Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
2.
Nat Neurosci ; 26(9): 1505-1515, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37563294

RESUMEN

Idiopathic autism spectrum disorder (ASD) is highly heterogeneous, and it remains unclear how convergent biological processes in affected individuals may give rise to symptoms. Here, using cortical organoids and single-cell transcriptomics, we modeled alterations in the forebrain development between boys with idiopathic ASD and their unaffected fathers in 13 families. Transcriptomic changes suggest that ASD pathogenesis in macrocephalic and normocephalic probands involves an opposite disruption of the balance between excitatory neurons of the dorsal cortical plate and other lineages such as early-generated neurons from the putative preplate. The imbalance stemmed from divergent expression of transcription factors driving cell fate during early cortical development. While we did not find genomic variants in probands that explained the observed transcriptomic alterations, a significant overlap between altered transcripts and reported ASD risk genes affected by rare variants suggests a degree of gene convergence between rare forms of ASD and the developmental transcriptome in idiopathic ASD.


Asunto(s)
Trastorno del Espectro Autista , Trastorno Autístico , Masculino , Humanos , Trastorno Autístico/genética , Trastorno del Espectro Autista/patología , Neuronas/metabolismo , Neurogénesis , Prosencéfalo/metabolismo , Organoides/metabolismo
3.
Nucleic Acids Res ; 51(10): e57, 2023 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-37026484

RESUMEN

Mosaic mutations can be used to track cell ancestries and reconstruct high-resolution lineage trees during cancer progression and during development, starting from the first cell divisions of the zygote. However, this approach requires sampling and analyzing the genomes of multiple cells, which can be redundant in lineage representation, limiting the scalability of the approach. We describe a strategy for cost- and time-efficient lineage reconstruction using clonal induced pluripotent stem cell lines from human skin fibroblasts. The approach leverages shallow sequencing coverage to assess the clonality of the lines, clusters redundant lines and sums their coverage to accurately discover mutations in the corresponding lineages. Only a fraction of lines needs to be sequenced to high coverage. We demonstrate the effectiveness of this approach for reconstructing lineage trees during development and in hematologic malignancies. We discuss and propose an optimal experimental design for reconstructing lineage trees.


Asunto(s)
Linaje de la Célula , Neoplasias , Programas Informáticos , Humanos , Células Germinativas , Mutación , Neoplasias/patología
5.
PLoS Comput Biol ; 18(4): e1009487, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35442945

RESUMEN

Accurate discovery of somatic mutations in a cell is a challenge that partially lays in immaturity of dedicated analytical approaches. Approaches comparing a cell's genome to a control bulk sample miss common mutations, while approaches to find such mutations from bulk suffer from low sensitivity. We developed a tool, All2, which enables accurate filtering of mutations in a cell without the need for data from bulk(s). It is based on pair-wise comparisons of all cells to each other where every call for base pair substitution and indel is classified as either a germline variant, mosaic mutation, or false positive. As All2 allows for considering dropped-out regions, it is applicable to whole genome and exome analysis of cloned and amplified cells. By applying the approach to a variety of available data, we showed that its application reduces false positives, enables sensitive discovery of high frequency mutations, and is indispensable for conducting high resolution cell lineage tracing.


Asunto(s)
Exoma , Programas Informáticos , Secuenciación de Nucleótidos de Alto Rendimiento , Mutación INDEL/genética , Mutación/genética , Secuenciación del Exoma
6.
Science ; 371(6535): 1245-1248, 2021 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-33737484

RESUMEN

Mosaic mutations can be used to track cell lineages in humans. We used cell cloning to analyze embryonic cell lineages in two living individuals and a postmortem human specimen. Of 10 reconstructed postzygotic divisions, none resulted in balanced contributions of daughter lineages to tissues. In both living individuals, one of two lineages from the first cleavage was dominant across tissues, with 90% frequency in blood. We propose that the efficiency of DNA repair contributes to lineage imbalance. Allocation of lineages in postmortem brain correlated with anterior-posterior axis, associating lineage history with cell fate choices in embryos. We establish a minimally invasive framework for defining cell lineages in any living individual, which paves the way for studying their relevance in health and disease.


Asunto(s)
Blastómeros/citología , División Celular , Linaje de la Célula , Desarrollo Embrionario , Adulto , Anciano , Blastocisto/citología , Células Sanguíneas , Diferenciación Celular , Línea Celular , Reparación del ADN , Femenino , Feto/citología , Variación Genética , Genoma Humano , Humanos , Mutación INDEL , Células Madre Pluripotentes Inducidas/citología , Masculino , Células-Madre Neurales/citología , Polimorfismo de Nucleótido Simple
7.
Genome Biol ; 22(1): 92, 2021 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-33781308

RESUMEN

BACKGROUND: Post-zygotic mutations incurred during DNA replication, DNA repair, and other cellular processes lead to somatic mosaicism. Somatic mosaicism is an established cause of various diseases, including cancers. However, detecting mosaic variants in DNA from non-cancerous somatic tissues poses significant challenges, particularly if the variants only are present in a small fraction of cells. RESULTS: Here, the Brain Somatic Mosaicism Network conducts a coordinated, multi-institutional study to examine the ability of existing methods to detect simulated somatic single-nucleotide variants (SNVs) in DNA mixing experiments, generate multiple replicates of whole-genome sequencing data from the dorsolateral prefrontal cortex, other brain regions, dura mater, and dural fibroblasts of a single neurotypical individual, devise strategies to discover somatic SNVs, and apply various approaches to validate somatic SNVs. These efforts lead to the identification of 43 bona fide somatic SNVs that range in variant allele fractions from ~ 0.005 to ~ 0.28. Guided by these results, we devise best practices for calling mosaic SNVs from 250× whole-genome sequencing data in the accessible portion of the human genome that achieve 90% specificity and sensitivity. Finally, we demonstrate that analysis of multiple bulk DNA samples from a single individual allows the reconstruction of early developmental cell lineage trees. CONCLUSIONS: This study provides a unified set of best practices to detect somatic SNVs in non-cancerous tissues. The data and methods are freely available to the scientific community and should serve as a guide to assess the contributions of somatic SNVs to neuropsychiatric diseases.


Asunto(s)
Encéfalo/metabolismo , Estudios de Asociación Genética , Variación Genética , Alelos , Mapeo Cromosómico , Biología Computacional/métodos , Estudios de Asociación Genética/métodos , Genómica/métodos , Células Germinativas/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Especificidad de Órganos/genética , Polimorfismo de Nucleótido Simple
8.
Genome Res ; 30(12): 1695-1704, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33122304

RESUMEN

Somatic mosaicism, manifesting as single nucleotide variants (SNVs), mobile element insertions, and structural changes in the DNA, is a common phenomenon in human brain cells, with potential functional consequences. Using a clonal approach, we previously detected 200-400 mosaic SNVs per cell in three human fetal brains (15-21 wk postconception). However, structural variation in the human fetal brain has not yet been investigated. Here, we discover and validate four mosaic structural variants (SVs) in the same brains and resolve their precise breakpoints. The SVs were of kilobase scale and complex, consisting of deletion(s) and rearranged genomic fragments, which sometimes originated from different chromosomes. Sequences at the breakpoints of these rearrangements had microhomologies, suggesting their origin from replication errors. One SV was found in two clones, and we timed its origin to ∼14 wk postconception. No large scale mosaic copy number variants (CNVs) were detectable in normal fetal human brains, suggesting that previously reported megabase-scale CNVs in neurons arise at later stages of development. By reanalysis of public single nuclei data from adult brain neurons, we detected an extrachromosomal circular DNA event. Our study reveals the existence of mosaic SVs in the developing human brain, likely arising from cell proliferation during mid-neurogenesis. Although relatively rare compared to SNVs and present in ∼10% of neurons, SVs in developing human brain affect a comparable number of bases in the genome (∼6200 vs. ∼4000 bp), implying that they may have similar functional consequences.


Asunto(s)
Encéfalo/embriología , ADN Circular/genética , Variación Estructural del Genoma , Análisis de Secuencia de ADN/métodos , Evolución Clonal , Femenino , Técnicas de Genotipaje , Edad Gestacional , Humanos , Mosaicismo , Neurogénesis , Embarazo
9.
Science ; 359(6375): 550-555, 2018 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-29217587

RESUMEN

Somatic mosaicism in the human brain may alter function of individual neurons. We analyzed genomes of single cells from the forebrains of three human fetuses (15 to 21 weeks postconception) using clonal cell populations. We detected 200 to 400 single-nucleotide variations (SNVs) per cell. SNV patterns resembled those found in cancer cell genomes, indicating a role of background mutagenesis in cancer. SNVs with a frequency of >2% in brain were also present in the spleen, revealing a pregastrulation origin. We reconstructed cell lineages for the first five postzygotic cleavages and calculated a mutation rate of ~1.3 mutations per division per cell. Later in development, during neurogenesis, the mutation spectrum shifted toward oxidative damage, and the mutation rate increased. Both neurogenesis and early embryogenesis exhibit substantially more mutagenesis than adulthood.


Asunto(s)
Encéfalo/embriología , Gastrulación/genética , Mosaicismo , Mutagénesis , Tasa de Mutación , Neurogénesis/genética , Linaje de la Célula/genética , Genoma Humano , Humanos , Mutación , Neoplasias/genética , Neuronas , Polimorfismo de Nucleótido Simple , Análisis de la Célula Individual
10.
Genome Res ; 27(4): 512-523, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28235832

RESUMEN

Few studies have been conducted to understand post-zygotic accumulation of mutations in cells of the healthy human body. We reprogrammed 32 skin fibroblast cells from families of donors into human induced pluripotent stem cell (hiPSC) lines. The clonal nature of hiPSC lines allows a high-resolution analysis of the genomes of the founder fibroblast cells without being confounded by the artifacts of single-cell whole-genome amplification. We estimate that on average a fibroblast cell in children has 1035 mostly benign mosaic SNVs. On average, 235 SNVs could be directly confirmed in the original fibroblast population by ultradeep sequencing, down to an allele frequency (AF) of 0.1%. More sensitive droplet digital PCR experiments confirmed more SNVs as mosaic with AF as low as 0.01%, suggesting that 1035 mosaic SNVs per fibroblast cell is the true average. Similar analyses in adults revealed no significant increase in the number of SNVs per cell, suggesting that a major fraction of mosaic SNVs in fibroblasts arises during development. Mosaic SNVs were distributed uniformly across the genome and were enriched in a mutational signature previously observed in cancers and in de novo variants and which, we hypothesize, is a hallmark of normal cell proliferation. Finally, AF distribution of mosaic SNVs had distinct narrow peaks, which could be a characteristic of clonal cell selection, clonal expansion, or both. These findings reveal a large degree of somatic mosaicism in healthy human tissues, link de novo and cancer mutations to somatic mosaicism, and couple somatic mosaicism with cell proliferation.


Asunto(s)
Evolución Clonal , Variaciones en el Número de Copia de ADN , Fibroblastos/citología , Mosaicismo , Acumulación de Mutaciones , Proliferación Celular , Células Cultivadas , Fibroblastos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Piel/citología
11.
Cell ; 162(2): 375-390, 2015 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-26186191

RESUMEN

Autism spectrum disorder (ASD) is a disorder of brain development. Most cases lack a clear etiology or genetic basis, and the difficulty of re-enacting human brain development has precluded understanding of ASD pathophysiology. Here we use three-dimensional neural cultures (organoids) derived from induced pluripotent stem cells (iPSCs) to investigate neurodevelopmental alterations in individuals with severe idiopathic ASD. While no known underlying genomic mutation could be identified, transcriptome and gene network analyses revealed upregulation of genes involved in cell proliferation, neuronal differentiation, and synaptic assembly. ASD-derived organoids exhibit an accelerated cell cycle and overproduction of GABAergic inhibitory neurons. Using RNA interference, we show that overexpression of the transcription factor FOXG1 is responsible for the overproduction of GABAergic neurons. Altered expression of gene network modules and FOXG1 are positively correlated with symptom severity. Our data suggest that a shift toward GABAergic neuron fate caused by FOXG1 is a developmental precursor of ASD.


Asunto(s)
Trastornos Generalizados del Desarrollo Infantil/genética , Trastornos Generalizados del Desarrollo Infantil/patología , Factores de Transcripción Forkhead/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis , Telencéfalo/embriología , Femenino , Perfilación de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas , Masculino , Megalencefalia/genética , Megalencefalia/patología , Modelos Biológicos , Neuronas/citología , Neuronas/metabolismo , Organoides/patología , Telencéfalo/patología
12.
Nature ; 492(7429): 438-42, 2012 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-23160490

RESUMEN

Reprogramming somatic cells into induced pluripotent stem cells (iPSCs) has been suspected of causing de novo copy number variation. To explore this issue, here we perform a whole-genome and transcriptome analysis of 20 human iPSC lines derived from the primary skin fibroblasts of seven individuals using next-generation sequencing. We find that, on average, an iPSC line manifests two copy number variants (CNVs) not apparent in the fibroblasts from which the iPSC was derived. Using PCR and digital droplet PCR, we show that at least 50% of those CNVs are present as low-frequency somatic genomic variants in parental fibroblasts (that is, the fibroblasts from which each corresponding human iPSC line is derived), and are manifested in iPSC lines owing to their clonal origin. Hence, reprogramming does not necessarily lead to de novo CNVs in iPSCs, because most of the line-manifested CNVs reflect somatic mosaicism in the human skin. Moreover, our findings demonstrate that clonal expansion, and iPSC lines in particular, can be used as a discovery tool to reliably detect low-frequency CNVs in the tissue of origin. Overall, we estimate that approximately 30% of the fibroblast cells have somatic CNVs in their genomes, suggesting widespread somatic mosaicism in the human body. Our study paves the way to understanding the fundamental question of the extent to which cells of the human body normally acquire structural alterations in their DNA post-zygotically.


Asunto(s)
Variaciones en el Número de Copia de ADN/genética , Células Madre Pluripotentes Inducidas/metabolismo , Mosaicismo , Piel/metabolismo , Diferenciación Celular , Células Cultivadas , Reprogramación Celular , Células Clonales , Fibroblastos/citología , Perfilación de la Expresión Génica , Genoma Humano/genética , Humanos , Células Madre Pluripotentes Inducidas/citología , Masculino , Neuronas/citología , Reacción en Cadena de la Polimerasa , Reproducibilidad de los Resultados , Piel/citología
13.
Proc Natl Acad Sci U S A ; 109(31): 12770-5, 2012 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-22761314

RESUMEN

Human induced pluripotent stem cells (hiPSCs) are emerging as a tool for understanding human brain development at cellular, molecular, and genomic levels. Here we show that hiPSCs grown in suspension in the presence of rostral neuralizing factors can generate 3D structures containing polarized radial glia, intermediate progenitors, and a spectrum of layer-specific cortical neurons reminiscent of their organization in vivo. The hiPSC-derived multilayered structures express a gene expression profile typical of the embryonic telencephalon but not that of other CNS regions. Their transcriptome is highly enriched in transcription factors controlling the specification, growth, and patterning of the dorsal telencephalon and displays highest correlation with that of the early human cerebral cortical wall at 8-10 wk after conception. Thus, hiPSC are capable of enacting a transcriptional program specifying human telencephalic (pallial) development. This model will allow the study of human brain development as well as disorders of the human cerebral cortex.


Asunto(s)
Corteza Cerebral , Células Madre Pluripotentes Inducidas , Modelos Biológicos , Neuronas , Transcriptoma/fisiología , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/embriología , Perfilación de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Neuronas/metabolismo , Factores de Transcripción/metabolismo
14.
Proc Natl Acad Sci U S A ; 107(8): 3511-6, 2010 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-20133602

RESUMEN

Mutations of the inositol 5' phosphatase oculocerebrorenal syndrome of Lowe (OCRL) give rise to the congenital X-linked disorders oculocerebrorenal syndrome of Lowe and Dent disease, two conditions giving rise to abnormal kidney proximal tubule reabsorption, and additional nervous system and ocular defects in the case of Lowe syndrome. Here, we identify two closely related endocytic proteins, Ses1 and Ses2, which interact with the ASH-RhoGAP-like (ASPM-SPD-2-Hydin homology and Rho-GTPase Activating Domain-like) domain of OCRL. The interaction is mediated by a short amino acid motif similar to that used by the rab-5 effector APPL1 (Adaptor Protein containing pleckstrin homology [PH] domain, PTB domain and Leucine zipper motif 1) APPL1 for OCRL binding. Ses binding is mutually exclusive with APPL1 binding, and is disrupted by the same missense mutations in the ASH-RhoGAP-like domain that also disrupt APPL1 binding. Like APPL1, Ses1 and -2 are localized on endosomes but reside on different endosomal subpopulations. These findings define a consensus motif (which we have called a phenylalanine and histidine [F&H] motif) for OCRL binding and are consistent with a scenario in which Lowe syndrome and Dent disease result from perturbations at multiple sites within the endocytic pathway.


Asunto(s)
Proteínas Portadoras/metabolismo , Endosomas/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Secuencia Conservada , Endocitosis , Humanos , Datos de Secuencia Molecular , Mutación Missense , Síndrome Oculocerebrorrenal/genética , Síndrome Oculocerebrorrenal/metabolismo , Monoéster Fosfórico Hidrolasas/genética , Estructura Terciaria de Proteína , Proteínas de Transporte Vesicular/genética
15.
EMBO J ; 28(13): 1831-42, 2009 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-19536138

RESUMEN

OCRL, whose mutations are responsible for Lowe syndrome and Dent disease, and INPP5B are two similar proteins comprising a central inositol 5-phosphatase domain followed by an ASH and a RhoGAP-like domain. Their divergent NH2-terminal portions remain uncharacterized. We show that the NH2-terminal region of OCRL, but not of INPP5B, binds clathrin heavy chain. OCRL, which in contrast to INPP5B visits late stage endocytic clathrin-coated pits, was earlier shown to contain another binding site for clathrin in its COOH-terminal region. NMR structure determination further reveals that despite their primary sequence dissimilarity, the NH2-terminal portions of both OCRL and INPP5B contain a PH domain. The novel clathrin-binding site in OCRL maps to an unusual clathrin-box motif located in a loop of the PH domain, whose mutations reduce recruitment efficiency of OCRL to coated pits. These findings suggest an evolutionary pressure for a specialized function of OCRL in bridging phosphoinositide metabolism to clathrin-dependent membrane trafficking.


Asunto(s)
Clatrina/metabolismo , Monoéster Fosfórico Hidrolasas/química , Monoéster Fosfórico Hidrolasas/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Clatrina/química , Vesículas Cubiertas/metabolismo , Vesículas Cubiertas/ultraestructura , Endocitosis , Células HeLa , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Resonancia Magnética Nuclear Biomolecular , Fosfatidilinositoles/metabolismo , Fosfolípidos , Monoéster Fosfórico Hidrolasas/genética , Conformación Proteica , Estructura Terciaria de Proteína , Ratas , Alineación de Secuencia
16.
Biochem Biophys Res Commun ; 369(2): 493-9, 2008 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-18307981

RESUMEN

Mutations in the inositol 5-phosphatase OCRL are responsible for Lowe syndrome, an X-linked disorder characterized by bilateral cataracts, mental retardation, neonatal hypotonia, and renal Fanconi syndrome, and for Dent disease, another X-linked condition characterized by kidney reabsorption defects. We have previously described an interaction of OCRL with the endocytic adaptor APPL1 that links OCRL to protein networks involved in the disease phenotype. Here, we provide new evidence showing that among the interactions which target OCRL to membranes of the endocytic pathway, binding to APPL1 is the only one abolished by all known disease-causing missense mutations in the ASH-RhoGAP domains of the protein. Furthermore, we demonstrate that APPL1 and rab5 independently contribute to recruit OCRL to enlarged endosomes induced by the expression of constitutively active Rab5. Thus, binding to APPL1 helps localize OCRL at specific cellular sites, and disruption of this interaction may play a role in disease.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteína Adaptadora GRB2/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Síndrome Oculocerebrorrenal/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Sitios de Unión , Células COS , Proteínas Portadoras/química , Proteínas Portadoras/genética , Chlorocebus aethiops , Proteína Adaptadora GRB2/química , Proteína Adaptadora GRB2/genética , Proteínas Activadoras de GTPasa/química , Proteínas Activadoras de GTPasa/genética , Humanos , Mutación , Síndrome Oculocerebrorrenal/genética , Monoéster Fosfórico Hidrolasas/química , Monoéster Fosfórico Hidrolasas/genética , Unión Proteica , Estructura Terciaria de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA