Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Nat Neurosci ; 25(12): 1714-1723, 2022 12.
Article in English | MEDLINE | ID: mdl-36424430

ABSTRACT

Aging is accompanied by a host of social and biological changes that correlate with behavior, cognitive health and susceptibility to neurodegenerative disease. To understand trajectories of brain aging in a primate, we generated a multiregion bulk (N = 527 samples) and single-nucleus (N = 24 samples) brain transcriptional dataset encompassing 15 brain regions and both sexes in a unique population of free-ranging, behaviorally phenotyped rhesus macaques. We demonstrate that age-related changes in the level and variance of gene expression occur in genes associated with neural functions and neurological diseases, including Alzheimer's disease. Further, we show that higher social status in females is associated with younger relative transcriptional ages, providing a link between the social environment and aging in the brain. Our findings lend insight into biological mechanisms underlying brain aging in a nonhuman primate model of human behavior, cognition and health.


Subject(s)
Neurodegenerative Diseases , Female , Male , Humans , Animals , Macaca mulatta , Transcriptome , Aging/genetics , Social Environment , Solitary Nucleus
2.
Science ; 370(6518)2020 11 13.
Article in English | MEDLINE | ID: mdl-33184180

ABSTRACT

The chromatin landscape underlying the specification of human cell types is of fundamental interest. We generated human cell atlases of chromatin accessibility and gene expression in fetal tissues. For chromatin accessibility, we devised a three-level combinatorial indexing assay and applied it to 53 samples representing 15 organs, profiling ~800,000 single cells. We leveraged cell types defined by gene expression to annotate these data and cataloged hundreds of thousands of candidate regulatory elements that exhibit cell type-specific chromatin accessibility. We investigated the properties of lineage-specific transcription factors (such as POU2F1 in neurons), organ-specific specializations of broadly distributed cell types (such as blood and endothelial), and cell type-specific enrichments of complex trait heritability. These data represent a rich resource for the exploration of in vivo human gene regulation in diverse tissues and cell types.


Subject(s)
Chromatin/metabolism , Fetus/cytology , Fetus/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Single-Cell Analysis , Atlases as Topic , Humans , Neurons/metabolism , Transcription Factors/metabolism
3.
Science ; 370(6518)2020 11 13.
Article in English | MEDLINE | ID: mdl-33184181

ABSTRACT

The gene expression program underlying the specification of human cell types is of fundamental interest. We generated human cell atlases of gene expression and chromatin accessibility in fetal tissues. For gene expression, we applied three-level combinatorial indexing to >110 samples representing 15 organs, ultimately profiling ~4 million single cells. We leveraged the literature and other atlases to identify and annotate hundreds of cell types and subtypes, both within and across tissues. Our analyses focused on organ-specific specializations of broadly distributed cell types (such as blood, endothelial, and epithelial), sites of fetal erythropoiesis (which notably included the adrenal gland), and integration with mouse developmental atlases (such as conserved specification of blood cells). These data represent a rich resource for the exploration of in vivo human gene expression in diverse tissues and cell types.


Subject(s)
Chromatin/metabolism , Fetus/cytology , Fetus/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Single-Cell Analysis , Atlases as Topic , Humans , Neurons/metabolism , Transcription Factors/metabolism
4.
Science ; 367(6473): 45-51, 2020 01 03.
Article in English | MEDLINE | ID: mdl-31806696

ABSTRACT

High-throughput chemical screens typically use coarse assays such as cell survival, limiting what can be learned about mechanisms of action, off-target effects, and heterogeneous responses. Here, we introduce "sci-Plex," which uses "nuclear hashing" to quantify global transcriptional responses to thousands of independent perturbations at single-cell resolution. As a proof of concept, we applied sci-Plex to screen three cancer cell lines exposed to 188 compounds. In total, we profiled ~650,000 single-cell transcriptomes across ~5000 independent samples in one experiment. Our results reveal substantial intercellular heterogeneity in response to specific compounds, commonalities in response to families of compounds, and insight into differential properties within families. In particular, our results with histone deacetylase inhibitors support the view that chromatin acts as an important reservoir of acetate in cancer cells.


Subject(s)
High-Throughput Screening Assays , Neoplasms/metabolism , RNA-Seq/methods , Single-Cell Analysis/methods , Transcriptome/drug effects , A549 Cells , Acetates/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Chromatin/metabolism , Genomics , Histone Deacetylase Inhibitors/pharmacology , Humans , K562 Cells , MCF-7 Cells , Neoplasms/genetics
5.
Nat Methods ; 16(10): 983-986, 2019 10.
Article in English | MEDLINE | ID: mdl-31501545

ABSTRACT

Single-cell molecular profiling technologies are gaining rapid traction, but the manual process by which resulting cell types are typically annotated is labor intensive and rate-limiting. We describe Garnett, a tool for rapidly annotating cell types in single-cell transcriptional profiling and single-cell chromatin accessibility datasets, based on an interpretable, hierarchical markup language of cell type-specific genes. Garnett successfully classifies cell types in tissue and whole organism datasets, as well as across species.


Subject(s)
Atlases as Topic , Single-Cell Analysis/methods , Algorithms , Animals , Datasets as Topic , Gene Expression Profiling , Humans , Sequence Analysis, RNA/methods , Transcription, Genetic
6.
Genome Res ; 29(5): 857-869, 2019 05.
Article in English | MEDLINE | ID: mdl-30936163

ABSTRACT

Here we present a comprehensive map of the accessible chromatin landscape of the mouse hippocampus at single-cell resolution. Substantial advances of this work include the optimization of a single-cell combinatorial indexing assay for transposase accessible chromatin (sci-ATAC-seq); a software suite, scitools, for the rapid processing and visualization of single-cell combinatorial indexing data sets; and a valuable resource of hippocampal regulatory networks at single-cell resolution. We used sci-ATAC-seq to produce 2346 high-quality single-cell chromatin accessibility maps with a mean unique read count per cell of 29,201 from both fresh and frozen hippocampi, observing little difference in accessibility patterns between the preparations. By using this data set, we identified eight distinct major clusters of cells representing both neuronal and nonneuronal cell types and characterized the driving regulatory factors and differentially accessible loci that define each cluster. Within pyramidal neurons, we identified four major clusters, including CA1 and CA3 neurons, and three additional subclusters. We then applied a recently described coaccessibility framework, Cicero, which identified 146,818 links between promoters and putative distal regulatory DNA. Identified coaccessibility networks showed cell-type specificity, shedding light on key dynamic loci that reconfigure to specify hippocampal cell lineages. Lastly, we performed an additional sci-ATAC-seq preparation from cultured hippocampal neurons (899 high-quality cells, 43,532 mean unique reads) that revealed substantial alterations in their epigenetic landscape compared with nuclei from hippocampal tissue. This data set and accompanying analysis tools provide a new resource that can guide subsequent studies of the hippocampus.


Subject(s)
Chromatin/genetics , Hippocampus/metabolism , Pyramidal Cells/metabolism , Animals , Cell Lineage/genetics , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cells, Cultured , Chromatin/metabolism , Epigenomics/methods , Mice , Neuronal Plasticity/genetics , Pyramidal Cells/cytology , Sequence Analysis, DNA , Single-Cell Analysis/methods , Transposases/genetics , Transposases/metabolism
7.
Science ; 361(6409): 1380-1385, 2018 09 28.
Article in English | MEDLINE | ID: mdl-30166440

ABSTRACT

Although we can increasingly measure transcription, chromatin, methylation, and other aspects of molecular biology at single-cell resolution, most assays survey only one aspect of cellular biology. Here we describe sci-CAR, a combinatorial indexing-based coassay that jointly profiles chromatin accessibility and mRNA (CAR) in each of thousands of single cells. As a proof of concept, we apply sci-CAR to 4825 cells, including a time series of dexamethasone treatment, as well as to 11,296 cells from the adult mouse kidney. With the resulting data, we compare the pseudotemporal dynamics of chromatin accessibility and gene expression, reconstruct the chromatin accessibility profiles of cell types defined by RNA profiles, and link cis-regulatory sites to their target genes on the basis of the covariance of chromatin accessibility and transcription across large numbers of single cells.


Subject(s)
Chromatin/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation , Genomics/methods , Single-Cell Analysis/methods , A549 Cells , Animals , Dexamethasone/pharmacology , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Kidney/cytology , Kidney/drug effects , Mice , NIH 3T3 Cells , Regulatory Elements, Transcriptional/drug effects , Transcription, Genetic/drug effects
8.
Cell ; 174(5): 1309-1324.e18, 2018 08 23.
Article in English | MEDLINE | ID: mdl-30078704

ABSTRACT

We applied a combinatorial indexing assay, sci-ATAC-seq, to profile genome-wide chromatin accessibility in ∼100,000 single cells from 13 adult mouse tissues. We identify 85 distinct patterns of chromatin accessibility, most of which can be assigned to cell types, and ∼400,000 differentially accessible elements. We use these data to link regulatory elements to their target genes, to define the transcription factor grammar specifying each cell type, and to discover in vivo correlates of heterogeneity in accessibility within cell types. We develop a technique for mapping single cell gene expression data to single-cell chromatin accessibility data, facilitating the comparison of atlases. By intersecting mouse chromatin accessibility with human genome-wide association summary statistics, we identify cell-type-specific enrichments of the heritability signal for hundreds of complex traits. These data define the in vivo landscape of the regulatory genome for common mammalian cell types at single-cell resolution.


Subject(s)
Chromatin/chemistry , Single-Cell Analysis/methods , Animals , Cluster Analysis , Epigenesis, Genetic , Epigenomics , Gene Expression Regulation , Genome, Human , Genome-Wide Association Study , Humans , Male , Mammals , Mice , Mice, Inbred C57BL , Transcription Factors
9.
Mol Cell ; 71(5): 858-871.e8, 2018 09 06.
Article in English | MEDLINE | ID: mdl-30078726

ABSTRACT

Linking regulatory DNA elements to their target genes, which may be located hundreds of kilobases away, remains challenging. Here, we introduce Cicero, an algorithm that identifies co-accessible pairs of DNA elements using single-cell chromatin accessibility data and so connects regulatory elements to their putative target genes. We apply Cicero to investigate how dynamically accessible elements orchestrate gene regulation in differentiating myoblasts. Groups of Cicero-linked regulatory elements meet criteria of "chromatin hubs"-they are enriched for physical proximity, interact with a common set of transcription factors, and undergo coordinated changes in histone marks that are predictive of changes in gene expression. Pseudotemporal analysis revealed that most DNA elements remain in chromatin hubs throughout differentiation. A subset of elements bound by MYOD1 in myoblasts exhibit early opening in a PBX1- and MEIS1-dependent manner. Our strategy can be applied to dissect the architecture, sequence determinants, and mechanisms of cis-regulation on a genome-wide scale.


Subject(s)
Chromatin Assembly and Disassembly/genetics , Chromatin/genetics , DNA/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Regulation/genetics , Adolescent , Cell Differentiation/genetics , Female , Genes, Homeobox/genetics , Histones/genetics , Humans , Myoblasts/physiology , Transcription Factors/genetics
10.
Nature ; 555(7697): 538-542, 2018 03 22.
Article in English | MEDLINE | ID: mdl-29539636

ABSTRACT

Understanding how gene regulatory networks control the progressive restriction of cell fates is a long-standing challenge. Recent advances in measuring gene expression in single cells are providing new insights into lineage commitment. However, the regulatory events underlying these changes remain unclear. Here we investigate the dynamics of chromatin regulatory landscapes during embryogenesis at single-cell resolution. Using single-cell combinatorial indexing assay for transposase accessible chromatin with sequencing (sci-ATAC-seq), we profiled chromatin accessibility in over 20,000 single nuclei from fixed Drosophila melanogaster embryos spanning three landmark embryonic stages: 2-4 h after egg laying (predominantly stage 5 blastoderm nuclei), when each embryo comprises around 6,000 multipotent cells; 6-8 h after egg laying (predominantly stage 10-11), to capture a midpoint in embryonic development when major lineages in the mesoderm and ectoderm are specified; and 10-12 h after egg laying (predominantly stage 13), when each of the embryo's more than 20,000 cells are undergoing terminal differentiation. Our results show that there is spatial heterogeneity in the accessibility of the regulatory genome before gastrulation, a feature that aligns with future cell fate, and that nuclei can be temporally ordered along developmental trajectories. During mid-embryogenesis, tissue granularity emerges such that individual cell types can be inferred by their chromatin accessibility while maintaining a signature of their germ layer of origin. Analysis of the data reveals overlapping usage of regulatory elements between cells of the endoderm and non-myogenic mesoderm, suggesting a common developmental program that is reminiscent of the mesendoderm lineage in other species. We identify 30,075 distal regulatory elements that exhibit tissue-specific accessibility. We validated the germ-layer specificity of a subset of these predicted enhancers in transgenic embryos, achieving an accuracy of 90%. Overall, our results demonstrate the power of shotgun single-cell profiling of embryos to resolve dynamic changes in the chromatin landscape during development, and to uncover the cis-regulatory programs of metazoan germ layers and cell types.


Subject(s)
Drosophila melanogaster/cytology , Drosophila melanogaster/embryology , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Single-Cell Analysis , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Chromatin/genetics , Chromatin/metabolism , Drosophila melanogaster/genetics , Endoderm/cytology , Endoderm/metabolism , Enhancer Elements, Genetic/genetics , Female , Gastrulation/genetics , Genome, Insect/genetics , Male , Mesoderm/cytology , Mesoderm/metabolism , Organ Specificity/genetics , Organisms, Genetically Modified/cytology , Organisms, Genetically Modified/genetics , Reproducibility of Results
11.
Neuron ; 97(6): 1268-1283.e6, 2018 03 21.
Article in English | MEDLINE | ID: mdl-29566793

ABSTRACT

To identify novel genes associated with ALS, we undertook two lines of investigation. We carried out a genome-wide association study comparing 20,806 ALS cases and 59,804 controls. Independently, we performed a rare variant burden analysis comparing 1,138 index familial ALS cases and 19,494 controls. Through both approaches, we identified kinesin family member 5A (KIF5A) as a novel gene associated with ALS. Interestingly, mutations predominantly in the N-terminal motor domain of KIF5A are causative for two neurodegenerative diseases: hereditary spastic paraplegia (SPG10) and Charcot-Marie-Tooth type 2 (CMT2). In contrast, ALS-associated mutations are primarily located at the C-terminal cargo-binding tail domain and patients harboring loss-of-function mutations displayed an extended survival relative to typical ALS cases. Taken together, these results broaden the phenotype spectrum resulting from mutations in KIF5A and strengthen the role of cytoskeletal defects in the pathogenesis of ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/diagnosis , Amyotrophic Lateral Sclerosis/genetics , Genome-Wide Association Study/methods , Kinesins/genetics , Loss of Function Mutation/genetics , Adult , Aged , Aged, 80 and over , Amino Acid Sequence , Amyotrophic Lateral Sclerosis/epidemiology , Cohort Studies , Female , Humans , Male , Middle Aged , Young Adult
12.
Nat Methods ; 14(10): 979-982, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28825705

ABSTRACT

Single-cell trajectories can unveil how gene regulation governs cell fate decisions. However, learning the structure of complex trajectories with multiple branches remains a challenging computational problem. We present Monocle 2, an algorithm that uses reversed graph embedding to describe multiple fate decisions in a fully unsupervised manner. We applied Monocle 2 to two studies of blood development and found that mutations in the genes encoding key lineage transcription factors divert cells to alternative fates.


Subject(s)
Algorithms , Cell Differentiation/physiology , Computer Simulation , Gene Expression Regulation, Developmental/physiology , Models, Biological , Animals , Mutation , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome/physiology
13.
Neurobiol Aging ; 43: 180.e1-5, 2016 07.
Article in English | MEDLINE | ID: mdl-27156075

ABSTRACT

Recently, mutations in the TANK-binding kinase 1 (TBK1) gene were identified as a cause for amyotrophic lateral sclerosis (ALS) with or without comorbid frontotemporal dementia. We have assessed the frequency and clinical characteristics of TBK1 mutations in a cohort of ALS patients of Sardinian ancestry. Whole-exome sequencing was performed on Hiseq2000 platform (Illumina). Genome analysis Toolkit was used to align and to code variants according to Human Genome (UCSC hg19). Mutation was confirmed with Sanger sequence. In our screening of 186 Sardinian ALS cases, we found 3 (1.6%) patients carrying 3 distinct novel genetic variants: a nonsynonymous SNV c.1150C>T leading to a p.Arg384Thr change in exon 9; a nonsynonymous SNV c.1331G>A causes a p.Arg444Gln change in exon 11; and a frameshift deletion c.2070delG (p.Met690fs) at the exon 20 of the gene leading to a stop at 693 codon. The latter patients also carried missense mutation c.98C>T of the SQSTM1 gene causing a substitution of an arginine with a valine at the position 33 (p.Arg33Val). All variants were found to be deleterious according to in silico predictions. All cases were apparently sporadic and one of them showed frontotemporal dementia associated to ALS. These mutations were not found in 2 cohorts of 6780 ethnic-matched controls. We have found that TBK1 mutations account for 1.6% of Sardinian ALS cases. Our data support the notion that TBK1 is a novel ALS gene, providing important evidence complementary to the first descriptions.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Frontotemporal Dementia/genetics , Genetic Association Studies , Mutation , Protein Serine-Threonine Kinases , Aged , Cohort Studies , Female , Humans , Italy , Male
14.
PLoS One ; 10(9): e0136929, 2015.
Article in English | MEDLINE | ID: mdl-26327321

ABSTRACT

Protein degradation via the multistep ubiquitin/26S proteasome pathway is a rapid way to alter the protein profile and drive cell processes and developmental changes. Many key regulators of embryonic development are targeted for degradation by E3 ubiquitin ligases. The most studied family of E3 ubiquitin ligases is the SCF ubiquitin ligases, which use F-box adaptor proteins to recognize and recruit target proteins. Here, we used a bioinformatics screen and phylogenetic analysis to identify and annotate the family of F-box proteins in the Xenopus tropicalis genome. To shed light on the function of the F-box proteins, we analyzed expression of F-box genes during early stages of Xenopus development. Many F-box genes are broadly expressed with expression domains localized to diverse tissues including brain, spinal cord, eye, neural crest derivatives, somites, kidneys, and heart. All together, our genome-wide identification and expression profiling of the Xenopus F-box family of proteins provide a foundation for future research aimed to identify the precise role of F-box dependent E3 ubiquitin ligases and their targets in the regulatory circuits of development.


Subject(s)
F-Box Proteins/genetics , Xenopus/genetics , Animals , Gene Expression Regulation, Developmental/genetics , Genome-Wide Association Study/methods , Phylogeny , Proteolysis , SKP Cullin F-Box Protein Ligases/genetics , Ubiquitin-Protein Ligases/genetics
15.
Science ; 348(6237): 910-4, 2015 May 22.
Article in English | MEDLINE | ID: mdl-25953818

ABSTRACT

Technical advances have enabled the collection of genome and transcriptome data sets with single-cell resolution. However, single-cell characterization of the epigenome has remained challenging. Furthermore, because cells must be physically separated before biochemical processing, conventional single-cell preparatory methods scale linearly. We applied combinatorial cellular indexing to measure chromatin accessibility in thousands of single cells per assay, circumventing the need for compartmentalization of individual cells. We report chromatin accessibility profiles from more than 15,000 single cells and use these data to cluster cells on the basis of chromatin accessibility landscapes. We identify modules of coordinately regulated chromatin accessibility at the level of single cells both between and within cell types, with a scalable method that may accelerate progress toward a human cell atlas.


Subject(s)
Chromatin/metabolism , Epigenesis, Genetic , Single-Cell Analysis/methods , HEK293 Cells , HL-60 Cells , Humans
16.
JAMA Neurol ; 72(4): 396-404, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25643325

ABSTRACT

IMPORTANCE: Myasthenia gravis is a chronic, autoimmune, neuromuscular disease characterized by fluctuating weakness of voluntary muscle groups. Although genetic factors are known to play a role in this neuroimmunological condition, the genetic etiology underlying myasthenia gravis is not well understood. OBJECTIVE: To identify genetic variants that alter susceptibility to myasthenia gravis, we performed a genome-wide association study. DESIGN, SETTING, AND PARTICIPANTS: DNA was obtained from 1032 white individuals from North America diagnosed as having acetylcholine receptor antibody-positive myasthenia gravis and 1998 race/ethnicity-matched control individuals from January 2010 to January 2011. These samples were genotyped on Illumina OmniExpress single-nucleotide polymorphism arrays. An independent cohort of 423 Italian cases and 467 Italian control individuals were used for replication. MAIN OUTCOMES AND MEASURES: We calculated P values for association between 8,114,394 genotyped and imputed variants across the genome and risk for developing myasthenia gravis using logistic regression modeling. A threshold P value of 5.0×10(-8) was set for genome-wide significance after Bonferroni correction for multiple testing. RESULTS: In the overall case-control cohort, we identified association signals at CTLA4 (rs231770; P=3.98×10(-8); odds ratio, 1.37; 95% CI, 1.25-1.49), HLA-DQA1 (rs9271871; P=1.08×10(-8); odds ratio, 2.31; 95% CI, 2.02-2.60), and TNFRSF11A (rs4263037; P=1.60×10(-9); odds ratio, 1.41; 95% CI, 1.29-1.53). These findings replicated for CTLA4 and HLA-DQA1 in an independent cohort of Italian cases and control individuals. Further analysis revealed distinct, but overlapping, disease-associated loci for early- and late-onset forms of myasthenia gravis. In the late-onset cases, we identified 2 association peaks: one was located in TNFRSF11A (rs4263037; P=1.32×10(-12); odds ratio, 1.56; 95% CI, 1.44-1.68) and the other was detected in the major histocompatibility complex on chromosome 6p21 (HLA-DQA1; rs9271871; P=7.02×10(-18); odds ratio, 4.27; 95% CI, 3.92-4.62). Association within the major histocompatibility complex region was also observed in early-onset cases (HLA-DQA1; rs601006; P=2.52×10(-11); odds ratio, 4.0; 95% CI, 3.57-4.43), although the set of single-nucleotide polymorphisms was different from that implicated among late-onset cases. CONCLUSIONS AND RELEVANCE: Our genetic data provide insights into aberrant cellular mechanisms responsible for this prototypical autoimmune disorder. They also suggest that clinical trials of immunomodulatory drugs related to CTLA4 and that are already Food and Drug Administration approved as therapies for other autoimmune diseases could be considered for patients with refractory disease.


Subject(s)
Genetic Predisposition to Disease , Genome-Wide Association Study , HLA-DQ alpha-Chains/genetics , Myasthenia Gravis/genetics , Adult , Age of Onset , CTLA-4 Antigen/genetics , Case-Control Studies , Female , Gene Frequency , Genotype , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide/genetics , United States
17.
Neurobiol Aging ; 35(12): 2882.e7-2882.e12, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25123918

ABSTRACT

Conserved populations, such as Sardinians, displaying elevated rates of familial or sporadic amyotrophic lateral sclerosis (ALS) provide unique information on the genetics of the disease. Our aim was to describe the genetic profile of a consecutive series of ALS patients of Sardinian ancestry. All ALS patients of Sardinian ancestry, identified between 2008 and 2013 through the Italian ALS Genetic Consortium, were eligible to be included in the study. Patients and controls underwent the analysis of TARDBP, C9ORF72, SOD1, and FUS genes. Genetic mutations were identified in 155 out of 375 Sardinian ALS cases (41.3%), more commonly the p.A382T and p.G295S mutations of TARDBP and the GGGGCC hexanucleotide repeat expansion of C9ORF72. One patient had both p.G295S and p.A382T mutations of TARDBP and 8 carried both the heterozygous p.A382T mutation of TARDBP and a repeat expansion of C9ORF72. Patients carrying the p.A382T and the p.G295S mutations of TARDBP and the C9ORF72 repeat expansion shared distinct haplotypes across these loci. Patients with cooccurrence of C9ORF72 and TARDBP p.A382T missense mutation had a significantly lower age at onset and shorter survival. More than 40% of all cases on the island of Sardinia carry a mutation of an ALS-related gene, representing the highest percentage of ALS cases genetically explained outside of Scandinavia. Clinical phenotypes associated with different genetic mutations show some distinctive characteristics, but the heterogeneity between and among families carrying the same mutations implies that ALS manifestation is influenced by other genetic and nongenetic factors.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Genetic Association Studies , Genetic Predisposition to Disease/genetics , Mutation , Age of Onset , Amyotrophic Lateral Sclerosis/epidemiology , C9orf72 Protein , DNA Repeat Expansion , DNA-Binding Proteins/genetics , Genotype , Humans , Italy/epidemiology , Penetrance , Phenotype , Proteins/genetics , RNA-Binding Protein FUS/genetics , Superoxide Dismutase/genetics , Superoxide Dismutase-1
18.
Nat Neurosci ; 17(5): 664-666, 2014 May.
Article in English | MEDLINE | ID: mdl-24686783

ABSTRACT

MATR3 is an RNA- and DNA-binding protein that interacts with TDP-43, a disease protein linked to amyotrophic lateral sclerosis (ALS) and frontotemporal dementia. Using exome sequencing, we identified mutations in MATR3 in ALS kindreds. We also observed MATR3 pathology in ALS-affected spinal cords with and without MATR3 mutations. Our data provide more evidence supporting the role of aberrant RNA processing in motor neuron degeneration.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Family Health , Genetic Predisposition to Disease/genetics , Mutation/genetics , Nuclear Matrix-Associated Proteins/genetics , RNA-Binding Proteins/genetics , Aged , Aged, 80 and over , Amyotrophic Lateral Sclerosis/pathology , Computational Biology , DNA Mutational Analysis , DNA-Binding Proteins/metabolism , Female , Genotype , Humans , Male , Middle Aged , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Neurologic Examination , Nuclear Matrix-Associated Proteins/metabolism , RNA-Binding Proteins/metabolism , Spinal Cord/metabolism , Spinal Cord/pathology
19.
Acta Neuropathol ; 127(3): 391-6, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24496499

ABSTRACT

Recent advances are uncovering more and more of the genetic architecture underlying amyotrophic lateral sclerosis (ALS), a fatal neurodegenerative condition that affects ~6,000 Americans annually. Chief among these was the discovery that a large repeat expansion in the C9ORF72 gene is responsible for an unprecedented portion of familial and sporadic ALS cases. Much has been published on how this expansion disrupts neuronal homeostasis and how gene-based therapy might be an effective treatment in the future. Nevertheless, it is instructive to look back at the origins of this important mutation. In this opinion piece, we attempt to answer three key questions concerning C9ORF72. First, how many times did the expansion occur throughout human history? Second, how old is the expansion? And finally and perhaps most importantly, how did the expansion spread throughout Europe? We speculate that the expansion occurred only once in the past, that this event took place in the Finnish population and that the Vikings and their descendants were responsible for disseminating this mutation throughout the rest of the continent.


Subject(s)
DNA Repeat Expansion , Evolution, Molecular , Proteins/genetics , Amyotrophic Lateral Sclerosis/genetics , C9orf72 Protein , Human Migration , Humans , Internationality , Time Factors , White People/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...