAR1-selective ligands have shown potential antipsychotic, antidepressant, and pro-cognitive effects in experimental animal models; however, it remains unclear whether TAAR1 can affect PFC-related processes and functions. In this study, we document a distinct pattern of expression of TAAR1 in the PFC, as well as altered subunit composition and deficient functionality of the glutamate N-methyl-D-aspartate (NMDA) receptors in the pyramidal neurons of layer V of PFC in mice lacking TAAR1. The dysregulated cortical glutamate transmission in TAAR1-KO mice was associated with aberrant behaviors in several tests, indicating a perseverative and impulsive phenotype of mutants. Conversely, pharmacological activation of TAAR1 with selective agonists reduced premature impulsive responses observed in the fixed-interval conditioning schedule in normal mice. Our study indicates that TAAR1 plays an important role in the modulation of NMDA receptor-mediated glutamate transmission in the PFC and related functions. Furthermore, these data suggest that the development of TAAR1-based drugs could provide a novel therapeutic approach for the treatment of disorders related to aberrant cortical functions.


Subject(s)
Excitatory Postsynaptic Potentials/genetics , Prefrontal Cortex/metabolism , Pyramidal Cells/physiology , Receptors, G-Protein-Coupled/metabolism , Receptors, Glutamate/metabolism , Animals , Conditioning, Operant/drug effects , Dose-Response Relationship, Drug , Excitatory Postsynaptic Potentials/drug effects , Female , Impulsive Behavior/drug effects , Impulsive Behavior/physiology , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxazoles/pharmacology , Patch-Clamp Techniques , Phenethylamines/pharmacology , Prefrontal Cortex/cytology , Prefrontal Cortex/drug effects , Pyramidal Cells/drug effects , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/genetics , Reinforcement Schedule
8.
J Alzheimers Dis ; 44(2): 613-24, 2015.
Article in English | MEDLINE | ID: mdl-25318543

ABSTRACT

The pathogenesis of Alzheimer's disease (AD) is characterized by the aggregation of amyloid-ß (Aß) peptides leading to deposition of senile plaques and a progressive decline of cognitive functions, which currently remains the main criterion for its diagnosis. Robust biomarkers for AD do not yet exist, although changes in the cerebrospinal fluid levels of tau and Aß represent promising candidates in addition to brain imaging and genetic risk profiling. Although concentrations of soluble Aß42 correlate with symptoms of AD, less is known about the biological activities of Aß peptides which are generated from the amyloid-ß protein precursor. An unbiased DNA microarray study showed that Aß42, at sub-lethal concentrations, specifically increases expression of several genes in neuroblastoma cells, notably the insulin-like growth factor binding proteins 3 and 5 (IGFBP3/5), the transcription regulator inhibitor of DNA binding, and the transcription factor Lim only domain protein 4. Using qRT-PCR, we confirmed that mRNA levels of the identified candidate genes were exclusively increased by the potentially neurotoxic Aß42 wild-type peptide, as both the less toxic Aß40 and a non-toxic substitution peptide Aß42 G33A did not affect mRNA levels. In vivo immunohistochemistry revealed a corresponding increase in both hippocampal and cortical IGFBP5 expression in an AD mouse model. Proteomic analyses of human AD cerebrospinal fluid displayed increased in vivo concentrations of IGFBPs. IGFBPs and transcription factors, as identified here, are modulated by soluble Aß42 and may represent useful early biomarkers.


Subject(s)
Amyloid beta-Peptides/metabolism , Cerebral Cortex/metabolism , Hippocampus/metabolism , Peptide Fragments/metabolism , Alzheimer Disease/cerebrospinal fluid , Amyloid beta-Peptides/genetics , Animals , Cell Line, Tumor , Gene Expression Regulation/physiology , Humans , Immunohistochemistry , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor Binding Protein 5/metabolism , Mice, Transgenic , Microarray Analysis , Peptide Fragments/genetics , Psychiatric Status Rating Scales , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction
9.
J Biol Chem ; 289(29): 20182-91, 2014 Jul 18.
Article in English | MEDLINE | ID: mdl-24878959

ABSTRACT

Although soluble species of the amyloid-ß peptide Aß42 correlate with disease symptoms in Alzheimer disease, little is known about the biological activities of amyloid-ß (Aß). Here, we show that Aß peptides varying in lengths from 38 to 43 amino acids are internalized by cultured neuroblastoma cells and can be found in the nucleus. By three independent methods, we demonstrate direct detection of nuclear Aß42 as follows: (i) biochemical analysis of nuclear fractions; (ii) detection of biotin-labeled Aß in living cells by confocal laser scanning microscopy; and (iii) transmission electron microscopy of Aß in cultured cells, as well as brain tissue of wild-type and transgenic APPPS1 mice (overexpression of amyloid precursor protein and presenilin 1 with Swedish and L166P mutations, respectively). Also, this study details a novel role for Aß42 in nuclear signaling, distinct from the amyloid precursor protein intracellular domain. Chromatin immunoprecipitation showed that Aß42 specifically interacts as a repressor of gene transcription with LRP1 and KAI1 promoters. By quantitative RT-PCR, we confirmed that mRNA levels of the examined candidate genes were exclusively decreased by the potentially neurotoxic Aß42 wild-type peptide. Shorter peptides (Aß38 or Aß40) and other longer peptides (nontoxic Aß42 G33A substitution or Aß43) did not affect mRNA levels. Overall, our data indicate that the nuclear translocation of Aß42 impacts gene regulation, and deleterious effects of Aß42 in Alzheimer disease pathogenesis may be influenced by altering the expression profiles of disease-modifying genes.


Subject(s)
Amyloid beta-Peptides/metabolism , Peptide Fragments/metabolism , Active Transport, Cell Nucleus , Alzheimer Disease/metabolism , Amino Acid Substitution , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/deficiency , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Cell Line , Gene Expression Regulation , HEK293 Cells , Humans , Mice , Mice, Knockout , Mice, Transgenic , Models, Molecular , Mutagenesis, Site-Directed , Neurons/metabolism , Neurons/ultrastructure , Peptide Fragments/chemistry , Peptide Fragments/genetics , Presenilin-1/deficiency , Presenilin-1/genetics , Presenilin-1/metabolism , Protein Multimerization , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Static Electricity
10.
Neuropharmacology ; 73: 56-65, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23707483

ABSTRACT

Protein kinase B (AKT) and glycogen synthase kinase 3 beta (GSK3ß) are two protein kinases involved in dopaminergic signaling. Dopamine-associated neuropsychiatric illnesses such as schizophrenia and bipolar disorder seem to be characterized by impairments in the AKT/GSK3ß network. Here, we sought evidence for the presence of molecular and functional changes in the AKT/GSK3ß pathway using an established infection-based mouse model of developmental neuropsychiatric disease that is based on prenatal administration of the viral mimetic poly(I:C) (=polyriboinosinic-polyribocytidilic acid). We found that adult offspring of poly(I:C)-exposed mothers displayed decreased total levels of AKT protein and reduced phosphorylation at AKT threonine residues in the medial prefrontal cortex. Prenatally immune challenged offspring also exhibited increased GSK3ß protein expression and activation status, the latter of which was evidenced by a decrease in the ratio between phosphorylated and total GSK3ß protein in the medial prefrontal cortex. These molecular changes were not associated with overt signs of inflammatory processes in the adult brain. We further found that acute pre-treatment with the selective GSK3ß inhibitor TDZD-8 dose-dependently normalized aberrant behavior typically emerging following prenatal immune activation, including deficient spontaneous alternation in the Y-maze and increased locomotor responses to systemic amphetamine treatment. Taken together, the present mouse model demonstrates that prenatal exposure to viral-like immune activation leads to long-term alterations in GSK3ß signaling, some of which are critically implicated in schizophrenia and bipolar disorder.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/psychology , Schizophrenia/metabolism , Schizophrenic Psychology , Amphetamine/antagonists & inhibitors , Amphetamine/pharmacology , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Inflammation Mediators/metabolism , Male , Maze Learning/drug effects , Mice , Motor Activity/drug effects , Phosphorylation , Poly I-C , Prefrontal Cortex/metabolism , Pregnancy , Proto-Oncogene Proteins c-akt/metabolism , Schizophrenia/chemically induced , Signal Transduction , Thiadiazoles/pharmacology
11.
Biol Psychiatry ; 72(11): 934-42, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-22705041

ABSTRACT

BACKGROUND: Trace amines, compounds structurally related to classical biogenic amines, represent endogenous ligands of the trace amine-associated receptor 1 (TAAR1). Because trace amines also influence the activity of other targets, selective ligands are needed for the elucidation of TAAR1 function. Here we report on the identification and characterization of the first selective and potent TAAR1 partial agonist. METHODS: The TAAR1 partial agonist RO5203648 was evaluated for its binding affinity and functional activity at rodent and primate TAAR1 receptors stably expressed in HEK293 cells, for its physicochemical and pharmacokinetic properties, for its effects on the firing frequency of monoaminergic neurons ex vivo, and for its properties in vivo with genetic and pharmacological models of central nervous system disorders. RESULTS: RO5203648 showed high affinity and potency at TAAR1, high selectivity versus other targets, and favorable pharmacokinetic properties. In mouse brain slices, RO5203648 increased the firing frequency of dopaminergic and serotonergic neurons in the ventral tegmental area and the dorsal raphe nucleus, respectively. In various behavioral paradigms in rodents and monkeys, RO5203648 demonstrated clear antipsychotic- and antidepressant-like activities as well as potential anxiolytic-like properties. Furthermore, it attenuated drug-taking behavior and was highly effective in promoting attention, cognitive performance, and wakefulness. CONCLUSIONS: With the first potent and selective TAAR1 partial agonist, RO5203648, we show that TAAR1 is implicated in a broad range of relevant physiological, behavioral, and cognitive neuropsychiatric dimensions. Collectively, these data uncover important neuromodulatory roles for TAAR1 and suggest that agonists at this receptor might have therapeutic potential in one or more neuropsychiatric domains.


Subject(s)
Dopaminergic Neurons/drug effects , Oxazoles/pharmacology , Raphe Nuclei/drug effects , Receptors, G-Protein-Coupled/agonists , Serotonergic Neurons/drug effects , Ventral Tegmental Area/drug effects , Action Potentials/drug effects , Action Potentials/physiology , Animals , Dopaminergic Neurons/physiology , Mice , Motor Activity/drug effects , Motor Activity/physiology , Raphe Nuclei/physiology , Serotonergic Neurons/physiology , Ventral Tegmental Area/physiology
12.
EMBO Mol Med ; 4(7): 647-59, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22514144

ABSTRACT

Here, we describe a novel missense mutation in the amyloid precursor protein (APP) causing a lysine-to-asparagine substitution at position 687 (APP770; herein, referred to as K16N according to amyloid-ß (Aß) numbering) resulting in an early onset dementia with an autosomal dominant inheritance pattern. The K16N mutation is located exactly at the α-secretase cleavage site and influences both APP and Aß. First, due to the K16N mutation APP secretion is affected and a higher amount of Aß peptides is being produced. Second, Aß peptides carrying the K16N mutation are unique in that the peptide itself is not harmful to neuronal cells. Severe toxicity, however, is evident upon equimolar mixture of wt and mutant peptides, mimicking the heterozygous state of the subject. Furthermore, Aß42 K16N inhibits fibril formation of Aß42 wild-type. Even more, Aß42 K16N peptides are protected against clearance activity by the major Aß-degrading enzyme neprilysin. Thus the mutation characterized here harbours a combination of risk factors that synergistically may contribute to the development of early onset Alzheimer disease.


Subject(s)
Amyloid beta-Peptides/metabolism , Peptide Fragments/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amino Acid Sequence , Amino Acid Substitution , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Peptides/genetics , Cell Line, Tumor , HEK293 Cells , Humans , Molecular Sequence Data , Mutation , Neprilysin/metabolism , Peptide Fragments/genetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Transfection
13.
Biol Chem ; 392(10): 919-26, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21801085

ABSTRACT

Amyloid plaques consisting of aggregated Aß peptide are a hallmark of Alzheimer's disease. Among the different forms of Aß, the one of 42aa length (Aß42) is most aggregation-prone and also the most neurotoxic. We find that eye-specific expression of human Aß42 in Drosophila results in a degeneration of eye structures that progresses with age. Dietary supplements of zinc or copper ions exacerbate eye damage. Positive effects are seen with zinc/copper chelators, or with elevated expression of MTF-1, a transcription factor with a key role in metal homeostasis and detoxification, or with human or fly transgenes encoding metallothioneins, metal scavenger proteins. These results show that a tight control of zinc and copper availability can minimize cellular damage associated with Aß42 expression.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Copper/metabolism , Eye/metabolism , Peptide Fragments/metabolism , Zinc/metabolism , Aging , Amyloid beta-Peptides/genetics , Animals , Chelating Agents/pharmacology , DNA-Binding Proteins/genetics , Drosophila , Eye/drug effects , Eye/pathology , Gene Expression , Humans , Metallothionein/genetics , Metallothionein/metabolism , Peptide Fragments/genetics , Transcription Factors/genetics , Transgenes , Up-Regulation , Transcription Factor MTF-1
14.
EMBO J ; 30(10): 2057-70, 2011 May 18.
Article in English | MEDLINE | ID: mdl-21441896

ABSTRACT

Formation of aberrant protein conformers is a common pathological denominator of different neurodegenerative disorders, such as Alzheimer's disease or prion diseases. Moreover, increasing evidence indicates that soluble oligomers are associated with early pathological alterations and that oligomeric assemblies of different disease-associated proteins may share common structural features. Previous studies revealed that toxic effects of the scrapie prion protein (PrP(Sc)), a ß-sheet-rich isoform of the cellular PrP (PrP(C)), are dependent on neuronal expression of PrP(C). In this study, we demonstrate that PrP(C) has a more general effect in mediating neurotoxic signalling by sensitizing cells to toxic effects of various ß-sheet-rich (ß) conformers of completely different origins, formed by (i) heterologous PrP, (ii) amyloid ß-peptide, (iii) yeast prion proteins or (iv) designed ß-peptides. Toxic signalling via PrP(C) requires the intrinsically disordered N-terminal domain (N-PrP) and the GPI anchor of PrP. We found that the N-terminal domain is important for mediating the interaction of PrP(C) with ß-conformers. Interestingly, a secreted version of N-PrP associated with ß-conformers and antagonized their toxic signalling via PrP(C). Moreover, PrP(C)-mediated toxic signalling could be blocked by an NMDA receptor antagonist or an oligomer-specific antibody. Our study indicates that PrP(C) can mediate toxic signalling of various ß-sheet-rich conformers independent of infectious prion propagation, suggesting a pathophysiological role of the prion protein beyond of prion diseases.


Subject(s)
Membrane Proteins/metabolism , Membrane Proteins/toxicity , PrPC Proteins/metabolism , PrPC Proteins/toxicity , Prion Diseases/pathology , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Cell Death , Humans , Membrane Proteins/chemistry , Neurons/drug effects , Neurons/physiology , PrPC Proteins/chemistry , Protein Conformation , Protein Interaction Mapping , Protein Structure, Tertiary , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/toxicity
15.
J Biol Chem ; 285(53): 41517-24, 2010 Dec 31.
Article in English | MEDLINE | ID: mdl-20971852

ABSTRACT

N-terminally truncated Aß peptides starting with pyroglutamate (AßpE3) represent a major fraction of all Aß peptides in the brain of Alzheimer disease (AD) patients. AßpE3 has a higher aggregation propensity and stability and shows increased toxicity compared with full-length Aß. In the present work, we generated a novel monoclonal antibody (9D5) that selectively recognizes oligomeric assemblies of AßpE3 and studied the potential involvement of oligomeric AßpE3 in vivo using transgenic mouse models as well as human brains from sporadic and familial AD cases. 9D5 showed an unusual staining pattern with almost nondetectable plaques in sporadic AD patients and non-demented controls. Interestingly, in sporadic and familial AD cases prominent intraneuronal and blood vessel staining was observed. Using a novel sandwich ELISA significantly decreased levels of oligomers in plasma samples from patients with AD compared with healthy controls were identified. Moreover, passive immunization of 5XFAD mice with 9D5 significantly reduced overall Aß plaque load and AßpE3 levels, and normalized behavioral deficits. These data indicate that 9D5 is a therapeutically and diagnostically effective monoclonal antibody targeting low molecular weight AßpE3 oligomers.


Subject(s)
Alzheimer Disease/metabolism , Pyrrolidonecarboxylic Acid/chemistry , Amyloid/chemistry , Animals , Behavior, Animal , Brain/metabolism , Cell Line, Tumor , Chromatography/methods , Enzyme-Linked Immunosorbent Assay/methods , Humans , Immunochemistry/methods , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Weight , Protein Structure, Tertiary , Transgenes
16.
J Neural Transm (Vienna) ; 117(1): 85-96, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19823761

ABSTRACT

The presence of Abeta(pE3) (N-terminal truncated Abeta starting with pyroglutamate) in Alzheimer's disease (AD) has received considerable attention since the discovery that this peptide represents a dominant fraction of Abeta peptides in senile plaques of AD brains. This was later confirmed by other reports investigating AD and Down's syndrome postmortem brain tissue. Importantly, Abeta(pE3) has a higher aggregation propensity, and stability, and shows an increased toxicity compared to full-length Abeta. We have recently shown that intraneuronal accumulation of Abeta(pE3) peptides induces a severe neuron loss and an associated neurological phenotype in the TBA2 mouse model for AD. Given the increasing interest in Abeta(pE3), we have generated two novel monoclonal antibodies which were characterized as highly specific for Abeta(pE3) peptides and herein used to analyze plaque deposition in APP/PS1KI mice, an AD model with severe neuron loss and learning deficits. This was compared with the plaque pattern present in brain tissue from sporadic and familial AD cases. Abundant plaques positive for Abeta(pE3) were present in patients with sporadic AD and familial AD including those carrying mutations in APP (arctic and Swedish) and PS1. Interestingly, in APP/PS1KI mice we observed a continuous increase in Abeta(pE3) plaque load with increasing age, while the density for Abeta(1-x ) plaques declined with aging. We therefore assume that, in particular, the peptides starting with position 1 of Abeta are N-truncated as disease progresses, and that, Abeta(pE3) positive plaques are resistant to age-dependent degradation likely due to their high stability and propensity to aggregate.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Brain/metabolism , Brain/pathology , Peptide Fragments/metabolism , Aged , Aged, 80 and over , Aging/metabolism , Aging/pathology , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Transgenic , Middle Aged , Mutation , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Presenilin-1/genetics , Protease Nexins , Pyrrolidonecarboxylic Acid/metabolism , Receptors, Cell Surface/genetics
17.
J Neurosci ; 29(23): 7582-90, 2009 Jun 10.
Article in English | MEDLINE | ID: mdl-19515926

ABSTRACT

The aggregation of the amyloid-beta (Abeta) peptide plays a pivotal role in the pathogenesis of Alzheimer's disease, as soluble oligomers are intimately linked to neuronal toxicity and inhibition of hippocampal long-term potentiation (LTP). In the C-terminal region of Abeta there are three consecutive GxxxG dimerization motifs, which we could previously demonstrate to play a critical role in the generation of Abeta. Here, we show that glycine 33 (G33) of the central GxxxG interaction motif within the hydrophobic Abeta sequence is important for the aggregation dynamics of the peptide. Abeta peptides with alanine or isoleucine substitutions of G33 displayed an increased propensity to form higher oligomers, which we could attribute to conformational changes. Importantly, the oligomers of G33 variants were much less toxic than Abeta(42) wild type (WT), in vitro and in vivo. Also, whereas Abeta(42) WT is known to inhibit LTP, Abeta(42) G33 variants had lost the potential to inhibit LTP. Our findings reveal that conformational changes induced by G33 substitutions unlink toxicity and oligomerization of Abeta on the molecular level and suggest that G33 is the key amino acid in the toxic activity of Abeta. Thus, a specific toxic conformation of Abeta exists, which represents a promising target for therapeutic interventions.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Glycine/chemistry , Neuronal Plasticity/physiology , Amino Acid Motifs , Amino Acid Sequence , Amino Acid Substitution , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/toxicity , Animals , Cell Death/physiology , Cell Line, Tumor , Cells, Cultured , Drosophila melanogaster , Excitatory Postsynaptic Potentials , Eye/metabolism , Hippocampus/physiology , Humans , Hydrophobic and Hydrophilic Interactions , Long-Term Potentiation/physiology , Models, Molecular , Molecular Sequence Data , Mutation, Missense , Neurons/physiology , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peptide Fragments/toxicity , Protein Conformation , Protein Multimerization , Rats , Rats, Wistar
18.
J Biol Chem ; 283(21): 14826-34, 2008 May 23.
Article in English | MEDLINE | ID: mdl-18362153

ABSTRACT

Sortilin-related receptor with A-type repeats (SORLA) is a sorting receptor that impairs processing of amyloid precursor protein (APP) to soluble (s) APP and to the amyloid beta-peptide in cultured neurons and is poorly expressed in patients with Alzheimer disease (AD). Here, we evaluated the consequences of Sorla gene defects on brain anatomy and function using mouse models of receptor deficiency. In line with a protective role for SORLA in APP metabolism, lack of the receptor results in increased amyloidogenic processing of endogenous APP and in aggravated plaque deposition when introduced into PDAPP mice expressing mutant human APP. Surprisingly, increased levels of sAPP caused by receptor deficiency correlate with pro-found stimulation of neuronal ERK signaling and with enhanced neurogenesis, providing in vivo support for neurotrophic functions of sAPP. Our data document a role for SORLA not only in control of plaque burden but also in APP-dependent neuronal signaling and suggest a molecular explanation for increased neurogenesis observed in some AD patients.


Subject(s)
Aging/physiology , Amyloid beta-Protein Precursor/metabolism , Cell Differentiation , MAP Kinase Signaling System , Membrane Transport Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Receptors, LDL/metabolism , Animals , Electrophysiology , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Membrane Transport Proteins/deficiency , Membrane Transport Proteins/genetics , Mice , Mice, Knockout , Patch-Clamp Techniques , Receptors, LDL/deficiency , Receptors, LDL/genetics
19.
EMBO J ; 26(6): 1702-12, 2007 Mar 21.
Article in English | MEDLINE | ID: mdl-17332749

ABSTRACT

Processing of the amyloid precursor protein (APP) by beta- and gamma-secretases leads to the generation of amyloid-beta (Abeta) peptides with varying lengths. Particularly Abeta42 contributes to cytotoxicity and amyloid accumulation in Alzheimer's disease (AD). However, the precise molecular mechanism of Abeta42 generation has remained unclear. Here, we show that an amino-acid motif GxxxG within the APP transmembrane sequence (TMS) has regulatory impact on the Abeta species produced. In a neuronal cell system, mutations of glycine residues G29 and G33 of the GxxxG motif gradually attenuate the TMS dimerization strength, specifically reduce the formation of Abeta42, leave the level of Abeta40 unaffected, but increase Abeta38 and shorter Abeta species. We show that glycine residues G29 and G33 are part of a dimerization site within the TMS, but do not impair oligomerization of the APP ectodomain. We conclude that gamma-secretase cleavages of APP are intimately linked to the dimerization strength of the substrate TMS. The results demonstrate that dimerization of APP TMS is a risk factor for AD due to facilitating Abeta42 production.


Subject(s)
Alzheimer Disease/genetics , Amino Acid Motifs/genetics , Amyloid beta-Peptides/biosynthesis , Amyloid beta-Protein Precursor/genetics , Models, Molecular , Peptide Fragments/biosynthesis , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/genetics , Blotting, Western , Dimerization , Enzyme-Linked Immunosorbent Assay , Fluorescence Resonance Energy Transfer , Humans , Immunoprecipitation , Mutation/genetics , Peptide Fragments/genetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
SELECTION OF CITATIONS
SEARCH DETAIL
...