Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
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
2.
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
3.
Nature ; 630(8017): 596-608, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38898293

ABSTRACT

The evolution of the modern human brain was accompanied by distinct molecular and cellular specializations, which underpin our diverse cognitive abilities but also increase our susceptibility to neurological diseases. These features, some specific to humans and others shared with related species, manifest during different stages of brain development. In this multi-stage process, neural stem cells proliferate to produce a large and diverse progenitor pool, giving rise to excitatory or inhibitory neurons that integrate into circuits during further maturation. This process unfolds over varying time scales across species and has progressively become slower in the human lineage, with differences in tempo correlating with differences in brain size, cell number and diversity, and connectivity. Here we introduce the terms 'bradychrony' and 'tachycrony' to describe slowed and accelerated developmental tempos, respectively. We review how recent technical advances across disciplines, including advanced engineering of in vitro models, functional comparative genetics and high-throughput single-cell profiling, are leading to a deeper understanding of how specializations of the human brain arise during bradychronic neurodevelopment. Emerging insights point to a central role for genetics, gene-regulatory networks, cellular innovations and developmental tempo, which together contribute to the establishment of human specializations during various stages of neurodevelopment and at different points in evolution.


Subject(s)
Biological Evolution , Brain , Humans , Brain/cytology , Brain/physiology , Brain/growth & development , Animals , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis , Time Factors , Neurons/cytology , Neurons/physiology , Single-Cell Analysis , Gene Regulatory Networks
4.
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
5.
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
6.
Bioessays ; : e2400105, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39101295

ABSTRACT

Organoids are quickly becoming an accepted model for understanding human biology and disease. Pluripotent stem cells (PSC) provide a starting point for many organs and enable modeling of the embryonic development and maturation of such organs. The foundation of PSC-derived organoids can be found in elegant developmental studies demonstrating the remarkable ability of immature cells to undergo histogenesis even when taken out of the embryo context. PSC-organoids are an evolution of earlier methods such as embryoid bodies, taken to a new level with finer control and in some cases going beyond tissue histogenesis to organ-like morphogenesis. But many of the discoveries that led to organoids were not necessarily planned, but rather the result of inquisitive minds with freedom to explore. Protecting such curiosity-led research through flexible funding will be important going forward if we are to see further ground-breaking discoveries.

7.
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
8.
EMBO J ; 36(10): 1316-1329, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28283582

ABSTRACT

Cerebral organoids recapitulate human brain development at a considerable level of detail, even in the absence of externally added signaling factors. The patterning events driving this self-organization are currently unknown. Here, we examine the developmental and differentiative capacity of cerebral organoids. Focusing on forebrain regions, we demonstrate the presence of a variety of discrete ventral and dorsal regions. Clearing and subsequent 3D reconstruction of entire organoids reveal that many of these regions are interconnected, suggesting that the entire range of dorso-ventral identities can be generated within continuous neuroepithelia. Consistent with this, we demonstrate the presence of forebrain organizing centers that express secreted growth factors, which may be involved in dorso-ventral patterning within organoids. Furthermore, we demonstrate the timed generation of neurons with mature morphologies, as well as the subsequent generation of astrocytes and oligodendrocytes. Our work provides the methodology and quality criteria for phenotypic analysis of brain organoids and shows that the spatial and temporal patterning events governing human brain development can be recapitulated in vitro.


Subject(s)
Brain/embryology , Cell Differentiation , Cell Proliferation , Organoids/growth & development , Body Patterning , Humans , Spatio-Temporal Analysis
9.
Nat Methods ; 15(7): 505-511, 2018 07.
Article in English | MEDLINE | ID: mdl-29867192

ABSTRACT

Specialized RNA-seq methods are required to identify the 5' ends of transcripts, which are critical for studies of gene regulation, but these methods have not been systematically benchmarked. We directly compared six such methods, including the performance of five methods on a single human cellular RNA sample and a new spike-in RNA assay that helps circumvent challenges resulting from uncertainties in annotation and RNA processing. We found that the 'cap analysis of gene expression' (CAGE) method performed best for mRNA and that most of its unannotated peaks were supported by evidence from other genomic methods. We applied CAGE to eight brain-related samples and determined sample-specific transcription start site (TSS) usage, as well as a transcriptome-wide shift in TSS usage between fetal and adult brain.


Subject(s)
RNA/chemistry , Sequence Analysis, RNA/methods , Base Sequence , Brain , Embryonic Stem Cells , Gene Library , Humans , RNA/genetics , RNA/metabolism
10.
Nat Methods ; 15(12): 1126, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30459407

ABSTRACT

The original version of this paper contained an incorrect primer sequence. In the Methods subsection "Rampage libraries," the text for modification 3 stated that the reverse primer used for library indexing was 5'-CAAGCAGAAGACGGCATACGAGATXXXXXXXXGTGACTGGAGT-3'. The correct sequence of the oligonucleotide used is 5'-CAAGCAGAAGACGGCATACGAGATXXXXXXXXGTGACTGGAGTTCAGACGTGTGCTCTTCCGATCT-3'. This error has been corrected in the PDF and HTML versions of the paper.

11.
Nature ; 501(7467): 373-9, 2013 Sep 19.
Article in English | MEDLINE | ID: mdl-23995685

ABSTRACT

The complexity of the human brain has made it difficult to study many brain disorders in model organisms, highlighting the need for an in vitro model of human brain development. Here we have developed a human pluripotent stem cell-derived three-dimensional organoid culture system, termed cerebral organoids, that develop various discrete, although interdependent, brain regions. These include a cerebral cortex containing progenitor populations that organize and produce mature cortical neuron subtypes. Furthermore, cerebral organoids are shown to recapitulate features of human cortical development, namely characteristic progenitor zone organization with abundant outer radial glial stem cells. Finally, we use RNA interference and patient-specific induced pluripotent stem cells to model microcephaly, a disorder that has been difficult to recapitulate in mice. We demonstrate premature neuronal differentiation in patient organoids, a defect that could help to explain the disease phenotype. Together, these data show that three-dimensional organoids can recapitulate development and disease even in this most complex human tissue.


Subject(s)
Brain/growth & development , Brain/pathology , Microcephaly/pathology , Models, Biological , Organoids/cytology , Organoids/growth & development , Tissue Culture Techniques/methods , Animals , Brain/anatomy & histology , Brain/cytology , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Cerebral Cortex/growth & development , Cerebral Cortex/pathology , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/pathology , Mice , Neural Stem Cells/cytology , Neural Stem Cells/pathology , Neurogenesis , Neurons/cytology , Neurons/pathology , Organoids/embryology , Organoids/pathology
13.
BMC Biol ; 15(1): 55, 2017 06 29.
Article in English | MEDLINE | ID: mdl-28662661

ABSTRACT

Model organisms are widely used in research as accessible and convenient systems to study a particular area or question in biology. Traditionally only a handful of organisms have been widely studied, but modern research tools are enabling researchers to extend the set of model organisms to include less-studied and more unusual systems. This Forum highlights a range of 'non-model model organisms' as emerging systems for tackling questions across the whole spectrum of biology (and beyond), the opportunities and challenges, and the outlook for the future.


Subject(s)
Biology , Eukaryota , Models, Animal , Animals , Plants
14.
Dev Biol ; 420(2): 199-209, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27402594

ABSTRACT

The ability to model human brain development in vitro represents an important step in our study of developmental processes and neurological disorders. Protocols that utilize human embryonic and induced pluripotent stem cells can now generate organoids which faithfully recapitulate, on a cell-biological and gene expression level, the early period of human embryonic and fetal brain development. In combination with novel gene editing tools, such as CRISPR, these methods represent an unprecedented model system in the field of mammalian neural development. In this review, we focus on the similarities of current organoid methods to in vivo brain development, discuss their limitations and potential improvements, and explore the future venues of brain organoid research.


Subject(s)
Brain/embryology , Organoids/embryology , Humans , Models, Neurological , Neurodevelopmental Disorders/etiology , Organ Culture Techniques/methods , Organ Culture Techniques/trends , Organogenesis
16.
Nat Genet ; 38(6): 623-5, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16682970

ABSTRACT

Joubert syndrome-related disorders (JSRD) are a group of syndromes sharing the neuroradiological features of cerebellar vermis hypoplasia and a peculiar brainstem malformation known as the 'molar tooth sign'. We identified mutations in the CEP290 gene in five families with variable neurological, retinal and renal manifestations. CEP290 expression was detected mostly in proliferating cerebellar granule neuron populations and showed centrosome and ciliary localization, linking JSRDs to other human ciliopathies.


Subject(s)
Antigens, Neoplasm/genetics , Brain/abnormalities , Mutation , Neoplasm Proteins/genetics , Animals , Antigens, Neoplasm/metabolism , Cell Cycle Proteins , Centrosome/metabolism , Cytoskeletal Proteins , Humans , Mice , Neoplasm Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Syndrome
17.
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.

18.
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.

19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL