Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 185(16): 3041-3055.e25, 2022 08 04.
Article in English | MEDLINE | ID: mdl-35917817

ABSTRACT

Rare copy-number variants (rCNVs) include deletions and duplications that occur infrequently in the global human population and can confer substantial risk for disease. In this study, we aimed to quantify the properties of haploinsufficiency (i.e., deletion intolerance) and triplosensitivity (i.e., duplication intolerance) throughout the human genome. We harmonized and meta-analyzed rCNVs from nearly one million individuals to construct a genome-wide catalog of dosage sensitivity across 54 disorders, which defined 163 dosage sensitive segments associated with at least one disorder. These segments were typically gene dense and often harbored dominant dosage sensitive driver genes, which we were able to prioritize using statistical fine-mapping. Finally, we designed an ensemble machine-learning model to predict probabilities of dosage sensitivity (pHaplo & pTriplo) for all autosomal genes, which identified 2,987 haploinsufficient and 1,559 triplosensitive genes, including 648 that were uniquely triplosensitive. This dosage sensitivity resource will provide broad utility for human disease research and clinical genetics.


Subject(s)
DNA Copy Number Variations , Genome, Human , DNA Copy Number Variations/genetics , Gene Dosage , Haploinsufficiency/genetics , Humans
2.
Cell ; 184(20): 5247-5260.e19, 2021 09 30.
Article in English | MEDLINE | ID: mdl-34534445

ABSTRACT

3' untranslated region (3'UTR) variants are strongly associated with human traits and diseases, yet few have been causally identified. We developed the massively parallel reporter assay for 3'UTRs (MPRAu) to sensitively assay 12,173 3'UTR variants. We applied MPRAu to six human cell lines, focusing on genetic variants associated with genome-wide association studies (GWAS) and human evolutionary adaptation. MPRAu expands our understanding of 3'UTR function, suggesting that simple sequences predominately explain 3'UTR regulatory activity. We adapt MPRAu to uncover diverse molecular mechanisms at base pair resolution, including an adenylate-uridylate (AU)-rich element of LEPR linked to potential metabolic evolutionary adaptations in East Asians. We nominate hundreds of 3'UTR causal variants with genetically fine-mapped phenotype associations. Using endogenous allelic replacements, we characterize one variant that disrupts a miRNA site regulating the viral defense gene TRIM14 and one that alters PILRB abundance, nominating a causal variant underlying transcriptional changes in age-related macular degeneration.


Subject(s)
3' Untranslated Regions/genetics , Biological Evolution , Disease/genetics , Genome-Wide Association Study , Algorithms , Alleles , Gene Expression Regulation , Genes, Reporter , Genetic Variation , Humans , Phenotype , Polymorphism, Single Nucleotide/genetics , Polyribosomes/metabolism , Quantitative Trait Loci/genetics , RNA/genetics
3.
Cell ; 176(6): 1325-1339.e22, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30827679

ABSTRACT

Lineage tracing provides key insights into the fate of individual cells in complex organisms. Although effective genetic labeling approaches are available in model systems, in humans, most approaches require detection of nuclear somatic mutations, which have high error rates, limited scale, and do not capture cell state information. Here, we show that somatic mutations in mtDNA can be tracked by single-cell RNA or assay for transposase accessible chromatin (ATAC) sequencing. We leverage somatic mtDNA mutations as natural genetic barcodes and demonstrate their utility as highly accurate clonal markers to infer cellular relationships. We track native human cells both in vitro and in vivo and relate clonal dynamics to gene expression and chromatin accessibility. Our approach should allow clonal tracking at a 1,000-fold greater scale than with nuclear genome sequencing, with simultaneous information on cell state, opening the way to chart cellular dynamics in human health and disease.


Subject(s)
DNA, Mitochondrial/genetics , Mitochondria/genetics , Base Sequence , Cell Lineage , Chromatin , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Genomics/methods , HEK293 Cells , Hematopoietic Stem Cells/physiology , High-Throughput Nucleotide Sequencing/methods , Humans , Mutation , Single-Cell Analysis , Transposases
4.
Cell ; 173(1): 90-103.e19, 2018 03 22.
Article in English | MEDLINE | ID: mdl-29551269

ABSTRACT

Blood cell formation is classically thought to occur through a hierarchical differentiation process, although recent studies have shown that lineage commitment may occur earlier in hematopoietic stem and progenitor cells (HSPCs). The relevance to human blood diseases and the underlying regulation of these refined models remain poorly understood. By studying a genetic blood disorder, Diamond-Blackfan anemia (DBA), where the majority of mutations affect ribosomal proteins and the erythroid lineage is selectively perturbed, we are able to gain mechanistic insight into how lineage commitment is programmed normally and disrupted in disease. We show that in DBA, the pool of available ribosomes is limited, while ribosome composition remains constant. Surprisingly, this global reduction in ribosome levels more profoundly alters translation of a select subset of transcripts. We show how the reduced translation of select transcripts in HSPCs can impair erythroid lineage commitment, illuminating a regulatory role for ribosome levels in cellular differentiation.


Subject(s)
Anemia, Diamond-Blackfan/pathology , Ribosomes/metabolism , 5' Untranslated Regions , Anemia, Diamond-Blackfan/genetics , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Bone Marrow Cells/metabolism , Cells, Cultured , Female , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Male , Mutation, Missense , RNA Interference , RNA, Small Interfering/metabolism , Ribosomal Proteins/antagonists & inhibitors , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Ribosomes/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Cell ; 168(6): 1053-1064.e15, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28283061

ABSTRACT

Cytokines are classically thought to stimulate downstream signaling pathways through monotonic activation of receptors. We describe a severe anemia resulting from a homozygous mutation (R150Q) in the cytokine erythropoietin (EPO). Surprisingly, the EPO R150Q mutant shows only a mild reduction in affinity for its receptor but has altered binding kinetics. The EPO mutant is less effective at stimulating erythroid cell proliferation and differentiation, even at maximally potent concentrations. While the EPO mutant can stimulate effectors such as STAT5 to a similar extent as the wild-type ligand, there is reduced JAK2-mediated phosphorylation of select downstream targets. This impairment in downstream signaling mechanistically arises from altered receptor dimerization dynamics due to extracellular binding changes. These results demonstrate how variation in a single cytokine can lead to biased downstream signaling and can thereby cause human disease. Moreover, we have defined a distinct treatable form of anemia through mutation identification and functional studies.


Subject(s)
Anemia, Diamond-Blackfan/genetics , Anemia, Diamond-Blackfan/pathology , Erythropoietin/genetics , Mutation, Missense , Signal Transduction , Anemia, Diamond-Blackfan/therapy , Child , Consanguinity , Enzyme Activation , Erythropoiesis , Erythropoietin/chemistry , Female , Humans , Janus Kinase 2/metabolism , Kinetics , Male , Receptors, Erythropoietin/chemistry , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/metabolism
6.
Cell ; 165(6): 1530-1545, 2016 Jun 02.
Article in English | MEDLINE | ID: mdl-27259154

ABSTRACT

Genome-wide association studies (GWAS) have successfully identified thousands of associations between common genetic variants and human disease phenotypes, but the majority of these variants are non-coding, often requiring genetic fine-mapping, epigenomic profiling, and individual reporter assays to delineate potential causal variants. We employ a massively parallel reporter assay (MPRA) to simultaneously screen 2,756 variants in strong linkage disequilibrium with 75 sentinel variants associated with red blood cell traits. We show that this assay identifies elements with endogenous erythroid regulatory activity. Across 23 sentinel variants, we conservatively identified 32 MPRA functional variants (MFVs). We used targeted genome editing to demonstrate endogenous enhancer activity across 3 MFVs that predominantly affect the transcription of SMIM1, RBM38, and CD164. Functional follow-up of RBM38 delineates a key role for this gene in the alternative splicing program occurring during terminal erythropoiesis. Finally, we provide evidence for how common GWAS-nominated variants can disrupt cell-type-specific transcriptional regulatory pathways.


Subject(s)
Erythrocytes , Genetic Techniques , Genetic Variation , Alternative Splicing , Cell Line , Cell Lineage/genetics , Erythropoiesis/genetics , Gene Library , Genes, Reporter , Humans , Regulatory Sequences, Nucleic Acid , Transcription, Genetic
7.
Nature ; 625(7996): 735-742, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38030727

ABSTRACT

Noncoding DNA is central to our understanding of human gene regulation and complex diseases1,2, and measuring the evolutionary sequence constraint can establish the functional relevance of putative regulatory elements in the human genome3-9. Identifying the genomic elements that have become constrained specifically in primates has been hampered by the faster evolution of noncoding DNA compared to protein-coding DNA10, the relatively short timescales separating primate species11, and the previously limited availability of whole-genome sequences12. Here we construct a whole-genome alignment of 239 species, representing nearly half of all extant species in the primate order. Using this resource, we identified human regulatory elements that are under selective constraint across primates and other mammals at a 5% false discovery rate. We detected 111,318 DNase I hypersensitivity sites and 267,410 transcription factor binding sites that are constrained specifically in primates but not across other placental mammals and validate their cis-regulatory effects on gene expression. These regulatory elements are enriched for human genetic variants that affect gene expression and complex traits and diseases. Our results highlight the important role of recent evolution in regulatory sequence elements differentiating primates, including humans, from other placental mammals.


Subject(s)
Conserved Sequence , Evolution, Molecular , Genome , Primates , Animals , Female , Humans , Pregnancy , Conserved Sequence/genetics , Deoxyribonuclease I/metabolism , DNA/genetics , DNA/metabolism , Genome/genetics , Mammals/classification , Mammals/genetics , Placenta , Primates/classification , Primates/genetics , Regulatory Sequences, Nucleic Acid/genetics , Reproducibility of Results , Transcription Factors/metabolism , Proteins/genetics , Gene Expression Regulation/genetics
8.
Nature ; 593(7858): 238-243, 2021 05.
Article in English | MEDLINE | ID: mdl-33828297

ABSTRACT

Genome-wide association studies (GWAS) have identified thousands of noncoding loci that are associated with human diseases and complex traits, each of which could reveal insights into the mechanisms of disease1. Many of the underlying causal variants may affect enhancers2,3, but we lack accurate maps of enhancers and their target genes to interpret such variants. We recently developed the activity-by-contact (ABC) model to predict which enhancers regulate which genes and validated the model using CRISPR perturbations in several cell types4. Here we apply this ABC model to create enhancer-gene maps in 131 human cell types and tissues, and use these maps to interpret the functions of GWAS variants. Across 72 diseases and complex traits, ABC links 5,036 GWAS signals to 2,249 unique genes, including a class of 577 genes that appear to influence multiple phenotypes through variants in enhancers that act in different cell types. In inflammatory bowel disease (IBD), causal variants are enriched in predicted enhancers by more than 20-fold in particular cell types such as dendritic cells, and ABC achieves higher precision than other regulatory methods at connecting noncoding variants to target genes. These variant-to-function maps reveal an enhancer that contains an IBD risk variant and that regulates the expression of PPIF to alter the membrane potential of mitochondria in macrophages. Our study reveals principles of genome regulation, identifies genes that affect IBD and provides a resource and generalizable strategy to connect risk variants of common diseases to their molecular and cellular functions.


Subject(s)
Enhancer Elements, Genetic/genetics , Genetic Predisposition to Disease , Genetic Variation/genetics , Genome, Human/genetics , Genome-Wide Association Study , Inflammatory Bowel Diseases/genetics , Cell Line , Chromosomes, Human, Pair 10/genetics , Cyclophilins/genetics , Dendritic Cells , Female , Humans , Macrophages/metabolism , Male , Mitochondria/metabolism , Organ Specificity/genetics , Phenotype
9.
PLoS Genet ; 19(9): e1010932, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37721944

ABSTRACT

The eQTL Catalogue is an open database of uniformly processed human molecular quantitative trait loci (QTLs). We are continuously updating the resource to further increase its utility for interpreting genetic associations with complex traits. Over the past two years, we have increased the number of uniformly processed studies from 21 to 31 and added X chromosome QTLs for 19 compatible studies. We have also implemented Leafcutter to directly identify splice-junction usage QTLs in all RNA sequencing datasets. Finally, to improve the interpretability of transcript-level QTLs, we have developed static QTL coverage plots that visualise the association between the genotype and average RNA sequencing read coverage in the region for all 1.7 million fine mapped associations. To illustrate the utility of these updates to the eQTL Catalogue, we performed colocalisation analysis between vitamin D levels in the UK Biobank and all molecular QTLs in the eQTL Catalogue. Although most GWAS loci colocalised both with eQTLs and transcript-level QTLs, we found that visual inspection could sometimes be used to distinguish primary splicing QTLs from those that appear to be secondary consequences of large-effect gene expression QTLs. While these visually confirmed primary splicing QTLs explain just 6/53 of the colocalising signals, they are significantly less pleiotropic than eQTLs and identify a prioritised causal gene in 4/6 cases.


Subject(s)
Multifactorial Inheritance , Quantitative Trait Loci , Humans , Quantitative Trait Loci/genetics , Genotype , Base Sequence , Genome-Wide Association Study , Polymorphism, Single Nucleotide
10.
Nature ; 575(7784): 652-657, 2019 11.
Article in English | MEDLINE | ID: mdl-31748747

ABSTRACT

Mosaic loss of chromosome Y (LOY) in circulating white blood cells is the most common form of clonal mosaicism1-5, yet our knowledge of the causes and consequences of this is limited. Here, using a computational approach, we estimate that 20% of the male population represented in the UK Biobank study (n = 205,011) has detectable LOY. We identify 156 autosomal genetic determinants of LOY, which we replicate in 757,114 men of European and Japanese ancestry. These loci highlight genes that are involved in cell-cycle regulation and cancer susceptibility, as well as somatic drivers of tumour growth and targets of cancer therapy. We demonstrate that genetic susceptibility to LOY is associated with non-haematological effects on health in both men and women, which supports the hypothesis that clonal haematopoiesis is a biomarker of genomic instability in other tissues. Single-cell RNA sequencing identifies dysregulated expression of autosomal genes in leukocytes with LOY and provides insights into why clonal expansion of these cells may occur. Collectively, these data highlight the value of studying clonal mosaicism to uncover fundamental mechanisms that underlie cancer and other ageing-related diseases.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Y/genetics , Genetic Predisposition to Disease/genetics , Genomic Instability/genetics , Leukocytes/pathology , Mosaicism , Adult , Aged , Computational Biology , Databases, Genetic , Female , Genetic Markers/genetics , Humans , Male , Middle Aged , Neoplasms/genetics , United Kingdom
11.
Blood ; 139(16): 2534-2546, 2022 04 21.
Article in English | MEDLINE | ID: mdl-35030251

ABSTRACT

Master regulators, such as the hematopoietic transcription factor (TF) GATA1, play an essential role in orchestrating lineage commitment and differentiation. However, the precise mechanisms by which such TFs regulate transcription through interactions with specific cis-regulatory elements remain incompletely understood. Here, we describe a form of congenital hemolytic anemia caused by missense mutations in an intrinsically disordered region of GATA1, with a poorly understood role in transcriptional regulation. Through integrative functional approaches, we demonstrate that these mutations perturb GATA1 transcriptional activity by partially impairing nuclear localization and selectively altering precise chromatin occupancy by GATA1. These alterations in chromatin occupancy and concordant chromatin accessibility changes alter faithful gene expression, with failure to both effectively silence and activate select genes necessary for effective terminal red cell production. We demonstrate how disease-causing mutations can reveal regulatory mechanisms that enable the faithful genomic targeting of master TFs during cellular differentiation.


Subject(s)
Anemia , GATA1 Transcription Factor , Cell Differentiation/genetics , Chromatin/genetics , Chromatin Immunoprecipitation , Erythropoiesis/genetics , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , Humans
12.
Nature ; 561(7721): 132-136, 2018 09.
Article in English | MEDLINE | ID: mdl-30150775

ABSTRACT

The human genome contains thousands of long non-coding RNAs1, but specific biological functions and biochemical mechanisms have been discovered for only about a dozen2-7. A specific long non-coding RNA-non-coding RNA activated by DNA damage (NORAD)-has recently been shown to be required for maintaining genomic stability8, but its molecular mechanism is unknown. Here we combine RNA antisense purification and quantitative mass spectrometry to identify proteins that directly interact with NORAD in living cells. We show that NORAD interacts with proteins involved in DNA replication and repair in steady-state cells and localizes to the nucleus upon stimulation with replication stress or DNA damage. In particular, NORAD interacts with RBMX, a component of the DNA-damage response, and contains the strongest RBMX-binding site in the transcriptome. We demonstrate that NORAD controls the ability of RBMX to assemble a ribonucleoprotein complex-which we term NORAD-activated ribonucleoprotein complex 1 (NARC1)-that contains the known suppressors of genomic instability topoisomerase I (TOP1), ALYREF and the PRPF19-CDC5L complex. Cells depleted for NORAD or RBMX display an increased frequency of chromosome segregation defects, reduced replication-fork velocity and altered cell-cycle progression-which represent phenotypes that are mechanistically linked to TOP1 and PRPF19-CDC5L function. Expression of NORAD in trans can rescue defects caused by NORAD depletion, but rescue is significantly impaired when the RBMX-binding site in NORAD is deleted. Our results demonstrate that the interaction between NORAD and RBMX is important for NORAD function, and that NORAD is required for the assembly of the previously unknown topoisomerase complex NARC1, which contributes to maintaining genomic stability. In addition, we uncover a previously unknown function for long non-coding RNAs in modulating the ability of an RNA-binding protein to assemble a higher-order ribonucleoprotein complex.


Subject(s)
DNA Topoisomerases, Type I/metabolism , Genomic Instability , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , RNA, Long Noncoding/metabolism , RNA-Binding Proteins/metabolism , Binding Sites , Cell Cycle , Cell Cycle Proteins/metabolism , Cell Nucleus/metabolism , Cell Survival , Chromosome Segregation , DNA Damage , DNA Repair , DNA Repair Enzymes/metabolism , DNA Replication , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , Mass Spectrometry , Nuclear Proteins/metabolism , Protein Binding , RNA Splicing Factors/metabolism , RNA, Long Noncoding/genetics , Ribonucleoproteins/metabolism , Transcription Factors/metabolism
14.
Am J Hum Genet ; 103(6): 930-947, 2018 12 06.
Article in English | MEDLINE | ID: mdl-30503522

ABSTRACT

Diamond-Blackfan anemia (DBA) is a rare bone marrow failure disorder that affects 7 out of 1,000,000 live births and has been associated with mutations in components of the ribosome. In order to characterize the genetic landscape of this heterogeneous disorder, we recruited a cohort of 472 individuals with a clinical diagnosis of DBA and performed whole-exome sequencing (WES). We identified relevant rare and predicted damaging mutations for 78% of individuals. The majority of mutations were singletons, absent from population databases, predicted to cause loss of function, and located in 1 of 19 previously reported ribosomal protein (RP)-encoding genes. Using exon coverage estimates, we identified and validated 31 deletions in RP genes. We also observed an enrichment for extended splice site mutations and validated their diverse effects using RNA sequencing in cell lines obtained from individuals with DBA. Leveraging the size of our cohort, we observed robust genotype-phenotype associations with congenital abnormalities and treatment outcomes. We further identified rare mutations in seven previously unreported RP genes that may cause DBA, as well as several distinct disorders that appear to phenocopy DBA, including nine individuals with biallelic CECR1 mutations that result in deficiency of ADA2. However, no new genes were identified at exome-wide significance, suggesting that there are no unidentified genes containing mutations readily identified by WES that explain >5% of DBA-affected case subjects. Overall, this report should inform not only clinical practice for DBA-affected individuals, but also the design and analysis of rare variant studies for heterogeneous Mendelian disorders.


Subject(s)
Anemia, Diamond-Blackfan/genetics , Adolescent , Child , Child, Preschool , Cohort Studies , Exome/genetics , Exons/genetics , Female , Gene Deletion , Genetic Association Studies/methods , Humans , Intercellular Signaling Peptides and Proteins/genetics , Male , Mutation/genetics , Phenotype , Ribosomal Proteins/genetics , Ribosomes/genetics , Sequence Analysis, RNA/methods , Exome Sequencing/methods
15.
PLoS Genet ; 14(3): e1007293, 2018 03.
Article in English | MEDLINE | ID: mdl-29590102

ABSTRACT

Co-inheritance of α-thalassemia has a significant protective effect on the severity of complications of sickle cell disease (SCD), including stroke. However, little information exists on the association and interactions for the common African ancestral α-thalassemia mutation (-α3.7 deletion) and ß-globin traits (HbS trait [SCT] and HbC trait) on important clinical phenotypes such as red blood cell parameters, anemia, and chronic kidney disease (CKD). In a community-based cohort of 2,916 African Americans from the Jackson Heart Study, we confirmed the expected associations between SCT, HbC trait, and the -α3.7 deletion with lower mean corpuscular volume/mean corpuscular hemoglobin and higher red blood cell count and red cell distribution width. In addition to the recently recognized association of SCT with lower estimated glomerular filtration rate and glycated hemoglobin (HbA1c), we observed a novel association of the -α3.7 deletion with higher HbA1c levels. Co-inheritance of each additional copy of the -α3.7 deletion significantly lowered the risk of anemia and chronic kidney disease among individuals with SCT (P-interaction = 0.031 and 0.019, respectively). Furthermore, co-inheritance of a novel α-globin regulatory variant was associated with normalization of red cell parameters in individuals with the -α3.7 deletion and significantly negated the protective effect of α-thalassemia on stroke in 1,139 patients with sickle cell anemia from the Cooperative Study of Sickle Cell Disease (CSSCD) (P-interaction = 0.0049). Functional assays determined that rs11865131, located in the major alpha-globin enhancer MCS-R2, was the most likely causal variant. These findings suggest that common α- and ß-globin variants interact to influence hematologic and clinical phenotypes in African Americans, with potential implications for risk-stratification and counseling of individuals with SCD and SCT.


Subject(s)
Anemia, Sickle Cell/genetics , Hemoglobin, Sickle/genetics , Sickle Cell Trait , alpha-Globins/genetics , Adult , Black or African American , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/physiopathology , Cohort Studies , DNA Copy Number Variations , Erythrocytes, Abnormal , Glomerular Filtration Rate , Glycated Hemoglobin/metabolism , Humans , Phenotype , Young Adult , alpha-Thalassemia/genetics
16.
Proc Natl Acad Sci U S A ; 114(3): E327-E336, 2017 01 17.
Article in English | MEDLINE | ID: mdl-28031487

ABSTRACT

Genetic variants affecting hematopoiesis can influence commonly measured blood cell traits. To identify factors that affect hematopoiesis, we performed association studies for blood cell traits in the population-based Estonian Biobank using high-coverage whole-genome sequencing (WGS) in 2,284 samples and SNP genotyping in an additional 14,904 samples. Using up to 7,134 samples with available phenotype data, our analyses identified 17 associations across 14 blood cell traits. Integration of WGS-based fine-mapping and complementary epigenomic datasets provided evidence for causal mechanisms at several loci, including at a previously undiscovered basophil count-associated locus near the master hematopoietic transcription factor CEBPA The fine-mapped variant at this basophil count association near CEBPA overlapped an enhancer active in common myeloid progenitors and influenced its activity. In situ perturbation of this enhancer by CRISPR/Cas9 mutagenesis in hematopoietic stem and progenitor cells demonstrated that it is necessary for and specifically regulates CEBPA expression during basophil differentiation. We additionally identified basophil count-associated variation at another more pleiotropic myeloid enhancer near GATA2, highlighting regulatory mechanisms for ordered expression of master hematopoietic regulators during lineage specification. Our study illustrates how population-based genetic studies can provide key insights into poorly understood cell differentiation processes of considerable physiologic relevance.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , Hematopoiesis/genetics , Base Sequence , Basophils/cytology , Cell Differentiation/genetics , Cell Lineage/genetics , Chromosome Mapping , Databases, Nucleic Acid , Enhancer Elements, Genetic , Epigenesis, Genetic , Estonia , Female , GATA2 Transcription Factor/genetics , Gene Expression Regulation, Developmental , Genome-Wide Association Study , Humans , Leukocyte Count , Male , Polymorphism, Single Nucleotide , Whole Genome Sequencing
17.
Proc Natl Acad Sci U S A ; 113(16): 4434-9, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27044088

ABSTRACT

Whole-exome sequencing has been incredibly successful in identifying causal genetic variants and has revealed a number of novel genes associated with blood and other diseases. One limitation of this approach is that it overlooks mutations in noncoding regulatory elements. Furthermore, the mechanisms by which mutations in transcriptionalcis-regulatory elements result in disease remain poorly understood. Here we used CRISPR/Cas9 genome editing to interrogate three such elements harboring mutations in human erythroid disorders, which in all cases are predicted to disrupt a canonical binding motif for the hematopoietic transcription factor GATA1. Deletions of as few as two to four nucleotides resulted in a substantial decrease (>80%) in target gene expression. Isolated deletions of the canonical GATA1 binding motif completely abrogated binding of the cofactor TAL1, which binds to a separate motif. Having verified the functionality of these three GATA1 motifs, we demonstrate strong evolutionary conservation of GATA1 motifs in regulatory elements proximal to other genes implicated in erythroid disorders, and show that targeted disruption of such elements results in altered gene expression. By modeling transcription factor binding patterns, we show that multiple transcription factors are associated with erythroid gene expression, and have created predictive maps modeling putative disruptions of their binding sites at key regulatory elements. Our study provides insight into GATA1 transcriptional activity and may prove a useful resource for investigating the pathogenicity of noncoding variants in human erythroid disorders.


Subject(s)
Anemia, Diamond-Blackfan/metabolism , GATA1 Transcription Factor/metabolism , Mutation , Response Elements , Transcription, Genetic , Anemia, Diamond-Blackfan/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , CRISPR-Cas Systems , GATA1 Transcription Factor/genetics , Humans , K562 Cells , Nucleotide Motifs , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , T-Cell Acute Lymphocytic Leukemia Protein 1
19.
Am J Hematol ; 92(9): E513-E519, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28568895

ABSTRACT

Immunodeficient mouse models have been valuable for studies of human hematopoiesis, but high-fidelity recapitulation of erythropoiesis in most xenograft recipients remains elusive. Recently developed immunodeficient and Kit mutant mice, however, have provided a suitable background to achieve higher-level human erythropoiesis after long-term hematopoietic engraftment. While there has been some characterization of human erythropoiesis in these models, a comprehensive analysis from various human developmental stages has not yet been reported. Here, we have utilized cell surface phenotypes, morphologic analyses, and molecular studies to fully characterize human erythropoiesis from multiple developmental stages in immunodeficient and Kit mutant mouse models following long-term hematopoietic stem and progenitor cell engraftment. We show that human erythropoiesis in such models demonstrates complete maturation and enucleation, as well as developmentally appropriate globin gene expression. These results provide a framework for future studies to utilize this model system for interrogating disorders affecting human erythropoiesis and for developing improved therapeutic approaches.


Subject(s)
Erythropoiesis , Hematopoietic Stem Cell Transplantation , Mutation , Proto-Oncogene Proteins c-kit/metabolism , Animals , Heterografts , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Proto-Oncogene Proteins c-kit/genetics
20.
PLoS Genet ; 10(12): e1004890, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25521328

ABSTRACT

Erythropoiesis is one of the best understood examples of cellular differentiation. Morphologically, erythroid differentiation proceeds in a nearly identical fashion between humans and mice, but recent evidence has shown that networks of gene expression governing this process are divergent between species. We undertook a systematic comparative analysis of six histone modifications and four transcriptional master regulators in primary proerythroblasts and erythroid cell lines to better understand the underlying basis of these transcriptional differences. Our analyses suggest that while chromatin structure across orthologous promoters is strongly conserved, subtle differences are associated with transcriptional divergence between species. Many transcription factor (TF) occupancy sites were poorly conserved across species (∼25% for GATA1, TAL1, and NFE2) but were more conserved between proerythroblasts and cell lines derived from the same species. We found that certain cis-regulatory modules co-occupied by GATA1, TAL1, and KLF1 are under strict evolutionary constraint and localize to genes necessary for erythroid cell identity. More generally, we show that conserved TF occupancy sites are indicative of active regulatory regions and strong gene expression that is sustained during maturation. Our results suggest that evolutionary turnover of TF binding sites associates with changes in the underlying chromatin structure, driving transcriptional divergence. We provide examples of how this framework can be applied to understand epigenomic variation in specific regulatory regions, such as the ß-globin gene locus. Our findings have important implications for understanding epigenomic changes that mediate variation in cellular differentiation across species, while also providing a valuable resource for studies of hematopoiesis.


Subject(s)
Cell Differentiation , Chromatin/genetics , Erythroid Precursor Cells/physiology , Erythropoiesis , Animals , Base Sequence , Chromatin/metabolism , Conserved Sequence , Epigenesis, Genetic , Evolution, Molecular , Histones/metabolism , Humans , K562 Cells , Promoter Regions, Genetic , Protein Binding , Protein Processing, Post-Translational , Species Specificity , Transcription Factors/physiology , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL