Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Animals (Basel) ; 14(11)2024 May 23.
Article in English | MEDLINE | ID: mdl-38891588

ABSTRACT

The documentation, preservation and rescue of biological diversity increasingly uses living biological samples. Persistent associations between species, biosamples, such as tissues and cell lines, and the accompanying data are indispensable for using, exchanging and benefiting from these valuable materials. Explicit authentication of such biosamples by assigning unique and robust identifiers is therefore required to allow for unambiguous referencing, avoid identification conflicts and maintain reproducibility in research. A predefined nomenclature based on uniform rules would facilitate this process. However, such a nomenclature is currently lacking for animal biological material. We here present a first, standardized, human-readable nomenclature design, which is sufficient to generate unique and stable identifying names for animal cellular material with a focus on wildlife species. A species-specific human- and machine-readable syntax is included in the proposed standard naming scheme, allowing for the traceability of donated material and cultured cells, as well as data FAIRification. Only when it is consistently applied in the public domain, as publications and inter-institutional samples and data are exchanged, distributed and stored centrally, can the risks of misidentification and loss of traceability be mitigated. This innovative globally applicable identification system provides a standard for a sustainable structure for the long-term storage of animal bio-samples in cryobanks and hence facilitates current as well as future species conservation and biomedical research.

2.
Elife ; 102021 10 26.
Article in English | MEDLINE | ID: mdl-34698018

ABSTRACT

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.


Subject(s)
Axons/ultrastructure , Electron Microscope Tomography , Organoids/cytology , Brain/cytology , Brain/ultrastructure , Cryoelectron Microscopy , HeLa Cells , Humans , Macromolecular Substances/metabolism , Microscopy , Microscopy, Fluorescence , Organoids/ultrastructure
3.
Cell ; 184(8): 2084-2102.e19, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33765444

ABSTRACT

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.


Subject(s)
Biological Evolution , Brain/cytology , Cell Shape/physiology , Animals , Brain/metabolism , Cell Differentiation , Cell Line , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Epithelial-Mesenchymal Transition/genetics , Gene Expression , Gorilla gorilla , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Neurogenesis , Neurons/cytology , Neurons/metabolism , Organoids/cytology , Organoids/metabolism , Pan troglodytes , Zinc Finger E-box Binding Homeobox 2/genetics , Zinc Finger E-box Binding Homeobox 2/metabolism
4.
Nat Protoc ; 16(2): 579-602, 2021 02.
Article in English | MEDLINE | ID: mdl-33328611

ABSTRACT

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.


Subject(s)
Cell Culture Techniques/methods , Neurons/cytology , Organoids/cytology , Brain/cytology , Embryoid Bodies/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Neurogenesis/physiology , Organoids/metabolism
5.
Nat Neurosci ; 22(4): 669-679, 2019 04.
Article in English | MEDLINE | ID: mdl-30886407

ABSTRACT

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.


Subject(s)
Cerebral Cortex/growth & development , Neurons/physiology , Organoids/growth & development , Tissue Culture Techniques/methods , Axons/physiology , Cell Survival , Cerebral Cortex/cytology , Female , Humans , Male , Neural Pathways/cytology , Neural Pathways/physiology , Neurons/cytology , Organoids/cytology , Pluripotent Stem Cells/physiology
6.
Methods Mol Biol ; 1576: 1-12, 2019.
Article in English | MEDLINE | ID: mdl-28361479

ABSTRACT

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.


Subject(s)
Brain/cytology , Cell Culture Techniques/methods , Cell Differentiation , Induced Pluripotent Stem Cells/cytology , Organoids/cytology , Cells, Cultured , Humans , Neurogenesis
SELECTION OF CITATIONS
SEARCH DETAIL
...