Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Elife ; 102021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34698018

RESUMO

During brain development, axons must extend over great distances in a relatively short amount of time. How the subcellular architecture of the growing axon sustains the requirements for such rapid build-up of cellular constituents has remained elusive. Human axons have been particularly poorly accessible to imaging at high resolution in a near-native context. Here, we present a method that combines cryo-correlative light microscopy and electron tomography with human cerebral organoid technology to visualize growing axon tracts. Our data reveal a wealth of structural details on the arrangement of macromolecules, cytoskeletal components, and organelles in elongating axon shafts. In particular, the intricate shape of the endoplasmic reticulum is consistent with its role in fulfilling the high demand for lipid biosynthesis to support growth. Furthermore, the scarcity of ribosomes within the growing shaft suggests limited translational competence during expansion of this compartment. These findings establish our approach as a powerful resource for investigating the ultrastructure of defined neuronal compartments.


Assuntos
Axônios/ultraestrutura , Tomografia com Microscopia Eletrônica , Organoides/citologia , Encéfalo/citologia , Encéfalo/ultraestrutura , Microscopia Crioeletrônica , Células HeLa , Humanos , Substâncias Macromoleculares/metabolismo , Microscopia , Microscopia de Fluorescência , Organoides/ultraestrutura
2.
Cell ; 184(8): 2084-2102.e19, 2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33765444

RESUMO

The human brain has undergone rapid expansion since humans diverged from other great apes, but the mechanism of this human-specific enlargement is still unknown. Here, we use cerebral organoids derived from human, gorilla, and chimpanzee cells to study developmental mechanisms driving evolutionary brain expansion. We find that neuroepithelial differentiation is a protracted process in apes, involving a previously unrecognized transition state characterized by a change in cell shape. Furthermore, we show that human organoids are larger due to a delay in this transition, associated with differences in interkinetic nuclear migration and cell cycle length. Comparative RNA sequencing (RNA-seq) reveals differences in expression dynamics of cell morphogenesis factors, including ZEB2, a known epithelial-mesenchymal transition regulator. We show that ZEB2 promotes neuroepithelial transition, and its manipulation and downstream signaling leads to acquisition of nonhuman ape architecture in the human context and vice versa, establishing an important role for neuroepithelial cell shape in human brain expansion.


Assuntos
Evolução Biológica , Encéfalo/citologia , Forma Celular/fisiologia , Animais , Encéfalo/metabolismo , Diferenciação Celular , Linhagem Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Transição Epitelial-Mesenquimal/genética , Expressão Gênica , Gorilla gorilla , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Neurogênese , Neurônios/citologia , Neurônios/metabolismo , Organoides/citologia , Organoides/metabolismo , Pan troglodytes , Homeobox 2 de Ligação a E-box com Dedos de Zinco/genética , Homeobox 2 de Ligação a E-box com Dedos de Zinco/metabolismo
3.
Nat Protoc ; 16(2): 579-602, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33328611

RESUMO

Cerebral organoids, or brain organoids, can be generated from a wide array of emerging technologies for modeling brain development and disease. The fact that they are cultured in vitro makes them easily accessible both genetically and for live assays such as fluorescence imaging. In this Protocol Extension, we describe a modified version of our original protocol (published in 2014) that can be used to reliably generate cerebral organoids of a telencephalic identity and maintain long-term viability for later stages of neural development, including axon outgrowth and neuronal maturation. The method builds upon earlier cerebral organoid methodology, with modifications of embryoid body size and shape to increase surface area and slice culture to maintain nutrient and oxygen access to the interior regions of the organoid, enabling long-term culture. We also describe approaches for introducing exogenous plasmid constructs and for sparse cell labeling to image neuronal axon outgrowth and maturation over time. Together, these methods allow for modeling of later events in cortical development, which are important for neurodevelopmental disease modeling. The protocols described can be easily performed by an experimenter with stem cell culture experience and take 2-3 months to complete, with long-term maturation occurring over several months.


Assuntos
Técnicas de Cultura de Células/métodos , Neurônios/citologia , Organoides/citologia , Encéfalo/citologia , Corpos Embrioides/citologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Neurogênese/fisiologia , Organoides/metabolismo
4.
Nat Neurosci ; 22(4): 669-679, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30886407

RESUMO

Neural organoids have the potential to improve our understanding of human brain development and neurological disorders. However, it remains to be seen whether these tissues can model circuit formation with functional neuronal output. Here we have adapted air-liquid interface culture to cerebral organoids, leading to improved neuronal survival and axon outgrowth. The resulting thick axon tracts display various morphologies, including long-range projection within and away from the organoid, growth-cone turning, and decussation. Single-cell RNA sequencing reveals various cortical neuronal identities, and retrograde tracing demonstrates tract morphologies that match proper molecular identities. These cultures exhibit active neuronal networks, and subcortical projecting tracts can innervate mouse spinal cord explants and evoke contractions of adjacent muscle in a manner dependent on intact organoid-derived innervating tracts. Overall, these results reveal a remarkable self-organization of corticofugal and callosal tracts with a functional output, providing new opportunities to examine relevant aspects of human CNS development and disease.


Assuntos
Córtex Cerebral/crescimento & desenvolvimento , Neurônios/fisiologia , Organoides/crescimento & desenvolvimento , Técnicas de Cultura de Tecidos/métodos , Axônios/fisiologia , Sobrevivência Celular , Córtex Cerebral/citologia , Feminino , Humanos , Masculino , Vias Neurais/citologia , Vias Neurais/fisiologia , Neurônios/citologia , Organoides/citologia , Células-Tronco Pluripotentes/fisiologia
5.
Methods Mol Biol ; 1576: 1-12, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-28361479

RESUMO

3D brain organoids are a powerful tool with prospective application for the study of neural development and disease. Here we describe the growth factor-free method of generating cerebral organoids from feeder-dependent or feeder-free human pluripotent stem cells using standard laboratory equipment. The protocol outlined below allows generation of 3D tissues, which replicate human early in vivo brain development up to the end of the first trimester, both in terms of morphology and gene expression pattern.


Assuntos
Encéfalo/citologia , Técnicas de Cultura de Células/métodos , Diferenciação Celular , Células-Tronco Pluripotentes Induzidas/citologia , Organoides/citologia , Células Cultivadas , Humanos , Neurogênese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...