Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters










Publication year range
1.
Science ; 384(6698): eadh0559, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38781390

ABSTRACT

Nucleotide changes in gene regulatory elements are important determinants of neuronal development and diseases. Using massively parallel reporter assays in primary human cells from mid-gestation cortex and cerebral organoids, we interrogated the cis-regulatory activity of 102,767 open chromatin regions, including thousands of sequences with cell type-specific accessibility and variants associated with brain gene regulation. In primary cells, we identified 46,802 active enhancer sequences and 164 variants that alter enhancer activity. Activity was comparable in organoids and primary cells, suggesting that organoids provide an adequate model for the developing cortex. Using deep learning we decoded the sequence basis and upstream regulators of enhancer activity. This work establishes a comprehensive catalog of functional gene regulatory elements and variants in human neuronal development.


Subject(s)
Cerebral Cortex , Neurogenesis , Organoids , Humans , Cerebral Cortex/embryology , Cerebral Cortex/metabolism , Chromatin/metabolism , Chromatin/genetics , Deep Learning , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Neurogenesis/genetics , Neurons/metabolism , Organoids/metabolism , Regulatory Sequences, Nucleic Acid , Promoter Regions, Genetic , Regulatory Elements, Transcriptional
2.
Cell ; 187(6): 1547-1562.e13, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38428424

ABSTRACT

We sequenced and assembled using multiple long-read sequencing technologies the genomes of chimpanzee, bonobo, gorilla, orangutan, gibbon, macaque, owl monkey, and marmoset. We identified 1,338,997 lineage-specific fixed structural variants (SVs) disrupting 1,561 protein-coding genes and 136,932 regulatory elements, including the most complete set of human-specific fixed differences. We estimate that 819.47 Mbp or ∼27% of the genome has been affected by SVs across primate evolution. We identify 1,607 structurally divergent regions wherein recurrent structural variation contributes to creating SV hotspots where genes are recurrently lost (e.g., CARD, C4, and OLAH gene families) and additional lineage-specific genes are generated (e.g., CKAP2, VPS36, ACBD7, and NEK5 paralogs), becoming targets of rapid chromosomal diversification and positive selection (e.g., RGPD gene family). High-fidelity long-read sequencing has made these dynamic regions of the genome accessible for sequence-level analyses within and between primate species.


Subject(s)
Genome , Primates , Animals , Humans , Base Sequence , Primates/classification , Primates/genetics , Biological Evolution , Sequence Analysis, DNA , Genomic Structural Variation
3.
Cell Stem Cell ; 31(3): 421-432.e8, 2024 03 07.
Article in English | MEDLINE | ID: mdl-38382530

ABSTRACT

Thalamic dysfunction has been implicated in multiple psychiatric disorders. We sought to study the mechanisms by which abnormalities emerge in the context of the 22q11.2 microdeletion, which confers significant genetic risk for psychiatric disorders. We investigated early stages of human thalamus development using human pluripotent stem cell-derived organoids and show that the 22q11.2 microdeletion underlies widespread transcriptional dysregulation associated with psychiatric disorders in thalamic neurons and glia, including elevated expression of FOXP2. Using an organoid co-culture model, we demonstrate that the 22q11.2 microdeletion mediates an overgrowth of thalamic axons in a FOXP2-dependent manner. Finally, we identify ROBO2 as a candidate molecular mediator of the effects of FOXP2 overexpression on thalamic axon overgrowth. Together, our study suggests that early steps in thalamic development are dysregulated in a model of genetic risk for schizophrenia and contribute to neural phenotypes in 22q11.2 deletion syndrome.


Subject(s)
DiGeorge Syndrome , Schizophrenia , Humans , Schizophrenia/genetics , DiGeorge Syndrome/genetics , DiGeorge Syndrome/psychology , Phenotype
4.
Nat Rev Neurosci ; 25(1): 7-29, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37996703

ABSTRACT

The delayed and prolonged postmitotic maturation of human neurons, compared with neurons from other species, may contribute to human-specific cognitive abilities and neurological disorders. Here we review the mechanisms of neuronal maturation, applying lessons from model systems to understand the specific features of protracted human cortical maturation and species differences. We cover cell-intrinsic features of neuronal maturation, including transcriptional, epigenetic and metabolic mechanisms, as well as cell-extrinsic features, including the roles of activity and synapses, the actions of glial cells and the contribution of the extracellular matrix. We discuss evidence for species differences in biochemical reaction rates, the proposed existence of an epigenetic maturation clock and the contributions of both general and modular mechanisms to species-specific maturation timing. Finally, we suggest approaches to measure, improve and accelerate the maturation of human neurons in culture, examine crosstalk and interactions among these different aspects of maturation and propose conceptual models to guide future studies.


Subject(s)
Neuroglia , Neurons , Humans , Species Specificity , Neurons/physiology , Neurogenesis/physiology , Synapses/physiology
5.
Nature ; 622(7981): 112-119, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37704727

ABSTRACT

The molecular mechanisms and evolutionary changes accompanying synapse development are still poorly understood1,2. Here we generate a cross-species proteomic map of synapse development in the human, macaque and mouse neocortex. By tracking the changes of more than 1,000 postsynaptic density (PSD) proteins from midgestation to young adulthood, we find that PSD maturation in humans separates into three major phases that are dominated by distinct pathways. Cross-species comparisons reveal that human PSDs mature about two to three times slower than those of other species and contain higher levels of Rho guanine nucleotide exchange factors (RhoGEFs) in the perinatal period. Enhancement of RhoGEF signalling in human neurons delays morphological maturation of dendritic spines and functional maturation of synapses, potentially contributing to the neotenic traits of human brain development. In addition, PSD proteins can be divided into four modules that exert stage- and cell-type-specific functions, possibly explaining their differential associations with cognitive functions and diseases. Our proteomic map of synapse development provides a blueprint for studying the molecular basis and evolutionary changes of synapse maturation.


Subject(s)
Proteomics , Synapses , Adolescent , Animals , Child , Child, Preschool , Humans , Infant , Infant, Newborn , Mice , Young Adult , Cognition/physiology , Dendritic Spines , Gestational Age , Macaca , Neurons/metabolism , Post-Synaptic Density/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Signal Transduction , Species Specificity , Synapses/metabolism , Synapses/physiology
6.
Cell ; 186(14): 2977-2994.e23, 2023 07 06.
Article in English | MEDLINE | ID: mdl-37343560

ABSTRACT

Comparative studies of great apes provide a window into our evolutionary past, but the extent and identity of cellular differences that emerged during hominin evolution remain largely unexplored. We established a comparative loss-of-function approach to evaluate whether human cells exhibit distinct genetic dependencies. By performing genome-wide CRISPR interference screens in human and chimpanzee pluripotent stem cells, we identified 75 genes with species-specific effects on cellular proliferation. These genes comprised coherent processes, including cell-cycle progression and lysosomal signaling, which we determined to be human-derived by comparison with orangutan cells. Human-specific robustness to CDK2 and CCNE1 depletion persisted in neural progenitor cells and cerebral organoids, supporting the G1-phase length hypothesis as a potential evolutionary mechanism in human brain expansion. Our findings demonstrate that evolutionary changes in human cells reshaped the landscape of essential genes and establish a platform for systematically uncovering latent cellular and molecular differences between species.


Subject(s)
Hominidae , Neural Stem Cells , Pluripotent Stem Cells , Stem Cells , Animals , Humans , Pan troglodytes/genetics
7.
Curr Opin Neurobiol ; 80: 102710, 2023 06.
Article in English | MEDLINE | ID: mdl-37003107

ABSTRACT

Comparative studies of hominids have long sought to identify mutational events that shaped the evolution of the human nervous system. However, functional genetic differences are outnumbered by millions of nearly neutral mutations, and the developmental mechanisms underlying human nervous system specializations are difficult to model and incompletely understood. Candidate-gene studies have attempted to map select human-specific genetic differences to neurodevelopmental functions, but it remains unclear how to contextualize the relative effects of genes that are investigated independently. Considering these limitations, we discuss scalable approaches for probing the functional contributions of human-specific genetic differences. We propose that a systems-level view will enable a more quantitative and integrative understanding of the genetic, molecular and cellular underpinnings of human nervous system evolution.


Subject(s)
Brain , Nervous System , Humans , Brain/physiology , Biological Evolution
8.
bioRxiv ; 2023 Mar 20.
Article in English | MEDLINE | ID: mdl-36993685

ABSTRACT

Comparative studies of great apes provide a window into our evolutionary past, but the extent and identity of cellular differences that emerged during hominin evolution remain largely unexplored. We established a comparative loss-of-function approach to evaluate whether changes in human cells alter requirements for essential genes. By performing genome-wide CRISPR interference screens in human and chimpanzee pluripotent stem cells, we identified 75 genes with species-specific effects on cellular proliferation. These genes comprised coherent processes, including cell cycle progression and lysosomal signaling, which we determined to be human-derived by comparison with orangutan cells. Human-specific robustness to CDK2 and CCNE1 depletion persisted in neural progenitor cells, providing support for the G1-phase length hypothesis as a potential evolutionary mechanism in human brain expansion. Our findings demonstrate that evolutionary changes in human cells can reshape the landscape of essential genes and establish a platform for systematically uncovering latent cellular and molecular differences between species.

9.
Nat Rev Genet ; 24(10): 687-711, 2023 10.
Article in English | MEDLINE | ID: mdl-36737647

ABSTRACT

Our ancestors acquired morphological, cognitive and metabolic modifications that enabled humans to colonize diverse habitats, develop extraordinary technologies and reshape the biosphere. Understanding the genetic, developmental and molecular bases for these changes will provide insights into how we became human. Connecting human-specific genetic changes to species differences has been challenging owing to an abundance of low-effect size genetic changes, limited descriptions of phenotypic differences across development at the level of cell types and lack of experimental models. Emerging approaches for single-cell sequencing, genetic manipulation and stem cell culture now support descriptive and functional studies in defined cell types with a human or ape genetic background. In this Review, we describe how the sequencing of genomes from modern and archaic hominins, great apes and other primates is revealing human-specific genetic changes and how new molecular and cellular approaches - including cell atlases and organoids - are enabling exploration of the candidate causal factors that underlie human-specific traits.


Subject(s)
Hominidae , Animals , Humans , Hominidae/genetics , Organoids , Biological Evolution , Evolution, Molecular
10.
Neuron ; 111(6): 857-873.e8, 2023 03 15.
Article in English | MEDLINE | ID: mdl-36640767

ABSTRACT

Using machine learning (ML), we interrogated the function of all human-chimpanzee variants in 2,645 human accelerated regions (HARs), finding 43% of HARs have variants with large opposing effects on chromatin state and 14% on neurodevelopmental enhancer activity. This pattern, consistent with compensatory evolution, was confirmed using massively parallel reporter assays in chimpanzee and human neural progenitor cells. The species-specific enhancer activity of HARs was accurately predicted from the presence and absence of transcription factor footprints in each species. Despite these striking cis effects, activity of a given HAR sequence was nearly identical in human and chimpanzee cells. This suggests that HARs did not evolve to compensate for changes in the trans environment but instead altered their ability to bind factors present in both species. Thus, ML prioritized variants with functional effects on human neurodevelopment and revealed an unexpected reason why HARs may have evolved so rapidly.


Subject(s)
Brain , Enhancer Elements, Genetic , Pan troglodytes , Animals , Humans , Chromatin , Machine Learning , Pan troglodytes/metabolism , Transcription Factors/genetics , Brain/growth & development
11.
bioRxiv ; 2023 Dec 28.
Article in English | MEDLINE | ID: mdl-38234800

ABSTRACT

Deletion of functional sequence is predicted to represent a fundamental mechanism of molecular evolution1,2. Comparative genetic studies of primates2,3 have identified thousands of human-specific deletions (hDels), and the cis-regulatory potential of short (≤31 base pairs) hDels has been assessed using reporter assays4. However, how structural variant-sized (≥50 base pairs) hDels influence molecular and cellular processes in their native genomic contexts remains unexplored. Here, we design genome-scale libraries of single-guide RNAs targeting 7.2 megabases of sequence in 6,358 hDels and present a systematic CRISPR interference (CRISPRi) screening approach to identify hDels that modify cellular proliferation in chimpanzee pluripotent stem cells. By intersecting hDels with chromatin state features and performing single-cell CRISPRi (Perturb-seq) to identify their cis- and trans-regulatory target genes, we discovered 19 hDels controlling gene expression. We highlight two hDels, hDel_2247 and hDel_585, with tissue-specific activity in the liver and brain, respectively. Our findings reveal a molecular and cellular role for sequences lost in the human lineage and establish a framework for functionally interrogating human-specific genetic variants.

12.
EMBO Rep ; 23(11): e56076, 2022 11 07.
Article in English | MEDLINE | ID: mdl-36161459

ABSTRACT

The human cerebral cortex has tripled in size since our divergence from a common ancestor with chimpanzees. This cortical expansion is driven by the increased proliferative capacity of radial glia (RG), a neural progenitor cell (NPC) population that generates cortical neurons. RG along the ventricular zone (VZ) produce neurons and also give rise to basal progenitors (BPs), which migrate to the embryonic subventricular zone (SVZ). Comparative studies suggest that the increased proliferative capacity of human NPCs involves cell-intrinsic mechanisms (Otani et al, 2016), and a number of human-specific genetic changes have recently been linked to NPC proliferation. In particular, overexpression studies in model organisms indicate that the human-specific gene ARHGAP11B is sufficient to increase BP abundance when introduced into the developing brain of non-human model organisms (Florio et al, 2015; Kalebic et al, 2018; Heide et al, 2020). However, studying human-specific mutations in a hominid genetic and developmental context, rather than in more divergent model organisms, could provide further insight into the evolutionary consequences and effect size of human mutations. Fischer et al (2022) now developed a novel organoid electroporation technique to establish the necessity and sufficiency of ARHGAP11B for BP proliferation in cells from humans and our closest living relative, chimpanzees (Fig 1).


Subject(s)
Neural Stem Cells , Pan troglodytes , Animals , Humans , Pan troglodytes/genetics , Neurons/physiology , Cerebral Cortex , GTPase-Activating Proteins/genetics
13.
Proc Natl Acad Sci U S A ; 119(30): e2122236119, 2022 07 26.
Article in English | MEDLINE | ID: mdl-35858406

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) readily infects a variety of cell types impacting the function of vital organ systems, with particularly severe impact on respiratory function. Neurological symptoms, which range in severity, accompany as many as one-third of COVID-19 cases, indicating a potential vulnerability of neural cell types. To assess whether human cortical cells can be directly infected by SARS-CoV-2, we utilized stem-cell-derived cortical organoids as well as primary human cortical tissue, both from developmental and adult stages. We find significant and predominant infection in cortical astrocytes in both primary tissue and organoid cultures, with minimal infection of other cortical populations. Infected and bystander astrocytes have a corresponding increase in inflammatory gene expression, reactivity characteristics, increased cytokine and growth factor signaling, and cellular stress. Although human cortical cells, particularly astrocytes, have no observable ACE2 expression, we find high levels of coronavirus coreceptors in infected astrocytes, including CD147 and DPP4. Decreasing coreceptor abundance and activity reduces overall infection rate, and increasing expression is sufficient to promote infection. Thus, we find tropism of SARS-CoV-2 for human astrocytes resulting in inflammatory gliosis-type injury that is dependent on coronavirus coreceptors.


Subject(s)
Astrocytes , Cerebral Cortex , SARS-CoV-2 , Viral Tropism , Angiotensin-Converting Enzyme 2/metabolism , Astrocytes/enzymology , Astrocytes/virology , Cerebral Cortex/virology , Humans , Organoids/virology , Primary Cell Culture , SARS-CoV-2/physiology
14.
Nature ; 603(7903): 871-877, 2022 03.
Article in English | MEDLINE | ID: mdl-35322231

ABSTRACT

Neuroanatomists have long speculated that expanded primate brains contain an increased morphological diversity of inhibitory neurons (INs)1, and recent studies have identified primate-specific neuronal populations at the molecular level2. However, we know little about the developmental mechanisms that specify evolutionarily novel cell types in the brain. Here, we reconstruct gene expression trajectories specifying INs generated throughout the neurogenic period in macaques and mice by analysing the transcriptomes of 250,181 cells. We find that the initial classes of INs generated prenatally are largely conserved among mammals. Nonetheless, we identify two contrasting developmental mechanisms for specifying evolutionarily novel cell types during prenatal development. First, we show that recently identified primate-specific TAC3 striatal INs are specified by a unique transcriptional programme in progenitors followed by induction of a distinct suite of neuropeptides and neurotransmitter receptors in new-born neurons. Second, we find that multiple classes of transcriptionally conserved olfactory bulb (OB)-bound precursors are redirected to expanded primate white matter and striatum. These classes include a novel peristriatal class of striatum laureatum neurons that resemble dopaminergic periglomerular cells of the OB. We propose an evolutionary model in which conserved initial classes of neurons supplying the smaller primate OB are reused in the enlarged striatum and cortex. Together, our results provide a unified developmental taxonomy of initial classes of mammalian INs and reveal multiple developmental mechanisms for neural cell type evolution.


Subject(s)
Biological Evolution , Corpus Striatum , Embryonic Development , Macaca , Neurogenesis , Neurons , Olfactory Bulb , Animals , Corpus Striatum/growth & development , Dopaminergic Neurons , Female , Macaca/growth & development , Mammals , Mice , Neurogenesis/physiology , Olfactory Bulb/physiology , Pregnancy , Primates
15.
Nat Rev Genet ; 23(5): 315-320, 2022 05.
Article in English | MEDLINE | ID: mdl-35082442

ABSTRACT

Cell fusions have a long history of supporting biomedical research. These experimental models, historically referred to as 'somatic cell hybrids', involve combining the plasma membranes of two cells and merging their nuclei within a single cytoplasm. Cell fusion studies involving human and chimpanzee pluripotent stem cells, rather than somatic cells, highlight the need for responsible communication and a revised nomenclature. Applying the terms 'hybrid' and 'parental' to the fused and source cell lines, respectively, evokes reproductive relationships that do not exist between humans and other species. These misnomers become more salient in the context of fused pluripotent stem cells derived from different but closely related species. Here, we propose a precise, versatile and generalizable framework to describe these fused cell lines. We recommend the term 'composite cell line', to distinguish cell lines that are experimentally created through fusions from both reproductive hybrids and natural cell fusion events without obscuring the model in overly technical terms. For scientific audiences, we further recommend technical nomenclature that describes the contributing species, ploidy and cell type.


Subject(s)
Pluripotent Stem Cells , Cell Communication , Cell Fusion , Cell Line , Cell Nucleus/genetics , Humans
16.
Article in English | MEDLINE | ID: mdl-37383277

ABSTRACT

The Internet of Things (IoT) provides a simple framework to control online devices easily. IoT is now a commonplace tool used by technology companies but is rarely used in biology experiments. IoT can benefit cloud biology research through alarm notifications, automation, and the real-time monitoring of experiments. We developed an IoT architecture to control biological devices and implemented it in lab experiments. Lab devices for electrophysiology, microscopy, and microfluidics were created from the ground up to be part of a unified IoT architecture. The system allows each device to be monitored and controlled from an online web tool. We present our IoT architecture so other labs can replicate it for their own experiments.

17.
Commun Biol ; 4(1): 1261, 2021 11 04.
Article in English | MEDLINE | ID: mdl-34737378

ABSTRACT

Simultaneous longitudinal imaging across multiple conditions and replicates has been crucial for scientific studies aiming to understand biological processes and disease. Yet, imaging systems capable of accomplishing these tasks are economically unattainable for most academic and teaching laboratories around the world. Here, we propose the Picroscope, which is the first low-cost system for simultaneous longitudinal biological imaging made primarily using off-the-shelf and 3D-printed materials. The Picroscope is compatible with standard 24-well cell culture plates and captures 3D z-stack image data. The Picroscope can be controlled remotely, allowing for automatic imaging with minimal intervention from the investigator. Here, we use this system in a range of applications. We gathered longitudinal whole organism image data for frogs, zebrafish, and planaria worms. We also gathered image data inside an incubator to observe 2D monolayers and 3D mammalian tissue culture models. Using this tool, we can measure the behavior of entire organisms or individual cells over long-time periods.


Subject(s)
Imaging, Three-Dimensional/methods , Mammals , Planarians , Xenopus , Zebrafish , Animals , Behavior, Animal , Mammals/physiology , Organoids/physiology , Planarians/anatomy & histology , Planarians/physiology , Xenopus/anatomy & histology , Xenopus/physiology , Zebrafish/anatomy & histology , Zebrafish/physiology
18.
J Neural Eng ; 18(6)2021 11 12.
Article in English | MEDLINE | ID: mdl-34666315

ABSTRACT

Objective.Neural activity represents a functional readout of neurons that is increasingly important to monitor in a wide range of experiments. Extracellular recordings have emerged as a powerful technique for measuring neural activity because these methods do not lead to the destruction or degradation of the cells being measured. Current approaches to electrophysiology have a low throughput of experiments due to manual supervision and expensive equipment. This bottleneck limits broader inferences that can be achieved with numerous long-term recorded samples.Approach.We developed Piphys, an inexpensive open source neurophysiological recording platform that consists of both hardware and software. It is easily accessed and controlled via a standard web interface through Internet of Things (IoT) protocols.Main results.We used a Raspberry Pi as the primary processing device along with an Intan bioamplifier. We designed a hardware expansion circuit board and software to enable voltage sampling and user interaction. This standalone system was validated with primary human neurons, showing reliability in collecting neural activity in near real-time.Significance.The hardware modules and cloud software allow for remote control of neural recording experiments as well as horizontal scalability, enabling long-term observations of development, organization, and neural activity at scale.


Subject(s)
Cloud Computing , Software , Computers , Electrophysiology/methods , Humans , Reproducibility of Results
19.
Nature ; 598(7881): 417-418, 2021 10.
Article in English | MEDLINE | ID: mdl-34588642

Subject(s)
Brain , Prefrontal Cortex
20.
Nat Neurosci ; 24(9): 1235-1242, 2021 09.
Article in English | MEDLINE | ID: mdl-34239128

ABSTRACT

Nuclear compartments are thought to play a role in three-dimensional genome organization and gene expression. In mammalian brain, the architecture and dynamics of nuclear compartment-associated genome organization is not known. In this study, we developed Genome Organization using CUT and RUN Technology (GO-CaRT) to map genomic interactions with two nuclear compartments-the nuclear lamina and nuclear speckles-from different regions of the developing mouse, macaque and human brain. Lamina-associated domain (LAD) architecture in cells in vivo is distinct from that of cultured cells, including major differences in LADs previously considered to be cell type invariant. In the mouse and human forebrain, dorsal and ventral neural precursor cells have differences in LAD architecture that correspond to their regional identity. LADs in the human and mouse cortex contain transcriptionally highly active sub-domains characterized by broad depletion of histone-3-lysine-9 dimethylation. Evolutionarily conserved LADs in human, macaque and mouse brain are enriched for transcriptionally active neural genes associated with synapse function. By integrating GO-CaRT maps with genome-wide association study data, we found speckle-associated domains to be enriched for schizophrenia risk loci, indicating a physical relationship between these disease-associated genetic variants and a specific nuclear structure. Our work provides a framework for understanding the relationship between distinct nuclear compartments and genome function in brain development and disease.


Subject(s)
Brain/physiology , Cell Nucleus/physiology , Gene Expression/genetics , Genome/genetics , Neurogenesis/physiology , Animals , Genetic Variation , Genome-Wide Association Study , Humans , Macaca , Mice , Mice, Inbred C57BL , Neural Stem Cells/physiology , Schizophrenia/genetics
SELECTION OF CITATIONS
SEARCH DETAIL