Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 454
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 187(5): 1024-1037, 2024 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-38290514

RESUMEN

This perspective focuses on advances in genome technology over the last 25 years and their impact on germline variant discovery within the field of human genetics. The field has witnessed tremendous technological advances from microarrays to short-read sequencing and now long-read sequencing. Each technology has provided genome-wide access to different classes of human genetic variation. We are now on the verge of comprehensive variant detection of all forms of variation for the first time with a single assay. We predict that this transition will further transform our understanding of human health and biology and, more importantly, provide novel insights into the dynamic mutational processes shaping our genomes.


Asunto(s)
Variación Estructural del Genoma , Genómica , Humanos , Genómica/métodos , Mutación de Línea Germinal , Mutación , Tecnología
2.
Cell ; 187(6): 1547-1562.e13, 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38428424

RESUMEN

We sequenced and assembled using multiple long-read sequencing technologies the genomes of chimpanzee, bonobo, gorilla, orangutan, gibbon, macaque, owl monkey, and marmoset. We identified 1,338,997 lineage-specific fixed structural variants (SVs) disrupting 1,561 protein-coding genes and 136,932 regulatory elements, including the most complete set of human-specific fixed differences. We estimate that 819.47 Mbp or ∼27% of the genome has been affected by SVs across primate evolution. We identify 1,607 structurally divergent regions wherein recurrent structural variation contributes to creating SV hotspots where genes are recurrently lost (e.g., CARD, C4, and OLAH gene families) and additional lineage-specific genes are generated (e.g., CKAP2, VPS36, ACBD7, and NEK5 paralogs), becoming targets of rapid chromosomal diversification and positive selection (e.g., RGPD gene family). High-fidelity long-read sequencing has made these dynamic regions of the genome accessible for sequence-level analyses within and between primate species.


Asunto(s)
Genoma , Primates , Animales , Humanos , Secuencia de Bases , Primates/clasificación , Primates/genética , Evolución Biológica , Análisis de Secuencia de ADN , Variación Estructural del Genoma
3.
Cell ; 185(11): 1986-2005.e26, 2022 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-35525246

RESUMEN

Unlike copy number variants (CNVs), inversions remain an underexplored genetic variation class. By integrating multiple genomic technologies, we discover 729 inversions in 41 human genomes. Approximately 85% of inversions <2 kbp form by twin-priming during L1 retrotransposition; 80% of the larger inversions are balanced and affect twice as many nucleotides as CNVs. Balanced inversions show an excess of common variants, and 72% are flanked by segmental duplications (SDs) or retrotransposons. Since flanking repeats promote non-allelic homologous recombination, we developed complementary approaches to identify recurrent inversion formation. We describe 40 recurrent inversions encompassing 0.6% of the genome, showing inversion rates up to 2.7 × 10-4 per locus per generation. Recurrent inversions exhibit a sex-chromosomal bias and co-localize with genomic disorder critical regions. We propose that inversion recurrence results in an elevated number of heterozygous carriers and structural SD diversity, which increases mutability in the population and predisposes specific haplotypes to disease-causing CNVs.


Asunto(s)
Inversión Cromosómica , Duplicaciones Segmentarias en el Genoma , Inversión Cromosómica/genética , Variaciones en el Número de Copia de ADN/genética , Genoma Humano , Genómica , Humanos
4.
Cell ; 176(3): 663-675.e19, 2019 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-30661756

RESUMEN

In order to provide a comprehensive resource for human structural variants (SVs), we generated long-read sequence data and analyzed SVs for fifteen human genomes. We sequence resolved 99,604 insertions, deletions, and inversions including 2,238 (1.6 Mbp) that are shared among all discovery genomes with an additional 13,053 (6.9 Mbp) present in the majority, indicating minor alleles or errors in the reference. Genotyping in 440 additional genomes confirms the most common SVs in unique euchromatin are now sequence resolved. We report a ninefold SV bias toward the last 5 Mbp of human chromosomes with nearly 55% of all VNTRs (variable number of tandem repeats) mapping to this portion of the genome. We identify SVs affecting coding and noncoding regulatory loci improving annotation and interpretation of functional variation. These data provide the framework to construct a canonical human reference and a resource for developing advanced representations capable of capturing allelic diversity.


Asunto(s)
Frecuencia de los Genes/genética , Genoma Humano/genética , Variación Estructural del Genoma/genética , Alelos , Eucromatina/genética , Genómica/métodos , Humanos , Repeticiones de Minisatélite/genética , Análisis de Secuencia de ADN/métodos
5.
Cell ; 176(4): 743-756.e17, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30735633

RESUMEN

Direct comparisons of human and non-human primate brains can reveal molecular pathways underlying remarkable specializations of the human brain. However, chimpanzee tissue is inaccessible during neocortical neurogenesis when differences in brain size first appear. To identify human-specific features of cortical development, we leveraged recent innovations that permit generating pluripotent stem cell-derived cerebral organoids from chimpanzee. Despite metabolic differences, organoid models preserve gene regulatory networks related to primary cell types and developmental processes. We further identified 261 differentially expressed genes in human compared to both chimpanzee organoids and macaque cortex, enriched for recent gene duplications, and including multiple regulators of PI3K-AKT-mTOR signaling. We observed increased activation of this pathway in human radial glia, dependent on two receptors upregulated specifically in human: INSR and ITGB8. Our findings establish a platform for systematic analysis of molecular changes contributing to human brain development and evolution.


Asunto(s)
Corteza Cerebral/citología , Organoides/metabolismo , Animales , Evolución Biológica , Encéfalo/citología , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/genética , Corteza Cerebral/metabolismo , Redes Reguladoras de Genes/genética , Humanos , Células Madre Pluripotentes Inducidas/citología , Macaca , Neurogénesis/genética , Organoides/crecimiento & desarrollo , Pan troglodytes , Células Madre Pluripotentes/citología , Análisis de la Célula Individual , Especificidad de la Especie , Transcriptoma/genética
6.
Cell ; 173(6): 1356-1369.e22, 2018 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-29856954

RESUMEN

Genetic changes causing brain size expansion in human evolution have remained elusive. Notch signaling is essential for radial glia stem cell proliferation and is a determinant of neuronal number in the mammalian cortex. We find that three paralogs of human-specific NOTCH2NL are highly expressed in radial glia. Functional analysis reveals that different alleles of NOTCH2NL have varying potencies to enhance Notch signaling by interacting directly with NOTCH receptors. Consistent with a role in Notch signaling, NOTCH2NL ectopic expression delays differentiation of neuronal progenitors, while deletion accelerates differentiation into cortical neurons. Furthermore, NOTCH2NL genes provide the breakpoints in 1q21.1 distal deletion/duplication syndrome, where duplications are associated with macrocephaly and autism and deletions with microcephaly and schizophrenia. Thus, the emergence of human-specific NOTCH2NL genes may have contributed to the rapid evolution of the larger human neocortex, accompanied by loss of genomic stability at the 1q21.1 locus and resulting recurrent neurodevelopmental disorders.


Asunto(s)
Encéfalo/embriología , Corteza Cerebral/fisiología , Neurogénesis/fisiología , Receptor Notch2/metabolismo , Transducción de Señal , Animales , Diferenciación Celular , Células Madre Embrionarias/metabolismo , Femenino , Eliminación de Gen , Genes Reporteros , Gorilla gorilla , Células HEK293 , Humanos , Neocórtex/citología , Células-Madre Neurales/metabolismo , Neuroglía/metabolismo , Neuronas/metabolismo , Pan troglodytes , Receptor Notch2/genética , Análisis de Secuencia de ARN
7.
Cell ; 172(5): 897-909.e21, 2018 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-29474918

RESUMEN

X-linked Dystonia-Parkinsonism (XDP) is a Mendelian neurodegenerative disease that is endemic to the Philippines and is associated with a founder haplotype. We integrated multiple genome and transcriptome assembly technologies to narrow the causal mutation to the TAF1 locus, which included a SINE-VNTR-Alu (SVA) retrotransposition into intron 32 of the gene. Transcriptome analyses identified decreased expression of the canonical cTAF1 transcript among XDP probands, and de novo assembly across multiple pluripotent stem-cell-derived neuronal lineages discovered aberrant TAF1 transcription that involved alternative splicing and intron retention (IR) in proximity to the SVA that was anti-correlated with overall TAF1 expression. CRISPR/Cas9 excision of the SVA rescued this XDP-specific transcriptional signature and normalized TAF1 expression in probands. These data suggest an SVA-mediated aberrant transcriptional mechanism associated with XDP and may provide a roadmap for layered technologies and integrated assembly-based analyses for other unsolved Mendelian disorders.


Asunto(s)
Trastornos Distónicos/genética , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Genoma Humano , Transcriptoma/genética , Empalme Alternativo/genética , Elementos Alu/genética , Secuencia de Bases , Sistemas CRISPR-Cas/genética , Estudios de Cohortes , Familia , Femenino , Sitios Genéticos , Haplotipos/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Histona Acetiltransferasas/genética , Histona Acetiltransferasas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Intrones/genética , Masculino , Repeticiones de Minisatélite/genética , Modelos Genéticos , Degeneración Nerviosa/genética , Degeneración Nerviosa/patología , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Elementos de Nucleótido Esparcido Corto , Factores Asociados con la Proteína de Unión a TATA/genética , Factores Asociados con la Proteína de Unión a TATA/metabolismo , Factor de Transcripción TFIID/genética , Factor de Transcripción TFIID/metabolismo
8.
Cell ; 171(3): 710-722.e12, 2017 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-28965761

RESUMEN

To further our understanding of the genetic etiology of autism, we generated and analyzed genome sequence data from 516 idiopathic autism families (2,064 individuals). This resource includes >59 million single-nucleotide variants (SNVs) and 9,212 private copy number variants (CNVs), of which 133,992 and 88 are de novo mutations (DNMs), respectively. We estimate a mutation rate of ∼1.5 × 10-8 SNVs per site per generation with a significantly higher mutation rate in repetitive DNA. Comparing probands and unaffected siblings, we observe several DNM trends. Probands carry more gene-disruptive CNVs and SNVs, resulting in severe missense mutations and mapping to predicted fetal brain promoters and embryonic stem cell enhancers. These differences become more pronounced for autism genes (p = 1.8 × 10-3, OR = 2.2). Patients are more likely to carry multiple coding and noncoding DNMs in different genes, which are enriched for expression in striatal neurons (p = 3 × 10-3), suggesting a path forward for genetically characterizing more complex cases of autism.


Asunto(s)
Trastorno Autístico/genética , Variaciones en el Número de Copia de ADN , Polimorfismo de Nucleótido Simple , Animales , Análisis Mutacional de ADN , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Mutación INDEL , Masculino , Ratones
9.
Nature ; 629(8010): 136-145, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38570684

RESUMEN

Human centromeres have been traditionally very difficult to sequence and assemble owing to their repetitive nature and large size1. As a result, patterns of human centromeric variation and models for their evolution and function remain incomplete, despite centromeres being among the most rapidly mutating regions2,3. Here, using long-read sequencing, we completely sequenced and assembled all centromeres from a second human genome and compared it to the finished reference genome4,5. We find that the two sets of centromeres show at least a 4.1-fold increase in single-nucleotide variation when compared with their unique flanks and vary up to 3-fold in size. Moreover, we find that 45.8% of centromeric sequence cannot be reliably aligned using standard methods owing to the emergence of new α-satellite higher-order repeats (HORs). DNA methylation and CENP-A chromatin immunoprecipitation experiments show that 26% of the centromeres differ in their kinetochore position by >500 kb. To understand evolutionary change, we selected six chromosomes and sequenced and assembled 31 orthologous centromeres from the common chimpanzee, orangutan and macaque genomes. Comparative analyses reveal a nearly complete turnover of α-satellite HORs, with characteristic idiosyncratic changes in α-satellite HORs for each species. Phylogenetic reconstruction of human haplotypes supports limited to no recombination between the short (p) and long (q) arms across centromeres and reveals that novel α-satellite HORs share a monophyletic origin, providing a strategy to estimate the rate of saltatory amplification and mutation of human centromeric DNA.


Asunto(s)
Centrómero , Evolución Molecular , Variación Genética , Animales , Humanos , Centrómero/genética , Centrómero/metabolismo , Proteína A Centromérica/metabolismo , Metilación de ADN/genética , ADN Satélite/genética , Cinetocoros/metabolismo , Macaca/genética , Pan troglodytes/genética , Polimorfismo de Nucleótido Simple/genética , Pongo/genética , Masculino , Femenino , Estándares de Referencia , Inmunoprecipitación de Cromatina , Haplotipos , Mutación , Amplificación de Genes , Alineación de Secuencia , Cromatina/genética , Cromatina/metabolismo , Especificidad de la Especie
10.
Cell ; 156(5): 872-7, 2014 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-24581488

RESUMEN

Medical genetics typically entails the detailed characterization of a patient's phenotypes followed by genotyping to discover the responsible gene or mutation. Here, we propose that the systematic discovery of genetic variants associated with complex diseases such as autism are progressing to a point where a reverse strategy may be fruitful in assigning the pathogenic effects of many different genes and in determining whether particular genotypes manifest as clinically recognizable phenotypes. This "genotype-first" approach for complex disease necessitates the development of large, highly integrated networks of researchers, clinicians, and patient families, with the promise of improved therapies for subsets of patients.


Asunto(s)
Trastorno Autístico/genética , Heterogeneidad Genética , Genotipo , Trastorno Autístico/clasificación , Trastorno Autístico/diagnóstico , Redes Comunitarias , Exoma , Humanos , Mutación , Fenotipo
11.
Cell ; 158(2): 263-276, 2014 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-24998929

RESUMEN

Autism spectrum disorder (ASD) is a heterogeneous disease in which efforts to define subtypes behaviorally have met with limited success. Hypothesizing that genetically based subtype identification may prove more productive, we resequenced the ASD-associated gene CHD8 in 3,730 children with developmental delay or ASD. We identified a total of 15 independent mutations; no truncating events were identified in 8,792 controls, including 2,289 unaffected siblings. In addition to a high likelihood of an ASD diagnosis among patients bearing CHD8 mutations, characteristics enriched in this group included macrocephaly, distinct faces, and gastrointestinal complaints. chd8 disruption in zebrafish recapitulates features of the human phenotype, including increased head size as a result of expansion of the forebrain/midbrain and impairment of gastrointestinal motility due to a reduction in postmitotic enteric neurons. Our findings indicate that CHD8 disruptions define a distinct ASD subtype and reveal unexpected comorbidities between brain development and enteric innervation.


Asunto(s)
Trastornos Generalizados del Desarrollo Infantil/genética , Trastornos Generalizados del Desarrollo Infantil/fisiopatología , Proteínas de Unión al ADN/genética , Factores de Transcripción/genética , Adolescente , Secuencia de Aminoácidos , Animales , Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Niño , Trastornos Generalizados del Desarrollo Infantil/clasificación , Trastornos Generalizados del Desarrollo Infantil/patología , Preescolar , Proteínas de Unión al ADN/metabolismo , Femenino , Tracto Gastrointestinal/inervación , Tracto Gastrointestinal/fisiopatología , Humanos , Macaca mulatta , Masculino , Megalencefalia/patología , Datos de Secuencia Molecular , Mutación , Alineación de Secuencia , Factores de Transcripción/metabolismo , Pez Cebra , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
12.
Nature ; 617(7960): 325-334, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37165237

RESUMEN

Single-nucleotide variants (SNVs) in segmental duplications (SDs) have not been systematically assessed because of the limitations of mapping short-read sequencing data1,2. Here we constructed 1:1 unambiguous alignments spanning high-identity SDs across 102 human haplotypes and compared the pattern of SNVs between unique and duplicated regions3,4. We find that human SNVs are elevated 60% in SDs compared to unique regions and estimate that at least 23% of this increase is due to interlocus gene conversion (IGC) with up to 4.3 megabase pairs of SD sequence converted on average per human haplotype. We develop a genome-wide map of IGC donors and acceptors, including 498 acceptor and 454 donor hotspots affecting the exons of about 800 protein-coding genes. These include 171 genes that have 'relocated' on average 1.61 megabase pairs in a subset of human haplotypes. Using a coalescent framework, we show that SD regions are slightly evolutionarily older when compared to unique sequences, probably owing to IGC. SNVs in SDs, however, show a distinct mutational spectrum: a 27.1% increase in transversions that convert cytosine to guanine or the reverse across all triplet contexts and a 7.6% reduction in the frequency of CpG-associated mutations when compared to unique DNA. We reason that these distinct mutational properties help to maintain an overall higher GC content of SD DNA compared to that of unique DNA, probably driven by GC-biased conversion between paralogous sequences5,6.


Asunto(s)
Conversión Génica , Mutación , Duplicaciones Segmentarias en el Genoma , Humanos , Conversión Génica/genética , Genoma Humano/genética , Polimorfismo de Nucleótido Simple/genética , Haplotipos/genética , Exones/genética , Citosina/química , Guanina/química , Islas de CpG/genética
13.
Nature ; 621(7978): 355-364, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37612510

RESUMEN

The prevalence of highly repetitive sequences within the human Y chromosome has prevented its complete assembly to date1 and led to its systematic omission from genomic analyses. Here we present de novo assemblies of 43 Y chromosomes spanning 182,900 years of human evolution and report considerable diversity in size and structure. Half of the male-specific euchromatic region is subject to large inversions with a greater than twofold higher recurrence rate compared with all other chromosomes2. Ampliconic sequences associated with these inversions show differing mutation rates that are sequence context dependent, and some ampliconic genes exhibit evidence for concerted evolution with the acquisition and purging of lineage-specific pseudogenes. The largest heterochromatic region in the human genome, Yq12, is composed of alternating repeat arrays that show extensive variation in the number, size and distribution, but retain a 1:1 copy-number ratio. Finally, our data suggest that the boundary between the recombining pseudoautosomal region 1 and the non-recombining portions of the X and Y chromosomes lies 500 kb away from the currently established1 boundary. The availability of fully sequence-resolved Y chromosomes from multiple individuals provides a unique opportunity for identifying new associations of traits with specific Y-chromosomal variants and garnering insights into the evolution and function of complex regions of the human genome.


Asunto(s)
Cromosomas Humanos Y , Evolución Molecular , Humanos , Masculino , Cromosomas Humanos Y/genética , Genoma Humano/genética , Genómica , Tasa de Mutación , Fenotipo , Eucromatina/genética , Seudogenes , Variación Genética/genética , Cromosomas Humanos X/genética , Regiones Pseudoautosómicas/genética
14.
Nature ; 604(7906): 437-446, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35444317

RESUMEN

The human reference genome is the most widely used resource in human genetics and is due for a major update. Its current structure is a linear composite of merged haplotypes from more than 20 people, with a single individual comprising most of the sequence. It contains biases and errors within a framework that does not represent global human genomic variation. A high-quality reference with global representation of common variants, including single-nucleotide variants, structural variants and functional elements, is needed. The Human Pangenome Reference Consortium aims to create a more sophisticated and complete human reference genome with a graph-based, telomere-to-telomere representation of global genomic diversity. Here we leverage innovations in technology, study design and global partnerships with the goal of constructing the highest-possible quality human pangenome reference. Our goal is to improve data representation and streamline analyses to enable routine assembly of complete diploid genomes. With attention to ethical frameworks, the human pangenome reference will contain a more accurate and diverse representation of global genomic variation, improve gene-disease association studies across populations, expand the scope of genomics research to the most repetitive and polymorphic regions of the genome, and serve as the ultimate genetic resource for future biomedical research and precision medicine.


Asunto(s)
Genoma Humano , Genómica , Genoma Humano/genética , Haplotipos/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Análisis de Secuencia de ADN
15.
Am J Hum Genet ; 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38991590

RESUMEN

The secreted mucins MUC5AC and MUC5B are large glycoproteins that play critical defensive roles in pathogen entrapment and mucociliary clearance. Their respective genes contain polymorphic and degenerate protein-coding variable number tandem repeats (VNTRs) that make the loci difficult to investigate with short reads. We characterize the structural diversity of MUC5AC and MUC5B by long-read sequencing and assembly of 206 human and 20 nonhuman primate (NHP) haplotypes. We find that human MUC5B is largely invariant (5,761-5,762 amino acids [aa]); however, seven haplotypes have expanded VNTRs (6,291-7,019 aa). In contrast, 30 allelic variants of MUC5AC encode 16 distinct proteins (5,249-6,325 aa) with cysteine-rich domain and VNTR copy-number variation. We group MUC5AC alleles into three phylogenetic clades: H1 (46%, ∼5,654 aa), H2 (33%, ∼5,742 aa), and H3 (7%, ∼6,325 aa). The two most common human MUC5AC variants are smaller than NHP gene models, suggesting a reduction in protein length during recent human evolution. Linkage disequilibrium and Tajima's D analyses reveal that East Asians carry exceptionally large blocks with an excess of rare variation (p < 0.05) at MUC5AC. To validate this result, we use Locityper for genotyping MUC5AC haplogroups in 2,600 unrelated samples from the 1000 Genomes Project. We observe a signature of positive selection in H1 among East Asians and a depletion of the likely ancestral haplogroup (H3). In Europeans, H3 alleles show an excess of common variation and deviate from Hardy-Weinberg equilibrium (p < 0.05), consistent with heterozygote advantage and balancing selection. This study provides a generalizable strategy to characterize complex protein-coding VNTRs for improved disease associations.

16.
Genome Res ; 34(3): 454-468, 2024 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-38627094

RESUMEN

Reference-free genome phasing is vital for understanding allele inheritance and the impact of single-molecule DNA variation on phenotypes. To achieve thorough phasing across homozygous or repetitive regions of the genome, long-read sequencing technologies are often used to perform phased de novo assembly. As a step toward reducing the cost and complexity of this type of analysis, we describe new methods for accurately phasing Oxford Nanopore Technologies (ONT) sequence data with the Shasta genome assembler and a modular tool for extending phasing to the chromosome scale called GFAse. We test using new variants of ONT PromethION sequencing, including those using proximity ligation, and show that newer, higher accuracy ONT reads substantially improve assembly quality.


Asunto(s)
Nanoporos , Humanos , Análisis de Secuencia de ADN/métodos , Secuenciación de Nanoporos/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Programas Informáticos , Genómica/métodos
17.
Cell ; 149(4): 912-22, 2012 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-22559943

RESUMEN

Gene duplication is an important source of phenotypic change and adaptive evolution. We leverage a haploid hydatidiform mole to identify highly identical sequences missing from the reference genome, confirming that the cortical development gene Slit-Robo Rho GTPase-activating protein 2 (SRGAP2) duplicated three times exclusively in humans. We show that the promoter and first nine exons of SRGAP2 duplicated from 1q32.1 (SRGAP2A) to 1q21.1 (SRGAP2B) ∼3.4 million years ago (mya). Two larger duplications later copied SRGAP2B to chromosome 1p12 (SRGAP2C) and to proximal 1q21.1 (SRGAP2D) ∼2.4 and ∼1 mya, respectively. Sequence and expression analyses show that SRGAP2C is the most likely duplicate to encode a functional protein and is among the most fixed human-specific duplicate genes. Our data suggest a mechanism where incomplete duplication created a novel gene function-antagonizing parental SRGAP2 function-immediately "at birth" 2-3 mya, which is a time corresponding to the transition from Australopithecus to Homo and the beginning of neocortex expansion.


Asunto(s)
Evolución Molecular , Proteínas Activadoras de GTPasa/genética , Primates/genética , Duplicaciones Segmentarias en el Genoma , Animales , Variaciones en el Número de Copia de ADN , Femenino , Genética Médica , Humanos , Mola Hidatiforme/genética , Hibridación Fluorescente in Situ , Mamíferos/genética , Datos de Secuencia Molecular , Embarazo
18.
Nature ; 598(7879): 205-213, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34616060

RESUMEN

During mammalian development, differences in chromatin state coincide with cellular differentiation and reflect changes in the gene regulatory landscape1. In the developing brain, cell fate specification and topographic identity are important for defining cell identity2 and confer selective vulnerabilities to neurodevelopmental disorders3. Here, to identify cell-type-specific chromatin accessibility patterns in the developing human brain, we used a single-cell assay for transposase accessibility by sequencing (scATAC-seq) in primary tissue samples from the human forebrain. We applied unbiased analyses to identify genomic loci that undergo extensive cell-type- and brain-region-specific changes in accessibility during neurogenesis, and an integrative analysis to predict cell-type-specific candidate regulatory elements. We found that cerebral organoids recapitulate most putative cell-type-specific enhancer accessibility patterns but lack many cell-type-specific open chromatin regions that are found in vivo. Systematic comparison of chromatin accessibility across brain regions revealed unexpected diversity among neural progenitor cells in the cerebral cortex and implicated retinoic acid signalling in the specification of neuronal lineage identity in the prefrontal cortex. Together, our results reveal the important contribution of chromatin state to the emerging patterns of cell type diversity and cell fate specification and provide a blueprint for evaluating the fidelity and robustness of cerebral organoids as a model for cortical development.


Asunto(s)
Encéfalo/citología , Epigenómica , Neurogénesis , Análisis de la Célula Individual , Atlas como Asunto , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Cromatina/química , Cromatina/genética , Cromatina/metabolismo , Susceptibilidad a Enfermedades , Elementos de Facilitación Genéticos , Humanos , Neuronas/citología , Neuronas/metabolismo , Organoides/citología , Tretinoina/metabolismo
19.
Nature ; 593(7857): 101-107, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33828295

RESUMEN

The complete assembly of each human chromosome is essential for understanding human biology and evolution1,2. Here we use complementary long-read sequencing technologies to complete the linear assembly of human chromosome 8. Our assembly resolves the sequence of five previously long-standing gaps, including a 2.08-Mb centromeric α-satellite array, a 644-kb copy number polymorphism in the ß-defensin gene cluster that is important for disease risk, and an 863-kb variable number tandem repeat at chromosome 8q21.2 that can function as a neocentromere. We show that the centromeric α-satellite array is generally methylated except for a 73-kb hypomethylated region of diverse higher-order α-satellites enriched with CENP-A nucleosomes, consistent with the location of the kinetochore. In addition, we confirm the overall organization and methylation pattern of the centromere in a diploid human genome. Using a dual long-read sequencing approach, we complete high-quality draft assemblies of the orthologous centromere from chromosome 8 in chimpanzee, orangutan and macaque to reconstruct its evolutionary history. Comparative and phylogenetic analyses show that the higher-order α-satellite structure evolved in the great ape ancestor with a layered symmetry, in which more ancient higher-order repeats locate peripherally to monomeric α-satellites. We estimate that the mutation rate of centromeric satellite DNA is accelerated by more than 2.2-fold compared to the unique portions of the genome, and this acceleration extends into the flanking sequence.


Asunto(s)
Cromosomas Humanos Par 8/química , Cromosomas Humanos Par 8/genética , Evolución Molecular , Animales , Línea Celular , Centrómero/química , Centrómero/genética , Centrómero/metabolismo , Cromosomas Humanos Par 8/fisiología , Metilación de ADN , ADN Satélite/genética , Epigénesis Genética , Femenino , Humanos , Macaca mulatta/genética , Masculino , Repeticiones de Minisatélite/genética , Pan troglodytes/genética , Filogenia , Pongo abelii/genética , Telómero/química , Telómero/genética , Telómero/metabolismo
20.
Nature ; 594(7861): 77-81, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33953399

RESUMEN

The divergence of chimpanzee and bonobo provides one of the few examples of recent hominid speciation1,2. Here we describe a fully annotated, high-quality bonobo genome assembly, which was constructed without guidance from reference genomes by applying a multiplatform genomics approach. We generate a bonobo genome assembly in which more than 98% of genes are completely annotated and 99% of the gaps are closed, including the resolution of about half of the segmental duplications and almost all of the full-length mobile elements. We compare the bonobo genome to those of other great apes1,3-5 and identify more than 5,569 fixed structural variants that specifically distinguish the bonobo and chimpanzee lineages. We focus on genes that have been lost, changed in structure or expanded in the last few million years of bonobo evolution. We produce a high-resolution map of incomplete lineage sorting and estimate that around 5.1% of the human genome is genetically closer to chimpanzee or bonobo and that more than 36.5% of the genome shows incomplete lineage sorting if we consider a deeper phylogeny including gorilla and orangutan. We also show that 26% of the segments of incomplete lineage sorting between human and chimpanzee or human and bonobo are non-randomly distributed and that genes within these clustered segments show significant excess of amino acid replacement compared to the rest of the genome.


Asunto(s)
Evolución Molecular , Genoma/genética , Genómica , Pan paniscus/genética , Filogenia , Animales , Factor 4A Eucariótico de Iniciación/genética , Femenino , Genes , Gorilla gorilla/genética , Anotación de Secuencia Molecular/normas , Pan troglodytes/genética , Pongo/genética , Duplicaciones Segmentarias en el Genoma , Análisis de Secuencia de ADN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA