Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
3.
bioRxiv ; 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39131314

ABSTRACT

The nuclear genome is spatially organized into a three-dimensional (3D) architecture by physical association of large chromosomal domains with subnuclear compartments including the nuclear lamina at the radial periphery and nuclear speckles within the nucleoplasm1-5. However, how spatial genome architecture regulates human brain development has been overlooked owing to technical limitations. Here, we generate high-resolution maps of genomic interactions with the lamina and speckles in cells of the neurogenic lineage isolated from midgestational human cortex, uncovering an intimate association between subnuclear genome compartmentalization, chromatin state and transcription. During cortical neurogenesis, spatial genome organization is extensively remodeled, relocating hundreds of neuronal genes from the lamina to speckles including key neurodevelopmental genes bivalent for H3K27me3 and H3K4me3. At the lamina, bivalent genes have exceptionally low expression, and relocation to speckles enhances resolution of bivalent chromatin to H3K4me3 and increases transcription >7-fold. We further demonstrate that proximity to the nuclear periphery - not the presence of H3K27me3 - is the dominant factor in maintaining the lowly expressed, poised state of bivalent genes embedded in the lamina. In addition to uncovering a critical role of subnuclear genome compartmentalization in neurogenic transcriptional regulation, our results establish a new paradigm in which knowing the spatial location of a gene is necessary to understanding its epigenomic regulation.

4.
Cell Genom ; 2(11)2022 Nov 09.
Article in English | MEDLINE | ID: mdl-36381608

ABSTRACT

Human chromosomes are pervasively transcribed, but systematic understanding of coding and lncRNA genome function in cell differentiation is lacking. Using CRISPR interference (CRISPRi) in human induced pluripotent stem cells, we performed dual genome-wide screens - assessing 18,905 protein-coding and 10,678 lncRNA loci - and identified 419 coding and 201 lncRNA genes that regulate neural induction. Integrative analyses revealed distinct properties of coding and lncRNA genome function, including a 10-fold enrichment of lncRNA genes for roles in differentiation compared to proliferation. Further, we applied Perturb-seq to obtain granular insights into neural induction phenotypes. While most coding hits stalled or aborted differentiation, lncRNA hits were enriched for the genesis of diverse cellular states, including those outside the neural lineage. In addition to providing a rich resource (danlimlab.shinyapps.io/dualgenomewide) for understanding coding and lncRNA gene function in development, these results indicate that the lncRNA genome regulates lineage commitment in a manner fundamentally distinct from coding genes.

5.
Nat Neurosci ; 24(9): 1235-1242, 2021 09.
Article in English | MEDLINE | ID: mdl-34239128

ABSTRACT

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


Subject(s)
Brain/physiology , Cell Nucleus/physiology , Gene Expression/genetics , Genome/genetics , Neurogenesis/physiology , Animals , Genetic Variation , Genome-Wide Association Study , Humans , Macaca , Mice , Mice, Inbred C57BL , Neural Stem Cells/physiology , Schizophrenia/genetics
6.
Nat Genet ; 53(5): 719-728, 2021 05.
Article in English | MEDLINE | ID: mdl-33859416

ABSTRACT

Known fetal hemoglobin (HbF) silencers have potential on-target liabilities for rational ß-hemoglobinopathy therapeutic inhibition. Here, through transcription factor (TF) CRISPR screening, we identify zinc-finger protein (ZNF) 410 as an HbF repressor. ZNF410 does not bind directly to the genes encoding γ-globins, but rather its chromatin occupancy is concentrated solely at CHD4, encoding the NuRD nucleosome remodeler, which is itself required for HbF repression. CHD4 has two ZNF410-bound regulatory elements with 27 combined ZNF410 binding motifs constituting unparalleled genomic clusters. These elements completely account for the effects of ZNF410 on fetal globin repression. Knockout of ZNF410 or its mouse homolog Zfp410 reduces CHD4 levels by 60%, enough to substantially de-repress HbF while eluding cellular or organismal toxicity. These studies suggest a potential target for HbF induction for ß-hemoglobin disorders with a wide therapeutic index. More broadly, ZNF410 represents a special class of gene regulator, a conserved TF with singular devotion to regulation of a chromatin subcomplex.


Subject(s)
Fetal Hemoglobin/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Transcription Factors/metabolism , Adult , Animals , Base Sequence , CRISPR-Cas Systems/genetics , Cells, Cultured , Chromatin/metabolism , DNA/metabolism , Erythroid Cells/metabolism , Erythropoiesis , Gene Editing , Gene Expression Regulation , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Humans , Mice , Mutagenesis/genetics , Protein Binding , Reproducibility of Results
7.
Nat Genet ; 51(7): 1149-1159, 2019 07.
Article in English | MEDLINE | ID: mdl-31253978

ABSTRACT

Developmental silencing of fetal globins serves as both a paradigm of spatiotemporal gene regulation and an opportunity for therapeutic intervention of ß-hemoglobinopathy. The nucleosome remodeling and deacetylase (NuRD) chromatin complex participates in γ-globin repression. We used pooled CRISPR screening to disrupt NuRD protein coding sequences comprehensively in human adult erythroid precursors. Essential for fetal hemoglobin (HbF) control is a non-redundant subcomplex of NuRD protein family paralogs, whose composition we corroborated by affinity chromatography and proximity labeling mass spectrometry proteomics. Mapping top functional guide RNAs identified key protein interfaces where in-frame alleles resulted in loss-of-function due to destabilization or altered function of subunits. We ascertained mutations of CHD4 that dissociate its requirement for cell fitness from HbF repression in both primary human erythroid precursors and transgenic mice. Finally we demonstrated that sequestering CHD4 from NuRD phenocopied these mutations. These results indicate a generalizable approach to discover protein complex features amenable to rational biochemical targeting.


Subject(s)
Chromatin/genetics , Erythroid Cells/metabolism , Fetal Hemoglobin/metabolism , Gene Expression Regulation , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Mutagenesis , Animals , Chromatin/metabolism , Erythroid Cells/cytology , Fetal Hemoglobin/genetics , Humans , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Mice , Mice, Transgenic , Protein Interaction Domains and Motifs
8.
Nat Med ; 25(5): 776-783, 2019 05.
Article in English | MEDLINE | ID: mdl-30911135

ABSTRACT

Re-expression of the paralogous γ-globin genes (HBG1/2) could be a universal strategy to ameliorate the severe ß-globin disorders sickle cell disease (SCD) and ß-thalassemia by induction of fetal hemoglobin (HbF, α2γ2)1. Previously, we and others have shown that core sequences at the BCL11A erythroid enhancer are required for repression of HbF in adult-stage erythroid cells but are dispensable in non-erythroid cells2-6. CRISPR-Cas9-mediated gene modification has demonstrated variable efficiency, specificity, and persistence in hematopoietic stem cells (HSCs). Here, we demonstrate that Cas9:sgRNA ribonucleoprotein (RNP)-mediated cleavage within a GATA1 binding site at the +58 BCL11A erythroid enhancer results in highly penetrant disruption of this motif, reduction of BCL11A expression, and induction of fetal γ-globin. We optimize conditions for selection-free on-target editing in patient-derived HSCs as a nearly complete reaction lacking detectable genotoxicity or deleterious impact on stem cell function. HSCs preferentially undergo non-homologous compared with microhomology-mediated end joining repair. Erythroid progeny of edited engrafting SCD HSCs express therapeutic levels of HbF and resist sickling, while those from patients with ß-thalassemia show restored globin chain balance. Non-homologous end joining repair-based BCL11A enhancer editing approaching complete allelic disruption in HSCs is a practicable therapeutic strategy to produce durable HbF induction.


Subject(s)
Gene Editing/methods , Hematopoietic Stem Cells/metabolism , Amino Acid Sequence , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/therapy , Base Sequence , CRISPR-Cas Systems , Carrier Proteins/genetics , Enhancer Elements, Genetic , Erythroid Precursor Cells/metabolism , Fetal Hemoglobin/biosynthesis , Fetal Hemoglobin/genetics , Hematopoietic Stem Cell Transplantation , Humans , INDEL Mutation , Nuclear Proteins/genetics , RNA, Guide, Kinetoplastida/genetics , Repressor Proteins , beta-Thalassemia/blood , beta-Thalassemia/genetics , beta-Thalassemia/therapy , gamma-Globins/biosynthesis , gamma-Globins/genetics
9.
Genome Biol ; 19(1): 169, 2018 10 19.
Article in English | MEDLINE | ID: mdl-30340514

ABSTRACT

CRISPR/Cas9 pooled screening permits parallel evaluation of comprehensive guide RNA libraries to systematically perturb protein coding sequences in situ and correlate with functional readouts. For the analysis and visualization of the resulting datasets, we develop CRISPRO, a computational pipeline that maps functional scores associated with guide RNAs to genomes, transcripts, and protein coordinates and structures. No currently available tool has similar functionality. The ensuing genotype-phenotype linear and three-dimensional maps raise hypotheses about structure-function relationships at discrete protein regions. Machine learning based on CRISPRO features improves prediction of guide RNA efficacy. The CRISPRO tool is freely available at gitlab.com/bauerlab/crispro .


Subject(s)
CRISPR-Cas Systems/genetics , Gene Editing , Genome , Mutagenesis/genetics , Open Reading Frames/genetics , Cell Line , Humans , Molecular Sequence Annotation , Protein Structure, Secondary , RNA, Guide, Kinetoplastida/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
10.
Cancer Cell ; 33(3): 386-400.e5, 2018 03 12.
Article in English | MEDLINE | ID: mdl-29478914

ABSTRACT

To identify novel targets for acute myeloid leukemia (AML) therapy, we performed genome-wide CRISPR-Cas9 screening using AML cell lines, followed by a second screen in vivo. Here, we show that the mRNA decapping enzyme scavenger (DCPS) gene is essential for AML cell survival. The DCPS enzyme interacted with components of pre-mRNA metabolic pathways, including spliceosomes, as revealed by mass spectrometry. RG3039, a DCPS inhibitor originally developed to treat spinal muscular atrophy, exhibited anti-leukemic activity via inducing pre-mRNA mis-splicing. Humans harboring germline biallelic DCPS loss-of-function mutations do not exhibit aberrant hematologic phenotypes, indicating that DCPS is dispensable for human hematopoiesis. Our findings shed light on a pre-mRNA metabolic pathway and identify DCPS as a target for AML therapy.


Subject(s)
CRISPR-Cas Systems/drug effects , Endoribonucleases/drug effects , Leukemia/drug therapy , Muscular Atrophy, Spinal/drug therapy , Quinazolines/pharmacology , Animals , CRISPR-Cas Systems/genetics , Cell Line , Endoribonucleases/genetics , Endoribonucleases/metabolism , Humans , Leukemia/genetics , Male , Metabolic Networks and Pathways/drug effects , Mice, Inbred C57BL , Muscular Atrophy, Spinal/genetics , RNA Precursors/drug effects , RNA Precursors/genetics , RNA Splicing/drug effects , RNA Splicing/genetics , RNA, Messenger/genetics
SELECTION OF CITATIONS
SEARCH DETAIL