Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38370637

ABSTRACT

Microelectrode array (MEA) recordings are commonly used to compare firing and burst rates in neuronal cultures. MEA recordings can also reveal microscale functional connectivity, topology, and network dynamics-patterns seen in brain networks across spatial scales. Network topology is frequently characterized in neuroimaging with graph theoretical metrics. However, few computational tools exist for analyzing microscale functional brain networks from MEA recordings. Here, we present a MATLAB MEA network analysis pipeline (MEA-NAP) for raw voltage time-series acquired from single- or multi-well MEAs. Applications to 3D human cerebral organoids or 2D human-derived or murine cultures reveal differences in network development, including topology, node cartography, and dimensionality. MEA-NAP incorporates multi-unit template-based spike detection, probabilistic thresholding for determining significant functional connections, and normalization techniques for comparing networks. MEA-NAP can identify network-level effects of pharmacologic perturbation and/or disease-causing mutations and, thus, can provide a translational platform for revealing mechanistic insights and screening new therapeutic approaches.

4.
Cell Stem Cell ; 30(10): 1351-1367.e10, 2023 10 05.
Article in English | MEDLINE | ID: mdl-37802039

ABSTRACT

Progression through fate decisions determines cellular composition and tissue architecture, but how that same architecture may impact cell fate is less clear. We took advantage of organoids as a tractable model to interrogate this interaction of form and fate. Screening methodological variations revealed that common protocol adjustments impacted various aspects of morphology, from macrostructure to tissue architecture. We examined the impact of morphological perturbations on cell fate through integrated single nuclear RNA sequencing (snRNA-seq) and spatial transcriptomics. Regardless of the specific protocol, organoids with more complex morphology better mimicked in vivo human fetal brain development. Organoids with perturbed tissue architecture displayed aberrant temporal progression, with cells being intermingled in both space and time. Finally, encapsulation to impart a simplified morphology led to disrupted tissue cytoarchitecture and a similar abnormal maturational timing. These data demonstrate that cells of the developing brain require proper spatial coordinates to undergo correct temporal progression.


Subject(s)
Brain , Organoids , Humans , Cell Differentiation , Sequence Analysis, RNA
5.
Stem Cell Reports ; 18(9): 1744-1752, 2023 09 12.
Article in English | MEDLINE | ID: mdl-37703820

ABSTRACT

The laboratory culture of human stem cells seeks to capture a cellular state as an in vitro surrogate of a biological system. For the results and outputs from this research to be accurate, meaningful, and durable, standards that ensure reproducibility and reliability of the data should be applied. Although such standards have been previously proposed for repositories and distribution centers, no widely accepted best practices exist for laboratory research with human pluripotent and tissue stem cells. To fill that void, the International Society for Stem Cell Research has developed a set of recommendations, including reporting criteria, for scientists in basic research laboratories. These criteria are designed to be technically and financially feasible and, when implemented, enhance the reproducibility and rigor of stem cell research.


Subject(s)
Stem Cell Research , Humans , Reproducibility of Results
6.
Nat Genet ; 55(9): 1483-1493, 2023 09.
Article in English | MEDLINE | ID: mdl-37592024

ABSTRACT

Our understanding of the genetics of the human cerebral cortex is limited both in terms of the diversity and the anatomical granularity of brain structural phenotypes. Here we conducted a genome-wide association meta-analysis of 13 structural and diffusion magnetic resonance imaging-derived cortical phenotypes, measured globally and at 180 bilaterally averaged regions in 36,663 individuals and identified 4,349 experiment-wide significant loci. These phenotypes include cortical thickness, surface area, gray matter volume, measures of folding, neurite density and water diffusion. We identified four genetic latent structures and causal relationships between surface area and some measures of cortical folding. These latent structures partly relate to different underlying gene expression trajectories during development and are enriched for different cell types. We also identified differential enrichment for neurodevelopmental and constrained genes and demonstrate that common genetic variants associated with cortical expansion are associated with cephalic disorders. Finally, we identified complex interphenotype and inter-regional genetic relationships among the 13 phenotypes, reflecting the developmental differences among them. Together, these analyses identify distinct genetic organizational principles of the cortex and their correlates with neurodevelopment.


Subject(s)
Cerebral Cortex , Genome-Wide Association Study , Humans , Cerebral Cortex/diagnostic imaging , Brain/diagnostic imaging , Neuroimaging , Phenotype
8.
Nature ; 609(7929): 907-910, 2022 09.
Article in English | MEDLINE | ID: mdl-36171373

ABSTRACT

Self-organizing three-dimensional cellular models derived from human pluripotent stem cells or primary tissue have great potential to provide insights into how the human nervous system develops, what makes it unique and how disorders of the nervous system arise, progress and could be treated. Here, to facilitate progress and improve communication with the scientific community and the public, we clarify and provide a basic framework for the nomenclature of human multicellular models of nervous system development and disease, including organoids, assembloids and transplants.


Subject(s)
Consensus , Nervous System , Organoids , Terminology as Topic , Humans , Models, Biological , Nervous System/cytology , Nervous System/pathology , Organoids/cytology , Organoids/pathology , Pluripotent Stem Cells/cytology
9.
Nature ; 602(7895): 112-116, 2022 02.
Article in English | MEDLINE | ID: mdl-35046577

ABSTRACT

The biological basis of male-female brain differences has been difficult to elucidate in humans. The most notable morphological difference is size, with male individuals having on average a larger brain than female individuals1,2, but a mechanistic understanding of how this difference arises remains unknown. Here we use brain organoids3 to show that although sex chromosomal complement has no observable effect on neurogenesis, sex steroids-namely androgens-lead to increased proliferation of cortical progenitors and an increased neurogenic pool. Transcriptomic analysis and functional studies demonstrate downstream effects on histone deacetylase activity and the mTOR pathway. Finally, we show that androgens specifically increase the neurogenic output of excitatory neuronal progenitors, whereas inhibitory neuronal progenitors are not increased. These findings reveal a role for androgens in regulating the number of excitatory neurons and represent a step towards understanding the origin of sex-related brain differences in humans.


Subject(s)
Androgens/pharmacology , Brain/cytology , Cortical Excitability/drug effects , Neurogenesis/drug effects , Organoids/cytology , Organoids/drug effects , Sex Characteristics , Action Potentials/drug effects , Androgens/metabolism , Brain/drug effects , Brain/enzymology , Brain/metabolism , Cell Count , Female , Gene Expression Profiling , Histone Deacetylases/genetics , Humans , Male , Neural Inhibition/drug effects , Neuroglia/cytology , Neuroglia/drug effects , Organ Size/drug effects , Organoids/enzymology , Organoids/metabolism , Stem Cells/cytology , Stem Cells/drug effects , TOR Serine-Threonine Kinases/genetics
10.
Dev Cell ; 56(23): 3185-3191, 2021 12 06.
Article in English | MEDLINE | ID: mdl-34875224

ABSTRACT

In our 20th anniversary year, we reflect on how fields have changed since our first issue and here look to the future. In this collection of Voices, our writers speculate on the future: in terms of philosophy, cell states, cell processes, and then how to model cell systems.


Subject(s)
Cell Biology , Developmental Biology , Periodicals as Topic/statistics & numerical data , Humans , Time Factors
11.
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
12.
Cell ; 184(17): 4377-4379, 2021 08 19.
Article in English | MEDLINE | ID: mdl-34416145

ABSTRACT

Greater understanding of the events preceding neurodegeneration is needed to design effective preventive and therapeutic strategies. In this issue of Cell, Bowles et al. (2021) report cerebral organoids that reveal early events in frontotemporal dementia pathogenesis due to mutations in microtubule-associated protein tau (MAPT), shedding light on a novel mechanism involving abnormal splicing and glutamate signaling.


Subject(s)
Frontotemporal Dementia , Organoids , Humans , Mutation , tau Proteins/genetics
13.
Curr Opin Cell Biol ; 73: 41-49, 2021 12.
Article in English | MEDLINE | ID: mdl-34182208

ABSTRACT

The choroid plexus is central to normal brain function by secreting cerebrospinal fluid and dynamically regulating its composition throughout development and homoeostasis. Much of our current understanding of this region of the brain comes from studies in animal models. These fundamental studies have shed light on choroid plexus mechanisms of secretion, barrier function and homoeostatic regulation. However, how these specific mechanisms are regulated in the human choroid plexus is much less understood, due to ethical and technical limitations. A number of recent breakthroughs have enabled a new range of techniques and tools for functional characterisation of choroid plexus development and physiology. With the advance of new technologies such as in vivo imaging, single-cell transcriptomics and in vitro three-dimensional cultures we are now able to address a number of outstanding questions in choroid plexus biology. Here, we discuss some of these recent breakthroughs and we focus in particular on how in vitro models can be a powerful tool to study human cerebrospinal fluid secretion and barrier function.


Subject(s)
Brain , Choroid Plexus , Animals , Biological Transport , Homeostasis , Humans , Models, Animal
14.
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
15.
Nat Genet ; 53(3): 304-312, 2021 03.
Article in English | MEDLINE | ID: mdl-33664506

ABSTRACT

Studying the function of common genetic variants in primary human tissues and during development is challenging. To address this, we use an efficient multiplexing strategy to differentiate 215 human induced pluripotent stem cell (iPSC) lines toward a midbrain neural fate, including dopaminergic neurons, and use single-cell RNA sequencing (scRNA-seq) to profile over 1 million cells across three differentiation time points. The proportion of neurons produced by each cell line is highly reproducible and is predictable by robust molecular markers expressed in pluripotent cells. Expression quantitative trait loci (eQTL) were characterized at different stages of neuronal development and in response to rotenone-induced oxidative stress. Of these, 1,284 eQTL colocalize with known neurological trait risk loci, and 46% are not found in the Genotype-Tissue Expression (GTEx) catalog. Our study illustrates how coupling scRNA-seq with long-term iPSC differentiation enables mechanistic studies of human trait-associated genetic variants in otherwise inaccessible cell states.


Subject(s)
Dopaminergic Neurons/cytology , Dopaminergic Neurons/physiology , Induced Pluripotent Stem Cells/cytology , Quantitative Trait Loci , Transcriptome , Cell Differentiation/genetics , Genetic Predisposition to Disease , Humans , Induced Pluripotent Stem Cells/physiology , Neurogenesis/genetics , Oxidative Stress/drug effects , Receptor, Fibroblast Growth Factor, Type 1/genetics , Rotenone/toxicity , Sequence Analysis, RNA , Single-Cell Analysis
16.
Nat Mater ; 20(2): 145-155, 2021 02.
Article in English | MEDLINE | ID: mdl-33199860

ABSTRACT

In recent years considerable progress has been made in the development of faithful procedures for the differentiation of human pluripotent stem cells (hPSCs). An important step in this direction has also been the derivation of organoids. This technology generally relies on traditional three-dimensional culture techniques that exploit cell-autonomous self-organization responses of hPSCs with minimal control over the external inputs supplied to the system. The convergence of stem cell biology and bioengineering offers the possibility to provide these stimuli in a controlled fashion, resulting in the development of naturally inspired approaches to overcome major limitations of this nascent technology. Based on the current developments, we emphasize the achievements and ongoing challenges of bringing together hPSC organoid differentiation, bioengineering and ethics. This Review underlines the need for providing engineering solutions to gain control of self-organization and functionality of hPSC-derived organoids. We expect that this knowledge will guide the community to generate higher-grade hPSC-derived organoids for further applications in developmental biology, drug screening, disease modelling and personalized medicine.


Subject(s)
Bioengineering , Organoids/growth & development , Pluripotent Stem Cells/metabolism , Humans , Organoids/cytology , Pluripotent Stem Cells/cytology
18.
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
19.
Nat Neurosci ; 23(12): 1496-1508, 2020 12.
Article in English | MEDLINE | ID: mdl-33139941

ABSTRACT

Brain development is an extraordinarily complex process achieved through the spatially and temporally regulated release of key patterning factors. In vitro neurodevelopmental models seek to mimic these processes to recapitulate the steps of tissue fate acquisition and morphogenesis. Classic two-dimensional neural cultures present higher homogeneity but lower complexity compared to the brain. Brain organoids instead have more advanced cell composition, maturation and tissue architecture. They can thus be considered at the interface of in vitro and in vivo neurobiology, and further improvements in organoid techniques are continuing to narrow the gap with in vivo brain development. Here we describe these efforts to recapitulate brain development in neural organoids and focus on their applicability for disease modeling, evolutionary studies and neural network research.


Subject(s)
Brain/physiology , Neurobiology/methods , Organoids/physiology , Animals , Brain/growth & development , Brain/ultrastructure , Humans , In Vitro Techniques
20.
Cell Stem Cell ; 27(6): 951-961.e5, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33113348

ABSTRACT

Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus, leads to respiratory symptoms that can be fatal. However, neurological symptoms have also been observed in some patients. The cause of these complications is currently unknown. Here, we use human-pluripotent-stem-cell-derived brain organoids to examine SARS-CoV-2 neurotropism. We find expression of viral receptor ACE2 in mature choroid plexus cells expressing abundant lipoproteins, but not in neurons or other cell types. We challenge organoids with SARS-CoV-2 spike pseudovirus and live virus to demonstrate viral tropism for choroid plexus epithelial cells but little to no infection of neurons or glia. We find that infected cells are apolipoprotein- and ACE2-expressing cells of the choroid plexus epithelial barrier. Finally, we show that infection with SARS-CoV-2 damages the choroid plexus epithelium, leading to leakage across this important barrier that normally prevents entry of pathogens, immune cells, and cytokines into cerebrospinal fluid and the brain.


Subject(s)
Blood-Brain Barrier/virology , Choroid Plexus/virology , SARS-CoV-2/physiology , Angiotensin-Converting Enzyme 2/metabolism , Animals , Chlorocebus aethiops , HEK293 Cells , Humans , Models, Biological , Organoids/virology , Vero Cells , Viral Tropism , Virus Internalization
SELECTION OF CITATIONS
SEARCH DETAIL
...