Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
Add more filters

Country/Region as subject
Publication year range
1.
PLoS Genet ; 19(5): e1010760, 2023 05.
Article in English | MEDLINE | ID: mdl-37200393

ABSTRACT

Heterozygous variants in the glucocerebrosidase (GBA) gene are common and potent risk factors for Parkinson's disease (PD). GBA also causes the autosomal recessive lysosomal storage disorder (LSD), Gaucher disease, and emerging evidence from human genetics implicates many other LSD genes in PD susceptibility. We have systemically tested 86 conserved fly homologs of 37 human LSD genes for requirements in the aging adult Drosophila brain and for potential genetic interactions with neurodegeneration caused by α-synuclein (αSyn), which forms Lewy body pathology in PD. Our screen identifies 15 genetic enhancers of αSyn-induced progressive locomotor dysfunction, including knockdown of fly homologs of GBA and other LSD genes with independent support as PD susceptibility factors from human genetics (SCARB2, SMPD1, CTSD, GNPTAB, SLC17A5). For several genes, results from multiple alleles suggest dose-sensitivity and context-dependent pleiotropy in the presence or absence of αSyn. Homologs of two genes causing cholesterol storage disorders, Npc1a / NPC1 and Lip4 / LIPA, were independently confirmed as loss-of-function enhancers of αSyn-induced retinal degeneration. The enzymes encoded by several modifier genes are upregulated in αSyn transgenic flies, based on unbiased proteomics, revealing a possible, albeit ineffective, compensatory response. Overall, our results reinforce the important role of lysosomal genes in brain health and PD pathogenesis, and implicate several metabolic pathways, including cholesterol homeostasis, in αSyn-mediated neurotoxicity.


Subject(s)
Parkinson Disease , alpha-Synuclein , Animals , Humans , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , Animals, Genetically Modified , Drosophila/genetics , Drosophila/metabolism , Glucosylceramidase/genetics , Glucosylceramidase/metabolism , Lysosomes/metabolism , Parkinson Disease/pathology , Transferases (Other Substituted Phosphate Groups)/metabolism , Aging/metabolism
2.
Hum Mol Genet ; 32(4): 685-695, 2023 01 27.
Article in English | MEDLINE | ID: mdl-36173927

ABSTRACT

Genome-wide association studies (GWAS) have markedly advanced our understanding of the genetics of Parkinson's disease (PD), but they currently do not account for the full heritability of PD. In many cases it is difficult to unambiguously identify a specific gene within each locus because GWAS does not provide functional information on the identified candidate loci. Here we present an integrative approach that combines transcriptome-wide association study (TWAS) with high-throughput neuronal dysfunction analyses in Drosophila to discover and validate candidate PD genes. We identified 160 candidate genes whose misexpression is associated with PD risk via TWAS. Candidates were validated using orthogonal in silico methods and found to be functionally related to PD-associated pathways (i.e. endolysosome). We then mimicked these TWAS-predicted transcriptomic alterations in a Drosophila PD model and discovered that 50 candidates can modulate α-Synuclein(α-Syn)-induced neurodegeneration, allowing us to nominate new genes in previously known PD loci. We also uncovered additional novel PD candidate genes within GWAS suggestive loci (e.g. TTC19, ADORA2B, LZTS3, NRBP1, HN1L), which are also supported by clinical and functional evidence. These findings deepen our understanding of PD, and support applying our integrative approach to other complex trait disorders.


Subject(s)
Parkinson Disease , Animals , Parkinson Disease/genetics , Transcriptome/genetics , Genome-Wide Association Study/methods , Genetic Predisposition to Disease , Genomics , Drosophila/genetics , Polymorphism, Single Nucleotide
3.
J Biol Chem ; 296: 100760, 2021.
Article in English | MEDLINE | ID: mdl-33965374

ABSTRACT

One of the defining pathological features of Alzheimer's disease (AD) is the deposition of neurofibrillary tangles (NFTs) composed of hyperphosphorylated tau in the brain. Aberrant activation of kinases in AD has been suggested to enhance phosphorylation and toxicity of tau, making the responsible tau kinases attractive therapeutic targets. The full complement of tau-interacting kinases in AD brain and their activity in disease remains incompletely defined. Here, immunoaffinity enrichment coupled with mass spectrometry (MS) identified TANK-binding kinase 1 (TBK1) as a tau-interacting partner in human AD cortical brain tissues. We validated this interaction in human AD, familial frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17) caused by mutations in MAPT (R406W & P301L) and corticobasal degeneration (CBD) postmortem brain tissues as well as human cell lines. Further, we document increased TBK1 activation in both AD and FTDP-17 and map TBK1 phosphorylation sites on tau based on in vitro kinase assays coupled to MS. Lastly, in a Drosophila tauopathy model, activating expression of a conserved TBK1 ortholog triggers tau hyperphosphorylation and enhanced neurodegeneration, whereas knockdown had the reciprocal effect, suppressing tau toxicity. Collectively, our findings suggest that increased TBK1 activation may promote tau hyperphosphorylation and neuronal loss in AD and related tauopathies.


Subject(s)
Alzheimer Disease/metabolism , Protein Interaction Maps , Protein Serine-Threonine Kinases/metabolism , Tauopathies/metabolism , tau Proteins/metabolism , Alzheimer Disease/pathology , Animals , Drosophila , Female , HEK293 Cells , Humans , Male , Tauopathies/pathology
4.
Annu Rev Neurosci ; 37: 137-59, 2014.
Article in English | MEDLINE | ID: mdl-24821430

ABSTRACT

Parkinson's disease (PD) is a common neurodegenerative disease, yet the underlying causative molecular mechanisms are ill defined. Numerous observations based on drug studies and mutations in genes that cause PD point to a complex set of rather subtle mitochondrial defects that may be causative. Indeed, intensive investigation of these genes in model organisms has revealed roles in the electron transport chain, mitochondrial protein homeostasis, mitophagy, and the fusion and fission of mitochondria. Here, we attempt to synthesize results from experimental studies in diverse systems to define the precise function of these PD genes, as well as their interplay with other genes that affect mitochondrial function. We propose that subtle mitochondrial defects in combination with other insults trigger the onset and progression of disease, in both familial and idiopathic PD.


Subject(s)
Mitochondria/physiology , Nerve Tissue Proteins/physiology , Parkinson Disease/physiopathology , Animals , Humans , Mitochondria/genetics , Models, Biological , Nerve Tissue Proteins/genetics , Neurons/physiology , Parkinson Disease/genetics
5.
J Proteome Res ; 20(5): 2266-2282, 2021 05 07.
Article in English | MEDLINE | ID: mdl-33900085

ABSTRACT

Proteinaceous aggregates containing α-synuclein protein called Lewy bodies in the substantia nigra is a hallmark of Parkinson's disease. The molecular mechanisms of Lewy body formation and associated neuronal loss remain largely unknown. To gain insights into proteins and pathways associated with Lewy body pathology, we performed quantitative profiling of the proteome. We analyzed substantia nigra tissue from 51 subjects arranged into three groups: cases with Lewy body pathology, Lewy body-negative controls with matching neuronal loss, and controls with no neuronal loss. Using a label-free liquid chromatography-tandem mass spectrometry (LC-MS/MS) approach, we characterized the proteome both in terms of protein abundances and peptide modifications. Statistical testing for differential abundance of the most abundant 2963 proteins, followed by pathway enrichment and Bayesian learning of the causal network structure, was performed to identify likely drivers of Lewy body formation and dopaminergic neuronal loss. The identified pathways include (1) Arp2/3 complex-mediated actin nucleation; (2) synaptic function; (3) poly(A) RNA binding; (4) basement membrane and endothelium; and (5) hydrogen peroxide metabolic process. According to the data, the endothelial/basement membrane pathway is tightly connected with both pathologies and likely to be one of the drivers of neuronal loss. The poly(A) RNA-binding proteins, including the ones relevant to other neurodegenerative disorders (e.g., TDP-43 and FUS), have a strong inverse correlation with Lewy bodies and may reflect an alternative mechanism of nigral neurodegeneration.


Subject(s)
Lewy Bodies , Proteomics , Bayes Theorem , Chromatography, Liquid , Humans , Neurons/metabolism , Substantia Nigra/metabolism , Tandem Mass Spectrometry , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
6.
J Neurosci ; 38(43): 9286-9301, 2018 10 24.
Article in English | MEDLINE | ID: mdl-30249792

ABSTRACT

Accumulation of α-Synuclein (α-Syn) causes Parkinson's disease (PD) as well as other synucleopathies. α-Syn is the major component of Lewy bodies and Lewy neurites, the proteinaceous aggregates that are a hallmark of sporadic PD. In familial forms of PD, mutations or copy number variations in SNCA (the α-Syn gene) result in a net increase of its protein levels. Furthermore, common risk variants tied to PD are associated with small increases of wild-type α-Syn levels. These findings are further bolstered by animal studies which show that overexpression of α-Syn is sufficient to cause PD-like features. Thus, increased α-Syn levels are intrinsically tied to PD pathogenesis and underscore the importance of identifying the factors that regulate its levels. In this study, we establish a pooled RNAi screening approach and validation pipeline to probe the druggable genome for modifiers of α-Syn levels and identify 60 promising targets. Using a cross-species, tiered validation approach, we validate six strong candidates that modulate α-Syn levels and toxicity in cell lines, Drosophila, human neurons, and mouse brain of both sexes. More broadly, this genetic strategy and validation pipeline can be applied for the identification of therapeutic targets for disorders driven by dosage-sensitive proteins.SIGNIFICANCE STATEMENT We present a research strategy for the systematic identification and validation of genes modulating the levels of α-Synuclein, a protein involved in Parkinson's disease. A cell-based screen of the druggable genome (>7,500 genes that are potential therapeutic targets) yielded many modulators of α-Synuclein that were subsequently confirmed and validated in Drosophila, human neurons, and mouse brain. This approach has broad applicability to the multitude of neurological diseases that are caused by mutations in genes whose dosage is critical for brain function.


Subject(s)
Genome/genetics , Neurons/physiology , RNA Interference/physiology , Sequence Analysis, RNA/methods , alpha-Synuclein/genetics , Animals , Animals, Newborn , Drosophila , Female , HEK293 Cells , Humans , Male , Mice , Reproducibility of Results , Species Specificity
7.
Mov Disord ; 34(5): 614-624, 2019 05.
Article in English | MEDLINE | ID: mdl-30726573

ABSTRACT

Lysosomal storage disorders comprise a clinically heterogeneous group of autosomal-recessive or X-linked genetic syndromes caused by disruption of lysosomal biogenesis or function resulting in accumulation of nondegraded substrates. Although lysosomal storage disorders are diagnosed predominantly in children, many show variable expressivity with clinical presentations possible later in life. Given the important role of lysosomes in neuronal homeostasis, neurological manifestations, including movement disorders, can accompany many lysosomal storage disorders. Over the last decade, evidence from genetics, clinical epidemiology, cell biology, and biochemistry have converged to implicate links between lysosomal storage disorders and adult-onset movement disorders. The strongest evidence comes from mutations in Glucocerebrosidase, which cause Gaucher's disease and are among the most common and potent risk factors for PD. However, recently, many additional lysosomal storage disorder genes have been similarly implicated, including SMPD1, ATP13A2, GALC, and others. Examination of these links can offer insight into pathogenesis of PD and guide development of new therapeutic strategies. We systematically review the emerging genetic links between lysosomal storage disorders and PD. © 2019 International Parkinson and Movement Disorder Society.


Subject(s)
Lysosomal Storage Diseases/genetics , Parkinsonian Disorders/genetics , Adult , Child , Galactosylceramidase/genetics , Gaucher Disease/genetics , Glucosylceramidase/genetics , Humans , Leukodystrophy, Globoid Cell/genetics , Mucopolysaccharidosis III/genetics , Mutation , Neuronal Ceroid-Lipofuscinoses/genetics , Niemann-Pick Diseases/genetics , Parkinson Disease/genetics , Phenotype , Proton-Translocating ATPases/genetics , Sandhoff Disease/genetics , Sphingomyelin Phosphodiesterase/genetics
8.
Mov Disord ; 34(6): 866-875, 2019 06.
Article in English | MEDLINE | ID: mdl-30957308

ABSTRACT

BACKGROUND: Increasing evidence supports an extensive and complex genetic contribution to PD. Previous genome-wide association studies (GWAS) have shed light on the genetic basis of risk for this disease. However, the genetic determinants of PD age at onset are largely unknown. OBJECTIVES: To identify the genetic determinants of PD age at onset. METHODS: Using genetic data of 28,568 PD cases, we performed a genome-wide association study based on PD age at onset. RESULTS: We estimated that the heritability of PD age at onset attributed to common genetic variation was ∼0.11, lower than the overall heritability of risk for PD (∼0.27), likely, in part, because of the subjective nature of this measure. We found two genome-wide significant association signals, one at SNCA and the other a protein-coding variant in TMEM175, both of which are known PD risk loci and a Bonferroni-corrected significant effect at other known PD risk loci, GBA, INPP5F/BAG3, FAM47E/SCARB2, and MCCC1. Notably, SNCA, TMEM175, SCARB2, BAG3, and GBA have all been shown to be implicated in α-synuclein aggregation pathways. Remarkably, other well-established PD risk loci, such as GCH1 and MAPT, did not show a significant effect on age at onset of PD. CONCLUSIONS: Overall, we have performed the largest age at onset of PD genome-wide association studies to date, and our results show that not all PD risk loci influence age at onset with significant differences between risk alleles for age at onset. This provides a compelling picture, both within the context of functional characterization of disease-linked genetic variability and in defining differences between risk alleles for age at onset, or frank risk for disease. © 2019 International Parkinson and Movement Disorder Society.


Subject(s)
Age of Onset , Genetic Loci , Parkinson Disease/genetics , alpha-Synuclein/genetics , Adult , Aged , Aged, 80 and over , Alleles , Databases, Genetic , Female , Genetic Predisposition to Disease , Genome-Wide Association Study , Glucosylceramidase/genetics , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Male , Middle Aged , Polymorphism, Single Nucleotide , Young Adult
9.
PLoS Biol ; 14(6): e1002472, 2016 06.
Article in English | MEDLINE | ID: mdl-27254664

ABSTRACT

Nicotinamide mononucleotide adenylyl transferase 2 (NMNAT2) is neuroprotective in numerous preclinical models of neurodegeneration. Here, we show that brain nmnat2 mRNA levels correlate positively with global cognitive function and negatively with AD pathology. In AD brains, NMNAT2 mRNA and protein levels are reduced. NMNAT2 shifts its solubility and colocalizes with aggregated Tau in AD brains, similar to chaperones, which aid in the clearance or refolding of misfolded proteins. Investigating the mechanism of this observation, we discover a novel chaperone function of NMNAT2, independent from its enzymatic activity. NMNAT2 complexes with heat shock protein 90 (HSP90) to refold aggregated protein substrates. NMNAT2's refoldase activity requires a unique C-terminal ATP site, activated in the presence of HSP90. Furthermore, deleting NMNAT2 function increases the vulnerability of cortical neurons to proteotoxic stress and excitotoxicity. Interestingly, NMNAT2 acts as a chaperone to reduce proteotoxic stress, while its enzymatic activity protects neurons from excitotoxicity. Taken together, our data indicate that NMNAT2 exerts its chaperone or enzymatic function in a context-dependent manner to maintain neuronal health.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , HSP90 Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Animals , Blotting, Western , Brain/pathology , Brain/physiopathology , COS Cells , Cells, Cultured , Chlorocebus aethiops , Cognition/physiology , Female , HSP90 Heat-Shock Proteins/genetics , Humans , Male , Mice, Transgenic , Microscopy, Fluorescence , Middle Aged , Molecular Chaperones/genetics , Mutation , Neurons/cytology , Neurons/metabolism , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Protein Binding , Protein Folding , Protein Stability , Reverse Transcriptase Polymerase Chain Reaction
10.
PLoS Genet ; 12(10): e1006327, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27764101

ABSTRACT

We performed an exome-wide association analysis in 1393 late-onset Alzheimer's disease (LOAD) cases and 8141 controls from the CHARGE consortium. We found that a rare variant (P155L) in TM2D3 was enriched in Icelanders (~0.5% versus <0.05% in other European populations). In 433 LOAD cases and 3903 controls from the Icelandic AGES sub-study, P155L was associated with increased risk and earlier onset of LOAD [odds ratio (95% CI) = 7.5 (3.5-15.9), p = 6.6x10-9]. Mutation in the Drosophila TM2D3 homolog, almondex, causes a phenotype similar to loss of Notch/Presenilin signaling. Human TM2D3 is capable of rescuing these phenotypes, but this activity is abolished by P155L, establishing it as a functionally damaging allele. Our results establish a rare TM2D3 variant in association with LOAD susceptibility, and together with prior work suggests possible links to the ß-amyloid cascade.


Subject(s)
Alzheimer Disease/genetics , Drosophila Proteins/genetics , Membrane Proteins/genetics , Receptors, Notch/genetics , Tropomyosin/genetics , Age of Onset , Aged , Alleles , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Animals , Apolipoproteins E/genetics , Drosophila melanogaster/genetics , Exome/genetics , Female , Genome-Wide Association Study , Genomics , Humans , Iceland , Intracellular Signaling Peptides and Proteins/genetics , Male , Mutation , Phenotype , White People
11.
Brain ; 140(12): 3191-3203, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29140481

ABSTRACT

Mutations in the glucocerebrosidase gene (GBA), which cause Gaucher disease, are also potent risk factors for Parkinson's disease. We examined whether a genetic burden of variants in other lysosomal storage disorder genes is more broadly associated with Parkinson's disease susceptibility. The sequence kernel association test was used to interrogate variant burden among 54 lysosomal storage disorder genes, leveraging whole exome sequencing data from 1156 Parkinson's disease cases and 1679 control subjects. We discovered a significant burden of rare, likely damaging lysosomal storage disorder gene variants in association with Parkinson's disease risk. The association signal was robust to the exclusion of GBA, and consistent results were obtained in two independent replication cohorts, including 436 cases and 169 controls with whole exome sequencing and an additional 6713 cases and 5964 controls with exome-wide genotyping. In secondary analyses designed to highlight the specific genes driving the aggregate signal, we confirmed associations at the GBA and SMPD1 loci and newly implicate CTSD, SLC17A5, and ASAH1 as candidate Parkinson's disease susceptibility genes. In our discovery cohort, the majority of Parkinson's disease cases (56%) have at least one putative damaging variant in a lysosomal storage disorder gene, and 21% carry multiple alleles. Our results highlight several promising new susceptibility loci and reinforce the importance of lysosomal mechanisms in Parkinson's disease pathogenesis. We suggest that multiple genetic hits may act in combination to degrade lysosomal function, enhancing Parkinson's disease susceptibility.


Subject(s)
Acid Ceramidase/genetics , Cathepsin D/genetics , Glucosylceramidase/genetics , Organic Anion Transporters/genetics , Parkinson Disease/genetics , Sphingomyelin Phosphodiesterase/genetics , Symporters/genetics , Adult , Aged , Aged, 80 and over , Case-Control Studies , Cohort Studies , Exome , Female , Genetic Predisposition to Disease , Genotype , Humans , Lysosomal Storage Diseases/genetics , Male , Middle Aged , Mutation
12.
Brain ; 139(Pt 12): 3163-3169, 2016 12.
Article in English | MEDLINE | ID: mdl-27797806

ABSTRACT

We conducted a genome-wide association study of essential tremor, a common movement disorder characterized mainly by a postural and kinetic tremor of the upper extremities. Twin and family history studies show a high heritability for essential tremor. The molecular genetic determinants of essential tremor are unknown. We included 2807 patients and 6441 controls of European descent in our two-stage genome-wide association study. The 59 most significantly disease-associated markers of the discovery stage were genotyped in the replication stage. After Bonferroni correction two markers, one (rs10937625) located in the serine/threonine kinase STK32B and one (rs17590046) in the transcriptional coactivator PPARGC1A were associated with essential tremor. Three markers (rs12764057, rs10822974, rs7903491) in the cell-adhesion molecule CTNNA3 were significant in the combined analysis of both stages. The expression of STK32B was increased in the cerebellar cortex of patients and expression quantitative trait loci database mining showed association between the protective minor allele of rs10937625 and reduced expression in cerebellar cortex. We found no expression differences related to disease status or marker genotype for the other two genes. Replication of two lead single nucleotide polymorphisms of previous small genome-wide association studies (rs3794087 in SLC1A2, rs9652490 in LINGO1) did not confirm the association with essential tremor.


Subject(s)
Essential Tremor/genetics , Genome-Wide Association Study , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Protein Serine-Threonine Kinases/genetics , alpha Catenin/genetics , Humans , Polymorphism, Single Nucleotide
13.
Proteomics ; 16(23): 3042-3053, 2016 12.
Article in English | MEDLINE | ID: mdl-27718298

ABSTRACT

Despite a key role of amyloid-beta (Aß) in Alzheimer's disease (AD), mechanisms that link Aß plaques to tau neurofibrillary tangles and cognitive decline still remain poorly understood. The purpose of this study was to quantify proteins in the sarkosyl-insoluble brain proteome correlated with Aß and tau insolubility in the asymptomatic phase of AD (AsymAD) and through mild cognitive impairment (MCI) and symptomatic AD. Employing label-free mass spectrometry-based proteomics, we quantified 2711 sarkosyl-insoluble proteins across the prefrontal cortex from 35 individual cases representing control, AsymAD, MCI and AD. Significant enrichment of Aß and tau in AD was observed, which correlated with neuropathological measurements of plaque and tau tangle density, respectively. Pairwise correlation coefficients were also determined for all quantified proteins to Aß and tau, across the 35 cases. Notably, six of the ten most correlated proteins to Aß were U1 small nuclear ribonucleoproteins (U1 snRNPs). Three of these U1 snRNPs (U1A, SmD and U1-70K) also correlated with tau consistent with their association with tangle pathology in AD. Thus, proteins that cross-correlate with both Aß and tau, including specific U1 snRNPs, may have potential mechanistic roles in linking Aß plaques to tau tangle pathology during AD progression.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Proteins/metabolism , tau Proteins/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Case-Control Studies , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Detergents/chemistry , Female , Humans , Male , Proteins/analysis , Proteins/chemistry , Proteome/analysis , Proteome/chemistry , Proteomics/methods , Ribonucleoprotein, U1 Small Nuclear/chemistry , Ribonucleoprotein, U1 Small Nuclear/metabolism , Tandem Mass Spectrometry/methods
14.
Hum Mol Genet ; 23(4): 870-7, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24067533

ABSTRACT

Using a Drosophila model of Alzheimer's disease (AD), we systematically evaluated 67 candidate genes based on AD-associated genomic loci (P < 10(-4)) from published human genome-wide association studies (GWAS). Genetic manipulation of 87 homologous fly genes was tested for modulation of neurotoxicity caused by human Tau, which forms neurofibrillary tangle pathology in AD. RNA interference (RNAi) targeting 9 genes enhanced Tau neurotoxicity, and in most cases reciprocal activation of gene expression suppressed Tau toxicity. Our screen implicates cindr, the fly ortholog of the human CD2AP AD susceptibility gene, as a modulator of Tau-mediated disease mechanisms. Importantly, we also identify the fly orthologs of FERMT2 and CELF1 as Tau modifiers, and these loci have been independently validated as AD susceptibility loci in the latest GWAS meta-analysis. Both CD2AP and FERMT2 have been previously implicated with roles in cell adhesion, and our screen additionally identifies a fly homolog of the human integrin adhesion receptors, ITGAM and ITGA9, as a modifier of Tau neurotoxicity. Our results highlight cell adhesion pathways as important in Tau toxicity and AD susceptibility and demonstrate the power of model organism genetic screens for the functional follow-up of human GWAS.


Subject(s)
Alzheimer Disease/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , tau Proteins/genetics , Animals , Animals, Genetically Modified , CD11b Antigen/genetics , Disease Models, Animal , Drosophila Proteins/metabolism , Gene Knockdown Techniques , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Integrins/genetics , RNA Interference
15.
Am J Hum Genet ; 90(4): 720-6, 2012 Apr 06.
Article in English | MEDLINE | ID: mdl-22482808

ABSTRACT

Recent genome-wide association studies have identified a number of susceptibility loci for Alzheimer disease (AD). To understand the functional consequences and potential interactions of the associated loci, we explored large-scale data sets interrogating the human genome for evidence of positive natural selection. Our findings provide significant evidence for signatures of recent positive selection acting on several haplotypes carrying AD susceptibility alleles; interestingly, the genes found in these selected haplotypes can be assembled, independently, into a molecular complex via a protein-protein interaction (PPI) network approach. These results suggest a possible coevolution of genes encoding physically-interacting proteins that underlie AD susceptibility and are coexpressed in different tissues. In particular, PICALM, BIN1, CD2AP, and EPHA1 are interconnected through multiple interacting proteins and appear to have coordinated evidence of selection in the same human population, suggesting that they may be involved in the execution of a shared molecular function. This observation may be AD-specific, as the 12 loci associated with Parkinson disease do not demonstrate excess evidence of natural selection. The context for selection is probably unrelated to AD itself; it is likely that these genes interact in another context, such as in immune cells, where we observe cis-regulatory effects at several of the selected AD loci.


Subject(s)
Alzheimer Disease/genetics , Genetic Loci , Selection, Genetic , Adaptor Proteins, Signal Transducing/genetics , Age of Onset , Cytoskeletal Proteins/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study , Haplotypes , Humans , Monomeric Clathrin Assembly Proteins/genetics , Nuclear Proteins/genetics , Polymorphism, Single Nucleotide , Protein Interaction Maps/genetics , Receptor, EphA1/genetics , Tumor Suppressor Proteins/genetics
16.
Hum Mol Genet ; 21(10): 2377-88, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22343410

ABSTRACT

Complement receptor 1 (CR1) is an Alzheimer's disease (AD) susceptibility locus that also influences AD-related traits such as episodic memory decline and neuritic amyloid plaque deposition. We implemented a functional fine-mapping approach, leveraging intermediate phenotypes to identify functional variant(s) within the CR1 locus. Using 1709 subjects (697 deceased) from the Religious Orders Study and the Rush Memory and Aging Project, we tested 41 single-nucleotide polymorphisms (SNPs) within the linkage disequilibrium block containing the published CR1 AD SNP (rs6656401) for associations with episodic memory decline, and then examined the functional consequences of the top result. We report that a coding variant in the LHR-D (long homologous repeat D) region of the CR1 gene, rs4844609 (Ser1610Thr, minor allele frequency = 0.02), is associated with episodic memory decline and accounts for the known effect of the index SNP rs6656401 (D' = 1, r(2)= 0.084) on this trait. Further, we demonstrate that the coding variant's effect is largely dependent on an interaction with APOE-ε4 and mediated by an increased burden of AD-related neuropathology. Finally, in our data, this coding variant is also associated with AD susceptibility (joint odds ratio = 1.4). Taken together, our analyses identify a CR1 coding variant that influences episodic memory decline; it is a variant known to alter the conformation of CR1 and points to LHR-D as the functional domain within the CR1 protein that mediates the effect on memory decline. We thus implicate C1q and MBL, which bind to LHR-D, as likely targets of the variant's effect and suggest that CR1 may be an important intermediate in the clearance of Aß42 particles by C1q.


Subject(s)
Apolipoprotein E4/metabolism , Cognition Disorders/genetics , Receptors, Complement/genetics , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Apolipoprotein E4/genetics , Cognition Disorders/metabolism , Female , Gene Frequency , Genome-Wide Association Study , Genotype , Haplotypes , Humans , Memory, Episodic , Middle Aged , Odds Ratio , Phenotype , Plaque, Amyloid/metabolism , Polymorphism, Single Nucleotide , Receptors, Complement/metabolism
17.
Am J Hum Genet ; 88(2): 232-8, 2011 Feb 11.
Article in English | MEDLINE | ID: mdl-21295279

ABSTRACT

We have leveraged a Drosophila model relevant to Alzheimer disease (AD) for functional screening of findings from a genome-wide scan for loci associated with a quantitative measure of AD pathology in humans. In six of the 15 genomic regions evaluated, we successfully identified a causal gene for the association, on the basis of in vivo interactions with the neurotoxicity of Tau, which forms neurofibrillary tangles in AD. Among the top results, rs10845990 within SLC2A14, encoding a glucose transporter, showed evidence of replication for association with AD pathology, and gain and loss of function in glut1, the Drosophila ortholog, was associated with suppression and enhancement of Tau toxicity, respectively. Our strategy of coupling genome-wide association in humans with functional screening in a model organism is likely to be a powerful approach for gene discovery in AD and other complex genetic disorders.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/pathology , Drosophila/genetics , Genome , Neurofibrillary Tangles/genetics , Neurofibrillary Tangles/pathology , Polymorphism, Single Nucleotide/genetics , Animals , Genetic Predisposition to Disease , Genome-Wide Association Study , Glucose Transporter Type 1/genetics , Humans , Phenotype , Quantitative Trait Loci , Signal Transduction , tau Proteins/genetics
19.
BMC Geriatr ; 14: 54, 2014 Apr 23.
Article in English | MEDLINE | ID: mdl-24754876

ABSTRACT

BACKGROUND: We tested the hypothesis that harm avoidance, a trait associated with behavioral inhibition, is associated with the rate of change in parkinsonism in older adults. METHODS: At baseline harm avoidance was assessed with a standard self-report instrument in 969 older people without dementia participating in the Rush Memory and Aging Project, a longitudinal community-based cohort study. Parkinsonism was assessed annually with a modified version of the motor section of the Unified Parkinson's Disease Rating Scale (mUPDRS). RESULTS: Average follow-up was 5 years. A linear mixed-effects model controlling for age, sex and education showed that for an average participant (female, 80 years old at baseline, with 14 years of education and a harm avoidance score of 10), the overall severity of parkinsonism increased by about 0.05 unit/ year (Estimate, 0.054, S.E., 0.007, p <0.001) and that the level of harm avoidance was associated with the progression of parkinsonism (Estimate, 0.004, S.E., 0.001, p <0.001). Thus, for an average participant, every 6 point (~1 SD) increase in harm avoidance score at baseline, the rate of progression of parkinsonism increased about 50% compared to an individual with an average harm avoidance score. This amount of change in parkinsonism over the course of the study was associated with about a 5% increased risk of death. The association between harm avoidance and progression of parkinsonism persisted when controlling for cognitive function, depressive symptoms, loneliness, neuroticism, late-life cognitive, social and physical activities and chronic health conditions. CONCLUSION: A higher level of the harm avoidance trait is associated with a more rapid progression of parkinsonism in older adults.


Subject(s)
Disease Progression , Harm Reduction/physiology , Parkinsonian Disorders/diagnosis , Parkinsonian Disorders/psychology , Residence Characteristics , Aged , Aged, 80 and over , Cohort Studies , Female , Follow-Up Studies , Humans , Longitudinal Studies , Male , Parkinsonian Disorders/physiopathology , Prospective Studies
20.
Alzheimers Dement ; 10(1): 45-52, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23535033

ABSTRACT

BACKGROUND: Substantial interindividual variability exists in the disease trajectories of Alzheimer's disease (AD) patients. Some decline rapidly whereas others decline slowly, and there are no known explanations for this variability. We describe the first genome-wide association study to examine rate of cognitive decline in a sample of AD patients with longitudinal measures of cognition. METHODS: The discovery sample was 303 AD cases recruited in the Alzheimer's Disease Neuroimaging Initiative and the replication sample was 323 AD cases from the Religious Orders Study and Rush Memory and Aging Project. In the discovery sample, Alzheimer's Disease Assessment Scale-cognitive subscale responses were tested for association with genome-wide single-nucleotide polymorphism (SNP) data using linear regression. We tested the 65 most significant SNPs from the discovery sample for association in the replication sample. RESULTS: We identified SNPs in the spondin 1 gene (SPON1), the minor alleles of which were significantly associated with a slower rate of decline (rs11023139, P = 7.0 × 10(-11)) in the discovery sample. A SPON1 SNP 5.5 kb upstream was associated with decline in the replication sample (rs11606345, P = .002). CONCLUSION: SPON1 has not been previously associated with AD risk, but is plausibly related because the gene product binds to the amyloid precursor protein and inhibits its cleavage by ß-secretase. These data suggest that SPON1 may be associated with the differential rate of cognitive decline in AD.


Subject(s)
Alzheimer Disease/complications , Cognition Disorders/etiology , Cognition Disorders/genetics , Extracellular Matrix Proteins/genetics , Polymorphism, Single Nucleotide/genetics , Aged , Aged, 80 and over , Apolipoprotein E4/genetics , Female , Follow-Up Studies , Genome-Wide Association Study , Genotype , Humans , Male , Neuropsychological Tests , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL