Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Feb 08.
Article in English | MEDLINE | ID: mdl-38370646

ABSTRACT

Prior evidence indicates that the erythroid cellular response to glucocorticoids (GC) has developmental specificity, namely, that developmentally more advanced cells that are undergoing or have undergone fetal to adult globin switching are more responsive to GC-induced expansion. To investigate the molecular underpinnings of this, we focused on the major developmental globin regulator BCL11A. We compared: a) levels of expression and nuclear content of BCL11A in adult erythroid cells upon GC stimulation; b) response to GC of CD34+ cells from patients with BCL11A microdeletions and reduced BCL11A expression, and; c) response to GC of two cellular models (HUDEP-2 and adult CD34+ cells) before and after reduction of BCL11A expression by shRNA. We observed that: a) GC-expanded erythroid cells from a large cohort of blood donors displayed amplified expression and nuclear accumulation of BCL11A; b) CD34+ cells from BCL11A microdeletion patients generated fewer erythroid cells when cultured with GC compared to their parents, while the erythroid expansion of the patients was similar to that of their parents in cultures without GC, and; c) adult CD34+ cells and HUDEP-2 cells with shRNA-depleted expression of BCL11A exhibit reduced expansion in response to GC. In addition, RNA-seq profiling of shRNA-BCL11A CD34+ cells cultured with and without GC was similar (very few differentially expressed genes), while GC-specific responses (differential expression of GILZ and of numerous additional genes) were observed only in controls cells with unperturbed BCL11A expression. These data indicate that BCL11A is an important participant of certain aspects of the stress pathway sustained by GC.

4.
Cell Rep ; 31(8): 107676, 2020 05 26.
Article in English | MEDLINE | ID: mdl-32460018

ABSTRACT

The human genome encodes millions of regulatory elements, of which only a small fraction are active within a given cell type. Little is known about the global impact of chromatin remodelers on regulatory DNA landscapes and how this translates to gene expression. We use precision genome engineering to reawaken homozygously inactivated SMARCA4, a central ATPase of the human SWI/SNF chromatin remodeling complex, in lung adenocarcinoma cells. Here, we combine DNase I hypersensitivity, histone modification, and transcriptional profiling to show that SMARCA4 dramatically increases both the number and magnitude of accessible chromatin sites genome-wide, chiefly by unmasking sites of low regulatory factor occupancy. By contrast, transcriptional changes are concentrated within well-demarcated remodeling domains wherein expression of specific genes is gated by both distal element activation and promoter chromatin configuration. Our results provide a perspective on how global chromatin remodeling activity is translated to gene expression via regulatory DNA.


Subject(s)
Chromatin Assembly and Disassembly/genetics , DNA Helicases/metabolism , DNA/genetics , Gene Expression/genetics , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Humans
5.
Bioessays ; 41(8): e1900041, 2019 08.
Article in English | MEDLINE | ID: mdl-31245868

ABSTRACT

Transcriptional silencing may not necessarily depend on the continuous residence of a sequence-specific repressor at a control element and may act via a "hit and run" mechanism. Due to limitations in assays that detect transcription factor (TF) binding, such as chromatin immunoprecipitation followed by high-throughput sequencing (ChIP-seq), this phenomenon may be challenging to detect and therefore its prevalence may be underappreciated. To explore this possibility, erythroid gene promoters that are regulated directly by GATA1 in an inducible system are analyzed. It is found that many regulated genes are bound immediately after induction of GATA1 but the residency of GATA1 decreases over time, particularly at repressed genes. Furthermore, it is shown that the repressive mark H3K27me3 is seldom associated with bound repressors, whereas, in contrast, the active (H3K4me3) histone mark is overwhelmingly associated with TF binding. It is hypothesized that during cellular differentiation and development, certain genes are silenced by repressive TFs that subsequently vacate the region. Catching such repressor TFs in the act of silencing via assays such as ChIP-seq is thus a temporally challenging prospect. The use of inducible systems, epitope tags, and alternative techniques may provide opportunities for detecting elusive "hit and run" transcriptional silencing. Also see the video abstract here https://youtu.be/vgrsoP_HF3g.


Subject(s)
Gene Silencing , Repressor Proteins/metabolism , Transcription Factors/metabolism , Binding Sites , Chromatin Immunoprecipitation Sequencing , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , Genetic Loci , Histones/metabolism , Humans , Promoter Regions, Genetic , Protein Binding , Transcriptional Activation
6.
Biotechnol Rep (Amst) ; 20: e00285, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30364711

ABSTRACT

The ability of transcriptional regulators to drive lineage conversion of somatic cells offers great potential for the treatment of human disease. To explore the concept of switching on specific target genes in heterologous cells, we developed a model system to screen candidate factors for their ability to activate the archetypal megakaryocyte-specific chemokine platelet factor 4 (PF4) in fibroblasts. We found that co-expression of the transcriptional regulators GATA1 and FLI1 resulted in a significant increase in levels of PF4, which became magnified over time. This finding demonstrates that such combinations can be used to produce potentially beneficial chemokines in readily available heterologous cell types.

7.
Trends Genet ; 34(12): 927-940, 2018 12.
Article in English | MEDLINE | ID: mdl-30287096

ABSTRACT

Disorders in hemoglobin (hemoglobinopathies) were the first monogenic diseases to be characterized and remain among the most common and best understood genetic conditions. Moreover, the study of the ß-globin locus provides a textbook example of developmental gene regulation. The fetal γ-globin genes (HBG1/HBG2) are ordinarily silenced around birth, whereupon their expression is replaced by the adult ß-globin genes (HBB primarily and HBD). Over 50 years ago it was recognized that mutations that cause lifelong persistence of fetal γ-globin expression ameliorate the debilitating effects of mutations in ß-globin. Since then, research has focused on therapeutically reactivating the fetal γ-globin genes. Here, we summarize recent discoveries, focusing on the influence of genome editing technologies, including CRISPR-Cas9, and emerging gene therapy approaches.


Subject(s)
Genetic Therapy/trends , Hemoglobinopathies/genetics , beta-Globins/genetics , gamma-Globins/genetics , Adult , CRISPR-Cas Systems/genetics , Gene Editing/trends , Hemoglobinopathies/blood , Hemoglobinopathies/pathology , Humans , Mutation , beta-Globins/therapeutic use , gamma-Globins/therapeutic use
8.
Nat Genet ; 50(4): 498-503, 2018 04.
Article in English | MEDLINE | ID: mdl-29610478

ABSTRACT

ß-hemoglobinopathies such as sickle cell disease (SCD) and ß-thalassemia result from mutations in the adult HBB (ß-globin) gene. Reactivating the developmentally silenced fetal HBG1 and HBG2 (γ-globin) genes is a therapeutic goal for treating SCD and ß-thalassemia 1 . Some forms of hereditary persistence of fetal hemoglobin (HPFH), a rare benign condition in which individuals express the γ-globin gene throughout adulthood, are caused by point mutations in the γ-globin gene promoter at regions residing ~115 and 200 bp upstream of the transcription start site. We found that the major fetal globin gene repressors BCL11A and ZBTB7A (also known as LRF) directly bound to the sites at -115 and -200 bp, respectively. Furthermore, introduction of naturally occurring HPFH-associated mutations into erythroid cells by CRISPR-Cas9 disrupted repressor binding and raised γ-globin gene expression. These findings clarify how these HPFH-associated mutations operate and demonstrate that BCL11A and ZBTB7A are major direct repressors of the fetal globin gene.


Subject(s)
Carrier Proteins/metabolism , DNA-Binding Proteins/metabolism , Fetal Hemoglobin/genetics , Mutation , Nuclear Proteins/metabolism , Transcription Factors/metabolism , gamma-Globins/genetics , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/therapy , Base Sequence , Binding Sites/genetics , CRISPR-Cas Systems , Cell Line , Humans , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/metabolism , Transcription Initiation Site , beta-Thalassemia/genetics , beta-Thalassemia/therapy
10.
Nat Med ; 24(2): 176-185, 2018 02.
Article in English | MEDLINE | ID: mdl-29334376

ABSTRACT

Metastasis results from a complex set of traits acquired by tumor cells, distinct from those necessary for tumorigenesis. Here, we investigate the contribution of enhancer elements to the metastatic phenotype of osteosarcoma. Through epigenomic profiling, we identify substantial differences in enhancer activity between primary and metastatic human tumors and between near isogenic pairs of highly lung metastatic and nonmetastatic osteosarcoma cell lines. We term these regions metastatic variant enhancer loci (Met-VELs). Met-VELs drive coordinated waves of gene expression during metastatic colonization of the lung. Met-VELs cluster nonrandomly in the genome, indicating that activity of these enhancers and expression of their associated gene targets are positively selected. As evidence of this causal association, osteosarcoma lung metastasis is inhibited by global interruptions of Met-VEL-associated gene expression via pharmacologic BET inhibition, by knockdown of AP-1 transcription factors that occupy Met-VELs, and by knockdown or functional inhibition of individual genes activated by Met-VELs, such as that encoding coagulation factor III/tissue factor (F3). We further show that genetic deletion of a single Met-VEL at the F3 locus blocks metastatic cell outgrowth in the lung. These findings indicate that Met-VELs and the genes they regulate play a functional role in metastasis and may be suitable targets for antimetastatic therapies.


Subject(s)
Carcinogenesis/genetics , Enhancer Elements, Genetic/genetics , Lung Neoplasms/genetics , Osteosarcoma/genetics , Cell Line, Tumor , Epigenomics , Gene Expression Regulation, Neoplastic , Genome, Human/genetics , Humans , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Neoplasm Metastasis/genetics , Osteosarcoma/pathology , Proteins/antagonists & inhibitors , Proteins/genetics , Selection, Genetic , Thromboplastin/genetics , Transcription Factor AP-1/antagonists & inhibitors , Transcription Factor AP-1/genetics , Tumor Microenvironment/genetics
11.
Sci Rep ; 7(1): 3137, 2017 06 09.
Article in English | MEDLINE | ID: mdl-28600522

ABSTRACT

The Krüppel-like factor (KLF) family of transcription factors play critical roles in haematopoiesis. KLF1, the founding member of the family, has been implicated in the control of both erythropoiesis and megakaryopoiesis. Here we describe a novel system using an artificial dominant negative isoform of KLF1 to investigate the role of KLF1 in the erythroid/megakaryocytic switch in vivo. We developed murine cell lines stably overexpressing a GST-KLF1 DNA binding domain fusion protein (GST-KLF1 DBD), as well as lines expressing GST only as a control. Interestingly, overexpression of GST-KLF1 DBD led to an overall reduction in erythroid features and an increase in megakaryocytic features indicative of a reduced function of endogenous KLF1. We simultaneously compared in vivo DNA occupancy of both endogenous KLF1 and GST-KLF1 DBD by ChIP qPCR. Here we found that GST-KLF1 DBD physically displaces endogenous KLF1 at a number of loci, providing novel in vivo evidence of direct competition between DNA binding proteins. These results highlight the role of KLF1 in the erythroid/megakaryocyte switch and suggest that direct competition between transcription factors with similar consensus sequences is an important mechanism in transcriptional regulation.


Subject(s)
Erythrocytes/cytology , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Megakaryocytes/cytology , Animals , Binding Sites , Cell Line, Tumor , DNA/metabolism , Erythrocytes/metabolism , Kruppel-Like Transcription Factors/chemistry , Megakaryocytes/metabolism , Mice , Phenotype , Recombinant Proteins/metabolism
12.
Blood Adv ; 1(11): 685-692, 2017 Apr 25.
Article in English | MEDLINE | ID: mdl-29296711

ABSTRACT

Genes encoding the human ß-like hemoglobin proteins undergo a developmental switch from fetal γ-globin to adult ß-globin expression around the time of birth. ß-hemoglobinopathies, such as sickle-cell disease and ß-thalassemia, result from mutations affecting the adult ß-globin gene. The only treatment options currently available carry significant adverse effects. Analyses of heritable variations in fetal hemoglobin (HbF) levels have provided evidence that reactivation of the silenced fetal γ-globin genes in adult erythroid cells is a promising therapy. The γ-globin repressor BCL11A has become the major focus, with several studies investigating its regulation and function as a first step to inhibiting its expression or activity. However, a second repression mechanism was recently shown to be mediated by the transcription factor ZBTB7A/LRF, suggesting that understanding the regulation of ZBTB7A may also be useful. Here we show that Krüppel-like factor 1 (KLF1) directly drives expression of ZBTB7A in erythroid cells by binding to its proximal promoter. We have also uncovered an erythroid-specific regulation mechanism, leading to the upregulation of a novel ZBTB7A transcript in the erythroid compartment. The demonstration that ZBTB7A, like BCL11A, is a KLF1 target gene also fits with the observation that reduced KLF1 expression or activity is associated with HbF derepression.

13.
J Biol Chem ; 291(31): 16048-58, 2016 07 29.
Article in English | MEDLINE | ID: mdl-27226561

ABSTRACT

The Lgals3 gene encodes a multifunctional ß-galactoside-binding protein, galectin-3. Galectin-3 has been implicated in a broad range of biological processes from chemotaxis and inflammation to fibrosis and apoptosis. The role of galectin-3 as a modulator of inflammation has been studied intensively, and recent evidence suggests that it may serve as a protective factor in obesity and other metabolic disorders. Despite considerable interest in galectin-3, little is known about its physiological regulation at the transcriptional level. Here, using knockout mice, chromatin immunoprecipitations, and cellular and molecular analyses, we show that the zinc finger transcription factor Krüppel-like factor 3 (KLF3) directly represses galectin-3 transcription. We find that galectin-3 is broadly up-regulated in KLF3-deficient mouse tissues, that KLF3 occupies regulatory regions of the Lgals3 gene, and that KLF3 directly binds its cognate elements (CACCC boxes) in the galectin-3 promoter and represses its activation in cellular assays. We also provide mechanistic insights into the regulation of Lgals3, demonstrating that C-terminal binding protein (CtBP) is required to drive optimal KLF3-mediated silencing. These findings help to enhance our understanding of how expression of the inflammatory modulator galectin-3 is controlled, opening up avenues for potential therapeutic interventions in the future.


Subject(s)
Galectin 3/biosynthesis , Gene Silencing , Inflammation Mediators/metabolism , Kruppel-Like Transcription Factors/metabolism , Repressor Proteins/metabolism , Response Elements , Transcription, Genetic , Animals , Galectin 3/genetics , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Knockout , Repressor Proteins/genetics
14.
Science ; 351(6270): 285-9, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26816381

ABSTRACT

Genes encoding human ß-type globin undergo a developmental switch from embryonic to fetal to adult-type expression. Mutations in the adult form cause inherited hemoglobinopathies or globin disorders, including sickle cell disease and thalassemia. Some experimental results have suggested that these diseases could be treated by induction of fetal-type hemoglobin (HbF). However, the mechanisms that repress HbF in adults remain unclear. We found that the LRF/ZBTB7A transcription factor occupies fetal γ-globin genes and maintains the nucleosome density necessary for γ-globin gene silencing in adults, and that LRF confers its repressive activity through a NuRD repressor complex independent of the fetal globin repressor BCL11A. Our study may provide additional opportunities for therapeutic targeting in the treatment of hemoglobinopathies.


Subject(s)
Carrier Proteins/metabolism , DNA-Binding Proteins/metabolism , Fetal Hemoglobin/genetics , Gene Silencing , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , gamma-Globins/genetics , Anemia, Sickle Cell/genetics , Animals , Carrier Proteins/genetics , Cell Line , Chromatin/metabolism , DNA-Binding Proteins/genetics , Erythroblasts/cytology , Erythropoiesis/genetics , Humans , Mice , Mice, Knockout , Nuclear Proteins/genetics , Repressor Proteins/genetics , Sequence Deletion , Thalassemia/genetics , Transcription Factors/genetics
15.
Nucleic Acids Res ; 44(7): 3118-30, 2016 Apr 20.
Article in English | MEDLINE | ID: mdl-26673701

ABSTRACT

Transcription factors are often regarded as having two separable components: a DNA-binding domain (DBD) and a functional domain (FD), with the DBD thought to determine target gene recognition. While this holds true for DNA bindingin vitro, it appears thatin vivoFDs can also influence genomic targeting. We fused the FD from the well-characterized transcription factor Krüppel-like Factor 3 (KLF3) to an artificial zinc finger (AZF) protein originally designed to target the Vascular Endothelial Growth Factor-A (VEGF-A) gene promoter. We compared genome-wide occupancy of the KLF3FD-AZF fusion to that observed with AZF. AZF bound to theVEGF-Apromoter as predicted, but was also found to occupy approximately 25,000 other sites, a large number of which contained the expected AZF recognition sequence, GCTGGGGGC. Interestingly, addition of the KLF3 FD re-distributes the fusion protein to new sites, with total DNA occupancy detected at around 50,000 sites. A portion of these sites correspond to known KLF3-bound regions, while others contained sequences similar but not identical to the expected AZF recognition sequence. These results show that FDs can influence and may be useful in directing AZF DNA-binding proteins to specific targets and provide insights into how natural transcription factors operate.


Subject(s)
DNA-Binding Proteins/chemistry , Transcription Factors/chemistry , Zinc Fingers , Binding Sites , Chromatin/metabolism , DNA/chemistry , DNA/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Genome, Human , HEK293 Cells , Humans , Kruppel-Like Transcription Factors/chemistry , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Vascular Endothelial Growth Factor A/genetics
16.
Nat Commun ; 6: 7085, 2015 May 14.
Article in English | MEDLINE | ID: mdl-25971621

ABSTRACT

Genetic disorders resulting from defects in the adult globin genes are among the most common inherited diseases. Symptoms worsen from birth as fetal γ-globin expression is silenced. Genome editing could permit the introduction of beneficial single-nucleotide variants to ameliorate symptoms. Here, as proof of concept, we introduce the naturally occurring Hereditary Persistance of Fetal Haemoglobin (HPFH) -175T>C point mutation associated with elevated fetal γ-globin into erythroid cell lines. We show that this mutation increases fetal globin expression through de novo recruitment of the activator TAL1 to promote chromatin looping of distal enhancers to the modified γ-globin promoter.


Subject(s)
Fetal Hemoglobin/genetics , Genome , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/genetics , Binding Sites , Chromatin/genetics , Dimerization , Gene Silencing , Humans , K562 Cells , Mice , Mice, Transgenic , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Point Mutation , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , T-Cell Acute Lymphocytic Leukemia Protein 1
17.
Blood ; 126(1): 89-93, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-26019277

ABSTRACT

Elevated fetal hemoglobin (HbF) ameliorates the clinical severity of hemoglobinopathies such as ß-thalassemia and sickle cell anemia. Currently, the only curative approach for individuals under chronic transfusion/chelation support therapy is allogeneic stem cell transplantation. However, recent analyses of heritable variations in HbF levels have provided a new therapeutic target for HbF reactivation: the transcriptional repressor BCL11A. Erythroid-specific BCL11A abrogation is now actively being sought as a therapeutic avenue, but the specific impact of such disruption in humans remains to be determined. Although single nucleotide polymorphisms in BCL11A erythroid regulatory elements have been reported, coding mutations are scarcer. It is thus of great interest that patients have recently been described with microdeletions encompassing BCL11A. These patients display neurodevelopmental abnormalities, but whether they show increased HbF has not been reported. We have examined the hematological phenotype, HbF levels, and erythroid BCL11A expression in 3 such patients. Haploinsufficiency of BCL11A induces only partial developmental γ-globin silencing. Of greater interest is that a patient with a downstream deletion exhibits reduced BCL11A expression and increased HbF. Novel erythroid-specific regulatory elements in this region may be required for normal erythroid BCL11A expression, whereas loss of separate elements in the developing brain may explain the neurological phenotype.


Subject(s)
Carrier Proteins/genetics , Chromosome Deletion , Chromosomes, Human, Pair 2 , Fetal Hemoglobin/metabolism , Nervous System Diseases/genetics , Nuclear Proteins/genetics , Adolescent , Child , Female , Humans , Male , Nervous System Diseases/blood , Repressor Proteins , Up-Regulation
18.
J Biol Chem ; 290(13): 8591-605, 2015 Mar 27.
Article in English | MEDLINE | ID: mdl-25659434

ABSTRACT

Krüppel-like factor 3 (KLF3/BKLF), a member of the Krüppel-like factor (KLF) family of transcription factors, is a widely expressed transcriptional repressor with diverse biological roles. Although there is considerable understanding of the molecular mechanisms that allow KLF3 to silence the activity of its target genes, less is known about the signal transduction pathways and post-translational modifications that modulate KLF3 activity in response to physiological stimuli. We observed that KLF3 is modified in a range of different tissues and found that the serine/threonine kinase homeodomain-interacting protein kinase 2 (HIPK2) can both bind and phosphorylate KLF3. Mass spectrometry identified serine 249 as the primary phosphorylation site. Mutation of this site reduces the ability of KLF3 to bind DNA and repress transcription. Furthermore, we also determined that HIPK2 can phosphorylate the KLF3 co-repressor C-terminal binding protein 2 (CtBP2) at serine 428. Finally, we found that phosphorylation of KLF3 and CtBP2 by HIPK2 strengthens the interaction between these two factors and increases transcriptional repression by KLF3. Taken together, our results indicate that HIPK2 potentiates the activity of KLF3.


Subject(s)
Carrier Proteins/physiology , DNA-Binding Proteins/metabolism , Kruppel-Like Transcription Factors/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/physiology , Alcohol Oxidoreductases , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , Co-Repressor Proteins , DNA/chemistry , Electrophoretic Mobility Shift Assay , Kruppel-Like Transcription Factors/chemistry , Mice , Molecular Sequence Data , NIH 3T3 Cells , Phosphorylation , Protein Binding , Protein Processing, Post-Translational , Transcription, Genetic , Transcriptional Activation
19.
Elife ; 3: e02626, 2014 Oct 03.
Article in English | MEDLINE | ID: mdl-25279814

ABSTRACT

As exome sequencing gives way to genome sequencing, the need to interpret the function of regulatory DNA becomes increasingly important. To test whether evolutionary conservation of cis-regulatory modules (CRMs) gives insight into human gene regulation, we determined transcription factor (TF) binding locations of four liver-essential TFs in liver tissue from human, macaque, mouse, rat, and dog. Approximately, two thirds of the TF-bound regions fell into CRMs. Less than half of the human CRMs were found as a CRM in the orthologous region of a second species. Shared CRMs were associated with liver pathways and disease loci identified by genome-wide association studies. Recurrent rare human disease causing mutations at the promoters of several blood coagulation and lipid metabolism genes were also identified within CRMs shared in multiple species. This suggests that multi-species analyses of experimentally determined combinatorial TF binding will help identify genomic regions critical for tissue-specific gene control.


Subject(s)
Liver/metabolism , Mammals/metabolism , Signal Transduction , Transcription Factors/metabolism , Animals , Blood Coagulation/genetics , Chromatin Immunoprecipitation , Gene Regulatory Networks , Genome-Wide Association Study , Genomics , Humans , Lipid Metabolism/genetics , Male , Molecular Sequence Annotation , Organ Specificity , Phylogeny , Polymorphism, Single Nucleotide/genetics , Protein Binding , Regulatory Sequences, Nucleic Acid/genetics , Species Specificity
20.
Genome Biol ; 15(4): R58, 2014 Apr 30.
Article in English | MEDLINE | ID: mdl-24946810

ABSTRACT

BACKGROUND: Retroviral elements are pervasively transcribed and dynamically regulated during development. While multiple histone- and DNA-modifying enzymes have broadly been associated with their global silencing, little is known about how the many diverse retroviral families are each selectively recognized. RESULTS: Here we show that the zinc finger protein Krüppel-like Factor 3 (KLF3) specifically silences transcription from the ORR1A0 long terminal repeat in murine fetal and adult erythroid cells. In the absence of KLF3, we detect widespread transcription from ORR1A0 elements driven by the master erythroid regulator KLF1. In several instances these aberrant transcripts are spliced to downstream genic exons. One such chimeric transcript produces a novel, dominant negative isoform of PU.1 that can induce erythroid differentiation. CONCLUSIONS: We propose that KLF3 ensures the integrity of the murine erythroid transcriptome through the selective repression of a particular retroelement and is likely one of multiple sequence-specific factors that cooperate to achieve global silencing.


Subject(s)
Kruppel-Like Transcription Factors/metabolism , Retroelements/genetics , Terminal Repeat Sequences/genetics , Animals , Cell Line , Erythroid Cells/metabolism , Kruppel-Like Transcription Factors/genetics , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL
...