864 results on '"Kriegstein, Arnold R."'
Search Results
2. Single-cell analysis of prenatal and postnatal human cortical development
- Author
-
Velmeshev, Dmitry, Perez, Yonatan, Yan, Zihan, Valencia, Jonathan E, Castaneda-Castellanos, David R, Wang, Li, Schirmer, Lucas, Mayer, Simone, Wick, Brittney, Wang, Shaohui, Nowakowski, Tomasz Jan, Paredes, Mercedes, Huang, Eric J, and Kriegstein, Arnold R
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Genetics ,Mental Health ,Pediatric ,Stem Cell Research - Nonembryonic - Human ,Biotechnology ,Human Genome ,Intellectual and Developmental Disabilities (IDD) ,Neurosciences ,Stem Cell Research ,Autism ,Brain Disorders ,Stem Cell Research - Nonembryonic - Non-Human ,Underpinning research ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,Aetiology ,Neurological ,Female ,Humans ,Infant ,Newborn ,Pregnancy ,Brain Diseases ,Cerebral Cortex ,Gene Regulatory Networks ,Interneurons ,Neurons ,Single-Cell Analysis ,Male ,Risk Factors ,General Science & Technology - Abstract
We analyzed >700,000 single-nucleus RNA sequencing profiles from 106 donors during prenatal and postnatal developmental stages and identified lineage-specific programs that underlie the development of specific subtypes of excitatory cortical neurons, interneurons, glial cell types, and brain vasculature. By leveraging single-nucleus chromatin accessibility data, we delineated enhancer gene regulatory networks and transcription factors that control commitment of specific cortical lineages. By intersecting our results with genetic risk factors for human brain diseases, we identified the cortical cell types and lineages most vulnerable to genetic insults of different brain disorders, especially autism. We find that lineage-specific gene expression programs up-regulated in female cells are especially enriched for the genetic risk factors of autism. Our study captures the molecular progression of cortical lineages across human development.
- Published
- 2023
3. A cross-species proteomic map reveals neoteny of human synapse development
- Author
-
Wang, Li, Pang, Kaifang, Zhou, Li, Cebrián-Silla, Arantxa, González-Granero, Susana, Wang, Shaohui, Bi, Qiuli, White, Matthew L, Ho, Brandon, Li, Jiani, Li, Tao, Perez, Yonatan, Huang, Eric J, Winkler, Ethan A, Paredes, Mercedes F, Kovner, Rothem, Sestan, Nenad, Pollen, Alex A, Liu, Pengyuan, Li, Jingjing, Piao, Xianhua, García-Verdugo, José Manuel, Alvarez-Buylla, Arturo, Liu, Zhandong, and Kriegstein, Arnold R
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Neurosciences ,Biotechnology ,1.1 Normal biological development and functioning ,Underpinning research ,Neurological ,Adolescent ,Animals ,Child ,Child ,Preschool ,Humans ,Infant ,Infant ,Newborn ,Mice ,Young Adult ,Cognition ,Dendritic Spines ,Gestational Age ,Macaca ,Neurons ,Post-Synaptic Density ,Proteomics ,Rho Guanine Nucleotide Exchange Factors ,Signal Transduction ,Species Specificity ,Synapses ,General Science & Technology - 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.
- Published
- 2023
4. Osteopontin drives retinal ganglion cell resiliency in glaucomatous optic neuropathy
- Author
-
Zhao, Mengya, Toma, Kenichi, Kinde, Benyam, Li, Liang, Patel, Amit K, Wu, Kong-Yan, Lum, Matthew R, Tan, Chengxi, Hooper, Jody E, Kriegstein, Arnold R, La Torre, Anna, Liao, Yaping Joyce, Welsbie, Derek S, Hu, Yang, Han, Ying, and Duan, Xin
- Subjects
Biological Sciences ,Neurosciences ,Neurodegenerative ,Eye Disease and Disorders of Vision ,Aging ,Eye ,Humans ,Retinal Ganglion Cells ,Osteopontin ,Optic Nerve ,Optic Nerve Diseases ,Glaucoma ,CP: Neuroscience ,glaucoma ,human retina ,neuronal types ,neuroprotection ,optic nerve crush ,retinal ganglion cell ,Biochemistry and Cell Biology ,Medical Physiology ,Biological sciences - Abstract
Chronic neurodegeneration and acute injuries lead to neuron losses via diverse processes. We compared retinal ganglion cell (RGC) responses between chronic glaucomatous conditions and the acute injury model. Among major RGC subclasses, αRGCs and intrinsically photosensitive RGCs (ipRGCs) preferentially survive glaucomatous conditions, similar to findings in the retina subject to axotomy. Focusing on an αRGC intrinsic factor, Osteopontin (secreted phosphoprotein 1 [Spp1]), we found an ectopic neuronal expression of Osteopontin (Spp1) in other RGCs subject to glaucomatous conditions. This contrasted with the Spp1 downregulation subject to axotomy. αRGC-specific Spp1 elimination led to significant αRGC loss, diminishing their resiliency. Spp1 overexpression led to robust neuroprotection of susceptible RGC subclasses under glaucomatous conditions. In contrast, Spp1 overexpression did not significantly protect RGCs subject to axotomy. Additionally, SPP1 marked adult human RGC subsets with large somata and SPP1 expression in the aqueous humor correlated with glaucoma severity. Our study reveals Spp1's role in mediating neuronal resiliency in glaucoma.
- Published
- 2023
5. Protracted neuronal recruitment in the temporal lobes of young children
- Author
-
Nascimento, Marcos Assis, Biagiotti, Sean, Herranz-Pérez, Vicente, Santiago, Samara, Bueno, Raymund, Ye, Chun J., Abel, Taylor J., Zhang, Zhuangzhi, Rubio-Moll, Juan S., Kriegstein, Arnold R., Yang, Zhengang, Garcia-Verdugo, Jose Manuel, Huang, Eric J., Alvarez-Buylla, Arturo, and Sorrells, Shawn F.
- Published
- 2024
- Full Text
- View/download PDF
6. Non-muscle myosins control the integrity of cortical radial glial endfeet
- Author
-
Wang, Li and Kriegstein, Arnold R
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Stem Cell Research ,Brain Disorders ,Neurosciences ,Stem Cell Research - Nonembryonic - Non-Human ,Cancer ,1.1 Normal biological development and functioning ,Underpinning research ,Ependymoglial Cells ,Cerebral Cortex ,Interneurons ,Stem Cells ,Myosins ,Agricultural and Veterinary Sciences ,Medical and Health Sciences ,Developmental Biology ,Agricultural ,veterinary and food sciences ,Biological sciences ,Biomedical and clinical sciences - Abstract
Radial glial cells, the stem cells of the cerebral cortex, extend a long basal fiber that ends in basal endfeet. A new study in PLOS Biology found that non-muscle myosins control basal endfoot integrity to regulate interneuron organization.
- Published
- 2023
7. Zika virus alters centrosome organization to suppress the innate immune response
- Author
-
Kodani, Andrew, Knopp, Kristeene A, Di Lullo, Elizabeth, Retallack, Hanna, Kriegstein, Arnold R, DeRisi, Joseph L, and Reiter, Jeremy F
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Infectious Diseases ,2.1 Biological and endogenous factors ,Aetiology ,Good Health and Well Being ,Animals ,Cell Cycle Proteins ,Centrosome ,Humans ,Immunity ,Innate ,Microcephaly ,Zika Virus ,Zika Virus Infection ,centrosome ,innate immunity ,microcephaly ,Zika virus ,Developmental Biology ,Biochemistry and cell biology - Abstract
Zika virus (ZIKV) is a flavivirus transmitted via mosquitoes and sex to cause congenital neurodevelopmental defects, including microcephaly. Inherited forms of microcephaly (MCPH) are associated with disrupted centrosome organization. Similarly, we found that ZIKV infection disrupted centrosome organization. ZIKV infection disrupted the organization of centrosomal proteins including CEP63, a MCPH-associated protein. The ZIKV nonstructural protein NS3 bound CEP63, and expression of NS3 was sufficient to alter centrosome architecture and CEP63 localization. Loss of CEP63 suppressed ZIKV-induced centrosome disorganization, indicating that ZIKV requires CEP63 to disrupt centrosome organization. ZIKV infection or CEP63 loss decreased the centrosomal localization and stability of TANK-binding kinase 1 (TBK1), a regulator of the innate immune response. ZIKV infection also increased the centrosomal accumulation of the CEP63 interactor DTX4, a ubiquitin ligase that degrades TBK1. Therefore, we propose that ZIKV disrupts CEP63 function to increase centrosomal DTX4 localization and destabilization of TBK1, thereby tempering the innate immune response.
- Published
- 2022
8. Ensembles of endothelial and mural cells promote angiogenesis in prenatal human brain
- Author
-
Crouch, Elizabeth E, Bhaduri, Aparna, Andrews, Madeline G, Cebrian-Silla, Arantxa, Diafos, Loukas N, Birrueta, Janeth Ochoa, Wedderburn-Pugh, Kaylee, Valenzuela, Edward J, Bennett, Neal K, Eze, Ugomma C, Sandoval-Espinosa, Carmen, Chen, Jiapei, Mora, Cristina, Ross, Jayden M, Howard, Clare E, Gonzalez-Granero, Susana, Lozano, Jaime Ferrer, Vento, Maximo, Haeussler, Maximilian, Paredes, Mercedes F, Nakamura, Ken, Garcia-Verdugo, Jose Manuel, Alvarez-Buylla, Arturo, Kriegstein, Arnold R, and Huang, Eric J
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Biological Sciences ,Pediatric ,Neurosciences ,Stem Cell Research - Embryonic - Human ,Stem Cell Research ,Stem Cell Research - Nonembryonic - Non-Human ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,Underpinning research ,Aetiology ,Brain ,Collagen ,Endothelial Cells ,Humans ,Laminin ,Midkine ,Neovascularization ,Pathologic ,Neovascularization ,Physiologic ,Pericytes ,angiogenesis ,arterial endothelial cells ,blood brain barrier ,cortical organoids ,endothelial cells ,human prenatal brain development ,mural cells ,pericytes ,smooth muscle cells ,tip cells ,venous and capillary endothelial cells ,ventricular zone ,Human prenatal brain development ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
Interactions between angiogenesis and neurogenesis regulate embryonic brain development. However, a comprehensive understanding of the stages of vascular cell maturation is lacking, especially in the prenatal human brain. Using fluorescence-activated cell sorting, single-cell transcriptomics, and histological and ultrastructural analyses, we show that an ensemble of endothelial and mural cell subtypes tile the brain vasculature during the second trimester. These vascular cells follow distinct developmental trajectories and utilize diverse signaling mechanisms, including collagen, laminin, and midkine, to facilitate cell-cell communication and maturation. Interestingly, our results reveal that tip cells, a subtype of endothelial cells, are highly enriched near the ventricular zone, the site of active neurogenesis. Consistent with these observations, prenatal vascular cells transplanted into cortical organoids exhibit restricted lineage potential that favors tip cells, promotes neurogenesis, and reduces cellular stress. Together, our results uncover important mechanisms into vascular maturation during this critical period of human brain development.
- Published
- 2022
9. Tropism of SARS-CoV-2 for human cortical astrocytes
- Author
-
Andrews, Madeline G, Mukhtar, Tanzila, Eze, Ugomma C, Simoneau, Camille R, Ross, Jayden, Parikshak, Neelroop, Wang, Shaohui, Zhou, Li, Koontz, Mark, Velmeshev, Dmitry, Siebert, Clara-Vita, Gemenes, Kaila M, Tabata, Takako, Perez, Yonatan, Wang, Li, Mostajo-Radji, Mohammed A, de Majo, Martina, Donohue, Kevin C, Shin, David, Salma, Jahan, Pollen, Alex A, Nowakowski, Tomasz J, Ullian, Erik, Kumar, G Renuka, Winkler, Ethan A, Crouch, Elizabeth E, Ott, Melanie, and Kriegstein, Arnold R
- Subjects
Biodefense ,Pneumonia & Influenza ,Infectious Diseases ,Neurosciences ,Emerging Infectious Diseases ,Clinical Research ,Stem Cell Research ,Lung ,Prevention ,Pneumonia ,Vaccine Related ,1.1 Normal biological development and functioning ,Underpinning research ,Aetiology ,2.1 Biological and endogenous factors ,Angiotensin-Converting Enzyme 2 ,Astrocytes ,Cerebral Cortex ,Humans ,Organoids ,Primary Cell Culture ,SARS-CoV-2 ,Viral Tropism ,SARS-CoV-2 tropism ,astrocyte reactivity ,organoid models - 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.
- Published
- 2022
10. Perivascular neurons instruct 3D vascular lattice formation via neurovascular contact
- Author
-
Toma, Kenichi, Zhao, Mengya, Zhang, Shaobo, Wang, Fei, Graham, Hannah K., Zou, Jun, Modgil, Shweta, Shang, Wenhao H., Tsai, Nicole Y., Cai, Zhishun, Liu, Liping, Hong, Guiying, Kriegstein, Arnold R., Hu, Yang, Körbelin, Jakob, Zhang, Ruobing, Liao, Yaping Joyce, Kim, Tyson N., Ye, Xin, and Duan, Xin
- Published
- 2024
- Full Text
- View/download PDF
11. Nests of dividing neuroblasts sustain interneuron production for the developing human brain
- Author
-
Paredes, Mercedes F, Mora, Cristina, Flores-Ramirez, Quetzal, Cebrian-Silla, Arantxa, Del Dosso, Ashley, Larimer, Phil, Chen, Jiapei, Kang, Gugene, Gonzalez Granero, Susana, Garcia, Eric, Chu, Julia, Delgado, Ryan, Cotter, Jennifer A, Tang, Vivian, Spatazza, Julien, Obernier, Kirsten, Ferrer Lozano, Jaime, Vento, Maximo, Scott, Julia, Studholme, Colin, Nowakowski, Tomasz J, Kriegstein, Arnold R, Oldham, Michael C, Hasenstaub, Andrea, Garcia-Verdugo, Jose Manuel, Alvarez-Buylla, Arturo, and Huang, Eric J
- Subjects
Neurosciences ,Stem Cell Research ,Regenerative Medicine ,Neurological ,Animals ,Animals ,Newborn ,Cell Movement ,Cell Proliferation ,Cerebral Cortex ,GABAergic Neurons ,Gene Expression Profiling ,Gestational Age ,Humans ,Interneurons ,Median Eminence ,Mice ,Neural Stem Cells ,Neurogenesis ,Prosencephalon ,Transplantation ,Heterologous ,General Science & Technology - Abstract
The human cortex contains inhibitory interneurons derived from the medial ganglionic eminence (MGE), a germinal zone in the embryonic ventral forebrain. How this germinal zone generates sufficient interneurons for the human brain remains unclear. We found that the human MGE (hMGE) contains nests of proliferative neuroblasts with ultrastructural and transcriptomic features that distinguish them from other progenitors in the hMGE. When dissociated hMGE cells are transplanted into the neonatal mouse brain, they reform into nests containing proliferating neuroblasts that generate young neurons that migrate extensively into the mouse forebrain and mature into different subtypes of functional interneurons. Together, these results indicate that the nest organization and sustained proliferation of neuroblasts in the hMGE provide a mechanism for the extended production of interneurons for the human forebrain.
- Published
- 2022
12. Identification of Lipid Heterogeneity and Diversity in the Developing Human Brain
- Author
-
Bhaduri, Aparna, Neumann, Elizabeth K, Kriegstein, Arnold R, and Sweedler, Jonathan V
- Subjects
Analytical Chemistry ,Chemical Sciences ,Neurosciences ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,single cell analysis ,neurons ,astrocytes ,MALDI ,mass spectrometry ,development ,brain ,Chemical sciences - Abstract
The lipidome is currently understudied but fundamental to life. Within the brain, little is known about cell-type lipid heterogeneity, and even less is known about cell-to-cell lipid diversity because it is difficult to study the lipids within individual cells. Here, we used single-cell mass spectrometry-based protocols to profile the lipidomes of 154 910 single cells across ten individuals consisting of five developmental ages and five brain regions, resulting in a unique lipid atlas available via a web browser of the developing human brain. From these data, we identify differentially expressed lipids across brain structures, cortical areas, and developmental ages. We inferred lipid profiles of several major cell types from this data set and additionally detected putative cell-type specific lipids. This data set will enable further interrogation of the developing human brain lipidome.
- Published
- 2021
13. An atlas of cortical arealization identifies dynamic molecular signatures
- Author
-
Bhaduri, Aparna, Sandoval-Espinosa, Carmen, Otero-Garcia, Marcos, Oh, Irene, Yin, Raymund, Eze, Ugomma C, Nowakowski, Tomasz J, and Kriegstein, Arnold R
- Subjects
Stem Cell Research ,Biotechnology ,Genetics ,Stem Cell Research - Nonembryonic - Non-Human ,Neurosciences ,Mental Health ,1.1 Normal biological development and functioning ,Underpinning research ,Neurological ,Atlases as Topic ,Base Sequence ,Biomarkers ,Gene Expression Regulation ,Developmental ,Humans ,Neocortex ,Neurogenesis ,Neuroglia ,Neurons ,Reproducibility of Results ,Single-Cell Analysis ,Time Factors ,General Science & Technology - Abstract
The human brain is subdivided into distinct anatomical structures, including the neocortex, which in turn encompasses dozens of distinct specialized cortical areas. Early morphogenetic gradients are known to establish early brain regions and cortical areas, but how early patterns result in finer and more discrete spatial differences remains poorly understood1. Here we use single-cell RNA sequencing to profile ten major brain structures and six neocortical areas during peak neurogenesis and early gliogenesis. Within the neocortex, we find that early in the second trimester, a large number of genes are differentially expressed across distinct cortical areas in all cell types, including radial glia, the neural progenitors of the cortex. However, the abundance of areal transcriptomic signatures increases as radial glia differentiate into intermediate progenitor cells and ultimately give rise to excitatory neurons. Using an automated, multiplexed single-molecule fluorescent in situ hybridization approach, we find that laminar gene-expression patterns are highly dynamic across cortical regions. Together, our data suggest that early cortical areal patterning is defined by strong, mutually exclusive frontal and occipital gene-expression signatures, with resulting gradients giving rise to the specification of areas between these two poles throughout successive developmental timepoints.
- Published
- 2021
14. Distinct nuclear compartment-associated genome architecture in the developing mammalian brain
- Author
-
Ahanger, Sajad Hamid, Delgado, Ryan N, Gil, Eugene, Cole, Mitchel A, Zhao, Jingjing, Hong, Sung Jun, Kriegstein, Arnold R, Nowakowski, Tomasz J, Pollen, Alex A, and Lim, Daniel A
- Subjects
Mental Health ,Genetics ,Stem Cell Research - Embryonic - Non-Human ,Biotechnology ,Human Genome ,Neurosciences ,Stem Cell Research ,1.1 Normal biological development and functioning ,Underpinning research ,Neurological ,Animals ,Brain ,Cell Nucleus ,Gene Expression ,Genetic Variation ,Genome ,Genome-Wide Association Study ,Humans ,Macaca ,Mice ,Mice ,Inbred C57BL ,Neural Stem Cells ,Neurogenesis ,Schizophrenia ,Psychology ,Cognitive Sciences ,Neurology & Neurosurgery - 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.
- Published
- 2021
15. Human intermediate progenitor diversity during cortical development
- Author
-
Pebworth, Mark-Phillip, Ross, Jayden, Andrews, Madeline, Bhaduri, Aparna, and Kriegstein, Arnold R
- Subjects
Neurosciences ,Stem Cell Research ,Brain Disorders ,Stem Cell Research - Nonembryonic - Non-Human ,Stem Cell Research - Nonembryonic - Human ,Neurological ,Cerebral Cortex ,Green Fluorescent Proteins ,Humans ,Neural Stem Cells ,Neurogenesis ,Reproducibility of Results ,Sequence Analysis ,RNA ,Single-Cell Analysis ,Time Factors ,Transcriptome ,neuronal ,human ,cortex ,development ,progenitor - Abstract
Studies of the spatiotemporal, transcriptomic, and morphological diversity of radial glia (RG) have spurred our current models of human corticogenesis. In the developing cortex, neural intermediate progenitor cells (nIPCs) are a neuron-producing transit-amplifying cell type born in the germinal zones of the cortex from RG. The potential diversity of the nIPC population, that produces a significant portion of excitatory cortical neurons, is understudied, particularly in the developing human brain. Here we explore the spatiotemporal, transcriptomic, and morphological variation that exists within the human nIPC population and provide a resource for future studies. We observe that the spatial distribution of nIPCs in the cortex changes abruptly around gestational week (GW) 19/20, marking a distinct shift in cellular distribution and organization during late neurogenesis. We also identify five transcriptomic subtypes, one of which appears at this spatiotemporal transition. Finally, we observe a diversity of nIPC morphologies that do not correlate with specific transcriptomic subtypes. These results provide an analysis of the spatiotemporal, transcriptional, and morphological diversity of nIPCs in developing brain tissue and provide an atlas of nIPC subtypes in the developing human cortex that can benchmark in vitro models of human development such as cerebral organoids and help inform future studies of how nIPCs contribute to cortical neurogenesis.
- Published
- 2021
16. Single-cell atlas of early human brain development highlights heterogeneity of human neuroepithelial cells and early radial glia.
- Author
-
Eze, Ugomma C, Bhaduri, Aparna, Haeussler, Maximilian, Nowakowski, Tomasz J, and Kriegstein, Arnold R
- Subjects
Cerebral Cortex ,Neuroepithelial Cells ,Animals ,Humans ,Neurogenesis ,Neural Stem Cells ,Single-Cell Analysis ,Ependymoglial Cells ,Neurosciences ,Psychology ,Cognitive Sciences ,Neurology & Neurosurgery - Abstract
The human cortex comprises diverse cell types that emerge from an initially uniform neuroepithelium that gives rise to radial glia, the neural stem cells of the cortex. To characterize the earliest stages of human brain development, we performed single-cell RNA-sequencing across regions of the developing human brain, including the telencephalon, diencephalon, midbrain, hindbrain and cerebellum. We identify nine progenitor populations physically proximal to the telencephalon, suggesting more heterogeneity than previously described, including a highly prevalent mesenchymal-like population that disappears once neurogenesis begins. Comparison of human and mouse progenitor populations at corresponding stages identifies two progenitor clusters that are enriched in the early stages of human cortical development. We also find that organoid systems display low fidelity to neuroepithelial and early radial glia cell types, but improve as neurogenesis progresses. Overall, we provide a comprehensive molecular and spatial atlas of early stages of human brain and cortical development.
- Published
- 2021
17. Neurotoxic microglia promote TDP-43 proteinopathy in progranulin deficiency
- Author
-
Zhang, Jiasheng, Velmeshev, Dmitry, Hashimoto, Kei, Huang, Yu-Hsin, Hofmann, Jeffrey W, Shi, Xiaoyu, Chen, Jiapei, Leidal, Andrew M, Dishart, Julian G, Cahill, Michelle K, Kelley, Kevin W, Liddelow, Shane A, Seeley, William W, Miller, Bruce L, Walther, Tobias C, Farese, Robert V, Taylor, J Paul, Ullian, Erik M, Huang, Bo, Debnath, Jayanta, Wittmann, Torsten, Kriegstein, Arnold R, and Huang, Eric J
- Subjects
Rare Diseases ,Dementia ,Acquired Cognitive Impairment ,Biotechnology ,Neurodegenerative ,Brain Disorders ,Neurosciences ,Genetics ,Aetiology ,2.1 Biological and endogenous factors ,Aging ,Animals ,Cell Nucleus ,Complement Activation ,Complement C1q ,Complement C3b ,Culture Media ,Conditioned ,DNA-Binding Proteins ,Disease Models ,Animal ,Female ,Male ,Mice ,Microglia ,Neurons ,Nuclear Pore ,Progranulins ,RNA-Seq ,Single-Cell Analysis ,TDP-43 Proteinopathies ,Thalamus ,Transcriptome ,General Science & Technology - Abstract
Aberrant aggregation of the RNA-binding protein TDP-43 in neurons is a hallmark of frontotemporal lobar degeneration caused by haploinsufficiency in the gene encoding progranulin1,2. However, the mechanism leading to TDP-43 proteinopathy remains unclear. Here we use single-nucleus RNA sequencing to show that progranulin deficiency promotes microglial transition from a homeostatic to a disease-specific state that causes endolysosomal dysfunction and neurodegeneration in mice. These defects persist even when Grn-/- microglia are cultured ex vivo. In addition, single-nucleus RNA sequencing reveals selective loss of excitatory neurons at disease end-stage, which is characterized by prominent nuclear and cytoplasmic TDP-43 granules and nuclear pore defects. Remarkably, conditioned media from Grn-/- microglia are sufficient to promote TDP-43 granule formation, nuclear pore defects and cell death in excitatory neurons via the complement activation pathway. Consistent with these results, deletion of the genes encoding C1qa and C3 mitigates microglial toxicity and rescues TDP-43 proteinopathy and neurodegeneration. These results uncover previously unappreciated contributions of chronic microglial toxicity to TDP-43 proteinopathy during neurodegeneration.
- Published
- 2020
18. Identification of amygdala-expressed genes associated with autism spectrum disorder
- Author
-
Herrero, Maria Jesus, Velmeshev, Dmitry, Hernandez-Pineda, David, Sethi, Saarthak, Sorrells, Shawn, Banerjee, Payal, Sullivan, Catherine, Gupta, Abha R, Kriegstein, Arnold R, and Corbin, Joshua G
- Subjects
Biological Psychology ,Biomedical and Clinical Sciences ,Psychology ,Genetics ,Human Genome ,Pediatric ,Brain Disorders ,Neurosciences ,Intellectual and Developmental Disabilities (IDD) ,Mental Health ,Pediatric Research Initiative ,Autism ,Behavioral and Social Science ,Biotechnology ,Aetiology ,2.1 Biological and endogenous factors ,Mental health ,Alleles ,Amygdala ,Animals ,Autism Spectrum Disorder ,Biomarkers ,Brain ,Computational Biology ,Databases ,Genetic ,Disease Susceptibility ,Gene Expression ,Gene Expression Profiling ,Gene Ontology ,Gene Regulatory Networks ,Genetic Predisposition to Disease ,Humans ,Mice ,Signal Transduction ,Transcriptome ,Single nucleus RNA sequencing ,Brain development ,Autism spectrum disorder ,ASD genes ,Clinical Sciences ,Clinical sciences ,Biological psychology - Abstract
BackgroundStudies of individuals with autism spectrum disorder (ASD) have revealed a strong multigenic basis with the identification of hundreds of ASD susceptibility genes. ASD is characterized by social deficits and a range of other phenotypes, implicating complex genetics and involvement of a variety of brain regions. However, how mutations and mis-expression of select gene sets are associated with the behavioral components of ASD remains unknown. We reasoned that for genes to be associated with ASD core behaviors they must be: (1) expressed in brain regions relevant to ASD social behaviors and (2) expressed during the ASD susceptible window of brain development.MethodsFocusing on the amygdala, a brain region whose dysfunction has been highly implicated in the social component of ASD, we mined publicly available gene expression databases to identify ASD-susceptibility genes expressed during human and mouse amygdala development. We found that a large cohort of known ASD susceptibility genes is expressed in the developing human and mouse amygdala. We further performed analysis of single-nucleus RNA-seq (snRNA-seq) data from microdissected amygdala tissue from five ASD and five control human postmortem brains ranging in age from 4 to 20 years to elucidate cell type specificity of amygdala-expressed genes and their dysregulation in ASD.ResultsOur analyses revealed that of the high-ranking ASD susceptibility genes, 80 are expressed in both human and mouse amygdala during fetal to early postnatal stages of development. Our human snRNA-seq analyses revealed cohorts of genes with altered expression in the ASD amygdala postnatally, especially within excitatory neurons, with dysregulated expression of seven genes predicted from our datamining pipeline.LimitationsWe were limited by the ages for which we were able to obtain human tissue; therefore, the results from our datamining pipeline approach will require validation, to the extent possible, in human tissue from earlier developmental stages.ConclusionsOur pipeline narrows down the number of amygdala-expressed genes possibly involved in the social pathophysiology of ASD. Our human single-nucleus gene expression analyses revealed that ASD is characterized by changes in gene expression in specific cell types in the early postnatal amygdala.
- Published
- 2020
19. Cell-type-specific 3D epigenomes in the developing human cortex
- Author
-
Song, Michael, Pebworth, Mark-Phillip, Yang, Xiaoyu, Abnousi, Armen, Fan, Changxu, Wen, Jia, Rosen, Jonathan D, Choudhary, Mayank NK, Cui, Xiekui, Jones, Ian R, Bergenholtz, Seth, Eze, Ugomma C, Juric, Ivan, Li, Bingkun, Maliskova, Lenka, Lee, Jerry, Liu, Weifang, Pollen, Alex A, Li, Yun, Wang, Ting, Hu, Ming, Kriegstein, Arnold R, and Shen, Yin
- Subjects
Human Genome ,Genetics ,Biotechnology ,Stem Cell Research ,Neurosciences ,1.1 Normal biological development and functioning ,Underpinning research ,CRISPR-Cas Systems ,Cell Lineage ,Cells ,Cells ,Cultured ,Cerebral Cortex ,Chromatin ,DNA Transposable Elements ,Epigenome ,Epigenomics ,Histones ,Humans ,Imaging ,Three-Dimensional ,Methylation ,Multifactorial Inheritance ,Organogenesis ,Polymorphism ,Single Nucleotide ,Promoter Regions ,Genetic ,Regulatory Elements ,Transcriptional ,Reproducibility of Results ,Transcription ,Genetic ,General Science & Technology - Abstract
Lineage-specific epigenomic changes during human corticogenesis have been difficult to study owing to challenges with sample availability and tissue heterogeneity. For example, previous studies using single-cell RNA sequencing identified at least 9 major cell types and up to 26 distinct subtypes in the dorsal cortex alone1,2. Here we characterize cell-type-specific cis-regulatory chromatin interactions, open chromatin peaks, and transcriptomes for radial glia, intermediate progenitor cells, excitatory neurons, and interneurons isolated from mid-gestational samples of the human cortex. We show that chromatin interactions underlie several aspects of gene regulation, with transposable elements and disease-associated variants enriched at distal interacting regions in a cell-type-specific manner. In addition, promoters with increased levels of chromatin interactivity-termed super-interactive promoters-are enriched for lineage-specific genes, suggesting that interactions at these loci contribute to the fine-tuning of transcription. Finally, we develop CRISPRview, a technique that integrates immunostaining, CRISPR interference, RNAscope, and image analysis to validate cell-type-specific cis-regulatory elements in heterogeneous populations of primary cells. Our findings provide insights into cell-type-specific gene expression patterns in the developing human cortex and advance our understanding of gene regulation and lineage specification during this crucial developmental window.
- Published
- 2020
20. Single-Cell Analyses Identify Brain Mural Cells Expressing CD19 as Potential Off-Tumor Targets for CAR-T Immunotherapies
- Author
-
Parker, Kevin R, Migliorini, Denis, Perkey, Eric, Yost, Kathryn E, Bhaduri, Aparna, Bagga, Puneet, Haris, Mohammad, Wilson, Neil E, Liu, Fang, Gabunia, Khatuna, Scholler, John, Montine, Thomas J, Bhoj, Vijay G, Reddy, Ravinder, Mohan, Suyash, Maillard, Ivan, Kriegstein, Arnold R, June, Carl H, Chang, Howard Y, Posey, Avery D, and Satpathy, Ansuman T
- Subjects
Biomedical and Clinical Sciences ,Immunology ,Immunization ,Neurosciences ,Vaccine Related ,Rare Diseases ,Biotechnology ,Genetics ,Aetiology ,2.1 Biological and endogenous factors ,Development of treatments and therapeutic interventions ,5.2 Cellular and gene therapies ,Neurological ,Animals ,Antibodies ,Bispecific ,Antigens ,CD19 ,B-Lymphocytes ,Blood-Brain Barrier ,Brain ,Cell Line ,Tumor ,Cytotoxicity ,Immunologic ,Epithelial Cells ,Humans ,Immunotherapy ,Immunotherapy ,Adoptive ,Mice ,Mice ,Inbred NOD ,Mice ,SCID ,Muscle ,Smooth ,Vascular ,Neoplasms ,Receptors ,Antigen ,T-Cell ,Receptors ,Chimeric Antigen ,Single-Cell Analysis ,T-Lymphocytes ,Xenograft Model Antitumor Assays ,Biological Sciences ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
CD19-directed immunotherapies are clinically effective for treating B cell malignancies but also cause a high incidence of neurotoxicity. A subset of patients treated with chimeric antigen receptor (CAR) T cells or bispecific T cell engager (BiTE) antibodies display severe neurotoxicity, including fatal cerebral edema associated with T cell infiltration into the brain. Here, we report that mural cells, which surround the endothelium and are critical for blood-brain-barrier integrity, express CD19. We identify CD19 expression in brain mural cells using single-cell RNA sequencing data and confirm perivascular staining at the protein level. CD19 expression in the brain begins early in development alongside the emergence of mural cell lineages and persists throughout adulthood across brain regions. Mouse mural cells demonstrate lower levels of Cd19 expression, suggesting limitations in preclinical animal models of neurotoxicity. These data suggest an on-target mechanism for neurotoxicity in CD19-directed therapies and highlight the utility of human single-cell atlases for designing immunotherapies.
- Published
- 2020
21. mTOR signaling regulates the morphology and migration of outer radial glia in developing human cortex.
- Author
-
Andrews, Madeline G, Subramanian, Lakshmi, and Kriegstein, Arnold R
- Subjects
human ,human cortex ,neuroscience ,organoids ,outer radial glia ,regenerative medicine ,stem cells ,Biochemistry and Cell Biology - Abstract
Outer radial glial (oRG) cells are a population of neural stem cells prevalent in the developing human cortex that contribute to its cellular diversity and evolutionary expansion. The mammalian Target of Rapamycin (mTOR) signaling pathway is active in human oRG cells. Mutations in mTOR pathway genes are linked to a variety of neurodevelopmental disorders and malformations of cortical development. We find that dysregulation of mTOR signaling specifically affects oRG cells, but not other progenitor types, by changing the actin cytoskeleton through the activity of the Rho-GTPase, CDC42. These effects change oRG cellular morphology, migration, and mitotic behavior, but do not affect proliferation or cell fate. Thus, mTOR signaling can regulate the architecture of the developing human cortex by maintaining the cytoskeletal organization of oRG cells and the radial glia scaffold. Our study provides insight into how mTOR dysregulation may contribute to neurodevelopmental disease.
- Published
- 2020
22. Are Organoids Ready for Prime Time?
- Author
-
Bhaduri, Aparna, Andrews, Madeline G, Kriegstein, Arnold R, and Nowakowski, Tomasz J
- Subjects
Organoids ,Humans ,Neurosciences ,Brain Disorders ,Good Health and Well Being ,Biological Sciences ,Medical and Health Sciences ,Developmental Biology - Abstract
Innovations in organoid-based models of human tissues have made them an exciting experimental platform for studying development and disease. However, these models require systematic benchmarking against primary tissue to establish their value. We discuss key parameters that impact the utility of organoid models, primarily focusing on cerebral organoids as examples.
- Published
- 2020
23. Origins and Proliferative States of Human Oligodendrocyte Precursor Cells.
- Author
-
Huang, Wei, Bhaduri, Aparna, Velmeshev, Dmitry, Wang, Shaohui, Wang, Li, Rottkamp, Catherine A, Alvarez-Buylla, Arturo, Rowitch, David H, and Kriegstein, Arnold R
- Subjects
Cerebral Cortex ,Cells ,Cultured ,Humans ,Cadherins ,RNA ,Small Interfering ,Immunohistochemistry ,Cell Proliferation ,Neurogenesis ,HEK293 Cells ,Single-Cell Analysis ,Ependymoglial Cells ,White Matter ,ErbB Receptors ,Cadherin Related Proteins ,Oligodendrocyte Precursor Cells ,RNA-Seq ,EGFR ,OPC ,PCDH15 ,cortical expansion ,oligodendrogenesis ,self-repulsion ,Neurosciences ,Regenerative Medicine ,Stem Cell Research - Nonembryonic - Human ,Stem Cell Research ,Stem Cell Research - Nonembryonic - Non-Human ,1.1 Normal biological development and functioning ,Underpinning research ,Neurological ,Biological Sciences ,Medical and Health Sciences ,Developmental Biology - Abstract
Human cerebral cortex size and complexity has increased greatly during evolution. While increased progenitor diversity and enhanced proliferative potential play important roles in human neurogenesis and gray matter expansion, the mechanisms of human oligodendrogenesis and white matter expansion remain largely unknown. Here, we identify EGFR-expressing "Pre-OPCs" that originate from outer radial glial cells (oRGs) and undergo mitotic somal translocation (MST) during division. oRG-derived Pre-OPCs provide an additional source of human cortical oligodendrocyte precursor cells (OPCs) and define a lineage trajectory. We further show that human OPCs undergo consecutive symmetric divisions to exponentially increase the progenitor pool size. Additionally, we find that the OPC-enriched gene, PCDH15, mediates daughter cell repulsion and facilitates proliferation. These findings indicate properties of OPC derivation, proliferation, and dispersion important for human white matter expansion and myelination.
- Published
- 2020
24. A Chromatin Accessibility Atlas of the Developing Human Telencephalon
- Author
-
Markenscoff-Papadimitriou, Eirene, Whalen, Sean, Przytycki, Pawel, Thomas, Reuben, Binyameen, Fadya, Nowakowski, Tomasz J, Kriegstein, Arnold R, Sanders, Stephan J, State, Matthew W, Pollard, Katherine S, and Rubenstein, John L
- Subjects
Neurosciences ,Genetics ,Brain Disorders ,Mental Health ,Underpinning research ,1.1 Normal biological development and functioning ,Animals ,Autistic Disorder ,Cell Line ,Chromatin ,Chromatin Immunoprecipitation Sequencing ,Enhancer Elements ,Genetic ,Euchromatin ,GABA Plasma Membrane Transport Proteins ,Gene Expression Regulation ,Developmental ,Gene Ontology ,Genetic Predisposition to Disease ,Gestational Age ,Humans ,Mice ,Mice ,Transgenic ,Nucleotide Motifs ,Point Mutation ,Prefrontal Cortex ,Repressor Proteins ,Spatio-Temporal Analysis ,Telencephalon ,Transcription Factors ,ATAC-seq ,autism ,chromatin ,enhancers ,gene regulation ,machine learning ,neurodevelopment ,neuropsychiatric disorders ,Biological Sciences ,Medical and Health Sciences ,Developmental Biology - Abstract
To discover regulatory elements driving the specificity of gene expression in different cell types and regions of the developing human brain, we generated an atlas of open chromatin from nine dissected regions of the mid-gestation human telencephalon, as well as microdissected upper and deep layers of the prefrontal cortex. We identified a subset of open chromatin regions (OCRs), termed predicted regulatory elements (pREs), that are likely to function as developmental brain enhancers. pREs showed temporal, regional, and laminar differences in chromatin accessibility and were correlated with gene expression differences across regions and gestational ages. We identified two functional de novo variants in a pRE for autism risk gene SLC6A1, and using CRISPRa, demonstrated that this pRE regulates SCL6A1. Additionally, mouse transgenic experiments validated enhancer activity for pREs proximal to FEZF2 and BCL11A. Thus, this atlas serves as a resource for decoding neurodevelopmental gene regulation in health and disease.
- Published
- 2020
25. Cortical Neural Stem Cell Lineage Progression Is Regulated by Extrinsic Signaling Molecule Sonic Hedgehog
- Author
-
Zhang, Yue, Liu, Guoping, Guo, Teng, Liang, Xiaoyi G, Du, Heng, Yang, Lin, Bhaduri, Aparna, Li, Xiaosu, Xu, Zhejun, Zhang, Zhuangzhi, Li, Zhenmeiyu, He, Miao, Tsyporin, Jeremiah, Kriegstein, Arnold R, Rubenstein, John L, Yang, Zhengang, and Chen, Bin
- Subjects
Neurosciences ,Stem Cell Research ,Stem Cell Research - Nonembryonic - Non-Human ,Regenerative Medicine ,1.1 Normal biological development and functioning ,Underpinning research ,Neurological ,Animals ,Astrocytes ,Biomarkers ,Cell Lineage ,Embryo ,Mammalian ,Hedgehog Proteins ,Homeodomain Proteins ,Interneurons ,Mice ,Inbred C57BL ,Neocortex ,Nerve Tissue Proteins ,Neural Stem Cells ,Neurogenesis ,Neuroglia ,Olfactory Bulb ,Oligodendroglia ,Pyramidal Cells ,Reproducibility of Results ,Signal Transduction ,Zinc Finger Protein Gli3 ,Gli3 ,Gsx2 ,Shh ,cerebral cortex ,neural stem cells ,olfactory bulb interneurons ,oligodendrocytes ,Biochemistry and Cell Biology ,Medical Physiology - Abstract
Neural stem cells (NSCs) in the prenatal neocortex progressively generate different subtypes of glutamatergic projection neurons. Following that, NSCs have a major switch in their progenitor properties and produce γ-aminobutyric acid (GABAergic) interneurons for the olfactory bulb (OB), cortical oligodendrocytes, and astrocytes. Herein, we provide evidence for the molecular mechanism that underlies this switch in the state of neocortical NSCs. We show that, at around E16.5, mouse neocortical NSCs start to generate GSX2-expressing (GSX2+) intermediate progenitor cells (IPCs). In vivo lineage-tracing study revealed that GSX2+ IPC population gives rise not only to OB interneurons but also to cortical oligodendrocytes and astrocytes, suggesting that they are a tri-potential population. We demonstrated that Sonic hedgehog signaling is both necessary and sufficient for the generation of GSX2+ IPCs by reducing GLI3R protein levels. Using single-cell RNA sequencing, we identify the transcriptional profile of GSX2+ IPCs and the process of the lineage switch of cortical NSCs.
- Published
- 2020
26. Cell stress in cortical organoids impairs molecular subtype specification
- Author
-
Bhaduri, Aparna, Andrews, Madeline G, Mancia Leon, Walter, Jung, Diane, Shin, David, Allen, Denise, Jung, Dana, Schmunk, Galina, Haeussler, Maximilian, Salma, Jahan, Pollen, Alex A, Nowakowski, Tomasz J, and Kriegstein, Arnold R
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Biological Sciences ,Biotechnology ,Stem Cell Research ,Pediatric ,Stem Cell Research - Nonembryonic - Non-Human ,Stem Cell Research - Nonembryonic - Human ,Neurosciences ,1.1 Normal biological development and functioning ,Underpinning research ,Neurological ,Good Health and Well Being ,Cerebral Cortex ,Humans ,Neurogenesis ,Neurons ,Organoids ,Single-Cell Analysis ,Stress ,Physiological ,Tissue Culture Techniques ,General Science & Technology - Abstract
Cortical organoids are self-organizing three-dimensional cultures that model features of the developing human cerebral cortex1,2. However, the fidelity of organoid models remains unclear3-5. Here we analyse the transcriptomes of individual primary human cortical cells from different developmental periods and cortical areas. We find that cortical development is characterized by progenitor maturation trajectories, the emergence of diverse cell subtypes and areal specification of newborn neurons. By contrast, organoids contain broad cell classes, but do not recapitulate distinct cellular subtype identities and appropriate progenitor maturation. Although the molecular signatures of cortical areas emerge in organoid neurons, they are not spatially segregated. Organoids also ectopically activate cellular stress pathways, which impairs cell-type specification. However, organoid stress and subtype defects are alleviated by transplantation into the mouse cortex. Together, these datasets and analytical tools provide a framework for evaluating and improving the accuracy of cortical organoids as models of human brain development.
- Published
- 2020
27. SMART-Q: An Integrative Pipeline Quantifying Cell Type-Specific RNA Transcription
- Author
-
Yang, Xiaoyu, Bergenholtz, Seth, Maliskova, Lenka, Pebworth, Mark-Phillip, Kriegstein, Arnold R, Li, Yun, and Shen, Yin
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Genetics ,Biotechnology ,Cell Nucleus ,Humans ,Image Processing ,Computer-Assisted ,Quality Control ,RNA ,RNA ,Messenger ,Software ,Transcription ,Genetic ,General Science & Technology - Abstract
Accurate RNA quantification at the single-cell level is critical for understanding the dynamics of gene expression and regulation across space and time. Single molecule FISH (smFISH), such as RNAscope, provides spatial and quantitative measurements of individual transcripts, therefore, can be used to explore differential gene expression among a heterogeneous cell population if combined with cell identify information. However, such analysis is not straightforward, and existing image analysis pipelines cannot integrate both RNA transcripts and cellular staining information to automatically output cell type-specific gene expression. We developed an efficient and customizable analysis method, Single-Molecule Automatic RNA Transcription Quantification (SMART-Q), to enable the analysis of gene transcripts in a cell type-specific manner. SMART-Q efficiently infers cell identity information from multiplexed immuno-staining and quantifies cell type-specific transcripts using a 3D Gaussian fitting algorithm. Furthermore, we have optimized SMART-Q for user experiences, such as flexible parameters specification, batch data outputs, and visualization of analysis results. SMART-Q meets the demands for efficient quantification of single-molecule RNA and can be widely used for cell type-specific RNA transcript analysis.
- Published
- 2020
28. How mechanisms of stem cell polarity shape the human cerebral cortex
- Author
-
Andrews, Madeline G., Subramanian, Lakshmi, Salma, Jahan, and Kriegstein, Arnold R.
- Published
- 2022
- Full Text
- View/download PDF
29. A nomenclature consensus for nervous system organoids and assembloids
- Author
-
Pașca, Sergiu P., Arlotta, Paola, Bateup, Helen S., Camp, J. Gray, Cappello, Silvia, Gage, Fred H., Knoblich, Jürgen A., Kriegstein, Arnold R., Lancaster, Madeline A., Ming, Guo-Li, Muotri, Alysson R., Park, In-Hyun, Reiner, Orly, Song, Hongjun, Studer, Lorenz, Temple, Sally, Testa, Giuseppe, Treutlein, Barbara, and Vaccarino, Flora M.
- Published
- 2022
- Full Text
- View/download PDF
30. Neuroglial stem cell-derived inflammatory pseudotumor (n-SCIPT): clinicopathologic characterization of a novel lesion of the lumbosacral spinal cord and nerve roots following intrathecal allogeneic stem cell intervention.
- Author
-
Sloan, Emily A, Sampognaro, Paul J, Junn, Jacqueline C, Chin, Cynthia, Jacques, Line, Ramachandran, Prashanth S, DeRisi, Joseph L, Wilson, Michael R, Kriegstein, Arnold R, Bollen, Andrew W, Solomon, David A, Margeta, Marta, and Engstrom, John W
- Subjects
Lumbosacral Region ,Neuroglia ,Spinal Nerve Roots ,Humans ,Spinal Cord Diseases ,Peripheral Nervous System Diseases ,Granuloma ,Plasma Cell ,Stem Cell Transplantation ,Transplantation ,Homologous ,Injections ,Spinal ,Middle Aged ,Male ,Neurology & Neurosurgery ,Clinical Sciences ,Neurosciences - Published
- 2019
31. Oligodendrocyte Death in Pelizaeus-Merzbacher Disease Is Rescued by Iron Chelation
- Author
-
Nobuta, Hiroko, Yang, Nan, Ng, Yi Han, Marro, Samuele G, Sabeur, Khalida, Chavali, Manideep, Stockley, John H, Killilea, David W, Walter, Patrick B, Zhao, Chao, Huie, Philip, Goldman, Steven A, Kriegstein, Arnold R, Franklin, Robin JM, Rowitch, David H, and Wernig, Marius
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Biological Sciences ,Autoimmune Disease ,Neurosciences ,Multiple Sclerosis ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Stem Cell Research - Embryonic - Human ,Stem Cell Research ,Genetics ,Neurodegenerative ,Rare Diseases ,Brain Disorders ,Stem Cell Research - Induced Pluripotent Stem Cell ,Aetiology ,2.1 Biological and endogenous factors ,Neurological ,Animals ,Cell Differentiation ,Cells ,Cultured ,Deferiprone ,Ferroptosis ,Humans ,Induced Pluripotent Stem Cells ,Iron ,Iron Chelating Agents ,Lipid Peroxidation ,Mice ,Mice ,Mutant Strains ,Mutation ,Myelin Proteolipid Protein ,Oligodendroglia ,Pelizaeus-Merzbacher Disease ,Stem Cell Transplantation ,Targeted Gene Repair ,ferroptosis ,gene correction ,induced pluripotent stem cells ,iron chelation ,leukodystrophy ,myelination ,oligodendrocyte ,patient models ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
Pelizaeus-Merzbacher disease (PMD) is an X-linked leukodystrophy caused by mutations in Proteolipid Protein 1 (PLP1), encoding a major myelin protein, resulting in profound developmental delay and early lethality. Previous work showed involvement of unfolded protein response (UPR) and endoplasmic reticulum (ER) stress pathways, but poor PLP1 genotype-phenotype associations suggest additional pathogenetic mechanisms. Using induced pluripotent stem cell (iPSC) and gene-correction, we show that patient-derived oligodendrocytes can develop to the pre-myelinating stage, but subsequently undergo cell death. Mutant oligodendrocytes demonstrated key hallmarks of ferroptosis including lipid peroxidation, abnormal iron metabolism, and hypersensitivity to free iron. Iron chelation rescued mutant oligodendrocyte apoptosis, survival, and differentiationin vitro, and post-transplantation in vivo. Finally, systemic treatment of Plp1 mutant Jimpy mice with deferiprone, a small molecule iron chelator, reduced oligodendrocyte apoptosis and enabled myelin formation. Thus, oligodendrocyte iron-induced cell death and myelination is rescued by iron chelation in PMD pre-clinical models.
- Published
- 2019
32. Neuroserpin expression during human brain development and in adult brain revealed by immunohistochemistry and single cell RNA sequencing
- Author
-
Adorjan, Istvan, Tyler, Teadora, Bhaduri, Aparna, Demharter, Samuel, Finszter, Cintia Klaudia, Bako, Maria, Sebok, Oliver Marcell, Nowakowski, Tomasz J, Khodosevich, Konstantin, Møllgård, Kjeld, Kriegstein, Arnold R, Shi, Lei, Hoerder‐Suabedissen, Anna, Ansorge, Olaf, and Molnár, Zoltán
- Subjects
Zoology ,Biological Sciences ,Neurosciences ,Epilepsy ,Stroke ,Pediatric ,Brain Disorders ,Neurodegenerative ,Neurological ,Brain ,Humans ,Immunohistochemistry ,Neuropeptides ,Sequence Analysis ,RNA ,Serpins ,Single-Cell Analysis ,human brain ,neurodevelopment ,neuroserpin ,subplate ,Biomedical Engineering ,Medical Physiology ,Anatomy & Morphology - Abstract
Neuroserpin is a serine-protease inhibitor mainly expressed in the CNS and involved in the inhibition of the proteolytic cascade. Animal models confirmed its neuroprotective role in perinatal hypoxia-ischaemia and adult stroke. Although neuroserpin may be a potential therapeutic target in the treatment of the aforementioned conditions, there is still no information in the literature on its distribution during human brain development. The present study provides a detailed description of the changing spatiotemporal patterns of neuroserpin focusing on physiological human brain development. Five stages were distinguished within our examined age range which spanned from the 7th gestational week until adulthood. In particular, subplate and deep cortical plate neurons were identified as the main sources of neuroserpin production between the 25th gestational week and the first postnatal month. Our immunohistochemical findings were substantiated by single cell RNA sequencing data showing specific neuronal and glial cell types expressing neuroserpin. The characterization of neuroserpin expression during physiological human brain development is essential for forthcoming studies which will explore its involvement in pathological conditions, such as perinatal hypoxia-ischaemia and adult stroke in human.
- Published
- 2019
33. Neuronal vulnerability and multilineage diversity in multiple sclerosis
- Author
-
Schirmer, Lucas, Velmeshev, Dmitry, Holmqvist, Staffan, Kaufmann, Max, Werneburg, Sebastian, Jung, Diane, Vistnes, Stephanie, Stockley, John H, Young, Adam, Steindel, Maike, Tung, Brian, Goyal, Nitasha, Bhaduri, Aparna, Mayer, Simone, Engler, Jan Broder, Bayraktar, Omer A, Franklin, Robin JM, Haeussler, Maximilian, Reynolds, Richard, Schafer, Dorothy P, Friese, Manuel A, Shiow, Lawrence R, Kriegstein, Arnold R, and Rowitch, David H
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Autoimmune Disease ,Human Genome ,Multiple Sclerosis ,Brain Disorders ,Neurosciences ,Genetics ,Neurodegenerative ,Biotechnology ,Aetiology ,2.1 Biological and endogenous factors ,Neurological ,Adult ,Animals ,Astrocytes ,Autopsy ,Cell Lineage ,Cryopreservation ,Female ,Homeodomain Proteins ,Humans ,Macrophages ,Male ,Mice ,Microglia ,Middle Aged ,Myelin Sheath ,Neurons ,Oligodendroglia ,Phagocytosis ,RNA ,Small Nuclear ,RNA-Seq ,Transcriptome ,General Science & Technology - Abstract
Multiple sclerosis (MS) is a neuroinflammatory disease with a relapsing-remitting disease course at early stages, distinct lesion characteristics in cortical grey versus subcortical white matter and neurodegeneration at chronic stages. Here we used single-nucleus RNA sequencing to assess changes in expression in multiple cell lineages in MS lesions and validated the results using multiplex in situ hybridization. We found selective vulnerability and loss of excitatory CUX2-expressing projection neurons in upper-cortical layers underlying meningeal inflammation; such MS neuron populations exhibited upregulation of stress pathway genes and long non-coding RNAs. Signatures of stressed oligodendrocytes, reactive astrocytes and activated microglia mapped most strongly to the rim of MS plaques. Notably, single-nucleus RNA sequencing identified phagocytosing microglia and/or macrophages by their ingestion and perinuclear import of myelin transcripts, confirmed by functional mouse and human culture assays. Our findings indicate lineage- and region-specific transcriptomic changes associated with selective cortical neuron damage and glial activation contributing to progression of MS lesions.
- Published
- 2019
34. Immature excitatory neurons develop during adolescence in the human amygdala.
- Author
-
Sorrells, Shawn F, Paredes, Mercedes F, Velmeshev, Dmitry, Herranz-Pérez, Vicente, Sandoval, Kadellyn, Mayer, Simone, Chang, Edward F, Insausti, Ricardo, Kriegstein, Arnold R, Rubenstein, John L, Manuel Garcia-Verdugo, Jose, Huang, Eric J, and Alvarez-Buylla, Arturo
- Subjects
Hippocampus ,Neurons ,Cell Nucleus ,Fetus ,Humans ,Sequence Analysis ,RNA ,Adolescent Development ,Neuronal Plasticity ,Adolescent ,Adult ,Aged ,Middle Aged ,Child ,Child ,Preschool ,Infant ,Infant ,Newborn ,Male ,Neurogenesis ,Young Adult ,Neural Stem Cells ,Single-Cell Analysis ,Basolateral Nuclear Complex - Abstract
The human amygdala grows during childhood, and its abnormal development is linked to mood disorders. The primate amygdala contains a large population of immature neurons in the paralaminar nuclei (PL), suggesting protracted development and possibly neurogenesis. Here we studied human PL development from embryonic stages to adulthood. The PL develops next to the caudal ganglionic eminence, which generates inhibitory interneurons, yet most PL neurons express excitatory markers. In children, most PL cells are immature (DCX+PSA-NCAM+), and during adolescence many transition into mature (TBR1+VGLUT2+) neurons. Immature PL neurons persist into old age, yet local progenitor proliferation sharply decreases in infants. Using single nuclei RNA sequencing, we identify the transcriptional profile of immature excitatory neurons in the human amygdala between 4-15 years. We conclude that the human PL contains excitatory neurons that remain immature for decades, a possible substrate for persistent plasticity at the interface of the hippocampus and amygdala.
- Published
- 2019
35. Single-cell genomics identifies cell type–specific molecular changes in autism
- Author
-
Velmeshev, Dmitry, Schirmer, Lucas, Jung, Diane, Haeussler, Maximilian, Perez, Yonatan, Mayer, Simone, Bhaduri, Aparna, Goyal, Nitasha, Rowitch, David H, and Kriegstein, Arnold R
- Subjects
Mental Health ,Brain Disorders ,Human Genome ,Pediatric ,Genetics ,Autism ,Neurosciences ,Behavioral and Social Science ,Intellectual and Developmental Disabilities (IDD) ,Biotechnology ,Aetiology ,2.1 Biological and endogenous factors ,Neurological ,Adolescent ,Autistic Disorder ,Cell Nucleus ,Child ,Child ,Preschool ,Female ,Gene Expression Profiling ,Gene Expression Regulation ,Genomics ,Humans ,Male ,Microglia ,Neocortex ,Neurons ,Sequence Analysis ,RNA ,Single-Cell Analysis ,Young Adult ,General Science & Technology - Abstract
Despite the clinical and genetic heterogeneity of autism, bulk gene expression studies show that changes in the neocortex of autism patients converge on common genes and pathways. However, direct assessment of specific cell types in the brain affected by autism has not been feasible until recently. We used single-nucleus RNA sequencing of cortical tissue from patients with autism to identify autism-associated transcriptomic changes in specific cell types. We found that synaptic signaling of upper-layer excitatory neurons and the molecular state of microglia are preferentially affected in autism. Moreover, our results show that dysregulation of specific groups of genes in cortico-cortical projection neurons correlates with clinical severity of autism. These findings suggest that molecular changes in upper-layer cortical circuits are linked to behavioral manifestations of autism.
- Published
- 2019
36. Multimodal Single-Cell Analysis Reveals Physiological Maturation in the Developing Human Neocortex.
- Author
-
Mayer, Simone, Chen, Jiadong, Velmeshev, Dmitry, Mayer, Andreas, Eze, Ugomma C, Bhaduri, Aparna, Cunha, Carlos E, Jung, Diane, Arjun, Arpana, Li, Emmy, Alvarado, Beatriz, Wang, Shaohui, Lovegren, Nils, Gonzales, Michael L, Szpankowski, Lukasz, Leyrat, Anne, West, Jay AA, Panagiotakos, Georgia, Alvarez-Buylla, Arturo, Paredes, Mercedes F, Nowakowski, Tomasz J, Pollen, Alex A, and Kriegstein, Arnold R
- Subjects
Brain ,Neocortex ,Animals ,Humans ,Mice ,Calcium ,Serotonin ,Receptor ,Serotonin ,5-HT2A ,Gene Expression Profiling ,Sequence Analysis ,RNA ,Gene Expression Regulation ,Developmental ,Cell Lineage ,Neurogenesis ,Single-Cell Analysis ,Ependymoglial Cells ,calcium imaging ,differentiation ,human neocortical development ,intermediate progenitor cells ,neurogenesis ,neurotransmitter ,radial glia ,radial glia scaffold ,serotonin ,single-cell RNA sequencing ,Neurosciences ,Stem Cell Research ,Pediatric ,Substance Misuse ,Stem Cell Research - Nonembryonic - Non-Human ,Underpinning research ,1.1 Normal biological development and functioning ,Psychology ,Cognitive Sciences ,Neurology & Neurosurgery - Abstract
In the developing human neocortex, progenitor cells generate diverse cell types prenatally. Progenitor cells and newborn neurons respond to signaling cues, including neurotransmitters. While single-cell RNA sequencing has revealed cellular diversity, physiological heterogeneity has yet to be mapped onto these developing and diverse cell types. By combining measurements of intracellular Ca2+ elevations in response to neurotransmitter receptor agonists and RNA sequencing of the same single cells, we show that Ca2+ responses are cell-type-specific and change dynamically with lineage progression. Physiological response properties predict molecular cell identity and additionally reveal diversity not captured by single-cell transcriptomics. We find that the serotonin receptor HTR2A selectively activates radial glia cells in the developing human, but not mouse, neocortex, and inhibiting HTR2A receptors in human radial glia disrupts the radial glial scaffold. We show highly specific neurotransmitter signaling during neurogenesis in the developing human neocortex and highlight evolutionarily divergent mechanisms of physiological signaling.
- Published
- 2019
37. Establishing Cerebral Organoids as Models of Human-Specific Brain Evolution.
- Author
-
Pollen, Alex A, Bhaduri, Aparna, Andrews, Madeline G, Nowakowski, Tomasz J, Meyerson, Olivia S, Mostajo-Radji, Mohammed A, Di Lullo, Elizabeth, Alvarado, Beatriz, Bedolli, Melanie, Dougherty, Max L, Fiddes, Ian T, Kronenberg, Zev N, Shuga, Joe, Leyrat, Anne A, West, Jay A, Bershteyn, Marina, Lowe, Craig B, Pavlovic, Bryan J, Salama, Sofie R, Haussler, David, Eichler, Evan E, and Kriegstein, Arnold R
- Subjects
Brain ,Cerebral Cortex ,Organoids ,Pluripotent Stem Cells ,Animals ,Macaca ,Humans ,Pan troglodytes ,Cell Culture Techniques ,Cell Differentiation ,Species Specificity ,Gene Regulatory Networks ,Neurogenesis ,Induced Pluripotent Stem Cells ,Biological Evolution ,Single-Cell Analysis ,Transcriptome ,cerebral organoids ,chimpanzee ,cortical development ,human-specific evolution ,mTOR ,macaque ,neural progenitor cells ,radial glia ,single-cell RNA sequencing ,Stem Cell Research - Induced Pluripotent Stem Cell ,Neurosciences ,Pediatric ,Stem Cell Research ,Stem Cell Research - Nonembryonic - Human ,Brain Disorders ,Regenerative Medicine ,Stem Cell Research - Embryonic - Human ,Stem Cell Research - Nonembryonic - Non-Human ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Underpinning research ,1.1 Normal biological development and functioning ,Neurological ,Biological Sciences ,Medical and Health Sciences ,Developmental Biology - Abstract
Direct comparisons of human and non-human primate brains can reveal molecular pathways underlying remarkable specializations of the human brain. However, chimpanzee tissue is inaccessible during neocortical neurogenesis when differences in brain size first appear. To identify human-specific features of cortical development, we leveraged recent innovations that permit generating pluripotent stem cell-derived cerebral organoids from chimpanzee. Despite metabolic differences, organoid models preserve gene regulatory networks related to primary cell types and developmental processes. We further identified 261 differentially expressed genes in human compared to both chimpanzee organoids and macaque cortex, enriched for recent gene duplications, and including multiple regulators of PI3K-AKT-mTOR signaling. We observed increased activation of this pathway in human radial glia, dependent on two receptors upregulated specifically in human: INSR and ITGB8. Our findings establish a platform for systematic analysis of molecular changes contributing to human brain development and evolution.
- Published
- 2019
38. Identification of cell types in a mouse brain single-cell atlas using low sampling coverage
- Author
-
Bhaduri, Aparna, Nowakowski, Tomasz J, Pollen, Alex A, and Kriegstein, Arnold R
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Genetics ,Bioengineering ,1.1 Normal biological development and functioning ,Underpinning research ,Animals ,Brain ,Gene Expression Profiling ,High-Throughput Screening Assays ,Mice ,Sampling Studies ,Single-Cell Analysis ,Single-cell analysis ,Downsampling ,Cell atlas studies ,Bioinformatics ,Developmental Biology ,Biological sciences - Abstract
BackgroundHigh throughput methods for profiling the transcriptomes of single cells have recently emerged as transformative approaches for large-scale population surveys of cellular diversity in heterogeneous primary tissues. However, the efficient generation of such atlases will depend on sufficient sampling of diverse cell types while remaining cost-effective to enable a comprehensive examination of organs, developmental stages, and individuals.ResultsTo examine the relationship between sampled cell numbers and transcriptional heterogeneity in the context of unbiased cell type classification, we explored the population structure of a publicly available 1.3 million cell dataset from E18.5 mouse brain and validated our findings in published data from adult mice. We propose a computational framework for inferring the saturation point of cluster discovery in a single-cell mRNA-seq experiment, centered around cluster preservation in downsampled datasets. In addition, we introduce a "complexity index," which characterizes the heterogeneity of cells in a given dataset. Using Cajal-Retzius cells as an example of a limited complexity dataset, we explored whether the detected biological distinctions relate to technical clustering. Surprisingly, we found that clustering distinctions carrying biologically interpretable meaning are achieved with far fewer cells than the originally sampled, though technical saturation of rare populations such as Cajal-Retzius cells is not achieved. We additionally validated these findings with a recently published atlas of cell types across mouse organs and again find using subsampling that a much smaller number of cells recapitulates the cluster distinctions of the complete dataset.ConclusionsTogether, these findings suggest that most of the biologically interpretable cell types from the 1.3 million cell database can be recapitulated by analyzing 50,000 randomly selected cells, indicating that instead of profiling few individuals at high "cellular coverage," cell atlas studies may instead benefit from profiling more individuals, or many time points at lower cellular coverage and then further enriching for populations of interest. This strategy is ideal for scenarios where cost and time are limited, though extremely rare populations of interest (
- Published
- 2018
39. Regulation of cell-type-specific transcriptomes by microRNA networks during human brain development.
- Author
-
Nowakowski, Tomasz J, Rani, Neha, Golkaram, Mahdi, Zhou, Hongjun R, Alvarado, Beatriz, Huch, Kylie, West, Jay A, Leyrat, Anne, Pollen, Alex A, Kriegstein, Arnold R, Petzold, Linda R, and Kosik, Kenneth S
- Subjects
Brain ,Humans ,MicroRNAs ,Cell Proliferation ,Gene Regulatory Networks ,High-Throughput Nucleotide Sequencing ,Transcriptome ,Genetics ,Neurosciences ,Biotechnology ,Underpinning research ,1.1 Normal biological development and functioning ,Psychology ,Cognitive Sciences ,Neurology & Neurosurgery - Abstract
MicroRNAs (miRNAs) regulate many cellular events during brain development by interacting with hundreds of mRNA transcripts. However, miRNAs operate nonuniformly upon the transcriptional profile with an as yet unknown logic. Shortcomings in defining miRNA-mRNA networks include limited knowledge of in vivo miRNA targets and their abundance in single cells. By combining multiple complementary approaches, high-throughput sequencing of RNA isolated by cross-linking immunoprecipitation with an antibody to AGO2 (AGO2-HITS-CLIP), single-cell profiling and computational analyses using bipartite and coexpression networks, we show that miRNA-mRNA interactions operate as functional modules that often correspond to cell-type identities and undergo dynamic transitions during brain development. These networks are highly dynamic during development and over the course of evolution. One such interaction is between radial-glia-enriched ORC4 and miR-2115, a great-ape-specific miRNA, which appears to control radial glia proliferation rates during human brain development.
- Published
- 2018
40. Secretagogin is Expressed by Developing Neocortical GABAergic Neurons in Humans but not Mice and Increases Neurite Arbor Size and Complexity.
- Author
-
Raju, Chandrasekhar S, Spatazza, Julien, Stanco, Amelia, Larimer, Phillip, Sorrells, Shawn F, Kelley, Kevin W, Nicholas, Cory R, Paredes, Mercedes F, Lui, Jan H, Hasenstaub, Andrea R, Kriegstein, Arnold R, Alvarez-Buylla, Arturo, Rubenstein, John L, and Oldham, Michael C
- Subjects
Neocortex ,Neurites ,Interneurons ,Animals ,Mice ,Inbred C57BL ,Humans ,Mice ,Neurogenesis ,Transcriptome ,GABAergic Neurons ,Secretagogins ,Stem Cell Research - Nonembryonic - Non-Human ,Stem Cell Research ,Genetics ,Neurosciences ,Neurological ,gene coexpression ,human brain development ,interneurons ,neuronal maturation ,species differences ,Psychology ,Cognitive Sciences ,Experimental Psychology - Abstract
The neocortex of primates, including humans, contains more abundant and diverse inhibitory neurons compared with rodents, but the molecular foundations of these observations are unknown. Through integrative gene coexpression analysis, we determined a consensus transcriptional profile of GABAergic neurons in mid-gestation human neocortex. By comparing this profile to genes expressed in GABAergic neurons purified from neonatal mouse neocortex, we identified conserved and distinct aspects of gene expression in these cells between the species. We show here that the calcium-binding protein secretagogin (SCGN) is robustly expressed by neocortical GABAergic neurons derived from caudal ganglionic eminences (CGE) and lateral ganglionic eminences during human but not mouse brain development. Through electrophysiological and morphometric analyses, we examined the effects of SCGN expression on GABAergic neuron function and form. Forced expression of SCGN in CGE-derived mouse GABAergic neurons significantly increased total neurite length and arbor complexity following transplantation into mouse neocortex, revealing a molecular pathway that contributes to morphological differences in these cells between rodents and primates.
- Published
- 2018
41. An analytical framework for whole-genome sequence association studies and its implications for autism spectrum disorder
- Author
-
Werling, Donna M, Brand, Harrison, An, Joon-Yong, Stone, Matthew R, Zhu, Lingxue, Glessner, Joseph T, Collins, Ryan L, Dong, Shan, Layer, Ryan M, Markenscoff-Papadimitriou, Eirene, Farrell, Andrew, Schwartz, Grace B, Wang, Harold Z, Currall, Benjamin B, Zhao, Xuefang, Dea, Jeanselle, Duhn, Clif, Erdman, Carolyn A, Gilson, Michael C, Yadav, Rachita, Handsaker, Robert E, Kashin, Seva, Klei, Lambertus, Mandell, Jeffrey D, Nowakowski, Tomasz J, Liu, Yuwen, Pochareddy, Sirisha, Smith, Louw, Walker, Michael F, Waterman, Matthew J, He, Xin, Kriegstein, Arnold R, Rubenstein, John L, Sestan, Nenad, McCarroll, Steven A, Neale, Benjamin M, Coon, Hilary, Willsey, A Jeremy, Buxbaum, Joseph D, Daly, Mark J, State, Matthew W, Quinlan, Aaron R, Marth, Gabor T, Roeder, Kathryn, Devlin, Bernie, Talkowski, Michael E, and Sanders, Stephan J
- Subjects
Biological Sciences ,Genetics ,Intellectual and Developmental Disabilities (IDD) ,Brain Disorders ,Pediatric ,Autism ,Biotechnology ,Mental Health ,Human Genome ,2.1 Biological and endogenous factors ,Aetiology ,Autism Spectrum Disorder ,Female ,Genetic Predisposition to Disease ,Genome ,Genome-Wide Association Study ,Humans ,INDEL Mutation ,Male ,Polymorphism ,Single Nucleotide ,Protein Isoforms ,Medical and Health Sciences ,Developmental Biology ,Agricultural biotechnology ,Bioinformatics and computational biology - Abstract
Genomic association studies of common or rare protein-coding variation have established robust statistical approaches to account for multiple testing. Here we present a comparable framework to evaluate rare and de novo noncoding single-nucleotide variants, insertion/deletions, and all classes of structural variation from whole-genome sequencing (WGS). Integrating genomic annotations at the level of nucleotides, genes, and regulatory regions, we define 51,801 annotation categories. Analyses of 519 autism spectrum disorder families did not identify association with any categories after correction for 4,123 effective tests. Without appropriate correction, biologically plausible associations are observed in both cases and controls. Despite excluding previously identified gene-disrupting mutations, coding regions still exhibited the strongest associations. Thus, in autism, the contribution of de novo noncoding variation is probably modest in comparison to that of de novo coding variants. Robust results from future WGS studies will require large cohorts and comprehensive analytical strategies that consider the substantial multiple-testing burden.
- Published
- 2018
42. Human hippocampal neurogenesis drops sharply in children to undetectable levels in adults.
- Author
-
Sorrells, Shawn F, Paredes, Mercedes F, Cebrian-Silla, Arantxa, Sandoval, Kadellyn, Qi, Dashi, Kelley, Kevin W, James, David, Mayer, Simone, Chang, Julia, Auguste, Kurtis I, Chang, Edward F, Gutierrez, Antonio J, Kriegstein, Arnold R, Mathern, Gary W, Oldham, Michael C, Huang, Eric J, Garcia-Verdugo, Jose Manuel, Yang, Zhengang, and Alvarez-Buylla, Arturo
- Subjects
Hippocampus ,Dentate Gyrus ,Neurons ,Animals ,Animals ,Newborn ,Macaca mulatta ,Humans ,Epilepsy ,Cell Count ,Cell Proliferation ,Fetal Development ,Adolescent ,Adult ,Aged ,Middle Aged ,Child ,Child ,Preschool ,Infant ,Female ,Male ,Neurogenesis ,Young Adult ,Neural Stem Cells ,Healthy Volunteers ,General Science & Technology - Abstract
New neurons continue to be generated in the subgranular zone of the dentate gyrus of the adult mammalian hippocampus. This process has been linked to learning and memory, stress and exercise, and is thought to be altered in neurological disease. In humans, some studies have suggested that hundreds of new neurons are added to the adult dentate gyrus every day, whereas other studies find many fewer putative new neurons. Despite these discrepancies, it is generally believed that the adult human hippocampus continues to generate new neurons. Here we show that a defined population of progenitor cells does not coalesce in the subgranular zone during human fetal or postnatal development. We also find that the number of proliferating progenitors and young neurons in the dentate gyrus declines sharply during the first year of life and only a few isolated young neurons are observed by 7 and 13 years of age. In adult patients with epilepsy and healthy adults (18-77 years; n = 17 post-mortem samples from controls; n = 12 surgical resection samples from patients with epilepsy), young neurons were not detected in the dentate gyrus. In the monkey (Macaca mulatta) hippocampus, proliferation of neurons in the subgranular zone was found in early postnatal life, but this diminished during juvenile development as neurogenesis decreased. We conclude that recruitment of young neurons to the primate hippocampus decreases rapidly during the first years of life, and that neurogenesis in the dentate gyrus does not continue, or is extremely rare, in adult humans. The early decline in hippocampal neurogenesis raises questions about how the function of the dentate gyrus differs between humans and other species in which adult hippocampal neurogenesis is preserved.
- Published
- 2018
43. Single-cell profiling of human gliomas reveals macrophage ontogeny as a basis for regional differences in macrophage activation in the tumor microenvironment.
- Author
-
Müller, Sören, Kohanbash, Gary, Liu, S John, Alvarado, Beatriz, Carrera, Diego, Bhaduri, Aparna, Watchmaker, Payal B, Yagnik, Garima, Di Lullo, Elizabeth, Malatesta, Martina, Amankulor, Nduka M, Kriegstein, Arnold R, Lim, Daniel A, Aghi, Manish, Okada, Hideho, and Diaz, Aaron
- Subjects
Macrophages ,Animals ,Humans ,Mice ,Glioma ,Prognosis ,Immunotherapy ,Survival Analysis ,Gene Expression Profiling ,Computational Biology ,Macrophage Activation ,Gene Expression Regulation ,Neoplastic ,Single-Cell Analysis ,High-Throughput Nucleotide Sequencing ,Tumor Microenvironment ,Transcriptome ,Gene Ontology ,Macrophage ,Single-cell sequencing ,Gene Expression Regulation ,Neoplastic ,Bioinformatics ,Environmental Sciences ,Biological Sciences ,Information and Computing Sciences - Abstract
BACKGROUND:Tumor-associated macrophages (TAMs) are abundant in gliomas and immunosuppressive TAMs are a barrier to emerging immunotherapies. It is unknown to what extent macrophages derived from peripheral blood adopt the phenotype of brain-resident microglia in pre-treatment gliomas. The relative proportions of blood-derived macrophages and microglia have been poorly quantified in clinical samples due to a paucity of markers that distinguish these cell types in malignant tissue. RESULTS:We perform single-cell RNA-sequencing of human gliomas and identify phenotypic differences in TAMs of distinct lineages. We isolate TAMs from patient biopsies and compare them with macrophages from non-malignant human tissue, glioma atlases, and murine glioma models. We present a novel signature that distinguishes TAMs by ontogeny in human gliomas. Blood-derived TAMs upregulate immunosuppressive cytokines and show an altered metabolism compared to microglial TAMs. They are also enriched in perivascular and necrotic regions. The gene signature of blood-derived TAMs, but not microglial TAMs, correlates with significantly inferior survival in low-grade glioma. Surprisingly, TAMs frequently co-express canonical pro-inflammatory (M1) and alternatively activated (M2) genes in individual cells. CONCLUSIONS:We conclude that blood-derived TAMs significantly infiltrate pre-treatment gliomas, to a degree that varies by glioma subtype and tumor compartment. Blood-derived TAMs do not universally conform to the phenotype of microglia, but preferentially express immunosuppressive cytokines and show an altered metabolism. Our results argue against status quo therapeutic strategies that target TAMs indiscriminately and in favor of strategies that specifically target immunosuppressive blood-derived TAMs.
- Published
- 2017
44. Spatiotemporal gene expression trajectories reveal developmental hierarchies of the human cortex
- Author
-
Nowakowski, Tomasz J, Bhaduri, Aparna, Pollen, Alex A, Alvarado, Beatriz, Mostajo-Radji, Mohammed A, Di Lullo, Elizabeth, Haeussler, Maximilian, Sandoval-Espinosa, Carmen, Liu, Siyuan John, Velmeshev, Dmitry, Ounadjela, Johain Ryad, Shuga, Joe, Wang, Xiaohui, Lim, Daniel A, West, Jay A, Leyrat, Anne A, Kent, W James, and Kriegstein, Arnold R
- Subjects
Neurosciences ,Stem Cell Research - Nonembryonic - Non-Human ,Stem Cell Research ,Genetics ,Pediatric ,1.1 Normal biological development and functioning ,Underpinning research ,Cerebral Cortex ,Gene Expression Regulation ,Developmental ,Humans ,Neurogenesis ,Neuroglia ,Neurons ,Telencephalon ,General Science & Technology - Abstract
Systematic analyses of spatiotemporal gene expression trajectories during organogenesis have been challenging because diverse cell types at different stages of maturation and differentiation coexist in the emerging tissues. We identified discrete cell types as well as temporally and spatially restricted trajectories of radial glia maturation and neurogenesis in developing human telencephalon. These lineage-specific trajectories reveal the expression of neurogenic transcription factors in early radial glia and enriched activation of mammalian target of rapamycin signaling in outer radial glia. Across cortical areas, modest transcriptional differences among radial glia cascade into robust typological distinctions among maturing neurons. Together, our results support a mixed model of topographical, typological, and temporal hierarchies governing cell-type diversity in the developing human telencephalon, including distinct excitatory lineages emerging in rostral and caudal cerebral cortex.
- Published
- 2017
45. The BRAIN Initiative Cell Census Consortium: Lessons Learned toward Generating a Comprehensive Brain Cell Atlas.
- Author
-
Ecker, Joseph R, Geschwind, Daniel H, Kriegstein, Arnold R, Ngai, John, Osten, Pavel, Polioudakis, Damon, Regev, Aviv, Sestan, Nenad, Wickersham, Ian R, and Zeng, Hongkui
- Subjects
Brain ,Nerve Net ,Animals ,Humans ,Brain Mapping ,Pilot Projects ,Atlases as Topic ,BRAIN initiative ,anatomy ,cell census ,connectivity ,electrophysiology ,human brain ,mouse brain ,single-cell RNA-seq ,single-cell epigenomics ,single-cell transcriptomics ,Neurosciences ,Pediatric Research Initiative ,Genetics ,Underpinning research ,1.1 Normal biological development and functioning ,Neurological ,Psychology ,Cognitive Sciences ,Neurology & Neurosurgery - Abstract
A comprehensive characterization of neuronal cell types, their distributions, and patterns of connectivity is critical for understanding the properties of neural circuits and how they generate behaviors. Here we review the experiences of the BRAIN Initiative Cell Census Consortium, ten pilot projects funded by the U.S. BRAIN Initiative, in developing, validating, and scaling up emerging genomic and anatomical mapping technologies for creating a complete inventory of neuronal cell types and their connections in multiple species and during development. These projects lay the foundation for a larger and longer-term effort to generate whole-brain cell atlases in species including mice and humans.
- Published
- 2017
46. The use of brain organoids to investigate neural development and disease
- Author
-
Di Lullo, Elizabeth and Kriegstein, Arnold R
- Subjects
Biomedical and Clinical Sciences ,Neurosciences ,Brain Disorders ,1.1 Normal biological development and functioning ,Underpinning research ,Neurological ,Good Health and Well Being ,Animals ,Brain ,Brain Diseases ,Humans ,Models ,Neurological ,Neurogenesis ,Organoids ,Pluripotent Stem Cells ,Psychology ,Cognitive Sciences ,Neurology & Neurosurgery ,Biological psychology - Abstract
Understanding the development and dysfunction of the human brain is a major goal of neurobiology. Much of our current understanding of human brain development has been derived from the examination of post-mortem and pathological specimens, bolstered by observations of developing non-human primates and experimental studies focused largely on mouse models. However, these tissue specimens and model systems cannot fully capture the unique and dynamic features of human brain development. Recent advances in stem cell technologies that enable the generation of human brain organoids from pluripotent stem cells (PSCs) promise to profoundly change our understanding of the development of the human brain and enable a detailed study of the pathogenesis of inherited and acquired brain diseases.
- Published
- 2017
47. Human iPSC-Derived Cerebral Organoids Model Cellular Features of Lissencephaly and Reveal Prolonged Mitosis of Outer Radial Glia
- Author
-
Bershteyn, Marina, Nowakowski, Tomasz J, Pollen, Alex A, Di Lullo, Elizabeth, Nene, Aishwarya, Wynshaw-Boris, Anthony, and Kriegstein, Arnold R
- Subjects
Medical Biotechnology ,Biomedical and Clinical Sciences ,Intellectual and Developmental Disabilities (IDD) ,Stem Cell Research - Nonembryonic - Human ,Clinical Research ,Stem Cell Research ,Brain Disorders ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Stem Cell Research - Embryonic - Human ,Stem Cell Research - Induced Pluripotent Stem Cell ,Regenerative Medicine ,Neurosciences ,2.1 Biological and endogenous factors ,Aetiology ,Neurological ,Adult ,Apoptosis ,Cell Movement ,Cerebrum ,Chromosome Duplication ,Classical Lissencephalies and Subcortical Band Heterotopias ,Cytokinesis ,Epithelium ,Female ,Humans ,Induced Pluripotent Stem Cells ,Infant ,Infant ,Newborn ,Lissencephaly ,Male ,Middle Aged ,Mitosis ,Neuroglia ,Neurons ,Organoids ,cerebral organoids ,human lissencephaly ,migration ,outer radial glia ,spindle orientation ,Biological Sciences ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
Classical lissencephaly is a genetic neurological disorder associated with mental retardation and intractable epilepsy, and Miller-Dieker syndrome (MDS) is the most severe form of the disease. In this study, to investigate the effects of MDS on human progenitor subtypes that control neuronal output and influence brain topology, we analyzed cerebral organoids derived from control and MDS-induced pluripotent stem cells (iPSCs) using time-lapse imaging, immunostaining, and single-cell RNA sequencing. We saw a cell migration defect that was rescued when we corrected the MDS causative chromosomal deletion and severe apoptosis of the founder neuroepithelial stem cells, accompanied by increased horizontal cell divisions. We also identified a mitotic defect in outer radial glia, a progenitor subtype that is largely absent from lissencephalic rodents but critical for human neocortical expansion. Our study, therefore, deepens our understanding of MDS cellular pathogenesis and highlights the broad utility of cerebral organoids for modeling human neurodevelopmental disorders.
- Published
- 2017
48. Dynamic behaviour of human neuroepithelial cells in the developing forebrain.
- Author
-
Subramanian, Lakshmi, Bershteyn, Marina, Paredes, Mercedes F, and Kriegstein, Arnold R
- Subjects
Neocortex ,Neuroglia ,Neurons ,Neuroepithelial Cells ,Organoids ,Fetus ,Humans ,Abortion ,Legal ,Cytokinesis ,Mitosis ,Cell Differentiation ,Cell Movement ,Pregnancy ,Pregnancy Trimester ,First ,Female ,Neurogenesis ,Induced Pluripotent Stem Cells ,Neural Stem Cells ,Abortion ,Legal ,Pregnancy Trimester ,First - Abstract
To understand how diverse progenitor cells contribute to human neocortex development, we examined forebrain progenitor behaviour using timelapse imaging. Here we find that cell cycle dynamics of human neuroepithelial (NE) cells differ from radial glial (RG) cells in both primary tissue and in stem cell-derived organoids. NE cells undergoing proliferative, symmetric divisions retract their basal processes, and both daughter cells regrow a new process following cytokinesis. The mitotic retraction of the basal process is recapitulated by NE cells in cerebral organoids generated from human-induced pluripotent stem cells. In contrast, RG cells undergoing vertical cleavage retain their basal fibres throughout mitosis, both in primary tissue and in older organoids. Our findings highlight developmentally regulated changes in mitotic behaviour that may relate to the role of RG cells to provide a stable scaffold for neuronal migration, and suggest that the transition in mitotic dynamics can be studied in organoid models.
- Published
- 2017
49. Astroglial toxicity promotes synaptic degeneration in the thalamocortical circuit in frontotemporal dementia with GRN mutations
- Author
-
Marsan, Elise, Velmeshev, Dmitry, Ramsey, Arren, Patel, Ravi K., Zhang, Jiasheng, Koontz, Mark, Andrews, Madeline G., de Majo, Martina, Mora, Cristina, Blumenfeld, Jessica, Li, Alissa N., Spina, Salvatore, Grinberg, Lea T., Seeley, William W., Miller, Bruce L., Ullian, Erik M., Krummel, Matthew F., Kriegstein, Arnold R., and Huang, Eric J.
- Subjects
Gene mutations -- Health aspects ,DNA testing ,Frontotemporal dementia -- Diagnosis -- Genetic aspects ,Health care industry - Abstract
Mutations in the human progranulin (GRN) gene are a leading cause of frontotemporal lobar degeneration (FTLD). While previous studies implicate aberrant microglial activation as a disease-driving factor in neurodegeneration in the thalamocortical circuit in [Grn.sup.-/-] mice, the exact mechanism for neurodegeneration in FTLD-GRN remains unclear. By performing comparative single-cell transcriptomics in the thalamus and frontal cortex of [Grn.sup.-/-] mice and patients with FTLD-GRN, we have uncovered a highly conserved astroglial pathology characterized by upregulation of gap junction protein GJA1, water channel AQP4, and lipid-binding protein APOE, and downregulation of glutamate transporter SLC1A2 that promoted profound synaptic degeneration across the two species. This astroglial toxicity could be recapitulated in mouse astrocyte-neuron cocultures and by transplanting induced pluripotent stem cell-derived astrocytes to cortical organoids, where progranulin-deficient astrocytes promoted synaptic degeneration, neuronal stress, and TDP-43 proteinopathy. Together, these results reveal a previously unappreciated astroglial pathology as a potential key mechanism in neurodegeneration in FTLD-GRN., Introduction Frontotemporal dementia (FTD) is a common neurodegenerative disease in patients under 65 years of age. The clinical manifestations of FTD include progressive behavioral changes and deterioration in language skills [...]
- Published
- 2023
- Full Text
- View/download PDF
50. Molecular and cellular dynamics of the developing human neocortex at single-cell resolution
- Author
-
Wang, Li, primary, Wang, Cheng, additional, Moriano, Juan A., additional, Chen, Songcang, additional, Zhang, Shaobo, additional, Mukhtar, Tanzila, additional, Wang, Shaohui, additional, Cebrián-Silla, Arantxa, additional, Bi, Qiuli, additional, Augustin, Jonathan J., additional, de Oliveira, Lilian Gomes, additional, Song, Mengyi, additional, Ge, Xinxin, additional, Zuo, Guolong, additional, Paredes, Mercedes F., additional, Huang, Eric J., additional, Alvarez-Buylla, Arturo, additional, Duan, Xin, additional, Li, Jingjing, additional, and Kriegstein, Arnold R., additional
- Published
- 2024
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.