Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 149
Filter
Add more filters

Publication year range
1.
Cell ; 186(17): 3659-3673.e23, 2023 08 17.
Article in English | MEDLINE | ID: mdl-37527660

ABSTRACT

Many regions in the human genome vary in length among individuals due to variable numbers of tandem repeats (VNTRs). To assess the phenotypic impact of VNTRs genome-wide, we applied a statistical imputation approach to estimate the lengths of 9,561 autosomal VNTR loci in 418,136 unrelated UK Biobank participants and 838 GTEx participants. Association and statistical fine-mapping analyses identified 58 VNTRs that appeared to influence a complex trait in UK Biobank, 18 of which also appeared to modulate expression or splicing of a nearby gene. Non-coding VNTRs at TMCO1 and EIF3H appeared to generate the largest known contributions of common human genetic variation to risk of glaucoma and colorectal cancer, respectively. Each of these two VNTRs associated with a >2-fold range of risk across individuals. These results reveal a substantial and previously unappreciated role of non-coding VNTRs in human health and gene regulation.


Subject(s)
Calcium Channels , Colorectal Neoplasms , Eukaryotic Initiation Factor-3 , Glaucoma , Minisatellite Repeats , Humans , Calcium Channels/genetics , Colorectal Neoplasms/genetics , Genome, Human , Glaucoma/genetics , Polymorphism, Genetic , Eukaryotic Initiation Factor-3/genetics
2.
Cell ; 177(2): 478-491.e20, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30929901

ABSTRACT

Genomic studies have identified hundreds of candidate genes near loci associated with risk for schizophrenia. To define candidates and their functions, we mutated zebrafish orthologs of 132 human schizophrenia-associated genes. We created a phenotype atlas consisting of whole-brain activity maps, brain structural differences, and profiles of behavioral abnormalities. Phenotypes were diverse but specific, including altered forebrain development and decreased prepulse inhibition. Exploration of these datasets identified promising candidates in more than 10 gene-rich regions, including the magnesium transporter cnnm2 and the translational repressor gigyf2, and revealed shared anatomical sites of activity differences, including the pallium, hypothalamus, and tectum. Single-cell RNA sequencing uncovered an essential role for the understudied transcription factor znf536 in the development of forebrain neurons implicated in social behavior and stress. This phenotypic landscape of schizophrenia-associated genes prioritizes more than 30 candidates for further study and provides hypotheses to bridge the divide between genetic association and biological mechanism.


Subject(s)
Schizophrenia/genetics , Schizophrenia/physiopathology , Animals , Brain , Cerebral Cortex , Disease Models, Animal , Gene Expression Regulation/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study , Phenotype , Polymorphism, Single Nucleotide/genetics , Zebrafish/genetics
3.
Cell ; 174(4): 1015-1030.e16, 2018 08 09.
Article in English | MEDLINE | ID: mdl-30096299

ABSTRACT

The mammalian brain is composed of diverse, specialized cell populations. To systematically ascertain and learn from these cellular specializations, we used Drop-seq to profile RNA expression in 690,000 individual cells sampled from 9 regions of the adult mouse brain. We identified 565 transcriptionally distinct groups of cells using computational approaches developed to distinguish biological from technical signals. Cross-region analysis of these 565 cell populations revealed features of brain organization, including a gene-expression module for synthesizing axonal and presynaptic components, patterns in the co-deployment of voltage-gated ion channels, functional distinctions among the cells of the vasculature and specialization of glutamatergic neurons across cortical regions. Systematic neuronal classifications for two complex basal ganglia nuclei and the striatum revealed a rare population of spiny projection neurons. This adult mouse brain cell atlas, accessible through interactive online software (DropViz), serves as a reference for development, disease, and evolution.


Subject(s)
Brain/metabolism , Cell Lineage , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Single-Cell Analysis/methods , Transcriptome , Animals , Brain/growth & development , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Male , Mice , Mice, Inbred C57BL
4.
Cell ; 166(5): 1308-1323.e30, 2016 Aug 25.
Article in English | MEDLINE | ID: mdl-27565351

ABSTRACT

Patterns of gene expression can be used to characterize and classify neuronal types. It is challenging, however, to generate taxonomies that fulfill the essential criteria of being comprehensive, harmonizing with conventional classification schemes, and lacking superfluous subdivisions of genuine types. To address these challenges, we used massively parallel single-cell RNA profiling and optimized computational methods on a heterogeneous class of neurons, mouse retinal bipolar cells (BCs). From a population of ∼25,000 BCs, we derived a molecular classification that identified 15 types, including all types observed previously and two novel types, one of which has a non-canonical morphology and position. We validated the classification scheme and identified dozens of novel markers using methods that match molecular expression to cell morphology. This work provides a systematic methodology for achieving comprehensive molecular classification of neurons, identifies novel neuronal types, and uncovers transcriptional differences that distinguish types within a class.


Subject(s)
Retinal Bipolar Cells/classification , Transcriptome , Amacrine Cells/cytology , Animals , Cluster Analysis , Female , Genetic Markers , Male , Mice , Mice, Inbred Strains , Mice, Transgenic , Retinal Bipolar Cells/cytology , Retinal Bipolar Cells/metabolism , Sequence Analysis, RNA , Single-Cell Analysis/methods , Transcription, Genetic
5.
Cell ; 161(5): 1202-1214, 2015 May 21.
Article in English | MEDLINE | ID: mdl-26000488

ABSTRACT

Cells, the basic units of biological structure and function, vary broadly in type and state. Single-cell genomics can characterize cell identity and function, but limitations of ease and scale have prevented its broad application. Here we describe Drop-seq, a strategy for quickly profiling thousands of individual cells by separating them into nanoliter-sized aqueous droplets, associating a different barcode with each cell's RNAs, and sequencing them all together. Drop-seq analyzes mRNA transcripts from thousands of individual cells simultaneously while remembering transcripts' cell of origin. We analyzed transcriptomes from 44,808 mouse retinal cells and identified 39 transcriptionally distinct cell populations, creating a molecular atlas of gene expression for known retinal cell classes and novel candidate cell subtypes. Drop-seq will accelerate biological discovery by enabling routine transcriptional profiling at single-cell resolution. VIDEO ABSTRACT.


Subject(s)
Gene Expression Profiling/methods , Genome-Wide Association Study , Microfluidic Analytical Techniques , Retina/cytology , Single-Cell Analysis , Animals , High-Throughput Nucleotide Sequencing , Mice , Sequence Analysis, RNA
6.
Nature ; 627(8004): 604-611, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38448582

ABSTRACT

Human brains vary across people and over time; such variation is not yet understood in cellular terms. Here we describe a relationship between people's cortical neurons and cortical astrocytes. We used single-nucleus RNA sequencing to analyse the prefrontal cortex of 191 human donors aged 22-97 years, including healthy individuals and people with schizophrenia. Latent-factor analysis of these data revealed that, in people whose cortical neurons more strongly expressed genes encoding synaptic components, cortical astrocytes more strongly expressed distinct genes with synaptic functions and genes for synthesizing cholesterol, an astrocyte-supplied component of synaptic membranes. We call this relationship the synaptic neuron and astrocyte program (SNAP). In schizophrenia and ageing-two conditions that involve declines in cognitive flexibility and plasticity1,2-cells divested from SNAP: astrocytes, glutamatergic (excitatory) neurons and GABAergic (inhibitory) neurons all showed reduced SNAP expression to corresponding degrees. The distinct astrocytic and neuronal components of SNAP both involved genes in which genetic risk factors for schizophrenia were strongly concentrated. SNAP, which varies quantitatively even among healthy people of similar age, may underlie many aspects of normal human interindividual differences and may be an important point of convergence for multiple kinds of pathophysiology.


Subject(s)
Aging , Astrocytes , Neurons , Prefrontal Cortex , Schizophrenia , Adult , Aged , Aged, 80 and over , Humans , Middle Aged , Young Adult , Aging/metabolism , Aging/pathology , Astrocytes/cytology , Astrocytes/metabolism , Astrocytes/pathology , Cholesterol/metabolism , Cognition , GABAergic Neurons/metabolism , Genetic Predisposition to Disease , Glutamine/metabolism , Health , Individuality , Neural Inhibition , Neuronal Plasticity , Neurons/cytology , Neurons/metabolism , Neurons/pathology , Prefrontal Cortex/cytology , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Schizophrenia/genetics , Schizophrenia/metabolism , Schizophrenia/pathology , Single-Cell Gene Expression Analysis , Synapses/genetics , Synapses/metabolism , Synapses/pathology , Synaptic Membranes/chemistry , Synaptic Membranes/metabolism
7.
Nature ; 631(8019): 134-141, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38867047

ABSTRACT

Mosaic loss of the X chromosome (mLOX) is the most common clonal somatic alteration in leukocytes of female individuals1,2, but little is known about its genetic determinants or phenotypic consequences. Here, to address this, we used data from 883,574 female participants across 8 biobanks; 12% of participants exhibited detectable mLOX in approximately 2% of leukocytes. Female participants with mLOX had an increased risk of myeloid and lymphoid leukaemias. Genetic analyses identified 56 common variants associated with mLOX, implicating genes with roles in chromosomal missegregation, cancer predisposition and autoimmune diseases. Exome-sequence analyses identified rare missense variants in FBXO10 that confer a twofold increased risk of mLOX. Only a small fraction of associations was shared with mosaic Y chromosome loss, suggesting that distinct biological processes drive formation and clonal expansion of sex chromosome missegregation. Allelic shift analyses identified X chromosome alleles that are preferentially retained in mLOX, demonstrating variation at many loci under cellular selection. A polygenic score including 44 allelic shift loci correctly inferred the retained X chromosomes in 80.7% of mLOX cases in the top decile. Our results support a model in which germline variants predispose female individuals to acquiring mLOX, with the allelic content of the X chromosome possibly shaping the magnitude of clonal expansion.


Subject(s)
Aneuploidy , Chromosomes, Human, X , Clone Cells , Leukocytes , Mosaicism , Adult , Female , Humans , Male , Middle Aged , Alleles , Autoimmune Diseases/genetics , Biological Specimen Banks , Chromosome Segregation/genetics , Chromosomes, Human, X/genetics , Chromosomes, Human, Y/genetics , Clone Cells/metabolism , Clone Cells/pathology , Exome/genetics , F-Box Proteins/genetics , Genetic Predisposition to Disease/genetics , Germ-Line Mutation , Leukemia/genetics , Leukocytes/metabolism , Models, Genetic , Multifactorial Inheritance/genetics , Mutation, Missense/genetics
8.
Cell ; 159(5): 1015-1026, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25416942

ABSTRACT

Genomic DNA replicates in a choreographed temporal order that impacts the distribution of mutations along the genome. We show here that DNA replication timing is shaped by genetic polymorphisms that act in cis upon megabase-scale DNA segments. In genome sequences from proliferating cells, read depth along chromosomes reflected DNA replication activity in those cells. We used this relationship to analyze variation in replication timing among 161 individuals sequenced by the 1000 Genomes Project. Genome-wide association of replication timing with genetic variation identified 16 loci at which inherited alleles associate with replication timing. We call these "replication timing quantitative trait loci" (rtQTLs). rtQTLs involved the differential use of replication origins, exhibited allele-specific effects on replication timing, and associated with gene expression variation at megabase scales. Our results show replication timing to be shaped by genetic polymorphism and identify a means by which inherited polymorphism regulates the mutability of nearby sequences.


Subject(s)
Polymorphism, Genetic , Quantitative Trait Loci , DNA Replication Timing , Genetics, Population , Genome, Human , Genome-Wide Association Study , Humans , Janus Kinase 2/metabolism , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , Replication Origin
9.
Nature ; 624(7991): 390-402, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38092918

ABSTRACT

Divergence of cis-regulatory elements drives species-specific traits1, but how this manifests in the evolution of the neocortex at the molecular and cellular level remains unclear. Here we investigated the gene regulatory programs in the primary motor cortex of human, macaque, marmoset and mouse using single-cell multiomics assays, generating gene expression, chromatin accessibility, DNA methylome and chromosomal conformation profiles from a total of over 200,000 cells. From these data, we show evidence that divergence of transcription factor expression corresponds to species-specific epigenome landscapes. We find that conserved and divergent gene regulatory features are reflected in the evolution of the three-dimensional genome. Transposable elements contribute to nearly 80% of the human-specific candidate cis-regulatory elements in cortical cells. Through machine learning, we develop sequence-based predictors of candidate cis-regulatory elements in different species and demonstrate that the genomic regulatory syntax is highly preserved from rodents to primates. Finally, we show that epigenetic conservation combined with sequence similarity helps to uncover functional cis-regulatory elements and enhances our ability to interpret genetic variants contributing to neurological disease and traits.


Subject(s)
Conserved Sequence , Evolution, Molecular , Gene Expression Regulation , Gene Regulatory Networks , Mammals , Neocortex , Animals , Humans , Mice , Callithrix/genetics , Chromatin/genetics , Chromatin/metabolism , Conserved Sequence/genetics , DNA Methylation , DNA Transposable Elements/genetics , Epigenome , Gene Expression Regulation/genetics , Macaca/genetics , Mammals/genetics , Motor Cortex/cytology , Motor Cortex/metabolism , Multiomics , Neocortex/cytology , Neocortex/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Single-Cell Analysis , Transcription Factors/metabolism , Genetic Variation/genetics
10.
Mol Cell ; 81(3): 426-441.e8, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33545059

ABSTRACT

Eukaryotic genomes replicate via spatially and temporally regulated origin firing. Cyclin-dependent kinase (CDK) and Dbf4-dependent kinase (DDK) promote origin firing, whereas the S phase checkpoint limits firing to prevent nucleotide and RPA exhaustion. We used chemical genetics to interrogate human DDK with maximum precision, dissect its relationship with the S phase checkpoint, and identify DDK substrates. We show that DDK inhibition (DDKi) leads to graded suppression of origin firing and fork arrest. S phase checkpoint inhibition rescued origin firing in DDKi cells and DDK-depleted Xenopus egg extracts. DDKi also impairs RPA loading, nascent-strand protection, and fork restart. Via quantitative phosphoproteomics, we identify the BRCA1-associated (BRCA1-A) complex subunit MERIT40 and the cohesin accessory subunit PDS5B as DDK effectors in fork protection and restart. Phosphorylation neutralizes autoinhibition mediated by intrinsically disordered regions in both substrates. Our results reveal mechanisms through which DDK controls the duplication of large vertebrate genomes.


Subject(s)
DNA Replication , Replication Origin , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Checkpoint Kinase 1/genetics , Checkpoint Kinase 1/metabolism , DNA Replication/drug effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , HCT116 Cells , HEK293 Cells , HeLa Cells , Humans , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , S Phase Cell Cycle Checkpoints , Substrate Specificity , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Xenopus laevis
11.
Immunity ; 50(1): 253-271.e6, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30471926

ABSTRACT

Microglia, the resident immune cells of the brain, rapidly change states in response to their environment, but we lack molecular and functional signatures of different microglial populations. Here, we analyzed the RNA expression patterns of more than 76,000 individual microglia in mice during development, in old age, and after brain injury. Our analysis uncovered at least nine transcriptionally distinct microglial states, which expressed unique sets of genes and were localized in the brain using specific markers. The greatest microglial heterogeneity was found at young ages; however, several states-including chemokine-enriched inflammatory microglia-persisted throughout the lifespan or increased in the aged brain. Multiple reactive microglial subtypes were also found following demyelinating injury in mice, at least one of which was also found in human multiple sclerosis lesions. These distinct microglia signatures can be used to better understand microglia function and to identify and manipulate specific subpopulations in health and disease.


Subject(s)
Aging/immunology , Brain Injuries/immunology , Brain/physiology , Microglia/physiology , Multiple Sclerosis/immunology , Adaptation, Physiological , Aging/genetics , Animals , Brain Injuries/genetics , Cell Differentiation , Demyelinating Diseases , Humans , Longevity , Mice , Mice, Inbred C57BL , Sequence Analysis, RNA , Single-Cell Analysis
12.
Cell ; 154(3): 481-3, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23911315

ABSTRACT

The selective breeding of rats as physiological, behavioral, and disease models generated a wealth of variation relevant to the genetics of complex traits. In this issue, Atanur and colleagues sequence the genomes of 25 inbred rat strains to understand how artificial selection shaped their genomes.


Subject(s)
Electrocardiography/methods , Intraoperative Care/methods , Humans
13.
Nature ; 584(7819): 136-141, 2020 08.
Article in English | MEDLINE | ID: mdl-32581363

ABSTRACT

Clonally expanded blood cells that contain somatic mutations (clonal haematopoiesis) are commonly acquired with age and increase the risk of blood cancer1-9. The blood clones identified so far contain diverse large-scale mosaic chromosomal alterations (deletions, duplications and copy-neutral loss of heterozygosity (CN-LOH)) on all chromosomes1,2,5,6,9, but the sources of selective advantage that drive the expansion of most clones remain unknown. Here, to identify genes, mutations and biological processes that give selective advantage to mutant clones, we analysed genotyping data from the blood-derived DNA of 482,789 participants from the UK Biobank10. We identified 19,632 autosomal mosaic chromosomal alterations and analysed these for relationships to inherited genetic variation. We found 52 inherited, rare, large-effect coding or splice variants in 7 genes that were associated with greatly increased vulnerability to clonal haematopoiesis with specific acquired CN-LOH mutations. Acquired mutations systematically replaced the inherited risk alleles (at MPL) or duplicated them to the homologous chromosome (at FH, NBN, MRE11, ATM, SH2B3 and TM2D3). Three of the genes (MRE11, NBN and ATM) encode components of the MRN-ATM pathway, which limits cell division after DNA damage and telomere attrition11-13; another two (MPL and SH2B3) encode proteins that regulate the self-renewal of stem cells14-16. In addition, we found that CN-LOH mutations across the genome tended to cause chromosomal segments with alleles that promote the expansion of haematopoietic cells to replace their homologous (allelic) counterparts, increasing polygenic drive for blood-cell proliferation traits. Readily acquired mutations that replace chromosomal segments with their homologous counterparts seem to interact with pervasive inherited variation to create a challenge for lifelong cytopoiesis.


Subject(s)
Clonal Evolution/genetics , Clone Cells/metabolism , Hematopoiesis/genetics , Multifactorial Inheritance/genetics , Adult , Aged , Alleles , Cardiovascular Diseases/genetics , Cardiovascular Diseases/pathology , Cell Division/genetics , Chromosome Aberrations , Clone Cells/cytology , Clone Cells/pathology , Female , Genetic Predisposition to Disease , Hematologic Neoplasms/genetics , Hematologic Neoplasms/pathology , Humans , Loss of Heterozygosity/genetics , Male , Middle Aged , Mosaicism , United Kingdom
14.
Nature ; 583(7815): 259-264, 2020 07.
Article in English | MEDLINE | ID: mdl-32494014

ABSTRACT

Meiosis, although essential for reproduction, is also variable and error-prone: rates of chromosome crossover vary among gametes, between the sexes, and among humans of the same sex, and chromosome missegregation leads to abnormal chromosome numbers (aneuploidy)1-8. To study diverse meiotic outcomes and how they covary across chromosomes, gametes and humans, we developed Sperm-seq, a way of simultaneously analysing the genomes of thousands of individual sperm. Here we analyse the genomes of 31,228 human gametes from 20 sperm donors, identifying 813,122 crossovers and 787 aneuploid chromosomes. Sperm donors had aneuploidy rates ranging from 0.01 to 0.05 aneuploidies per gamete; crossovers partially protected chromosomes from nondisjunction at the meiosis I cell division. Some chromosomes and donors underwent more-frequent nondisjunction during meiosis I, and others showed more meiosis II segregation failures. Sperm genomes also manifested many genomic anomalies that could not be explained by simple nondisjunction. Diverse recombination phenotypes-from crossover rates to crossover location and separation, a measure of crossover interference-covaried strongly across individuals and cells. Our results can be incorporated with earlier observations into a unified model in which a core mechanism, the variable physical compaction of meiotic chromosomes, generates interindividual and cell-to-cell variation in diverse meiotic phenotypes.


Subject(s)
Genome, Human/genetics , Meiosis/genetics , Spermatozoa/cytology , Spermatozoa/metabolism , Adolescent , Adult , Alleles , Aneuploidy , Crossing Over, Genetic/genetics , Haplotypes/genetics , Humans , Male , Nondisjunction, Genetic , Single-Cell Analysis , Tissue Donors , Young Adult
15.
Nature ; 584(7819): 130-135, 2020 08.
Article in English | MEDLINE | ID: mdl-32581364

ABSTRACT

The extent to which the biology of oncogenesis and ageing are shaped by factors that distinguish human populations is unknown. Haematopoietic clones with acquired mutations become common with advancing age and can lead to blood cancers1-10. Here we describe shared and population-specific patterns of genomic mutations and clonal selection in haematopoietic cells on the basis of 33,250 autosomal mosaic chromosomal alterations that we detected in 179,417 Japanese participants in the BioBank Japan cohort and compared with analogous data from the UK Biobank. In this long-lived Japanese population, mosaic chromosomal alterations were detected in more than 35.0% (s.e.m., 1.4%) of individuals older than 90 years, which suggests that such clones trend towards inevitability with advancing age. Japanese and European individuals exhibited key differences in the genomic locations of mutations in their respective haematopoietic clones; these differences predicted the relative rates of chronic lymphocytic leukaemia (which is more common among European individuals) and T cell leukaemia (which is more common among Japanese individuals) in these populations. Three different mutational precursors of chronic lymphocytic leukaemia (including trisomy 12, loss of chromosomes 13q and 13q, and copy-neutral loss of heterozygosity) were between two and six times less common among Japanese individuals, which suggests that the Japanese and European populations differ in selective pressures on clones long before the development of clinically apparent chronic lymphocytic leukaemia. Japanese and British populations also exhibited very different rates of clones that arose from B and T cell lineages, which predicted the relative rates of B and T cell cancers in these populations. We identified six previously undescribed loci at which inherited variants predispose to mosaic chromosomal alterations that duplicate or remove the inherited risk alleles, including large-effect rare variants at NBN, MRE11 and CTU2 (odds ratio, 28-91). We suggest that selective pressures on clones are modulated by factors that are specific to human populations. Further genomic characterization of clonal selection and cancer in populations from around the world is therefore warranted.


Subject(s)
Aging/genetics , Chromosome Aberrations , Chromosomes, Human/genetics , Clone Cells/metabolism , Genome, Human/genetics , Hematopoietic Stem Cells/metabolism , Mutation , Aged, 80 and over , Alleles , Cell Lineage , Clone Cells/cytology , Clone Cells/pathology , Cohort Studies , Female , Genetic Loci/genetics , Hematopoiesis/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/pathology , Humans , Japan , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Leukemia, T-Cell/genetics , Leukemia, T-Cell/pathology , Male , Mosaicism , United Kingdom
16.
Nature ; 586(7828): 262-269, 2020 10.
Article in English | MEDLINE | ID: mdl-32999462

ABSTRACT

Primates and rodents, which descended from a common ancestor around 90 million years ago1, exhibit profound differences in behaviour and cognitive capacity; the cellular basis for these differences is unknown. Here we use single-nucleus RNA sequencing to profile RNA expression in 188,776 individual interneurons across homologous brain regions from three primates (human, macaque and marmoset), a rodent (mouse) and a weasel (ferret). Homologous interneuron types-which were readily identified by their RNA-expression patterns-varied in abundance and RNA expression among ferrets, mice and primates, but varied less among primates. Only a modest fraction of the genes identified as 'markers' of specific interneuron subtypes in any one species had this property in another species. In the primate neocortex, dozens of genes showed spatial expression gradients among interneurons of the same type, which suggests that regional variation in cortical contexts shapes the RNA expression patterns of adult neocortical interneurons. We found that an interneuron type that was previously associated with the mouse hippocampus-the 'ivy cell', which has neurogliaform characteristics-has become abundant across the neocortex of humans, macaques and marmosets but not mice or ferrets. We also found a notable subcortical innovation: an abundant striatal interneuron type in primates that had no molecularly homologous counterpart in mice or ferrets. These interneurons expressed a unique combination of genes that encode transcription factors, receptors and neuropeptides and constituted around 30% of striatal interneurons in marmosets and humans.


Subject(s)
Interneurons/cytology , Primates , Animals , Callithrix , Cerebral Cortex/cytology , Female , Ferrets , Hippocampus/cytology , Humans , Interneurons/metabolism , LIM-Homeodomain Proteins/metabolism , Lysosomal Membrane Proteins/metabolism , Macaca , Male , Mice , Neostriatum/cytology , Nerve Tissue Proteins/metabolism , RNA/genetics , Species Specificity , Transcription Factors/metabolism
18.
Nature ; 582(7813): 577-581, 2020 06.
Article in English | MEDLINE | ID: mdl-32499649

ABSTRACT

Many common illnesses, for reasons that have not been identified, differentially affect men and women. For instance, the autoimmune diseases systemic lupus erythematosus (SLE) and Sjögren's syndrome affect nine times more women than men1, whereas schizophrenia affects men with greater frequency and severity relative to women2. All three illnesses have their strongest common genetic associations in the major histocompatibility complex (MHC) locus, an association that in SLE and Sjögren's syndrome has long been thought to arise from alleles of the human leukocyte antigen (HLA) genes at that locus3-6. Here we show that variation of the complement component 4 (C4) genes C4A and C4B, which are also at the MHC locus and have been linked to increased risk for schizophrenia7, generates 7-fold variation in risk for SLE and 16-fold variation in risk for Sjögren's syndrome among individuals with common C4 genotypes, with C4A protecting more strongly than C4B in both illnesses. The same alleles that increase risk for schizophrenia greatly reduce risk for SLE and Sjögren's syndrome. In all three illnesses, C4 alleles act more strongly in men than in women: common combinations of C4A and C4B generated 14-fold variation in risk for SLE, 31-fold variation in risk for Sjögren's syndrome, and 1.7-fold variation in schizophrenia risk among men (versus 6-fold, 15-fold and 1.26-fold variation in risk among women, respectively). At a protein level, both C4 and its effector C3 were present at higher levels in cerebrospinal fluid and plasma8,9 in men than in women among adults aged between 20 and 50 years, corresponding to the ages of differential disease vulnerability. Sex differences in complement protein levels may help to explain the more potent effects of C4 alleles in men, women's greater risk of SLE and Sjögren's syndrome and men's greater vulnerability to schizophrenia. These results implicate the complement system as a source of sexual dimorphism in vulnerability to diverse illnesses.


Subject(s)
Complement C3/genetics , Complement C4/genetics , Lupus Erythematosus, Systemic/genetics , Sex Characteristics , Sjogren's Syndrome/genetics , Adult , Alleles , Complement C3/analysis , Complement C3/cerebrospinal fluid , Complement C4/analysis , Complement C4/cerebrospinal fluid , Female , Genetic Predisposition to Disease , HLA Antigens/genetics , Haplotypes , Humans , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/cerebrospinal fluid , Major Histocompatibility Complex/genetics , Male , Middle Aged , Sjogren's Syndrome/blood , Sjogren's Syndrome/cerebrospinal fluid , Young Adult
19.
Bioinformatics ; 40(2)2024 01 02.
Article in English | MEDLINE | ID: mdl-38261650

ABSTRACT

MOTIVATION: Many genetics studies report results tied to genomic coordinates of a legacy genome assembly. However, as assemblies are updated and improved, researchers are faced with either realigning raw sequence data using the updated coordinate system or converting legacy datasets to the updated coordinate system to be able to combine results with newer datasets. Currently available tools to perform the conversion of genetic variants have numerous shortcomings, including poor support for indels and multi-allelic variants, that lead to a higher rate of variants being dropped or incorrectly converted. As a result, many researchers continue to work with and publish using legacy genomic coordinates. RESULTS: Here we present BCFtools/liftover, a tool to convert genomic coordinates across genome assemblies for variants encoded in the variant call format with improved support for indels represented by different reference alleles across genome assemblies and full support for multi-allelic variants. It further supports variant annotation fields updates whenever the reference allele changes across genome assemblies. The tool has the lowest rate of variants being dropped with an order of magnitude less indels dropped or incorrectly converted and is an order of magnitude faster than other tools typically used for the same task. It is particularly suited for converting variant callsets from large cohorts to novel telomere-to-telomere assemblies as well as summary statistics from genome-wide association studies tied to legacy genome assemblies. AVAILABILITY AND IMPLEMENTATION: The tool is written in C and freely available under the MIT open source license as a BCFtools plugin available at http://github.com/freeseek/score.


Subject(s)
Genome-Wide Association Study , Software , Genomics/methods , Alleles , INDEL Mutation
SELECTION OF CITATIONS
SEARCH DETAIL