Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
EMBO Rep ; 23(9): e52211, 2022 09 05.
Article in English | MEDLINE | ID: mdl-35793002

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.


Subject(s)
Microcephaly , Zika Virus Infection , Zika Virus , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Centrosome/metabolism , Humans , Immunity, Innate , Microcephaly/metabolism , Zika Virus/physiology
2.
Cell Stem Cell ; 26(1): 48-63.e6, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31901251

ABSTRACT

Glioblastoma is a devastating form of brain cancer. To identify aspects of tumor heterogeneity that may illuminate drivers of tumor invasion, we created a glioblastoma tumor cell atlas with single-cell transcriptomics of cancer cells mapped onto a reference framework of the developing and adult human brain. We find that multiple GSC subtypes exist within a single tumor. Within these GSCs, we identify an invasive cell population similar to outer radial glia (oRG), a fetal cell type that expands the stem cell niche in normal human cortex. Using live time-lapse imaging of primary resected tumors, we discover that tumor-derived oRG-like cells undergo characteristic mitotic somal translocation behavior previously only observed in human development, suggesting a reactivation of developmental programs. In addition, we show that PTPRZ1 mediates both mitotic somal translocation and glioblastoma tumor invasion. These data suggest that the presence of heterogeneous GSCs may underlie glioblastoma's rapid progression and invasion.


Subject(s)
Brain Neoplasms , Glioblastoma , Adult , Brain Neoplasms/genetics , Cell Line, Tumor , Ependymoglial Cells , Glioblastoma/genetics , Humans , Neoplastic Stem Cells , Receptor-Like Protein Tyrosine Phosphatases, Class 5
3.
Cancer Discov ; 9(12): 1708-1719, 2019 12.
Article in English | MEDLINE | ID: mdl-31554641

ABSTRACT

Although tumor-propagating cells can be derived from glioblastomas (GBM) of the proneural and mesenchymal subtypes, a glioma stem-like cell (GSC) of the classic subtype has not been identified. It is unclear whether mesenchymal GSCs (mGSC) and/or proneural GSCs (pGSC) alone are sufficient to generate the heterogeneity observed in GBM. We performed single-cell/single-nucleus RNA sequencing of 28 gliomas, and single-cell ATAC sequencing for 8 cases. We found that GBM GSCs reside on a single axis of variation, ranging from proneural to mesenchymal. In silico lineage tracing using both transcriptomics and genetics supports mGSCs as the progenitors of pGSCs. Dual inhibition of pGSC-enriched and mGSC-enriched growth and survival pathways provides a more complete treatment than combinations targeting one GSC phenotype alone. This study sheds light on a long-standing debate regarding lineage relationships among GSCs and presents a paradigm by which personalized combination therapies can be derived from single-cell RNA signatures, to overcome intratumor heterogeneity. SIGNIFICANCE: Tumor-propagating cells can be derived from mesenchymal and proneural glioblastomas. However, a stem cell of the classic subtype has yet to be demonstrated. We show that classic-subtype gliomas are comprised of proneural and mesenchymal cells. This study sheds light on a long-standing debate regarding lineage relationships between glioma cell types.See related commentary by Fine, p. 1650.This article is highlighted in the In This Issue feature, p. 1631.


Subject(s)
Brain Neoplasms/genetics , Gene Regulatory Networks , Glioblastoma/genetics , Neoplastic Stem Cells/chemistry , Sequence Analysis, RNA/methods , Cell Line, Tumor , Cell Lineage , Cell Proliferation , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans
4.
Cell ; 176(4): 743-756.e17, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30735633

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.


Subject(s)
Cerebral Cortex/cytology , Organoids/metabolism , Animals , Biological Evolution , Brain/cytology , Cell Culture Techniques/methods , Cell Differentiation/genetics , Cerebral Cortex/metabolism , Gene Regulatory Networks/genetics , Humans , Induced Pluripotent Stem Cells/cytology , Macaca , Neurogenesis/genetics , Organoids/growth & development , Pan troglodytes , Pluripotent Stem Cells/cytology , Single-Cell Analysis , Species Specificity , Transcriptome/genetics
5.
Science ; 358(6368): 1318-1323, 2017 12 08.
Article in English | MEDLINE | ID: mdl-29217575

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.


Subject(s)
Cerebral Cortex/growth & development , Gene Expression Regulation, Developmental , Neurogenesis/genetics , Telencephalon/growth & development , Cerebral Cortex/anatomy & histology , Cerebral Cortex/cytology , Humans , Neuroglia/physiology , Neurons , Telencephalon/anatomy & histology , Telencephalon/cytology
6.
Genome Biol ; 18(1): 234, 2017 Dec 20.
Article in English | MEDLINE | ID: mdl-29262845

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.


Subject(s)
Glioma/genetics , Glioma/pathology , Macrophage Activation/genetics , Macrophages/metabolism , Macrophages/pathology , Tumor Microenvironment/genetics , Animals , Computational Biology/methods , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Ontology , Glioma/immunology , Glioma/therapy , High-Throughput Nucleotide Sequencing , Humans , Immunotherapy/methods , Macrophage Activation/immunology , Mice , Prognosis , Single-Cell Analysis , Survival Analysis , Transcriptome , Tumor Microenvironment/immunology
7.
Nat Rev Neurosci ; 18(10): 573-584, 2017 10.
Article in English | MEDLINE | ID: mdl-28878372

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.


Subject(s)
Brain Diseases/physiopathology , Brain/growth & development , Neurogenesis/physiology , Organoids/physiology , Organoids/physiopathology , Animals , Humans , Models, Neurological , Organoids/cytology , Pluripotent Stem Cells
8.
Cell Stem Cell ; 20(4): 435-449.e4, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28111201

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.


Subject(s)
Cerebrum/pathology , Induced Pluripotent Stem Cells/pathology , Lissencephaly/pathology , Mitosis , Neuroglia/pathology , Organoids/pathology , Adult , Apoptosis , Cell Movement , Chromosome Duplication , Classical Lissencephalies and Subcortical Band Heterotopias/pathology , Cytokinesis , Epithelium/pathology , Female , Humans , Infant , Infant, Newborn , Male , Middle Aged , Neurons/pathology
9.
Curr Opin Neurobiol ; 42: 61-67, 2017 02.
Article in English | MEDLINE | ID: mdl-27978479

ABSTRACT

The evolution of the human brain has been characterized by an increase in the size of the neocortex. Underlying this expansion is a significant increase in the number of neurons produced by neural stem cells during early stages of cortical development. Here we highlight recent advances in our understating of these cell populations, consisting of ventricular radial glia and outer radial glia. We highlight how gene expression studies have identified molecular signatures for radial glial cell populations and outline what has been learned about the mechanisms underlying the characteristic mode of division observed in outer radial glia cells, mitotic somal translocation. Understanding the significance of this behavior may help us explain human cortical expansion and further elucidate neurodevelopmental diseases.


Subject(s)
Mitosis/genetics , Neocortex/embryology , Neuroglia/cytology , Cell Division/genetics , Cell Movement , Gene Expression Regulation , Humans , Neocortex/cytology , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/physiopathology , Neuroglia/metabolism
10.
Proc Natl Acad Sci U S A ; 113(50): 14408-14413, 2016 12 13.
Article in English | MEDLINE | ID: mdl-27911847

ABSTRACT

The rapid spread of Zika virus (ZIKV) and its association with abnormal brain development constitute a global health emergency. Congenital ZIKV infection produces a range of mild to severe pathologies, including microcephaly. To understand the pathophysiology of ZIKV infection, we used models of the developing brain that faithfully recapitulate the tissue architecture in early to midgestation. We identify the brain cell populations that are most susceptible to ZIKV infection in primary human tissue, provide evidence for a mechanism of viral entry, and show that a commonly used antibiotic protects cultured brain cells by reducing viral proliferation. In the brain, ZIKV preferentially infected neural stem cells, astrocytes, oligodendrocyte precursor cells, and microglia, whereas neurons were less susceptible to infection. These findings suggest mechanisms for microcephaly and other pathologic features of infants with congenital ZIKV infection that are not explained by neural stem cell infection alone, such as calcifications in the cortical plate. Furthermore, we find that blocking the glia-enriched putative viral entry receptor AXL reduced ZIKV infection of astrocytes in vitro, and genetic knockdown of AXL in a glial cell line nearly abolished infection. Finally, we evaluate 2,177 compounds, focusing on drugs safe in pregnancy. We show that the macrolide antibiotic azithromycin reduced viral proliferation and virus-induced cytopathic effects in glial cell lines and human astrocytes. Our characterization of infection in the developing human brain clarifies the pathogenesis of congenital ZIKV infection and provides the basis for investigating possible therapeutic strategies to safely alleviate or prevent the most severe consequences of the epidemic.


Subject(s)
Azithromycin/pharmacology , Brain/embryology , Brain/virology , Viral Tropism/drug effects , Zika Virus Infection/drug therapy , Zika Virus/drug effects , Zika Virus/physiology , Brain/pathology , Cell Line , Cytopathogenic Effect, Viral/drug effects , Female , Humans , Infant, Newborn , Microbial Sensitivity Tests , Microcephaly/drug therapy , Microcephaly/embryology , Microcephaly/pathology , Neuroglia/drug effects , Neuroglia/pathology , Neuroglia/virology , Pregnancy , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/physiology , Viral Tropism/physiology , Virus Internalization/drug effects , Virus Replication/drug effects , Zika Virus/pathogenicity , Zika Virus Infection/embryology , Zika Virus Infection/pathology , Axl Receptor Tyrosine Kinase
11.
Mol Syst Biol ; 12(11): 889, 2016 Nov 25.
Article in English | MEDLINE | ID: mdl-27888226

ABSTRACT

Glioblastoma multiforme (GBM) is the most common and aggressive type of primary brain tumor. Epidermal growth factor (EGF) and platelet-derived growth factor (PDGF) receptors are frequently amplified and/or possess gain-of-function mutations in GBM However, clinical trials of tyrosine-kinase inhibitors have shown disappointing efficacy, in part due to intra-tumor heterogeneity. To assess the effect of clonal heterogeneity on gene expression, we derived an approach to map single-cell expression profiles to sequentially acquired mutations identified from exome sequencing. Using 288 single cells, we constructed high-resolution phylogenies of EGF-driven and PDGF-driven GBMs, modeling transcriptional kinetics during tumor evolution. Descending the phylogenetic tree of a PDGF-driven tumor corresponded to a progressive induction of an oligodendrocyte progenitor-like cell type, expressing pro-angiogenic factors. In contrast, phylogenetic analysis of an EGFR-amplified tumor showed an up-regulation of pro-invasive genes. An in-frame deletion in a specific dimerization domain of PDGF receptor correlates with an up-regulation of growth pathways in a proneural GBM and enhances proliferation when ectopically expressed in glioma cell lines. In-frame deletions in this domain are frequent in public GBM data.


Subject(s)
ErbB Receptors/genetics , Gene Expression Profiling/methods , Receptors, Platelet-Derived Growth Factor/genetics , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Brain Neoplasms , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Genetic Heterogeneity , Glioblastoma , Humans , Mutation
12.
Cell Stem Cell ; 18(5): 591-6, 2016 05 05.
Article in English | MEDLINE | ID: mdl-27038591

ABSTRACT

The recent outbreak of Zika virus (ZIKV) in Brazil has been linked to substantial increases in fetal abnormalities and microcephaly. However, information about the underlying molecular and cellular mechanisms connecting viral infection to these defects remains limited. In this study we have examined the expression of receptors implicated in cell entry of several enveloped viruses including ZIKV across diverse cell types in the developing brain. Using single-cell RNA-seq and immunohistochemistry, we found that the candidate viral entry receptor AXL is highly expressed by human radial glial cells, astrocytes, endothelial cells, and microglia in developing human cortex and by progenitor cells in developing retina. We also show that AXL expression in radial glia is conserved in developing mouse and ferret cortex and in human stem cell-derived cerebral organoids, highlighting multiple experimental systems that could be applied to study mechanisms of ZIKV infectivity and effects on brain development.


Subject(s)
Neural Stem Cells/metabolism , Neural Stem Cells/virology , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Virus/metabolism , Virus Internalization , Zika Virus/physiology , Animals , Blood Vessels/metabolism , Cerebral Cortex/embryology , Cerebral Cortex/metabolism , Disease Models, Animal , Ferrets , Mice , Neurogenesis , Neuroglia/metabolism , Pluripotent Stem Cells/cytology , Axl Receptor Tyrosine Kinase
13.
Pharmacol Rev ; 65(1): 90-104, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23300132

ABSTRACT

Homeoproteins constitute a major class of transcription factors active throughout development and in adulthood. Their membrane transduction properties were discovered over 20 years ago, opening an original field of research in the domain of vector peptides and signal transduction. In early development, homeoprotein transfer participates in tissue patterning, cell/axon guidance, and migration. In the axon guidance model, homeoproteins exert their non-cell autonomous activity through the regulation of translation, in particular, that of nuclear-transcribed mitochondrial mRNAs. An important aspect of these studies on patterning and migration is that homeoproteins sensitize the cells to the action of other growth factors, thus cooperating with established signaling pathways. The role of homeoprotein signaling at later developmental stages is also of interest. In particular, the transfer of homeoprotein Otx2 into parvalbumin-expressing inhibitory neurons (PV-cells) in the visual cortex regulates cortical plasticity. The molecular deciphering of the interaction of Otx2 with binding sites at the surface of PV-cells has allowed the development of a specific Otx2 antagonist that reopens plasticity in the adult cortex and cures mice from experimental amblyopia, a neurodevelopmental disease. Finally, the use of homeoproteins as therapeutic proteins in mouse models of glaucoma and Parkinson disease is reviewed. In the latter case, engrailed homeoproteins protect mesencephalic dopaminergic neurons by increasing the local translation of complex I mitochondrial mRNAs. In conclusion, this review synthesizes 20 years of work on the fundamental and potentially translational aspects of homeoprotein signaling.


Subject(s)
Homeodomain Proteins/physiology , Animals , Axons/physiology , Carrier Proteins/metabolism , Cell Movement , Cell-Penetrating Peptides , Neuronal Plasticity , Signal Transduction , Visual Cortex/physiology
14.
Development ; 138(22): 4991-5001, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22028031

ABSTRACT

Homeoprotein transcription factors play fundamental roles in development, ranging from embryonic polarity to cell differentiation and migration. Research in recent years has underscored the physiological importance of homeoprotein intercellular transfer in eye field development, axon guidance and retino-tectal patterning, and visual cortex plasticity. Here, we have used the embryonic chick neural tube to investigate a possible role for homeoprotein Pax6 transfer in oligodendrocyte precursor cell (OPC) migration. We report the extracellular expression of Pax6 and the effects of gain and loss of extracellular Pax6 activity on OPCs. Open book cultures with recombinant Pax6 protein or Pax6 blocking antibodies, as well as in ovo gene transfer experiments involving expression of secreted Pax6 protein or secreted Pax6 antibodies, provide converging evidences that OPC migration is promoted by extracellular Pax6. The paracrine effect of Pax6 on OPC migration is thus a new example of direct non-cell autonomous homeoprotein activity.


Subject(s)
Cell Movement/genetics , Eye Proteins/physiology , Homeodomain Proteins/physiology , Neural Tube/embryology , Oligodendroglia/physiology , Paired Box Transcription Factors/physiology , Paracrine Communication , Repressor Proteins/physiology , Animals , Animals, Genetically Modified , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Movement/drug effects , Chick Embryo , Extracellular Space/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Eye Proteins/pharmacology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Homeodomain Proteins/pharmacology , Nerve Tissue Proteins/metabolism , Neural Tube/cytology , Neural Tube/metabolism , Neural Tube/physiology , Oligodendroglia/drug effects , Oligodendroglia/metabolism , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Paired Box Transcription Factors/pharmacology , Paracrine Communication/physiology , Protein Transport/genetics , Protein Transport/physiology , Repressor Proteins/genetics , Repressor Proteins/metabolism , Repressor Proteins/pharmacology , Stem Cells/drug effects , Stem Cells/metabolism , Stem Cells/physiology , Substrate Specificity , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...