Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Blood ; 142(25): 2198-2215, 2023 12 21.
Article in English | MEDLINE | ID: mdl-37738561

ABSTRACT

ABSTRACT: Regulation of RNA polymerase II (RNAPII) activity is an essential process that governs gene expression; however, its contribution to the fundamental process of erythropoiesis remains unclear. hexamethylene bis-acetamide inducible 1 (HEXIM1) regulates RNAPII activity by controlling the location and activity of positive transcription factor ß. We identified a key role for HEXIM1 in controlling erythroid gene expression and function, with overexpression of HEXIM1 promoting erythroid proliferation and fetal globin expression. HEXIM1 regulated erythroid proliferation by enforcing RNAPII pausing at cell cycle check point genes and increasing RNAPII occupancy at genes that promote cycle progression. Genome-wide profiling of HEXIM1 revealed that it was increased at both repressed and activated genes. Surprisingly, there were also genome-wide changes in the distribution of GATA-binding factor 1 (GATA1) and RNAPII. The most dramatic changes occurred at the ß-globin loci, where there was loss of RNAPII and GATA1 at ß-globin and gain of these factors at γ-globin. This resulted in increased expression of fetal globin, and BGLT3, a long noncoding RNA in the ß-globin locus that regulates fetal globin expression. GATA1 was a key determinant of the ability of HEXIM1 to repress or activate gene expression. Genes that gained both HEXIM1 and GATA1 had increased RNAPII and increased gene expression, whereas genes that gained HEXIM1 but lost GATA1 had an increase in RNAPII pausing and decreased expression. Together, our findings reveal a central role for universal transcription machinery in regulating key aspects of erythropoiesis, including cell cycle progression and fetal gene expression, which could be exploited for therapeutic benefit.


Subject(s)
Erythropoiesis , Transcription Factors , Humans , Erythropoiesis/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Gene Expression Regulation , Transcription, Genetic , beta-Globins/genetics , beta-Globins/metabolism , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , RNA-Binding Proteins/genetics
2.
Am J Kidney Dis ; 83(6): 829-833, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38211685

ABSTRACT

The etiologies of newborn deaths in neonatal intensive care units usually remain unknown, even after genetic testing. Whole-genome sequencing, combined with artificial intelligence-based methods for predicting the effects of non-coding variants, provide an avenue for resolving these deaths. Using one such method, SpliceAI, we identified a maternally inherited deep intronic PKHD1 splice variant (chr6:52030169T>C), in trans with a pathogenic missense variant (p.Thr36Met), in a newborn who died of autosomal recessive polycystic kidney disease at age 2 days. We validated the deep intronic variant's impact in maternal urine-derived cells expressing PKHD1. Reverse transcription polymerase chain reaction followed by Sanger sequencing showed that the variant causes inclusion of 147bp of the canonical intron between exons 29 and 30 of PKHD1 into the mRNA, including a premature stop codon. Allele-specific expression analysis at a heterozygous site in the mother showed that the mutant allele completely suppresses canonical splicing. In an unrelated healthy control, there was no evidence of transcripts including the novel splice junction. We returned a diagnostic report to the parents, who underwent in vitro embryo selection.


Subject(s)
Introns , Polycystic Kidney, Autosomal Recessive , Receptors, Cell Surface , Humans , Infant, Newborn , Male , Introns/genetics , Mutation, Missense , Polycystic Kidney, Autosomal Recessive/genetics , Polycystic Kidney, Autosomal Recessive/diagnosis , Receptors, Cell Surface/genetics
3.
Blood ; 139(21): 3181-3193, 2022 05 26.
Article in English | MEDLINE | ID: mdl-35040907

ABSTRACT

Anemia of inflammation, also known as anemia of chronic disease, is refractory to erythropoietin (EPO) treatment, but the mechanisms underlying the EPO refractory state are unclear. Here, we demonstrate that high mobility group box-1 protein (HMGB1), a damage-associated molecular pattern molecule recently implicated in anemia development during sepsis, leads to reduced expansion and increased death of EPO-sensitive erythroid precursors in human models of erythropoiesis. HMGB1 significantly attenuates EPO-mediated phosphorylation of the Janus kinase 2/STAT5 and mTOR signaling pathways. Genetic ablation of receptor for advanced glycation end products, the only known HMGB1 receptor expressed by erythroid precursors, does not rescue the deleterious effects of HMGB1 on EPO signaling, either in human or murine precursors. Furthermore, surface plasmon resonance studies highlight the ability of HMGB1 to interfere with the binding between EPO and the EPOR. Administration of a monoclonal anti-HMGB1 antibody after sepsis onset in mice partially restores EPO signaling in vivo. Thus, HMGB1-mediated restriction of EPO signaling contributes to the chronic phase of anemia of inflammation.


Subject(s)
Anemia , Erythropoietin , HMGB1 Protein , Sepsis , Anemia/genetics , Animals , Erythropoiesis/genetics , Erythropoietin/metabolism , Inflammation , Mice , Receptors, Erythropoietin/metabolism , Sepsis/complications
4.
Am J Hematol ; 99(1): 99-112, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37929634

ABSTRACT

Human erythropoiesis is a complex process leading to the production of 2.5 million red blood cells per second. Following commitment of hematopoietic stem cells to the erythroid lineage, this process can be divided into three distinct stages: erythroid progenitor differentiation, terminal erythropoiesis, and reticulocyte maturation. We recently resolved the heterogeneity of erythroid progenitors into four different subpopulations termed EP1-EP4. Here, we characterized the growth factor(s) responsiveness of these four progenitor populations in terms of proliferation and differentiation. Using mass spectrometry-based proteomics on sorted erythroid progenitors, we quantified the absolute expression of ~5500 proteins from EP1 to EP4. Further functional analyses highlighted dynamic changes in cell cycle in these populations with an acceleration of the cell cycle during erythroid progenitor differentiation. The finding that E2F4 expression was increased from EP1 to EP4 is consistent with the noted changes in cell cycle. Finally, our proteomic data suggest that the protein machinery necessary for both oxidative phosphorylation and glycolysis is present in these progenitor cells. Together, our data provide comprehensive insights into growth factor-dependence of erythroid progenitor proliferation and the proteome of four distinct populations of human erythroid progenitors which will be a useful framework for the study of erythroid disorders.


Subject(s)
Hematopoietic Stem Cells , Proteomics , Humans , Cell Differentiation , Cell Cycle , Erythropoiesis , Metabolic Networks and Pathways , Intercellular Signaling Peptides and Proteins/metabolism , Erythroid Precursor Cells
5.
Genet Med ; 25(8): 100856, 2023 08.
Article in English | MEDLINE | ID: mdl-37092537

ABSTRACT

PURPOSE: Dominant variants in the retinoic acid receptor beta (RARB) gene underlie a syndromic form of microphthalmia, known as MCOPS12, which is associated with other birth anomalies and global developmental delay with spasticity and/or dystonia. Here, we report 25 affected individuals with 17 novel pathogenic or likely pathogenic variants in RARB. This study aims to characterize the functional impact of these variants and describe the clinical spectrum of MCOPS12. METHODS: We used in vitro transcriptional assays and in silico structural analysis to assess the functional relevance of RARB variants in affecting the normal response to retinoids. RESULTS: We found that all RARB variants tested in our assays exhibited either a gain-of-function or a loss-of-function activity. Loss-of-function variants disrupted RARB function through a dominant-negative effect, possibly by disrupting ligand binding and/or coactivators' recruitment. By reviewing clinical data from 52 affected individuals, we found that disruption of RARB is associated with a more variable phenotype than initially suspected, with the absence in some individuals of cardinal features of MCOPS12, such as developmental eye anomaly or motor impairment. CONCLUSION: Our study indicates that pathogenic variants in RARB are functionally heterogeneous and associated with extensive clinical heterogeneity.


Subject(s)
Microphthalmos , Receptors, Retinoic Acid , Humans , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Retinoids
6.
Blood ; 138(18): 1740-1756, 2021 11 04.
Article in English | MEDLINE | ID: mdl-34075391

ABSTRACT

The terminal maturation of human erythroblasts requires significant changes in gene expression in the context of dramatic nuclear condensation. Defects in this process are associated with inherited anemias and myelodysplastic syndromes. The progressively dense appearance of the condensing nucleus in maturing erythroblasts led to the assumption that heterochromatin accumulation underlies this process, but despite extensive study, the precise mechanisms underlying this essential biologic process remain elusive. To delineate the epigenetic changes associated with the terminal maturation of human erythroblasts, we performed mass spectrometry of histone posttranslational modifications combined with chromatin immunoprecipitation coupled with high-throughput sequencing, Assay for Transposase Accessible Chromatin, and RNA sequencing. Our studies revealed that the terminal maturation of human erythroblasts is associated with a dramatic decline in histone marks associated with active transcription elongation, without accumulation of heterochromatin. Chromatin structure and gene expression were instead correlated with dynamic changes in occupancy of elongation competent RNA polymerase II, suggesting that terminal erythroid maturation is controlled largely at the level of transcription. We further demonstrate that RNA polymerase II "pausing" is highly correlated with transcriptional repression, with elongation competent RNA polymerase II becoming a scare resource in late-stage erythroblasts, allocated to erythroid-specific genes. Functional studies confirmed an essential role for maturation stage-specific regulation of RNA polymerase II activity during erythroid maturation and demonstrate a critical role for HEXIM1 in the regulation of gene expression and RNA polymerase II activity in maturing erythroblasts. Taken together, our findings reveal important insights into the mechanisms that regulate terminal erythroid maturation and provide a novel paradigm for understanding normal and perturbed erythropoiesis.


Subject(s)
Erythroblasts/metabolism , Erythroid Cells/metabolism , RNA Polymerase II/metabolism , Cell Line , Chromatin/genetics , Chromatin/metabolism , Erythroblasts/cytology , Erythroid Cells/cytology , Erythropoiesis , Gene Expression Regulation, Developmental , Histones/genetics , Histones/metabolism , Humans , RNA Polymerase II/genetics , Transcription, Genetic
7.
Am J Med Genet A ; 191(3): 760-769, 2023 03.
Article in English | MEDLINE | ID: mdl-36495114

ABSTRACT

Arthrogryposis multiplex congenita (AMC) [also known as multiple joints contracture or Fetal Akinesia Deformation Sequence (FADS)] is etiologically a heterogeneous condition with an estimated incidence of approximately 1 in 3000 live births and much higher incidence when prenatally diagnosed cases are included. The condition can be acquired or secondary to fetal exposures and can also be caused by a variety of single-gene disorders affecting the brain, spinal cord, peripheral nerves, neuromuscular junction, muscle, and a variety of disorders affecting the connective tissues (Niles et al., Prenatal Diagnosis, 2019; 39:720-731). The introduction of next-generation gene sequencing uncovered many genes and causative variants of AMC but also identified genes that cause both dominant and recessive inherited conditions with the variability of clinical manifestations depending on the genes and variants. Molecular diagnosis in these cases is not only important for prognostication but also for the determination of recurrence risk and for providing reproductive options including preimplantation and prenatal diagnosis. TTN, the largest known gene in the human genome, has been known to be associated with autosomal dominant dilated cardiomyopathy. However, homozygote and compound heterozygote pathogenic variants with recessive inheritance have rarely been reported. We report the effect of recessive variants located within the fetal IC and/or N2BA isoforms in association with severe FADS in three families. All parents were healthy obligate carriers and none of them had cardiac or skeletal muscle abnormalities. This report solidifies FADS as an alternative phenotypic presentation associated with homozygote/compound heterozygous pathogenic variants in the TTN.


Subject(s)
Arthrogryposis , Pregnancy , Female , Humans , Arthrogryposis/diagnosis , Arthrogryposis/genetics , Prenatal Diagnosis , Homozygote , Prenatal Care , Syndrome , Connectin/genetics
8.
J Med Genet ; 57(5): 296-300, 2020 05.
Article in English | MEDLINE | ID: mdl-31662342

ABSTRACT

BACKGROUND: Alveolar capillary dysplasia with misalignment of the pulmonary veins (ACDMPV) is a lethal disorder of lung development. ACDMPV is associated with haploinsufficiency of the transcription factor FOXF1, which plays an important role in the development of the lung and intestine. CNVs upstream of the FOXF1 gene have also been associated with an ACDMPV phenotype, but mechanism(s) by which these deletions disrupt lung development are not well understood. The objective of our study is to gain insights into the mechanisms by which CNVs contribute to an ACDMPV phenotype. METHODS: We analysed primary lung tissue from an infant with classic clinical and histological findings of ACDMPV and harboured a 340 kb deletion on chromosome 16q24.1 located 250 kb upstream of FOXF1. RESULTS: In RNA generated from paraffin-fixed lung sections, our patient had lower expression of FOXF1 than age-matched controls. He also had an abnormal pattern of FOXF1 protein expression, with a dramatic loss of FOXF1 expression in the lung. To gain insights into the mechanisms underlying these changes, we assessed the epigenetic landscape using chromatin immunoprecipitation, which demonstrated loss of histone H3 lysine 27 acetylation (H3K27Ac), an epigenetic mark of active enhancers, in the region of the deletion. CONCLUSIONS: Together, these data suggest that the deletion disrupts an enhancer responsible for directing FOXF1 expression in the developing lung and provide novel insights into the mechanisms underlying a fatal developmental lung disorder.


Subject(s)
Forkhead Transcription Factors/genetics , Genetic Predisposition to Disease , Lung/metabolism , Persistent Fetal Circulation Syndrome/genetics , Chromosomes, Human, Pair 16/genetics , Enhancer Elements, Genetic/genetics , Gene Deletion , Gene Expression Regulation/genetics , Haploinsufficiency/genetics , Humans , Infant , Infant, Newborn , Lung/growth & development , Lung/pathology , Persistent Fetal Circulation Syndrome/pathology
9.
Gut ; 68(7): 1245-1258, 2019 07.
Article in English | MEDLINE | ID: mdl-30228219

ABSTRACT

OBJECTIVE: Here, we evaluate the contribution of AT-rich interaction domain-containing protein 1A (ARID1A), the most frequently mutated member of the SWItch/sucrose non-fermentable (SWI/SNF) complex, in pancreatic homeostasis and pancreatic ductal adenocarcinoma (PDAC) pathogenesis using mouse models. DESIGN: Mice with a targeted deletion of Arid1a in the pancreas by itself and in the context of two common genetic alterations in PDAC, Kras and p53, were followed longitudinally. Pancreases were examined and analysed for proliferation, response to injury and tumourigenesis. Cancer cell lines derived from these models were analysed for clonogenic, migratory, invasive and transcriptomic changes. RESULTS: Arid1a deletion in the pancreas results in progressive acinar-to-ductal metaplasia (ADM), loss of acinar mass, diminished acinar regeneration in response to injury and ductal cell expansion. Mutant Kras cooperates with homozygous deletion of Arid1a, leading to intraductal papillary mucinous neoplasm (IPMN). Arid1a loss in the context of mutant Kras and p53 leads to shorter tumour latency, with the resulting tumours being poorly differentiated. Cancer cell lines derived from Arid1a-mutant tumours are more mesenchymal, migratory, invasive and capable of anchorage-independent growth; gene expression analysis showed activation of epithelial-mesenchymal transition (EMT) and stem cell identity pathways that are partially dependent on Arid1a loss for dysregulation. CONCLUSIONS: ARID1A plays a key role in pancreatic acinar homeostasis and response to injury. Furthermore, ARID1A restrains oncogenic KRAS-driven formation of premalignant proliferative IPMN. Arid1a-deficient PDACs are poorly differentiated and have mesenchymal features conferring migratory/invasive and stem-like properties.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , DNA-Binding Proteins/genetics , Epithelial-Mesenchymal Transition/physiology , Nuclear Proteins/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Acinar Cells/pathology , Acinar Cells/physiology , Animals , Cell Proliferation , Disease Models, Animal , Homeostasis , Mice , Proto-Oncogene Proteins p21(ras)/genetics , Transcription Factors
11.
Nucleic Acids Res ; 44(15): 7173-88, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27141965

ABSTRACT

The modulation of chromatin structure is a key step in transcription regulation in mammalian cells and eventually determines lineage commitment and differentiation. USF1/2, Setd1a and NURF complexes interact to regulate chromatin architecture in erythropoiesis, but the mechanistic basis for this regulation is hitherto unknown. Here we showed that Setd1a and NURF complexes bind to promoters to control chromatin structural alterations and gene activation in a cell context dependent manner. In human primary erythroid cells USF1/2, H3K4me3 and the NURF complex were significantly co-enriched at transcription start sites of erythroid genes, and their binding was associated with promoter/enhancer accessibility that resulted from nucleosome repositioning. Mice deficient for Setd1a, an H3K4 trimethylase, in the erythroid compartment exhibited reduced Ter119/CD71 positive erythroblasts, peripheral blood RBCs and hemoglobin levels. Loss of Setd1a led to a reduction of promoter-associated H3K4 methylation, inhibition of gene transcription and blockade of erythroid differentiation. This was associated with alterations in NURF complex occupancy at erythroid gene promoters and reduced chromatin accessibility. Setd1a deficiency caused decreased associations between enhancer and promoter looped interactions as well as reduced expression of erythroid genes such as the adult ß-globin gene. These data indicate that Setd1a and NURF complexes are specifically targeted to and coordinately regulate erythroid promoter chromatin dynamics during erythroid lineage differentiation.


Subject(s)
Cell Lineage , Chromatin Assembly and Disassembly , Erythrocytes/cytology , Erythropoiesis , Gene Expression Regulation/genetics , Histone-Lysine N-Methyltransferase/metabolism , Multiprotein Complexes/metabolism , Animals , Antigens, Nuclear/metabolism , Cell Lineage/genetics , Cells, Cultured , Chromatin/genetics , Chromatin/metabolism , Chromatin Immunoprecipitation , Erythroblasts/cytology , Erythroblasts/metabolism , Erythrocyte Count , Erythrocytes/metabolism , Erythropoiesis/genetics , Female , Hemoglobins/metabolism , Histone-Lysine N-Methyltransferase/deficiency , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , Humans , Lysine/metabolism , Male , Methylation , Mice , Mice, Knockout , Micrococcal Nuclease/metabolism , Multiprotein Complexes/chemistry , Nerve Tissue Proteins/metabolism , Promoter Regions, Genetic/genetics , Spleen/cytology , Transcription Factors/metabolism , Upstream Stimulatory Factors/metabolism
12.
Hum Genet ; 135(5): 569-586, 2016 May.
Article in English | MEDLINE | ID: mdl-27071622

ABSTRACT

Alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV) is a lethal lung developmental disorder caused by heterozygous point mutations or genomic deletion copy-number variants (CNVs) of FOXF1 or its upstream enhancer involving fetal lung-expressed long noncoding RNA genes LINC01081 and LINC01082. Using custom-designed array comparative genomic hybridization, Sanger sequencing, whole exome sequencing (WES), and bioinformatic analyses, we studied 22 new unrelated families (20 postnatal and two prenatal) with clinically diagnosed ACDMPV. We describe novel deletion CNVs at the FOXF1 locus in 13 unrelated ACDMPV patients. Together with the previously reported cases, all 31 genomic deletions in 16q24.1, pathogenic for ACDMPV, for which parental origin was determined, arose de novo with 30 of them occurring on the maternally inherited chromosome 16, strongly implicating genomic imprinting of the FOXF1 locus in human lungs. Surprisingly, we have also identified four ACDMPV families with the pathogenic variants in the FOXF1 locus that arose on paternal chromosome 16. Interestingly, a combination of the severe cardiac defects, including hypoplastic left heart, and single umbilical artery were observed only in children with deletion CNVs involving FOXF1 and its upstream enhancer. Our data demonstrate that genomic imprinting at 16q24.1 plays an important role in variable ACDMPV manifestation likely through long-range regulation of FOXF1 expression, and may be also responsible for key phenotypic features of maternal uniparental disomy 16. Moreover, in one family, WES revealed a de novo missense variant in ESRP1, potentially implicating FGF signaling in the etiology of ACDMPV.


Subject(s)
Genome, Human , Genomic Imprinting , Persistent Fetal Circulation Syndrome/pathology , Pulmonary Alveoli/abnormalities , Pulmonary Veins/pathology , Chromosomes, Human, Pair 16/genetics , Comparative Genomic Hybridization , Female , Forkhead Transcription Factors/genetics , Genes, Lethal , High-Throughput Nucleotide Sequencing , Humans , Infant, Newborn , Male , Pedigree , Persistent Fetal Circulation Syndrome/genetics , Pulmonary Alveoli/pathology , Sequence Deletion
13.
J Biol Chem ; 288(12): 8433-8444, 2013 Mar 22.
Article in English | MEDLINE | ID: mdl-23341446

ABSTRACT

Identification of cell type-specific enhancers is important for understanding the regulation of programs controlling cellular development and differentiation. Enhancers are typically marked by the co-transcriptional activator protein p300 or by groups of cell-expressed transcription factors. We hypothesized that a unique set of enhancers regulates gene expression in human erythroid cells, a highly specialized cell type evolved to provide adequate amounts of oxygen throughout the body. Using chromatin immunoprecipitation followed by massively parallel sequencing, genome-wide maps of candidate enhancers were constructed for p300 and four transcription factors, GATA1, NF-E2, KLF1, and SCL, using primary human erythroid cells. These data were combined with gene expression analyses, and candidate enhancers were identified. Consistent with their predicted function as candidate enhancers, there was statistically significant enrichment of p300 and combinations of co-localizing erythroid transcription factors within 1-50 kb of the transcriptional start site (TSS) of genes highly expressed in erythroid cells. Candidate enhancers were also enriched near genes with known erythroid cell function or phenotype. Candidate enhancers exhibited moderate conservation with mouse and minimal conservation with nonplacental vertebrates. Candidate enhancers were mapped to a set of erythroid-associated, biologically relevant, SNPs from the genome-wide association studies (GWAS) catalogue of NHGRI, National Institutes of Health. Fourteen candidate enhancers, representing 10 genetic loci, mapped to sites associated with biologically relevant erythroid traits. Fragments from these loci directed statistically significant expression in reporter gene assays. Identification of enhancers in human erythroid cells will allow a better understanding of erythroid cell development, differentiation, structure, and function and provide insights into inherited and acquired hematologic disease.


Subject(s)
Enhancer Elements, Genetic , Erythroid Cells/metabolism , Gene Expression Regulation , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/physiology , Cells, Cultured , Chromatin/genetics , Chromatin/metabolism , Chromatin Immunoprecipitation , Conserved Sequence , E1A-Associated p300 Protein/metabolism , GATA1 Transcription Factor/metabolism , GATA1 Transcription Factor/physiology , Genes, Reporter , High-Throughput Nucleotide Sequencing , Humans , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/physiology , Luciferases, Firefly/biosynthesis , Luciferases, Firefly/genetics , Molecular Sequence Annotation , NF-E2 Transcription Factor, p45 Subunit/metabolism , NF-E2 Transcription Factor, p45 Subunit/physiology , Oligonucleotide Array Sequence Analysis , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Analysis, DNA , T-Cell Acute Lymphocytic Leukemia Protein 1 , Transcriptome
14.
Blood ; 120(17): 3586-93, 2012 Oct 25.
Article in English | MEDLINE | ID: mdl-22968456

ABSTRACT

The human ankyrin-1 gene (ANK1) contains 3 tissue-specific alternative promoters. We have shown previously that the erythroid-specific ankyrin 1 (ANK1E) core promoter contains a 5' DNase I hypersensitive site (HS) with barrier insulator function that prevents gene silencing in vitro and in vivo. Mutations in the ANK1E barrier region lead to decreased ANK1 mRNA levels and hereditary spherocytosis. In this report, we demonstrate a second ANK1E regulatory element located in an adjacent pair of DNase I HS located 5.6 kb 3' of the ANK1E promoter at the 3' boundary of an erythroid-specific DNase I-sensitive chromatin domain. The 3' regulatory element exhibits enhancer activity in vitro and in transgenic mice, and it has the histone modifications associated with an enhancer element. One of the ANK1E 3'HS contains an NF-E2 binding site that is required for enhancer function. We show that a chromatin loop brings the 3' enhancer and NF-E2 into proximity with the 5' barrier region including the ANK1E core promoter. These observations demonstrate a model for the tissue-specific activation of alternative promoters that may be applicable to the ∼ 30% of mammalian genes with alternative promoters that exhibit distinct expression patterns.


Subject(s)
Ankyrins/genetics , Chromatin/genetics , Enhancer Elements, Genetic , Insulator Elements , NF-E2 Transcription Factor, p45 Subunit/genetics , Promoter Regions, Genetic , Spherocytosis, Hereditary/genetics , 3' Untranslated Regions , 5' Untranslated Regions , Animals , Ankyrins/metabolism , Binding Sites , Cell Line, Tumor , Chromatin/chemistry , Chromatin/metabolism , Deoxyribonuclease I/genetics , Deoxyribonuclease I/metabolism , Histones/genetics , Histones/metabolism , Humans , K562 Cells , Mice , Mice, Transgenic , NF-E2 Transcription Factor, p45 Subunit/metabolism , Organ Specificity , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Spherocytosis, Hereditary/metabolism
15.
bioRxiv ; 2024 Feb 06.
Article in English | MEDLINE | ID: mdl-38370741

ABSTRACT

The limited proliferative capacity of erythroid precursors is a major obstacle to generate sufficient numbers of in vitro-derived red blood cells (RBC) for clinical purposes. We and others have determined that BMI1, a member of the polycomb repressive complex 1 (PRC1), is both necessary and sufficient to drive extensive proliferation of self-renewing erythroblasts (SREs). However, the mechanisms of BMI1 action remain poorly understood. BMI1 overexpression led to 10 billion-fold increase BMI1-induced (i)SRE self-renewal. Despite prolonged culture and BMI1 overexpression, human iSREs can terminally mature and agglutinate with typing reagent monoclonal antibodies against conventional RBC antigens. BMI1 and RING1B occupancy, along with repressive histone marks, were identified at known BMI1 target genes, including the INK-ARF locus, consistent with an altered cell cycle following BMI1 inhibition. We also identified upregulated BMI1 target genes with low repressive histone modifications, including key regulator of cholesterol homeostasis. Functional studies suggest that both cholesterol import and synthesis are essential for BMI1-associated self-renewal. These findings support the hypothesis that BMI1 regulates erythroid self-renewal not only through gene repression but also through gene activation and offer a strategy to expand the pool of immature erythroid precursors for eventual clinical uses.

16.
Blood ; 118(5): 1386-94, 2011 Aug 04.
Article in English | MEDLINE | ID: mdl-21653943

ABSTRACT

Chromatin insulators protect erythroid genes from being silenced during erythropoiesis, and the disruption of barrier insulator function in erythroid membrane gene loci results in mild or severe anemia. We showed previously that the USF1/2-bound 5'HS4 insulator mediates chromatin barrier activity in the erythroid-specific chicken ß-globin locus. It is currently not known how insulators establish such a barrier. To understand the function of USF1, we purified USF1-associated protein complexes and found that USF1 forms a multiprotein complex with hSET1 and NURF, thus exhibiting histone H3K4 methyltransferase- and ATP-dependent nucleosome remodeling activities, respectively. Both SET1 and NURF are recruited to the 5'HS4 insulator by USF1 to retain the active chromatin structure in erythrocytes. Knock-down of NURF resulted in a rapid loss of barrier activity accompanied by an alteration of nucleosome positioning, increased occupancy of the nucleosome-free linker region at the insulator site, and increased repressive H3K27me3 levels in the vicinity of the HS4 insulator. Furthermore, suppression of SET1 reduced barrier activity, decreased H3K4me2 and acH3K9/K14, and diminished the recruitment of BPTF at several erythroid-specific barrier insulator sites. Therefore, our data reveal a synergistic role of hSET1 and NURF in regulating the USF-bound barrier insulator to prevent erythroid genes from encroachment of heterochromatin.


Subject(s)
Chromatin Assembly and Disassembly/genetics , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/physiology , Histone-Lysine N-Methyltransferase/physiology , Animals , Chickens , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , HeLa Cells , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Insulator Elements/physiology , K562 Cells , Models, Biological , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Multiprotein Complexes/physiology , Protein Binding/genetics , Protein Binding/physiology , Tumor Cells, Cultured , Upstream Stimulatory Factors/metabolism
17.
Blood ; 118(17): e139-48, 2011 Oct 27.
Article in English | MEDLINE | ID: mdl-21900194

ABSTRACT

Erythropoiesis is dependent on the activity of transcription factors, including the erythroid-specific erythroid Kruppel-like factor (EKLF). ChIP followed by massively parallel sequencing (ChIP-Seq) is a powerful, unbiased method to map trans-factor occupancy. We used ChIP-Seq to study the interactome of EKLF in mouse erythroid progenitor cells and more differentiated erythroblasts. We correlated these results with the nuclear distribution of EKLF, RNA-Seq analysis of the transcriptome, and the occupancy of other erythroid transcription factors. In progenitor cells, EKLF is found predominantly at the periphery of the nucleus, where EKLF primarily occupies the promoter regions of genes and acts as a transcriptional activator. In erythroblasts, EKLF is distributed throughout the nucleus, and erythroblast-specific EKLF occupancy is predominantly in intragenic regions. In progenitor cells, EKLF modulates general cell growth and cell cycle regulatory pathways, whereas in erythroblasts EKLF is associated with repression of these pathways. The EKLF interactome shows very little overlap with the interactomes of GATA1, GATA2, or TAL1, leading to a model in which EKLF directs programs that are independent of those regulated by the GATA factors or TAL1.


Subject(s)
Chromatin Immunoprecipitation , Chromosome Mapping/methods , Erythrocytes/physiology , Erythroid Precursor Cells/physiology , Kruppel-Like Transcription Factors/physiology , Animals , Binding Sites/genetics , Cells, Cultured , Chromatin Immunoprecipitation/methods , Embryo, Mammalian , Erythrocytes/metabolism , Erythroid Precursor Cells/metabolism , Erythropoiesis/genetics , Erythropoiesis/physiology , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Transgenic , Protein Binding , Sequence Analysis, DNA/methods , Transcription Factors/metabolism
18.
J Biol Chem ; 286(45): 39457-65, 2011 Nov 11.
Article in English | MEDLINE | ID: mdl-21937433

ABSTRACT

Post-translational histone modifications, acting alone or in a context-dependent manner, influence numerous cellular processes via their regulation of gene expression. Monomethylation of histone H3 lysine 27 (K27me1) is a poorly understood histone modification. Some reports describe depletion of K27Me1 at promoters and transcription start sites (TSS), implying its depletion at TSS is necessary for active transcription, while others have associated enrichment of H3K27me1 at TSS with increased levels of mRNA expression. Tissue- and gene-specific patterns of H3K27me1 enrichment and their correlation with gene expression were determined via chromatin immunoprecipitation on chip microarray (ChIP-chip) and human mRNA expression array analyses. Results from erythroid cells were compared with those in neural and muscle cells. H3K27me1 enrichment varied depending on levels of cell-type specific gene expression, with highest enrichment over transcriptionally active genes. Over individual genes, the highest levels of H3K27me1 enrichment were found over the gene bodies of highly expressed genes. In contrast to H3K4me3, which was highly enriched at the TSS of actively transcribing genes, H3K27me1 was selectively depleted at the TSS of actively transcribed genes. There was markedly decreased to no H3K27me1 enrichment in genes with low expression. At some locations, H3K27 monomethylation was also found to be associated with chromatin signatures of gene enhancers.


Subject(s)
Chromatin/metabolism , Erythroid Cells/metabolism , Gene Expression Regulation/physiology , Histones/metabolism , Transcription, Genetic/physiology , Base Sequence , Chromatin/genetics , Enhancer Elements, Genetic/physiology , Erythroid Cells/cytology , Gene Deletion , Histones/genetics , Humans , K562 Cells , Methylation , Molecular Sequence Data , Organ Specificity/physiology
19.
Front Genet ; 13: 805265, 2022.
Article in English | MEDLINE | ID: mdl-35330735

ABSTRACT

Erythropoiesis is a process of enormous magnitude, with the average person generating two to three million red cells every second. Erythroid progenitors start as large cells with large nuclei, and over the course of three to four cell divisions they undergo a dramatic decrease in cell size accompanied by profound nuclear condensation, which culminates in enucleation. As maturing erythroblasts are undergoing these dramatic phenotypic changes, they accumulate hemoglobin and express high levels of other erythroid-specific genes, while silencing much of the non-erythroid transcriptome. These phenotypic and gene expression changes are associated with distinct changes in the chromatin landscape, and require close coordination between transcription factors and epigenetic regulators, as well as precise regulation of RNA polymerase II activity. Disruption of these processes are associated with inherited anemias and myelodysplastic syndromes. Here, we review the epigenetic mechanisms that govern terminal erythroid maturation, and their role in human disease.

20.
Cell Rep ; 40(9): 111253, 2022 08 30.
Article in English | MEDLINE | ID: mdl-36044839

ABSTRACT

Activating KRAS mutations and functional loss of members of the SWI/SNF complex, including ARID1A, are found together in the primary liver tumor cholangiocarcinoma (CC). How these mutations cooperate to promote CC has not been established. Using murine models of hepatocyte and biliary-specific lineage tracing, we show that Kras and Arid1a mutations drive the formation of CC and tumor precursors from the biliary compartment, which are accelerated by liver inflammation. Using cultured cells, we find that Arid1a loss causes cellular proliferation, escape from cell-cycle control, senescence, and widespread changes in chromatin structure. Notably, we show that the biliary proliferative response elicited by Kras/Arid1a cooperation and tissue injury in CC is caused by failed engagement of the TGF-ß-Smad4 tumor suppressor pathway. We thus identify an ARID1A-TGF-ß-Smad4 axis as essential in limiting the biliary epithelial response to oncogenic insults, while its loss leads to biliary pre-neoplasia and CC.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Animals , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/metabolism , Bile Ducts, Intrahepatic/pathology , Cholangiocarcinoma/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Mice , Mutation/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL