Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
Cell ; 186(26): 5766-5783.e25, 2023 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-38134874

RESUMEN

The enhanced cognitive abilities characterizing the human species result from specialized features of neurons and circuits. Here, we report that the hominid-specific gene LRRC37B encodes a receptor expressed in human cortical pyramidal neurons (CPNs) and selectively localized to the axon initial segment (AIS), the subcellular compartment triggering action potentials. Ectopic expression of LRRC37B in mouse CPNs in vivo leads to reduced intrinsic excitability, a distinctive feature of some classes of human CPNs. Molecularly, LRRC37B binds to the secreted ligand FGF13A and to the voltage-gated sodium channel (Nav) ß-subunit SCN1B. LRRC37B concentrates inhibitory effects of FGF13A on Nav channel function, thereby reducing excitability, specifically at the AIS level. Electrophysiological recordings in adult human cortical slices reveal lower neuronal excitability in human CPNs expressing LRRC37B. LRRC37B thus acts as a species-specific modifier of human neuron excitability, linking human genome and cell evolution, with important implications for human brain function and diseases.


Asunto(s)
Neuronas , Células Piramidales , Canales de Sodio Activados por Voltaje , Animales , Humanos , Ratones , Potenciales de Acción/fisiología , Axones/metabolismo , Neuronas/metabolismo , Canales de Sodio Activados por Voltaje/genética , Canales de Sodio Activados por Voltaje/metabolismo
2.
Cell ; 185(26): 4869-4872, 2022 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-36563661

RESUMEN

Despite its importance to understanding human brain (dys)function, it has remained challenging to study human neurons in vivo. Recent approaches, using transplantation of human cortical neurons into the rodent brain, offer new prospects for the study of human neural function and disease in vivo, from molecular to circuit levels.


Asunto(s)
Encéfalo , Neuronas , Humanos , Neuronas/fisiología , Encéfalo/fisiología , Células Madre
3.
Cell ; 173(6): 1370-1384.e16, 2018 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-29856955

RESUMEN

The cerebral cortex underwent rapid expansion and increased complexity during recent hominid evolution. Gene duplications constitute a major evolutionary force, but their impact on human brain development remains unclear. Using tailored RNA sequencing (RNA-seq), we profiled the spatial and temporal expression of hominid-specific duplicated (HS) genes in the human fetal cortex and identified a repertoire of 35 HS genes displaying robust and dynamic patterns during cortical neurogenesis. Among them NOTCH2NL, human-specific paralogs of the NOTCH2 receptor, stood out for their ability to promote cortical progenitor maintenance. NOTCH2NL promote the clonal expansion of human cortical progenitors, ultimately leading to higher neuronal output. At the molecular level, NOTCH2NL function by activating the Notch pathway through inhibition of cis Delta/Notch interactions. Our study uncovers a large repertoire of recently evolved genes active during human corticogenesis and reveals how human-specific NOTCH paralogs may have contributed to the expansion of the human cortex.


Asunto(s)
Corteza Cerebral/metabolismo , Regulación de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/metabolismo , Neurogénesis , Neuronas/metabolismo , Receptor Notch2/genética , Secuencia de Aminoácidos , Proteínas de Unión al Calcio , Diferenciación Celular/genética , Análisis por Conglomerados , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Hibridación in Situ , Células-Madre Neurales/metabolismo , Transducción de Señal
4.
Cell ; 164(3): 460-75, 2016 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-26824657

RESUMEN

Neurogenesis is initiated by the transient expression of the highly conserved proneural proteins, bHLH transcriptional regulators. Here, we discover a conserved post-translational switch governing the duration of proneural protein activity that is required for proper neuronal development. Phosphorylation of a single Serine at the same position in Scute and Atonal proneural proteins governs the transition from active to inactive forms by regulating DNA binding. The equivalent Neurogenin2 Threonine also regulates DNA binding and proneural activity in the developing mammalian neocortex. Using genome editing in Drosophila, we show that Atonal outlives its mRNA but is inactivated by phosphorylation. Inhibiting the phosphorylation of the conserved proneural Serine causes quantitative changes in expression dynamics and target gene expression resulting in neuronal number and fate defects. Strikingly, even a subtle change from Serine to Threonine appears to shift the duration of Atonal activity in vivo, resulting in neuronal fate defects.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/química , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Neurogénesis , Secuencia de Aminoácidos , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Drosophila , Proteínas de Drosophila , Ojo/crecimiento & desarrollo , Ojo/ultraestructura , Discos Imaginales/metabolismo , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/metabolismo , Fosforilación , Retina/crecimiento & desarrollo , Alineación de Secuencia
5.
Annu Rev Genet ; 55: 555-581, 2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34535062

RESUMEN

The cerebral cortex is at the core of brain functions that are thought to be particularly developed in the human species. Human cortex specificities stem from divergent features of corticogenesis, leading to increased cortical size and complexity. Underlying cellular mechanisms include prolonged patterns of neuronal generation and maturation, as well as the amplification of specific types of stem/progenitor cells. While the gene regulatory networks of corticogenesis appear to be largely conserved among all mammals including humans, they have evolved in primates, particularly in the human species, through the emergence of rapidly divergent transcriptional regulatory elements, as well as recently duplicated novel genes. These human-specific molecular features together control key cellular milestones of human corticogenesis and are often affected in neurodevelopmental disorders, thus linking human neural development, evolution, and diseases.


Asunto(s)
Corteza Cerebral , Neurogénesis , Animales , Corteza Cerebral/fisiología , Redes Reguladoras de Genes/genética , Humanos , Mamíferos , Neurogénesis/genética
6.
Nat Rev Neurosci ; 24(4): 213-232, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36792753

RESUMEN

The brain of modern humans has evolved remarkable computational abilities that enable higher cognitive functions. These capacities are tightly linked to an increase in the size and connectivity of the cerebral cortex, which is thought to have resulted from evolutionary changes in the mechanisms of cortical development. Convergent progress in evolutionary genomics, developmental biology and neuroscience has recently enabled the identification of genomic changes that act as human-specific modifiers of cortical development. These modifiers influence most aspects of corticogenesis, from the timing and complexity of cortical neurogenesis to synaptogenesis and the assembly of cortical circuits. Mutations of human-specific genetic modifiers of corticogenesis have started to be linked to neurodevelopmental disorders, providing evidence for their physiological relevance and suggesting potential relationships between the evolution of the human brain and its sensitivity to specific diseases.


Asunto(s)
Corteza Cerebral , Neurogénesis , Humanos , Corteza Cerebral/fisiología , Encéfalo
7.
Cell ; 149(4): 923-35, 2012 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-22559944

RESUMEN

Structural genomic variations represent a major driving force of evolution, and a burst of large segmental gene duplications occurred in the human lineage during its separation from nonhuman primates. SRGAP2, a gene recently implicated in neocortical development, has undergone two human-specific duplications. Here, we find that both duplications (SRGAP2B and SRGAP2C) are partial and encode a truncated F-BAR domain. SRGAP2C is expressed in the developing and adult human brain and dimerizes with ancestral SRGAP2 to inhibit its function. In the mouse neocortex, SRGAP2 promotes spine maturation and limits spine density. Expression of SRGAP2C phenocopies SRGAP2 deficiency. It underlies sustained radial migration and leads to the emergence of human-specific features, including neoteny during spine maturation and increased density of longer spines. These results suggest that inhibition of SRGAP2 function by its human-specific paralogs has contributed to the evolution of the human neocortex and plays an important role during human brain development.


Asunto(s)
Encéfalo/citología , Encéfalo/embriología , Proteínas Activadoras de GTPasa/genética , Duplicación de Gen , Neuronas/citología , Duplicaciones Segmentarias en el Genoma , Animales , Movimiento Celular , Espinas Dendríticas/metabolismo , Evolución Molecular , Humanos , Ratones , Datos de Secuencia Molecular , Neuronas/metabolismo , Estructura Terciaria de Proteína , Especificidad de la Especie
8.
Development ; 142(18): 3138-50, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26395142

RESUMEN

The human brain is arguably the most complex structure among living organisms. However, the specific mechanisms leading to this complexity remain incompletely understood, primarily because of the poor experimental accessibility of the human embryonic brain. Over recent years, technologies based on pluripotent stem cells (PSCs) have been developed to generate neural cells of various types. While the translational potential of PSC technologies for disease modeling and/or cell replacement therapies is usually put forward as a rationale for their utility, they are also opening novel windows for direct observation and experimentation of the basic mechanisms of human brain development. PSC-based studies have revealed that a number of cardinal features of neural ontogenesis are remarkably conserved in human models, which can be studied in a reductionist fashion. They have also revealed species-specific features, which constitute attractive lines of investigation to elucidate the mechanisms underlying the development of the human brain, and its link with evolution.


Asunto(s)
Evolución Biológica , Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , Inducción Embrionaria/fisiología , Modelos Neurológicos , Células Madre Pluripotentes/fisiología , Encéfalo/citología , Humanos , Neuritas/fisiología , Retina/fisiología , Especificidad de la Especie
9.
PLoS Genet ; 9(10): e1003888, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24204302

RESUMEN

We describe a new syndrome of young onset diabetes, short stature and microcephaly with intellectual disability in a large consanguineous family with three affected children. Linkage analysis and whole exome sequencing were used to identify the causal nonsense mutation, which changed an arginine codon into a stop at position 127 of the tRNA methyltransferase homolog gene TRMT10A (also called RG9MTD2). TRMT10A mRNA and protein were absent in lymphoblasts from the affected siblings. TRMT10A is ubiquitously expressed but enriched in brain and pancreatic islets, consistent with the tissues affected in this syndrome. In situ hybridization studies showed that TRMT10A is expressed in human embryonic and fetal brain. TRMT10A is the mammalian ortholog of S. cerevisiae TRM10, previously shown to catalyze the methylation of guanine 9 (m(1)G9) in several tRNAs. Consistent with this putative function, in silico topology prediction indicated that TRMT10A has predominant nuclear localization, which we experimentally confirmed by immunofluorescence and confocal microscopy. TRMT10A localizes to the nucleolus of ß- and non-ß-cells, where tRNA modifications occur. TRMT10A silencing induces rat and human ß-cell apoptosis. Taken together, we propose that TRMT10A deficiency negatively affects ß-cell mass and the pool of neurons in the developing brain. This is the first study describing the impact of TRMT10A deficiency in mammals, highlighting a role in the pathogenesis of microcephaly and early onset diabetes. In light of the recent report that the type 2 diabetes candidate gene CDKAL1 is a tRNA methylthiotransferase, the findings in this family suggest broader relevance of tRNA methyltransferases in the pathogenesis of type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Discapacidad Intelectual/genética , Metiltransferasas/genética , Microcefalia/genética , ARNt Metiltransferasas/genética , Adulto , Edad de Inicio , Animales , Apoptosis/genética , Diabetes Mellitus Tipo 2/complicaciones , Femenino , Ligamiento Genético , Humanos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Discapacidad Intelectual/complicaciones , Discapacidad Intelectual/patología , Masculino , Microcefalia/complicaciones , Microcefalia/patología , Mutación , Linaje , Ratas , Proteínas de Saccharomyces cerevisiae/genética , ARNt Metiltransferasas/deficiencia
10.
EMBO Rep ; 13(4): 355-62, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22402664

RESUMEN

The transcription factor Eomesodermin (Eomes) is involved in early embryonic patterning, but the range of cell fates that it controls as well as its mechanisms of action remain unclear. Here we show that transient expression of Eomes promotes cardiovascular fate during embryonic stem cell differentiation. Eomes also rapidly induces the expression of Mesp1, a key regulator of cardiovascular differentiation, and directly binds to regulatory sequences of Mesp1. Eomes effects are strikingly modulated by Activin signalling: high levels of Activin inhibit the promotion of cardiac mesoderm by Eomes, while they enhance Eomes-dependent endodermal specification. These results place Eomes upstream of the Mesp1-dependent programme of cardiogenesis, and at the intersection of mesodermal and endodermal specification, depending on the levels of Activin/Nodal signalling.


Asunto(s)
Activinas , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular/efectos de los fármacos , Células Madre Embrionarias/citología , Regulación de la Expresión Génica/efectos de los fármacos , Miocardio/citología , Proteínas de Dominio T Box/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/genética , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Organogénesis/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteínas de Dominio T Box/genética
11.
Nature ; 455(7211): 351-7, 2008 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-18716623

RESUMEN

The cerebral cortex develops through the coordinated generation of dozens of neuronal subtypes, but the mechanisms involved remain unclear. Here we show that mouse embryonic stem cells, cultured without any morphogen but in the presence of a sonic hedgehog inhibitor, recapitulate in vitro the major milestones of cortical development, leading to the sequential generation of a diverse repertoire of neurons that display most salient features of genuine cortical pyramidal neurons. When grafted into the cerebral cortex, these neurons develop patterns of axonal projections corresponding to a wide range of cortical layers, but also to highly specific cortical areas, in particular visual and limbic areas, thereby demonstrating that the identity of a cortical area can be specified without any influence from the brain. The discovery of intrinsic corticogenesis sheds new light on the mechanisms of neuronal specification, and opens new avenues for the modelling and treatment of brain diseases.


Asunto(s)
Diferenciación Celular , Corteza Cerebral/citología , Corteza Cerebral/embriología , Células Madre Embrionarias/citología , Animales , Axones/efectos de los fármacos , Axones/fisiología , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Células Madre Embrionarias/efectos de los fármacos , Ratones , Células Piramidales/efectos de los fármacos , Alcaloides de Veratrum/farmacología
12.
Dev Cell ; 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38906137

RESUMEN

Development consists of a highly ordered suite of steps and transitions, like choreography. Although these sequences are often evolutionarily conserved, they can display species variations in duration and speed, thereby modifying final organ size or function. Despite their evolutionary significance, the mechanisms underlying species-specific scaling of developmental tempo have remained unclear. Here, we will review recent findings that implicate global cellular mechanisms, particularly intermediary and protein metabolism, as species-specific modifiers of developmental tempo. In various systems, from somitic cell oscillations to neuronal development, metabolic pathways display species differences. These have been linked to mitochondrial metabolism, which can influence the species-specific speed of developmental transitions. Thus, intermediary metabolic pathways regulate developmental tempo together with other global processes, including proteostasis and chromatin remodeling. By linking metabolism and the evolution of developmental trajectories, these findings provide opportunities to decipher how species-specific cellular timing can influence organism fitness.

13.
Curr Opin Genet Dev ; 86: 102182, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38555796

RESUMEN

Changes in developmental timing are an important factor of evolution in organ shape and function. This is particularly striking for human brain development, which, compared with other mammals, is considerably prolonged at the level of the cerebral cortex, resulting in brain neoteny. Here, we review recent findings that indicate that mitochondria and metabolism contribute to species differences in the tempo of cortical neuron development. Mitochondria display species-specific developmental timeline and metabolic activity patterns that are highly correlated with the speed of neuron maturation. Enhancing mitochondrial activity in human cortical neurons results in their accelerated maturation, while its reduction leads to decreased maturation rates in mouse neurons. Together with other global and gene-specific mechanisms, mitochondria thus act as a cellular hourglass of neuronal developmental tempo and may thereby contribute to species-specific features of human brain ontogeny.


Asunto(s)
Evolución Biológica , Encéfalo , Mitocondrias , Neuronas , Humanos , Mitocondrias/metabolismo , Mitocondrias/genética , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Animales , Neuronas/metabolismo , Neuronas/citología , Especificidad de la Especie , Neurogénesis/genética , Ratones
14.
Cell Rep ; 43(1): 113576, 2024 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-38128530

RESUMEN

Neuronal activity-dependent transcription plays a key role in plasticity and pathology in the brain. An intriguing question is how neuronal activity controls gene expression via interactions of transcription factors with DNA and chromatin modifiers in the nucleus. By utilizing single-molecule imaging in human embryonic stem cell (ESC)-derived cortical neurons, we demonstrate that neuronal activity increases repetitive emergence of cAMP response element-binding protein (CREB) at histone acetylation sites in the nucleus, where RNA polymerase II (RNAPII) accumulation and FOS expression occur rapidly. Neuronal activity also enhances co-localization of CREB and CREB-binding protein (CBP). Increased binding of a constitutively active CREB to CBP efficiently induces CREB repetitive emergence. On the other hand, the formation of histone acetylation sites is dependent on CBP histone modification via acetyltransferase (HAT) activity but is not affected by neuronal activity. Taken together, our results suggest that neuronal activity promotes repetitive CREB-CRE and CREB-CBP interactions at predetermined histone acetylation sites, leading to rapid gene expression.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico , Histonas , Humanos , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Histonas/metabolismo , ADN/metabolismo , Proteína de Unión a CREB/genética , Proteína de Unión a CREB/metabolismo , Expresión Génica , Neuronas/metabolismo , Acetilación , Histona Acetiltransferasas/genética , Histona Acetiltransferasas/metabolismo
15.
Cereb Cortex ; 22(7): 1678-89, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21940705

RESUMEN

The patterning of cortical areas is controlled by a combination of intrinsic factors that are expressed in the cortex and external signals such as inputs from the thalamus. EphA7 is a guidance receptor that is involved in key aspects of cortical development and is expressed in gradients within developing cortical areas. Here, we identified a regulatory element of the EphA7 promoter, named pA7, that can recapitulate salient features of the pattern of expression of EphA7, including cortical gradients. Using a pA7-Green fluorescent Protein (GFP) mouse reporter line, we isolated cortical neuron populations displaying different levels of EphA7/GFP expression. Transcriptome analysis of these populations enabled to identify many differentially expressed genes, including 26 transcription factors with putative binding sites in the pA7 element. Among these, Pbx1 was found to bind directly to the EphA7 promoter in the developing cortex. All genes validated further were confirmed to be expressed differentially in the developing cortex, similarly to EphA7. Their expression was unchanged in mutant mice defective for thalamocortical projections, indicating a transcriptional control largely intrinsic to the cortex. Our study identifies a novel repertoire of cortical neuron genes that may act upstream of, or together with EphA7, to control the patterning of cortical areas.


Asunto(s)
Corteza Cerebral/embriología , Corteza Cerebral/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Receptor EphA7/metabolismo , Factores de Transcripción/metabolismo , Activación Transcripcional/fisiología , Transcriptoma/fisiología , Animales , Ratones
16.
Neuron ; 111(1): 65-80.e6, 2023 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-36334595

RESUMEN

The primary cilium is a central signaling component during embryonic development. Here we focus on CROCCP2, a hominid-specific gene duplicate from ciliary rootlet coiled coil (CROCC), also known as rootletin, that encodes the major component of the ciliary rootlet. We find that CROCCP2 is highly expressed in the human fetal brain and not in other primate species. CROCCP2 gain of function in the mouse embryonic cortex and human cortical cells and organoids results in decreased ciliogenesis and increased cortical progenitor amplification, particularly basal progenitors. CROCCP2 decreases ciliary dynamics by inhibition of the IFT20 ciliary trafficking protein, which then impacts neurogenesis through increased mTOR signaling. Loss of function of CROCCP2 in human cortical cells and organoids leads to increased ciliogenesis, decreased mTOR signaling, and impaired basal progenitor amplification. These data identify CROCCP2 as a human-specific modifier of cortical neurogenesis that acts through modulation of ciliary dynamics and mTOR signaling.


Asunto(s)
Cilios , Transducción de Señal , Animales , Humanos , Ratones , Cilios/metabolismo , Citoesqueleto/metabolismo , Neurogénesis , Serina-Treonina Quinasas TOR/metabolismo
17.
Neuron ; 111(6): 839-856.e5, 2023 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-36924763

RESUMEN

mRNA localization and local translation enable exquisite spatial and temporal control of gene expression, particularly in polarized, elongated cells. These features are especially prominent in radial glial cells (RGCs), which are neural and glial precursors of the developing cerebral cortex and scaffolds for migrating neurons. Yet the mechanisms by which subcellular RGC compartments accomplish their diverse functions are poorly understood. Here, we demonstrate that mRNA localization and local translation of the RhoGAP ARHGAP11A in the basal endfeet of RGCs control their morphology and mediate neuronal positioning. Arhgap11a transcript and protein exhibit conserved localization to RGC basal structures in mice and humans, conferred by the 5' UTR. Proper RGC morphology relies upon active Arhgap11a mRNA transport and localization to the basal endfeet, where ARHGAP11A is locally synthesized. This translation is essential for positioning interneurons at the basement membrane. Thus, local translation spatially and acutely activates Rho signaling in RGCs to compartmentalize neural progenitor functions.


Asunto(s)
Células Ependimogliales , Neuroglía , Humanos , Ratones , Animales , Células Ependimogliales/metabolismo , ARN Mensajero/metabolismo , Neuroglía/metabolismo , Neurogénesis , Corteza Cerebral , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo
18.
Science ; 379(6632): eabn4705, 2023 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-36705539

RESUMEN

Neuronal development in the human cerebral cortex is considerably prolonged compared with that of other mammals. We explored whether mitochondria influence the species-specific timing of cortical neuron maturation. By comparing human and mouse cortical neuronal maturation at high temporal and cell resolution, we found a slower mitochondria development in human cortical neurons compared with that in the mouse, together with lower mitochondria metabolic activity, particularly that of oxidative phosphorylation. Stimulation of mitochondria metabolism in human neurons resulted in accelerated development in vitro and in vivo, leading to maturation of cells weeks ahead of time, whereas its inhibition in mouse neurons led to decreased rates of maturation. Mitochondria are thus important regulators of the pace of neuronal development underlying human-specific brain neoteny.


Asunto(s)
Mitocondrias , Neurogénesis , Neuronas , Animales , Humanos , Ratones , Corteza Cerebral/citología , Corteza Cerebral/crecimiento & desarrollo , Metabolismo Energético , Mitocondrias/metabolismo , Neuronas/metabolismo
19.
Nature ; 443(7108): 167-72, 2006 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-16915236

RESUMEN

The developmental and evolutionary mechanisms behind the emergence of human-specific brain features remain largely unknown. However, the recent ability to compare our genome to that of our closest relative, the chimpanzee, provides new avenues to link genetic and phenotypic changes in the evolution of the human brain. We devised a ranking of regions in the human genome that show significant evolutionary acceleration. Here we report that the most dramatic of these 'human accelerated regions', HAR1, is part of a novel RNA gene (HAR1F) that is expressed specifically in Cajal-Retzius neurons in the developing human neocortex from 7 to 19 gestational weeks, a crucial period for cortical neuron specification and migration. HAR1F is co-expressed with reelin, a product of Cajal-Retzius neurons that is of fundamental importance in specifying the six-layer structure of the human cortex. HAR1 and the other human accelerated regions provide new candidates in the search for uniquely human biology.


Asunto(s)
Corteza Cerebral/embriología , Corteza Cerebral/metabolismo , Evolución Molecular , Regulación del Desarrollo de la Expresión Génica , ARN no Traducido/genética , Envejecimiento/genética , Animales , Secuencia de Bases , Moléculas de Adhesión Celular Neuronal/genética , Corteza Cerebral/anatomía & histología , Proteínas de la Matriz Extracelular/genética , Perfilación de la Expresión Génica , Humanos , Macaca/genética , Datos de Secuencia Molecular , Mutación/genética , Neocórtex/anatomía & histología , Neocórtex/embriología , Neocórtex/metabolismo , Proteínas del Tejido Nervioso/genética , Conformación de Ácido Nucleico , Especificidad de Órganos , Estabilidad del ARN , ARN no Traducido/química , ARN no Traducido/metabolismo , Proteína Reelina , Serina Endopeptidasas/genética , Factores de Tiempo
20.
PLoS Biol ; 6(6): e140, 2008 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-18547142

RESUMEN

Gene duplication was prevalent during hominoid evolution, yet little is known about the functional fate of new ape gene copies. We characterized the CDC14B cell cycle gene and the functional evolution of its hominoid-specific daughter gene, CDC14Bretro. We found that CDC14B encodes four different splice isoforms that show different subcellular localizations (nucleus or microtubule-associated) and functional properties. A microtubular CDC14B variant spawned CDC14Bretro through retroposition in the hominoid ancestor 18-25 million years ago (Mya). CDC14Bretro evolved brain-/testis-specific expression after the duplication event and experienced a short period of intense positive selection in the African ape ancestor 7-12 Mya. Using resurrected ancestral protein variants, we demonstrate that by virtue of amino acid substitutions in distinct protein regions during this time, the subcellular localization of CDC14Bretro progressively shifted from the association with microtubules (stabilizing them) to an association with the endoplasmic reticulum. CDC14Bretro evolution represents a paradigm example of rapid, selectively driven subcellular relocalization, thus revealing a novel mode for the emergence of new gene function.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Fosfatasas de Especificidad Dual/genética , Fosfatasas de Especificidad Dual/metabolismo , Evolución Molecular , Secuencia de Aminoácidos , Animales , Encéfalo/metabolismo , Proteínas de Ciclo Celular/análisis , Línea Celular , Fosfatasas de Especificidad Dual/análisis , Duplicación de Gen , Genes Duplicados , Hominidae/fisiología , Humanos , Datos de Secuencia Molecular , Isoformas de Proteínas/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda