Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
J Neurosci ; 44(27)2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38692734

ABSTRACT

Aberrant condensation and localization of the RNA-binding protein (RBP) fused in sarcoma (FUS) occur in variants of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Changes in RBP function are commonly associated with changes in axonal cytoskeletal organization and branching in neurodevelopmental disorders. Here, we asked whether branching defects also occur in vivo in a model of FUS-associated disease. We use two reported Xenopus models of ALS/FTD (of either sex), the ALS-associated mutant FUS(P525L) and a mimic of hypomethylated FUS, FUS(16R). Both mutants strongly reduced axonal complexity in vivo. We also observed an axon looping defect for FUS(P525L) in the target area, which presumably arises due to errors in stop cue signaling. To assess whether the loss of axon complexity also had a cue-independent component, we assessed axonal cytoskeletal integrity in vitro. Using a novel combination of fluorescence and atomic force microscopy, we found that mutant FUS reduced actin density in the growth cone, altering its mechanical properties. Therefore, FUS mutants may induce defects during early axonal development.


Subject(s)
Amyotrophic Lateral Sclerosis , Axons , Frontotemporal Dementia , Mutation , RNA-Binding Protein FUS , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/metabolism , Axons/pathology , Axons/metabolism , Animals , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/metabolism , Frontotemporal Dementia/genetics , Frontotemporal Dementia/pathology , Frontotemporal Dementia/metabolism , Female , Male , Xenopus laevis , Growth Cones/metabolism , Humans , Disease Models, Animal
2.
EMBO J ; 41(19): e110777, 2022 10 04.
Article in English | MEDLINE | ID: mdl-35993436

ABSTRACT

The regulation of membrane lipid composition is critical for cellular homeostasis. Cells are particularly sensitive to phospholipid saturation, with increased saturation causing membrane rigidification and lipotoxicity. How mammalian cells sense membrane lipid composition and reverse fatty acid (FA)-induced membrane rigidification is poorly understood. Here we systematically identify proteins that differ between mammalian cells fed saturated versus unsaturated FAs. The most differentially expressed proteins were two ER-resident polytopic membrane proteins: the E3 ubiquitin ligase RNF145 and the lipid hydrolase ADIPOR2. In unsaturated lipid membranes, RNF145 is stable, promoting its lipid-sensitive interaction, ubiquitination and degradation of ADIPOR2. When membranes become enriched in saturated FAs, RNF145 is rapidly auto-ubiquitinated and degraded, stabilising ADIPOR2, whose hydrolase activity restores lipid homeostasis and prevents lipotoxicity. We therefore identify RNF145 as a FA-responsive ubiquitin ligase which, together with ADIPOR2, defines an autoregulatory pathway that controls cellular membrane lipid homeostasis and prevents acute lipotoxic stress.


Subject(s)
Hydrolases , Membrane Fluidity , Animals , Fatty Acids/metabolism , Hydrolases/metabolism , Mammals , Membrane Proteins/metabolism , Phospholipids , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
3.
Alzheimers Dement ; 17(2): 149-163, 2021 02.
Article in English | MEDLINE | ID: mdl-33314529

ABSTRACT

INTRODUCTION: Microglial TYROBP (DAP12) is a network hub and driver in sporadic late-onset Alzheimer's disease (AD). TYROBP is a cytoplasmic adaptor for TREM2 and other receptors, but little is known about its roles and actions in AD. Herein, we demonstrate that endogenous Tyrobp transcription is specifically increased in recruited microglia. METHODS: Using a novel transgenic mouse overexpressing TYROBP in microglia, we observed a decrease of the amyloid burden and an increase of TAU phosphorylation stoichiometry when crossed with APP/PSEN1 or MAPTP301S mice, respectively. Characterization of these mice revealed Tyrobp-related modulation of apolipoprotein E (Apoe) transcription. We also showed that Tyrobp and Apoe mRNAs were increased in Trem2-null microglia recruited around either amyloid beta deposits or a cortical stab injury. Conversely, microglial Apoe transcription was dramatically diminished when Tyrobp was absent. CONCLUSIONS: Our results provide evidence that TYROBP-APOE signaling does not require TREM2 and could be an initiating step in establishment of the disease-associated microglia (DAM) phenotype.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Alzheimer Disease/metabolism , Apolipoproteins E/genetics , Membrane Glycoproteins/genetics , Mice, Transgenic , Microglia/metabolism , Receptors, Immunologic/genetics , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/physiology , Amyloidosis/prevention & control , Animals , Brain/metabolism , Brain/pathology , Disease Models, Animal , Humans , Mice , Mice, Knockout , Phosphorylation , Presenilin-1/physiology , Signal Transduction , tau Proteins/metabolism
4.
Hum Mol Genet ; 28(R2): R187-R196, 2019 11 21.
Article in English | MEDLINE | ID: mdl-31595953

ABSTRACT

Recent work on the biophysics of proteins with low complexity, intrinsically disordered domains that have the capacity to form biological condensates has profoundly altered the concepts about the pathogenesis of inherited and sporadic neurodegenerative disorders associated with pathological accumulation of these proteins. In the present review, we use the FUS, TDP-43 and A11 proteins as examples to illustrate how missense mutations and aberrant post-translational modifications of these proteins cause amyotrophic lateral sclerosis (ALS) and fronto-temporal lobar degeneration (FTLD).


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Annexins/genetics , DNA-Binding Proteins/genetics , Frontotemporal Lobar Degeneration/genetics , RNA-Binding Protein FUS/chemistry , Temporal Lobe/physiopathology , Annexins/chemistry , Annexins/metabolism , Biological Transport/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Humans , Intracellular Membranes/chemistry , Intracellular Membranes/metabolism , Mutation, Missense , Neurodegenerative Diseases/physiopathology , Neurons/chemistry , Neurons/metabolism , Protein Processing, Post-Translational/genetics , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/metabolism
5.
Mol Psychiatry ; 24(9): 1383-1397, 2019 09.
Article in English | MEDLINE | ID: mdl-30283031

ABSTRACT

TYROBP/DAP12 forms complexes with ectodomains of immune receptors (TREM2, SIRPß1, CR3) associated with Alzheimer's disease (AD) and is a network hub and driver in the complement subnetwork identified by multi-scale gene network studies of postmortem human AD brain. Using transgenic or viral approaches, we characterized in mice the effects of TYROBP deficiency on the phenotypic and pathological evolution of tauopathy. Biomarkers usually associated with worsening clinical phenotype (i.e., hyperphosphorylation and increased tauopathy spreading) were unexpectedly increased in MAPTP301S;Tyrobp-/- mice despite the improved learning behavior and synaptic function relative to controls with normal levels of TYROBP. Notably, levels of complement cascade initiator C1q were reduced in MAPTP301S;Tyrobp-/- mice, consistent with the prediction that C1q reduction exerts a neuroprotective effect. These observations suggest a model wherein TYROBP-KO-(knock-out)-associated reduction in C1q is associated with normalized learning behavior and electrophysiological properties in tauopathy model mice despite a paradoxical evolution of biomarker signatures usually associated with neurological decline.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Alzheimer Disease/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/physiology , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/metabolism , Animals , Animals, Genetically Modified , Brain/metabolism , Complement C1q/metabolism , Complement C1q/physiology , Disease Models, Animal , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Proteins/physiology , Mice , Mice, Knockout , Mice, Transgenic , Microglia/metabolism , Phenotype , Phosphorylation , Plaque, Amyloid/metabolism , Tauopathies/genetics , tau Proteins/metabolism
6.
EMBO Mol Med ; 9(10): 1366-1378, 2017 10.
Article in English | MEDLINE | ID: mdl-28855301

ABSTRACT

We have characterised the proteolytic cleavage events responsible for the shedding of triggering receptor expressed on myeloid cells 2 (TREM2) from primary cultures of human macrophages, murine microglia and TREM2-expressing human embryonic kidney (HEK293) cells. In all cell types, a soluble 17 kDa N-terminal cleavage fragment was shed into the conditioned media in a constitutive process that is inhibited by G1254023X and metalloprotease inhibitors and siRNA targeting ADAM10. Inhibitors of serine proteases and matrix metalloproteinases 2/9, and ADAM17 siRNA did not block TREM2 shedding. Peptidomimetic protease inhibitors highlighted a possible cleavage site, and mass spectrometry confirmed that shedding occurred predominantly at the H157-S158 peptide bond for both wild-type and H157Y human TREM2 and for the wild-type murine orthologue. Crucially, we also show that the Alzheimer's disease-associated H157Y TREM2 variant was shed more rapidly than wild type from HEK293 cells, possibly by a novel, batimastat- and ADAM10-siRNA-independent, sheddase activity. These insights offer new therapeutic targets for modulating the innate immune response in Alzheimer's and other neurological diseases.


Subject(s)
Alzheimer Disease/genetics , Membrane Glycoproteins/metabolism , Proteolysis , Receptors, Immunologic/metabolism , ADAM10 Protein/genetics , ADAM10 Protein/metabolism , ADAM17 Protein/genetics , ADAM17 Protein/metabolism , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , Animals, Newborn , Culture Media, Conditioned , HEK293 Cells , Humans , Ketocholesterols/pharmacology , Macrophages/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Membrane Glycoproteins/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Microglia/metabolism , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Immunologic/genetics
7.
Science ; 356(6343): 1123-1124, 2017 06 16.
Article in English | MEDLINE | ID: mdl-28619900
8.
Article in English | MEDLINE | ID: mdl-28320827

ABSTRACT

The presenilin proteins are the catalytic subunits of a tetrameric complex containing presenilin 1 or 2, anterior pharynx defective 1 (APH1), nicastrin, and PEN-2. Other components such as TMP21 may exist in a subset of specialized complexes. The presenilin complex is the founding member of a unique class of aspartyl proteases that catalyze the γ, ɛ, ζ site cleavage of the transmembrane domains of Type I membrane proteins including amyloid precursor protein (APP) and Notch. Here, we detail the structural and chemical biology of this unusual enzyme. Taken together, these studies suggest that the complex exists in several conformations, and subtle long-range (allosteric) shifts in the conformation of the complex underpin substrate access to the catalytic site and the mechanism of action for allosteric inhibitors and modulators. Understanding the mechanics of these shifts will facilitate the design of γ-secretase modulator (GSM) compounds that modulate the relative efficiency of γ, ɛ, ζ site cleavage and/or substrate specificity.


Subject(s)
Amyloid beta-Protein Precursor/chemistry , Presenilin-1/chemistry , Presenilin-2/chemistry , Amyloid beta-Protein Precursor/metabolism , Animals , Humans , Presenilin-1/metabolism , Presenilin-2/metabolism
9.
Alzheimers Dement ; 13(7): 727-738, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28183528

ABSTRACT

INTRODUCTION: Genetic loci for Alzheimer's disease (AD) have been identified in whites of European ancestry, but the genetic architecture of AD among other populations is less understood. METHODS: We conducted a transethnic genome-wide association study (GWAS) for late-onset AD in Stage 1 sample including whites of European Ancestry, African-Americans, Japanese, and Israeli-Arabs assembled by the Alzheimer's Disease Genetics Consortium. Suggestive results from Stage 1 from novel loci were followed up using summarized results in the International Genomics Alzheimer's Project GWAS dataset. RESULTS: Genome-wide significant (GWS) associations in single-nucleotide polymorphism (SNP)-based tests (P < 5 × 10-8) were identified for SNPs in PFDN1/HBEGF, USP6NL/ECHDC3, and BZRAP1-AS1 and for the interaction of the (apolipoprotein E) APOE ε4 allele with NFIC SNP. We also obtained GWS evidence (P < 2.7 × 10-6) for gene-based association in the total sample with a novel locus, TPBG (P = 1.8 × 10-6). DISCUSSION: Our findings highlight the value of transethnic studies for identifying novel AD susceptibility loci.


Subject(s)
Alzheimer Disease/genetics , Genome-Wide Association Study , Polymorphism, Single Nucleotide , Adaptor Proteins, Signal Transducing/genetics , Apolipoprotein E4/genetics , GTPase-Activating Proteins/genetics , Genetic Predisposition to Disease , Heparin-binding EGF-like Growth Factor/genetics , Humans , Membrane Glycoproteins/genetics , Molecular Chaperones/genetics , NFI Transcription Factors/genetics , Peroxisomal Bifunctional Enzyme/genetics , Receptors, GABA/genetics
10.
Brain ; 139(Pt 1): 73-85, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26556829

ABSTRACT

Charcot-Marie-Tooth disease is a group of hereditary peripheral neuropathies that share clinical characteristics of progressive distal muscle weakness and atrophy, foot deformities, distal sensory loss, as well as diminished tendon reflexes. Hundreds of causative DNA changes have been found, but much of the genetic basis of the disease is still unexplained. Mutations in the ALS5/SPG11/KIAA1840 gene are a frequent cause of autosomal recessive hereditary spastic paraplegia with thin corpus callosum and peripheral axonal neuropathy, and account for ∼ 40% of autosomal recessive juvenile amyotrophic lateral sclerosis. The overlap of axonal Charcot-Marie-Tooth disease with both diseases, as well as the common autosomal recessive inheritance pattern of thin corpus callosum and axonal Charcot-Marie-Tooth disease in three related patients, prompted us to analyse the ALS5/SPG11/KIAA1840 gene in affected individuals with autosomal recessive axonal Charcot-Marie-Tooth disease. We investigated 28 unrelated families with autosomal recessive axonal Charcot-Marie-Tooth disease defined by clinical, electrophysiological, as well as pathological evaluation. Besides, we screened for all the known genes related to axonal autosomal recessive Charcot-Marie-Tooth disease (CMT2A2/HMSN2A2/MFN2, CMT2B1/LMNA, CMT2B2/MED25, CMT2B5/NEFL, ARCMT2F/dHMN2B/HSPB1, CMT2K/GDAP1, CMT2P/LRSAM1, CMT2R/TRIM2, CMT2S/IGHMBP2, CMT2T/HSJ1, CMTRID/COX6A1, ARAN-NM/HINT and GAN/GAN), for the genes related to autosomal recessive hereditary spastic paraplegia with thin corpus callosum and axonal peripheral neuropathy (SPG7/PGN, SPG15/ZFYVE26, SPG21/ACP33, SPG35/FA2H, SPG46/GBA2, SPG55/C12orf65 and SPG56/CYP2U1), as well as for the causative gene of peripheral neuropathy with or without agenesis of the corpus callosum (SLC12A6). Mitochondrial disorders related to Charcot-Marie-Tooth disease type 2 were also excluded by sequencing POLG and TYMP genes. An additional locus for autosomal recessive Charcot-Marie-Tooth disease type 2H on chromosome 8q13-21.1 was excluded by linkage analysis. Pedigrees originated in Italy, Brazil, Canada, England, Iran, and Japan. Interestingly, we identified 15 ALS5/SPG11/KIAA1840 mutations in 12 families (two sequence variants were never reported before, p.Gln198* and p.Pro2212fs*5). No large deletions/duplications were detected in these patients. The novel mutations seemed to be pathogenic since they co-segregated with the disease in all pedigrees and were absent in 300 unrelated controls. Furthermore, in silico analysis predicted their pathogenic effect. Our results indicate that ALS5/SPG11/KIAA1840 is the causative gene of a wide spectrum of clinical features, including autosomal recessive axonal Charcot-Marie-Tooth disease.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Genes, Recessive/genetics , Mutation , Proteins/genetics , Case-Control Studies , Female , Humans , Male , Pedigree , Peripheral Nervous System Diseases/genetics , Spastic Paraplegia, Hereditary/genetics
11.
Mol Neurodegener ; 9: 59, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25523933

ABSTRACT

The presenilin genes were first identified as the site of missense mutations causing early onset autosomal dominant familial Alzheimer's disease. Subsequent work has shown that the presenilin proteins are the catalytic subunits of a hetero-tetrameric complex containing APH1, nicastrin and PEN-2. This complex (variously termed presenilin complex or gamma-secretase complex) performs an unusual type of proteolysis in which the transmembrane domains of Type I proteins are cleaved within the hydrophobic compartment of the membrane. This review describes some of the molecular and structural biology of this unusual enzyme complex. The presenilin complex is a bilobed structure. The head domain contains the ectodomain of nicastrin. The base domain contains a central cavity with a lateral cleft that likely provides the route for access of the substrate to the catalytic cavity within the centre of the base domain. There are reciprocal allosteric interactions between various sites in the complex that affect its function. For instance, binding of Compound E, a peptidomimetic inhibitor to the PS1 N-terminus, induces significant conformational changes that reduces substrate binding at the initial substrate docking site, and thus inhibits substrate cleavage. However, there is a reciprocal allosteric interaction between these sites such that prior binding of the substrate to the initial docking site paradoxically increases the binding of the Compound E peptidomimetic inhibitor. Such reciprocal interactions are likely to form the basis of a gating mechanism that underlies access of substrate to the catalytic site. An increasingly detailed understanding of the structural biology of the presenilin complex is an essential step towards rational design of substrate- and/or cleavage site-specific modulators of presenilin complex function.


Subject(s)
Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Cell Membrane/metabolism , Presenilin-1/metabolism , Proteolysis , Animals , Humans , Molecular Docking Simulation/methods , Presenilin-1/chemistry , Protein Binding
12.
JAMA Neurol ; 71(11): 1394-404, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25199842

ABSTRACT

IMPORTANCE: Because APOE locus variants contribute to risk of late-onset Alzheimer disease (LOAD) and to differences in age at onset (AAO), it is important to know whether other established LOAD risk loci also affect AAO in affected participants. OBJECTIVES: To investigate the effects of known Alzheimer disease risk loci in modifying AAO and to estimate their cumulative effect on AAO variation using data from genome-wide association studies in the Alzheimer Disease Genetics Consortium. DESIGN, SETTING, AND PARTICIPANTS: The Alzheimer Disease Genetics Consortium comprises 14 case-control, prospective, and family-based data sets with data on 9162 participants of white race/ethnicity with Alzheimer disease occurring after age 60 years who also had complete AAO information, gathered between 1989 and 2011 at multiple sites by participating studies. Data on genotyped or imputed single-nucleotide polymorphisms most significantly associated with risk at 10 confirmed LOAD loci were examined in linear modeling of AAO, and individual data set results were combined using a random-effects, inverse variance-weighted meta-analysis approach to determine whether they contribute to variation in AAO. Aggregate effects of all risk loci on AAO were examined in a burden analysis using genotype scores weighted by risk effect sizes. MAIN OUTCOMES AND MEASURES: Age at disease onset abstracted from medical records among participants with LOAD diagnosed per standard criteria. RESULTS: Analysis confirmed the association of APOE with earlier AAO (P = 3.3 × 10(-96)), with associations in CR1 (rs6701713, P = 7.2 × 10(-4)), BIN1 (rs7561528, P = 4.8 × 10(-4)), and PICALM (rs561655, P = 2.2 × 10(-3)) reaching statistical significance (P < .005). Risk alleles individually reduced AAO by 3 to 6 months. Burden analyses demonstrated that APOE contributes to 3.7% of the variation in AAO (R(2) = 0.256) over baseline (R(2) = 0.221), whereas the other 9 loci together contribute to 2.2% of the variation (R(2) = 0.242). CONCLUSIONS AND RELEVANCE: We confirmed an association of APOE (OMIM 107741) variants with AAO among affected participants with LOAD and observed novel associations of CR1 (OMIM 120620), BIN1 (OMIM 601248), and PICALM (OMIM 603025) with AAO. In contrast to earlier hypothetical modeling, we show that the combined effects of Alzheimer disease risk variants on AAO are on the scale of, but do not exceed, the APOE effect. While the aggregate effects of risk loci on AAO may be significant, additional genetic contributions to AAO are individually likely to be small.


Subject(s)
Alzheimer Disease/genetics , Genetic Loci/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study , Adult , Age of Onset , Aged , Aged, 80 and over , Alzheimer Disease/epidemiology , Female , Genotype , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide/genetics , Prospective Studies , Risk Factors
13.
J Comp Neurol ; 522(10): 2319-35, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24415002

ABSTRACT

Alzheimer's disease (AD) is a complex and slowly progressing dementing disorder that results in neuronal and synaptic loss, deposition in brain of aberrantly folded proteins, and impairment of spatial and episodic memory. Most studies of mouse models of AD have employed analyses of cognitive status and assessment of amyloid burden, gliosis, and molecular pathology during disease progression. Here we sought to understand the behavioral, cellular, ultrastructural, and molecular changes that occur at a pathological stage equivalent to the early stages of human AD. We studied the TgCRND8 mouse, a model of aggressive AD amyloidosis, at an early stage of plaque pathology (3 months of age) in comparison to their wildtype littermates and assessed changes in cognition, neuron and spine structure, and expression of synaptic glutamate receptor proteins. We found that, at this age, TgCRND8 mice display substantial plaque deposition in the neocortex and hippocampus and impairment on cued and contextual memory tasks. Of particular interest, we also observed a significant decrease in the number of neurons in the hippocampus. Furthermore, analysis of CA1 neurons revealed significant changes in apical and basal dendritic spine types, as well as altered expression of GluN1 and GluA2 receptors. This change in molecular architecture within the hippocampus may reflect a rising representation of inherently less stable thin spine populations, which can cause cognitive decline. These changes, taken together with toxic insults from amyloid-ß protein, may underlie the observed neuronal loss.


Subject(s)
Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Fear , Memory Disorders/pathology , Memory Disorders/physiopathology , Neuronal Plasticity/physiology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Amyloidosis/metabolism , Amyloidosis/pathology , Animals , Brain/metabolism , Brain/pathology , Cell Count , Cues , Dendrites/metabolism , Dendrites/pathology , Dendritic Spines/metabolism , Dendritic Spines/pathology , Disease Models, Animal , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology
14.
Structure ; 22(1): 125-35, 2014 Jan 07.
Article in English | MEDLINE | ID: mdl-24210759

ABSTRACT

Presenilin-mediated endoproteolysis of transmembrane proteins plays a key role in physiological signaling and in the pathogenesis of Alzheimer disease and some cancers. Numerous inhibitors have been found via library screens, but their structural mechanisms remain unknown. We used several biophysical techniques to investigate the structure of human presenilin complexes and the effects of peptidomimetic γ-secretase inhibitors. The complexes are bilobed. The head contains nicastrin ectodomain. The membrane-embedded base has a central channel and a lateral cleft, which may represent the initial substrate docking site. Inhibitor binding induces widespread structural changes, including rotation of the head and closure of the lateral cleft. These changes block substrate access to the catalytic pocket and inhibit the enzyme. Intriguingly, peptide substrate docking has reciprocal effects on the inhibitor binding site. Similar reciprocal shifts may underlie the mechanisms of other inhibitors and of the "lateral gate" through which substrates access to the catalytic site.


Subject(s)
Benzodiazepinones/chemistry , Enzyme Inhibitors/chemistry , Peptidomimetics/chemistry , Presenilin-1/chemistry , Recombinant Fusion Proteins/chemistry , Allosteric Regulation , Amyloid Precursor Protein Secretases/chemistry , Catalytic Domain , HEK293 Cells , Humans , Ligands , Membrane Glycoproteins/chemistry , Molecular Docking Simulation , Presenilin-1/antagonists & inhibitors , Presenilin-1/genetics , Protein Structure, Tertiary , Proteolysis , Recombinant Fusion Proteins/genetics , Structure-Activity Relationship
15.
Mol Neurodegener ; 7: 58, 2012 Nov 22.
Article in English | MEDLINE | ID: mdl-23173713

ABSTRACT

BACKGROUND: The accumulation of amyloid beta (Aß) oligomers or fibrils is thought to be one of the main causes of synaptic and neuron loss, believed to underlie cognitive dysfunction in Alzheimer's disease (AD). Neuron loss has rarely been documented in amyloid precursor protein (APP) transgenic mouse models. We investigated whether two APP mouse models characterized by different folding states of amyloid showed different neuronal densities using an accurate method of cell counting. FINDINGS: We examined total cell and neuronal populations in Swedish/Indiana APP mutant mice (TgCRND8) with severe Aß pathology that includes fibrils, plaques, and oligomers, and Dutch APP mutant mice with only Aß oligomer pathology. Using the isotropic fractionator, we found no differences from control mice in regional total cell populations in either TgCRND8 or Dutch mice. However, there were 31.8% fewer hippocampal neurons in TgCRND8 compared to controls, while no such changes were observed in Dutch mice. CONCLUSIONS: We show that the isotropic fractionator is a convenient method for estimating neuronal content in milligram quantities of brain tissue and represents a useful tool to assess cell loss efficiently in transgenic models with different types of neuropathology. Our data support the hypothesis that TgCRND8 mice with a spectrum of Aß plaque, fibril, and oligomer pathology exhibit neuronal loss whereas Dutch mice with only oligomers, showed no evidence for neuronal loss. This suggests that the combination of plaques, fibrils, and oligomers causes more damage to mouse hippocampal neurons than Aß oligomers alone.


Subject(s)
Alzheimer Disease/pathology , Cell Count/methods , Cell Fractionation/methods , Hippocampus/pathology , Neurons/pathology , Animals , Disease Models, Animal , Mice , Mice, Transgenic
17.
Arch Neurol ; 68(1): 99-106, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21220680

ABSTRACT

OBJECTIVE: To reexamine the association between the neuronal sortilin-related receptor gene (SORL1) and Alzheimer disease (AD). DESIGN: Comprehensive and unbiased meta-analysis of all published and unpublished data from case-control studies for the SORL1 single-nucleotide polymorphisms (SNPs) that had been repeatedly assessed across studies. SETTING: Academic research institutions in the United States, the Netherlands, Canada, Belgium, the United Kingdom, Singapore, Japan, Sweden, Germany, France, and Italy. PARTICIPANTS: All published white and Asian case-control data sets, which included a total of 12,464 cases and 17,929 controls. MAIN OUTCOME MEASURES: Alzheimer disease according to the Diagnostic and Statistical Manual of Mental Disorders (Fourth Edition) and the National Institute of Neurological and Communicative Disorders and Stroke and the Alzheimer's Disease and Related Disorders Association (now known as the Alzheimer's Association). RESULTS: In the white data sets, several markers were associated with AD after correction for multiple testing, including previously reported SNPs 8, 9, and 10 (P < .001). In addition, the C-G-C haplotype at SNPs 8 through 10 was associated with AD risk (P < .001). In the combined Asian data sets, SNPs 19 and 23 through 25 were associated with AD risk (P < .001). The disease-associated alleles at SNPs 8, 9, and 10 (120,873,131-120,886,175 base pairs [bp]; C-G-C alleles), at SNP 19 (120,953,300 bp; G allele), and at SNPs 24 through 25 (120,988,611 bp; T and C alleles) were the same previously reported alleles. The SNPs 4 through 5, 8 through 10, 12, and 19 through 25 belong to distinct linkage disequilibrium blocks. The same alleles at SNPs 8 through 10 (C-G-C), 19 (G), and 24 and 25 (T and C) have also been associated with AD endophenotypes, including white matter hyperintensities and hippocampal atrophy on magnetic resonance imaging, cerebrospinal fluid measures of amyloid ß-peptide 42, and full-length SORL1 expression in the human brain. CONCLUSION: This comprehensive meta-analysis provides confirmatory evidence that multiple SORL1 variants in distinct linkage disequilibrium blocks are associated with AD.


Subject(s)
Alzheimer Disease/genetics , Genetic Variation/genetics , LDL-Receptor Related Proteins/genetics , Membrane Transport Proteins/genetics , Nerve Tissue Proteins/genetics , Alleles , Alzheimer Disease/epidemiology , Alzheimer Disease/etiology , Case-Control Studies , Humans , Polymorphism, Single Nucleotide/genetics
18.
Prog Neurobiol ; 93(3): 405-20, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21163327

ABSTRACT

Prion diseases are fatal neurodegenerative diseases of humans and animals which, in addition to sporadic and familial modes of manifestation, can be acquired via an infectious route of propagation. In disease, the prion protein (PrP(C)) undergoes a structural transition to its disease-causing form (PrP(Sc)) with profoundly different physicochemical properties. Surprisingly, despite intense interest in the prion protein, its function in the context of other cellular activities has largely remained elusive. We recently employed quantitative mass spectrometry to characterize the interactome of the prion protein in a murine neuroblastoma cell line (N2a), an established cell model for prion replication. Extensive bioinformatic analyses subsequently established an evolutionary link between the prion gene family and the family of ZIP (Zrt-, Irt-like protein) metal ion transporters. More specifically, sequence alignments, structural threading data and multiple additional pieces of evidence placed a ZIP5/ZIP6/ZIP10-like ancestor gene at the root of the PrP gene family. In this review we examine the biology of prion proteins and ZIP transporters from the viewpoint of a shared phylogenetic origin. We summarize and compare available data that shed light on genetics, function, expression, signaling, post-translational modifications and metal binding preferences of PrP and ZIP family members. Finally, we explore data indicative of retropositional origins of the prion gene founder and discuss a possible function for the prion-like (PL) domain within ZIP transporters. While throughout the article emphasis is placed on ZIP proteins, the intent is to highlight connections between PrP and ZIP transporters and uncover promising directions for future research.


Subject(s)
Multigene Family , Prion Diseases/pathology , Prion Diseases/physiopathology , Prions/genetics , Prions/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Animals , Cations, Divalent/metabolism , Evolution, Molecular , Humans , Phylogeny , Prions/chemistry , Prions/classification , Protein Processing, Post-Translational , Repressor Proteins/chemistry , Repressor Proteins/classification
19.
Arch Neurol ; 68(3): 320-8, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21059989

ABSTRACT

OBJECTIVES: To identify novel loci for late-onset Alzheimer disease (LOAD) in Caribbean Hispanic individuals and to replicate the findings in a publicly available data set from the National Institute on Aging Late-Onset Alzheimer's Disease Family Study. DESIGN: Nested case-control genome-wide association study. SETTING: The Washington Heights-Inwood Columbia Aging Project and the Estudio Familiar de Influencia Genetica de Alzheimer study. PARTICIPANTS: Five hundred forty-nine affected and 544 unaffected individuals of Caribbean Hispanic ancestry. INTERVENTION: The Illumina HumanHap 650Y chip for genotyping. MAIN OUTCOME MEASURE: Clinical diagnosis or pathologically confirmed diagnosis of LOAD. RESULTS: The strongest support for allelic association was for rs9945493 on 18q23 (P=1.7×10(-7)), but 22 additional single-nucleotide polymorphisms (SNPs) had a P value less than 9×10(-6) under 3 different analyses: unadjusted and stratified by the presence or absence of the APOE ε4 allele. Of these SNPs, 5 SNPs (rs4669573 and rs10197851 on 2p25.1; rs11711889 on 3q25.2; rs1117750 on 7p21.1; and rs7908652 on 10q23.1) were associated with LOAD in an independent cohort from the National Institute on Aging Late-Onset Alzheimer's Disease Family Study. We also replicated genetic associations for CLU, PICALM, and BIN1. CONCLUSIONS: Our genome-wide search of Caribbean Hispanic individuals identified several novel genetic variants associated with LOAD and replicated these associations in a white cohort. We also replicated associations in CLU, PICALM, and BIN1 in the Caribbean Hispanic cohort.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Alzheimer Disease/epidemiology , Alzheimer Disease/genetics , Clusterin/genetics , Monomeric Clathrin Assembly Proteins/genetics , Nuclear Proteins/genetics , Tumor Suppressor Proteins/genetics , Age of Onset , Aged , Aged, 80 and over , Alleles , Alzheimer Disease/psychology , Apolipoproteins E/genetics , Black People , Caribbean Region , Cohort Studies , Data Interpretation, Statistical , Female , Gene Frequency , Genome-Wide Association Study , Genotype , Hispanic or Latino , Humans , Male , Polymorphism, Single Nucleotide , White People
20.
J Neurosci ; 30(11): 3870-5, 2010 Mar 17.
Article in English | MEDLINE | ID: mdl-20237257

ABSTRACT

Aberrant accumulation of amyloid beta (Abeta) oligomers may underlie the cognitive failure of Alzheimer's disease (AD). All species of Abeta peptides are produced physiologically during normal brain activity. Therefore, elucidation of mechanisms that interconnect excitatory glutamatergic neurotransmission, synaptic amyloid precursor protein (APP) processing and production of its metabolite, Abeta, may reveal synapse-specific strategies for suppressing the pathological accumulation of Abeta oligomers and fibrils that characterize AD. To study synaptic APP processing, we used isolated intact nerve terminals (cortical synaptoneurosomes) from TgCRND8 mice, which express a human APP with familial AD mutations. Potassium chloride depolarization caused sustained release from synaptoneurosomes of Abeta(42) as well as Abeta(40), and appeared to coactivate alpha-, beta- and gamma-secretases, which are known to generate a family of released peptides, including Abeta(40) and Abeta(42). Stimulation of postsynaptic group I metabotropic glutamate receptor (mGluRs) with DHPG (3,5-dihydroxyphenylglycine) induced a rapid accumulation of APP C-terminal fragments (CTFs) in the synaptoneurosomes, a family of membrane-bound intermediates generated from APP metabolized by alpha- and beta-secretases. Following stimulation with the group II mGluR agonist DCG-IV, levels of APP CTFs in the synaptoneurosomes initially increased but then returned to baseline by 10 min after stimulation. This APP CTF degradation phase was accompanied by sustained accumulation of Abeta(42) in the releasate, which was blocked by the group II mGluR antagonist LY341495. These data suggest that group II mGluR may trigger synaptic activation of all three secretases and that suppression of group II mGluR signaling may be a therapeutic strategy for selectively reducing synaptic generation of Abeta(42).


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/biosynthesis , Amyloid beta-Peptides/metabolism , Nerve Endings/metabolism , Peptide Fragments/biosynthesis , Peptide Fragments/metabolism , Receptors, Metabotropic Glutamate/physiology , Alzheimer Disease/enzymology , Amino Acids/pharmacology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/biosynthesis , Amyloid beta-Protein Precursor/genetics , Animals , Humans , Methoxyhydroxyphenylglycol/analogs & derivatives , Methoxyhydroxyphenylglycol/pharmacology , Mice , Mice, Transgenic , Nerve Endings/drug effects , Nerve Endings/enzymology , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/physiology , Xanthenes/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...