Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Mol Ther ; 30(8): 2693-2708, 2022 08 03.
Article in English | MEDLINE | ID: mdl-35526095

ABSTRACT

A promising treatment for ß-hemoglobinopathies is the de-repression of γ-globin expression leading to increased fetal hemoglobin (HbF) by targeting BCL11A. Here, we aim to improve a lentivirus vector (LV) containing a single BCL11A shmiR (SS) to further increase γ-globin induction. We engineered a novel LV to express two shmiRs simultaneously targeting BCL11A and the γ-globin repressor ZNF410. Erythroid cells derived from human HSCs transduced with the double shmiR (DS) showed up to a 70% reduction of both BCL11A and ZNF410 proteins. There was a consistent and significant additional 10% increase in HbF compared to targeting BCL11A alone in erythroid cells. Erythrocytes differentiated from SCD HSCs transduced with the DS demonstrated significantly reduced in vitro sickling phenotype compared to the SS. Erythrocytes differentiated from transduced HSCs from ß-thalassemia major patients demonstrated improved globin chain balance by increased γ-globin with reduced microcytosis. Reconstitution of DS-transduced cells from Berkeley SCD mice was associated with a statistically larger reduction in peripheral blood hemolysis markers compared with the SS vector. Overall, these results indicate that the DS LV targeting BCL11A and ZNF410 can enhance HbF induction for treating ß-hemoglobinopathies and could be used as a model to simultaneously and efficiently target multiple gene products.


Subject(s)
Fetal Hemoglobin , Hemoglobinopathies , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , Hemoglobinopathies/genetics , Hemoglobinopathies/therapy , Humans , Lentivirus/genetics , Lentivirus/metabolism , Mice , Nuclear Proteins/genetics , Repressor Proteins/genetics , Transcription Factors/metabolism , gamma-Globins/genetics
2.
Blood Adv ; 5(9): 2339-2349, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33938942

ABSTRACT

The BCL11A gene encodes a transcriptional repressor with essential functions in multiple tissues during human development. Haploinsufficiency for BCL11A causes Dias-Logan syndrome (OMIM 617101), an intellectual developmental disorder with hereditary persistence of fetal hemoglobin (HPFH). Due to the severe phenotype, disease-causing variants in BCL11A occur de novo. We describe a patient with a de novo heterozygous variant, c.1453G>T, in the BCL11A gene, resulting in truncation of the BCL11A-XL protein (p.Glu485X). The truncated protein lacks the 3 C-terminal DNA-binding zinc fingers and the nuclear localization signal, rendering it inactive. The patient displayed high fetal hemoglobin (HbF) levels (12.1-18.7% of total hemoglobin), in contrast to the parents who had HbF levels of 0.3%. We used cultures of patient-derived erythroid progenitors to determine changes in gene expression and chromatin accessibility. In addition, we investigated DNA methylation of the promoters of the γ-globin genes HBG1 and HBG2. HUDEP1 and HUDEP2 cells were used as models for fetal and adult human erythropoiesis, respectively. Similar to HUDEP1 cells, the patient's cells displayed Assay for Transposase-Accessible Chromatin (ATAC) peaks at the HBG1/2 promoters and significant expression of HBG1/2 genes. In contrast, HBG1/2 promoter methylation and genome-wide gene expression profiling were consistent with normal adult erythropoiesis. We conclude that HPFH is the major erythroid phenotype of constitutive BCL11A haploinsufficiency. Given the essential functions of BCL11A in other hematopoietic lineages and the neuronal system, erythroid-specific targeting of the BCL11A gene has been proposed for reactivation of γ-globin expression in ß-hemoglobinopathy patients. Our data strongly support this approach.


Subject(s)
Haploinsufficiency , Nuclear Proteins , Adult , Carrier Proteins/genetics , Humans , Nuclear Proteins/genetics , Phenotype , Repressor Proteins/genetics
3.
Nat Genet ; 53(5): 719-728, 2021 05.
Article in English | MEDLINE | ID: mdl-33859416

ABSTRACT

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


Subject(s)
Fetal Hemoglobin/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Transcription Factors/metabolism , Adult , Animals , Base Sequence , CRISPR-Cas Systems/genetics , Cells, Cultured , Chromatin/metabolism , DNA/metabolism , Erythroid Cells/metabolism , Erythropoiesis , Gene Editing , Gene Expression Regulation , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Humans , Mice , Mutagenesis/genetics , Protein Binding , Reproducibility of Results
4.
Nat Genet ; 52(12): 1333-1345, 2020 12.
Article in English | MEDLINE | ID: mdl-33230299

ABSTRACT

Genome-wide association studies identify genomic variants associated with human traits and diseases. Most trait-associated variants are located within cell-type-specific enhancers, but the molecular mechanisms governing phenotypic variation are less well understood. Here, we show that many enhancer variants associated with red blood cell (RBC) traits map to enhancers that are co-bound by lineage-specific master transcription factors (MTFs) and signaling transcription factors (STFs) responsive to extracellular signals. The majority of enhancer variants reside on STF and not MTF motifs, perturbing DNA binding by various STFs (BMP/TGF-ß-directed SMADs or WNT-induced TCFs) and affecting target gene expression. Analyses of engineered human blood cells and expression quantitative trait loci verify that disrupted STF binding leads to altered gene expression. Our results propose that the majority of the RBC-trait-associated variants that reside on transcription-factor-binding sequences fall in STF target sequences, suggesting that the phenotypic variation of RBC traits could stem from altered responsiveness to extracellular stimuli.


Subject(s)
Erythrocytes/physiology , Gene Expression Regulation/genetics , Phenotype , Polymorphism, Single Nucleotide/genetics , Transcription Factors/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic/genetics , Erythrocytes/cytology , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Humans , Quantitative Trait Loci/genetics , Smad1 Protein/genetics , Smad1 Protein/metabolism , Transcription Factors/metabolism , Transcription, Genetic/genetics
5.
Nat Genet ; 51(7): 1149-1159, 2019 07.
Article in English | MEDLINE | ID: mdl-31253978

ABSTRACT

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


Subject(s)
Chromatin/genetics , Erythroid Cells/metabolism , Fetal Hemoglobin/metabolism , Gene Expression Regulation , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Mutagenesis , Animals , Chromatin/metabolism , Erythroid Cells/cytology , Fetal Hemoglobin/genetics , Humans , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Mice , Mice, Transgenic , Protein Interaction Domains and Motifs
6.
Cancer Cell ; 35(4): 664-676.e7, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30991026

ABSTRACT

Resistance to asparaginase, an antileukemic enzyme that depletes asparagine, is a common clinical problem. Using a genome-wide CRISPR/Cas9 screen, we found a synthetic lethal interaction between Wnt pathway activation and asparaginase in acute leukemias resistant to this enzyme. Wnt pathway activation induced asparaginase sensitivity in distinct treatment-resistant subtypes of acute leukemia, but not in normal hematopoietic progenitors. Sensitization to asparaginase was mediated by Wnt-dependent stabilization of proteins (Wnt/STOP), which inhibits glycogen synthase kinase 3 (GSK3)-dependent protein ubiquitination and proteasomal degradation, a catabolic source of asparagine. Inhibiting the alpha isoform of GSK3 phenocopied this effect, and pharmacologic GSK3α inhibition profoundly sensitized drug-resistant leukemias to asparaginase. Our findings provide a molecular rationale for activation of Wnt/STOP signaling to improve the therapeutic index of asparaginase.


Subject(s)
Antineoplastic Agents/pharmacology , Asparaginase/pharmacology , Drug Resistance, Neoplasm , Leukemia/drug therapy , Polyethylene Glycols/pharmacology , Synthetic Lethal Mutations , Wnt Signaling Pathway/genetics , Wnt3A Protein/genetics , Animals , Cell Death/drug effects , Dose-Response Relationship, Drug , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Jurkat Cells , Leukemia/genetics , Leukemia/metabolism , Leukemia/pathology , Male , Mice, Inbred NOD , Mice, Transgenic , Proteasome Endopeptidase Complex/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Stability , Proteolysis , THP-1 Cells , Ubiquitination , Wnt3A Protein/metabolism , Xenograft Model Antitumor Assays
7.
Methods Mol Biol ; 1698: 275-284, 2018.
Article in English | MEDLINE | ID: mdl-29076097

ABSTRACT

The recently established human umbilical cord blood-derived erythroid progenitor (HUDEP) cell lines have equipped red blood cell researchers with valuable in vitro models of erythroid development. Of the three established HUDEP cell lines, HUDEP-2 cells express predominantly adult ß-globin and most closely resemble adult erythroid cells. This chapter describes culture protocols for the maintenance and erythroid differentiation of HUDEP-2 cells. Methods to genetically manipulate HUDEP-2 cells using a CRISPR/Cas9 nuclease-based approach are also discussed.


Subject(s)
Cell Culture Techniques , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Fetal Blood/cytology , Genetic Engineering , Biomarkers , CRISPR-Cas Systems , Cell Differentiation , Cell Line , Cell Self Renewal , Flow Cytometry , Genetic Engineering/methods , Genetic Vectors/genetics , Hemoglobins/genetics , Histocytochemistry , Humans , Karyotype , Lentivirus/genetics , Transduction, Genetic
8.
Br J Haematol ; 180(5): 630-643, 2018 03.
Article in English | MEDLINE | ID: mdl-29193029

ABSTRACT

The major ß-haemoglobinopathies, sickle cell disease and ß-thalassaemia, represent the most common monogenic disorders worldwide and a steadily increasing global disease burden. Allogeneic haematopoietic stem cell transplantation, the only curative therapy, is only applied to a small minority of patients. Common clinical management strategies act mainly downstream of the root causes of disease. The observation that elevated fetal haemoglobin expression ameliorates these disorders has motivated longstanding investigations into the mechanisms of haemoglobin switching. Landmark studies over the last decade have led to the identification of two potent transcriptional repressors of γ-globin, BCL11A and ZBTB7A. These regulators act with additional trans-acting epigenetic repressive complexes, lineage-defining factors and developmental programs to silence fetal haemoglobin by working on cis-acting sequences at the globin gene loci. Rapidly advancing genetic technology is enabling researchers to probe deeply the interplay between the molecular players required for γ-globin (HBG1/HBG2) silencing. Gene therapies may enable permanent cures with autologous modified haematopoietic stem cells that generate persistent fetal haemoglobin expression. Ultimately rational small molecule pharmacotherapies to reactivate HbF could extend benefits widely to patients.


Subject(s)
Hemoglobinopathies/genetics , Hemoglobins/genetics , Animals , Carrier Proteins/genetics , DNA-Binding Proteins/genetics , Disease Models, Animal , Epigenesis, Genetic/genetics , Fetal Globulins/genetics , Genes, Switch/genetics , Globins/genetics , Humans , Kruppel-Like Transcription Factors/genetics , Mice , Nuclear Proteins/genetics , Oncogene Proteins v-myb/genetics , Repressor Proteins , Transcription Factors/genetics
9.
PLoS One ; 11(2): e0146802, 2016.
Article in English | MEDLINE | ID: mdl-26840243

ABSTRACT

In human adult erythroid cells, lower than normal levels of Krüppel-like transcription factor 1 (KLF1) are generally associated with decreased adult ß- and increased fetal γ-globin gene expression. KLF1 also regulates BCL11A, a known repressor of adult γ-globin expression. In seeming contrast to the findings in adult cells, lower amounts of KLF1 correlate with both reduced embryonic and reduced fetal ß-like globin mRNA in mouse embryonic erythroid cells. The role of KLF1 in primary human fetal erythroid cells, which express both γ- and ß-globin mRNA, is less well understood. Therefore, we studied the role of KLF1 in ex vivo differentiated CD34+ umbilical cord blood cells (UCB erythroblasts), representing the fetal milieu. In UCB erythroblasts, KLF1 binds to the ß-globin locus control region (LCR), and the ß-globin promoter. There is very little KLF1 binding detectable at the γ-globin promoter. Correspondingly, when cultured fetal UCB erythroblasts are subjected to lentiviral KLF1 knockdown, the active histone mark H3K4me3 and RNA pol II recruitment are diminished at the ß- but not the γ-globin gene. The amount of KLF1 expression strongly positively correlates with ß-globin mRNA and weakly positively correlates with BCL11A mRNA. With modest KLF1 knockdown, mimicking haploinsufficiency, γ-globin mRNA is increased in UCB erythroblasts, as is common in adult cells. However, a threshold level of KLF1 is evidently required, or there is no absolute increase in γ-globin mRNA in UCB erythroblasts. Therefore, the role of KLF1 in γ-globin regulation in fetal erythroblasts is complex, with both positive and negative facets. Furthermore, in UCB erythroblasts, diminished BCL11A is not sufficient to induce γ-globin in the absence of KLF1. These findings have implications for the manipulation of BCL11A and/or KLF1 to induce γ-globin for therapy of the ß-hemoglobinopathies.


Subject(s)
Erythroblasts/metabolism , Gene Expression Regulation, Developmental , Kruppel-Like Transcription Factors/physiology , beta-Globins/genetics , gamma-Globins/genetics , Carrier Proteins/physiology , Fetal Blood , Gene Knockdown Techniques , Humans , Immunoglobulin Class Switching/genetics , Nuclear Proteins/physiology , Repressor Proteins
10.
Nature ; 527(7577): 192-7, 2015 Nov 12.
Article in English | MEDLINE | ID: mdl-26375006

ABSTRACT

Enhancers, critical determinants of cellular identity, are commonly recognized by correlative chromatin marks and gain-of-function potential, although only loss-of-function studies can demonstrate their requirement in the native genomic context. Previously, we identified an erythroid enhancer of human BCL11A, subject to common genetic variation associated with the fetal haemoglobin level, the mouse orthologue of which is necessary for erythroid BCL11A expression. Here we develop pooled clustered regularly interspaced palindromic repeat (CRISPR)-Cas9 guide RNA libraries to perform in situ saturating mutagenesis of the human and mouse enhancers. This approach reveals critical minimal features and discrete vulnerabilities of these enhancers. Despite conserved function of the composite enhancers, their architecture diverges. The crucial human sequences appear to be primate-specific. Through editing of primary human progenitors and mouse transgenesis, we validate the BCL11A erythroid enhancer as a target for fetal haemoglobin reinduction. The detailed enhancer map will inform therapeutic genome editing, and the screening approach described here is generally applicable to functional interrogation of non-coding genomic elements.


Subject(s)
CRISPR-Associated Proteins/metabolism , Carrier Proteins/genetics , Enhancer Elements, Genetic/genetics , Genetic Engineering , Mutagenesis/genetics , Nuclear Proteins/genetics , Animals , Base Sequence , CRISPR-Cas Systems/genetics , Cells, Cultured , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , DNA-Binding Proteins , Erythroblasts/metabolism , Fetal Hemoglobin/genetics , Genome/genetics , Humans , Mice , Molecular Sequence Data , Organ Specificity , RNA, Guide, Kinetoplastida/genetics , Repressor Proteins , Reproducibility of Results , Species Specificity
11.
Haematologica ; 99(10): 1565-73, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25150253

ABSTRACT

The Krüppel-like transcription factors KLF1 and KLF2 are essential for embryonic erythropoiesis. They can partially compensate for each other during mouse development, and coordinately regulate numerous erythroid genes, including the ß-like globins. Simultaneous ablation of KLF1 and KLF2 results in earlier embryonic lethality and severe anemia. In this study, we determine that this anemia is caused by a paucity of blood cells, and exacerbated by diminished ß-like globin gene expression. The anemia phenotype is dose-dependent, and, interestingly, can be ameliorated by a single copy of the KLF2, but not the KLF1 gene. The roles of KLF1 and KLF2 in maintaining normal peripheral blood cell numbers and globin mRNA amounts are erythroid cell-specific. Mechanistic studies led to the discovery that KLF2 has an essential function in erythroid precursor maintenance. KLF1 can partially compensate for KLF2 in this role, but is uniquely crucial for erythroid precursor proliferation through its regulation of G1- to S-phase cell cycle transition. A more drastic impairment of primitive erythroid colony formation from embryonic progenitor cells occurs with simultaneous loss of KLF1 and KLF2 than with loss of a single factor. KLF1 and KLF2 coordinately regulate several proliferation-associated genes, including Foxm1. Differential expression of FoxM1, in particular, correlates with the observed KLF1 and KLF2 gene dosage effects on anemia. Furthermore, KLF1 binds to the FoxM1 gene promoter in blood cells. Thus KLF1 and KLF2 coordinately regulate embryonic erythroid precursor maturation through the regulation of multiple homeostasis-associated genes, and KLF2 has a novel and essential role in this process.


Subject(s)
Cell Differentiation/genetics , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Erythropoiesis/genetics , Kruppel-Like Transcription Factors/genetics , Anemia/genetics , Anemia/metabolism , Animals , Cell Cycle/genetics , Embryo, Mammalian , Gene Expression Regulation, Developmental , Genotype , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Knockout , Phenotype
12.
J Biol Chem ; 286(28): 24819-27, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21610079

ABSTRACT

Krüppel-like factors (KLFs) control cell differentiation and embryonic development. KLF1 (erythroid Krüppel-like factor) plays essential roles in embryonic and adult erythropoiesis. KLF2 is a positive regulator of the mouse and human embryonic ß-globin genes. KLF1 and KLF2 have highly homologous zinc finger DNA-binding domains. They have overlapping roles in embryonic erythropoiesis, as demonstrated using single and double KO mouse models. Ablation of the KLF1 or KLF2 gene causes embryonic lethality, but double KO embryos are more anemic and die sooner than either single KO. In this work, a dual human ß-globin locus transgenic and KLF knockout mouse model was used. The results demonstrate that the human ε- (embryonic) and γ-globin (fetal) genes are positively regulated by KLF1 and KLF2 in embryos. Conditional KO mouse experiments indicate that the effect of KLF2 on embryonic globin gene regulation is at least partly erythroid cell-autonomous. KLF1 and KLF2 bind directly to the promoters of the human ε- and γ-globin genes, the mouse embryonic Ey- and ßh1-globin genes, and also to the ß-globin locus control region, as demonstrated by ChIP assays with mouse embryonic blood cells. H3K9Ac and H3K4me3 marks indicate open chromatin and active transcription, respectively. These marks are diminished at the Ey-, ßh1-, ε- and γ-globin genes and locus control region in KLF1(-/-) embryos, correlating with reduced gene expression. Therefore, KLF1 and KLF2 positively regulate the embryonic and fetal ß-globin genes through direct promoter binding. KLF1 is required for normal histone modifications in the ß-globin locus in mouse embryos.


Subject(s)
Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental/physiology , Genetic Loci/physiology , Kruppel-Like Transcription Factors/metabolism , Promoter Regions, Genetic/physiology , beta-Globins/biosynthesis , Animals , Embryo, Mammalian/cytology , Erythropoiesis/physiology , Humans , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Knockout , Protein Binding , beta-Globins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...