Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Stem Cell Res ; 78: 103448, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38810502

ABSTRACT

Mutations in the eyes shut homolog (EYS) gene are one of the common causes of autosomal recessive retinitis pigmentosa (RP). The lack of suitable animal models hampers progress understanding of the disease mechanism and drug development. This study reported the reprogramming of CD34+ hematopoietic stem/progenitor cells from a patient with compound heterozygous EYS mutations (c.6416 G > A and c.7228 + 1 G > A) into the induced pluripotent stem cell line, MUi038-A, using non-integrating vectors. The MUi038-A demonstrates pluripotency, tri-lineage differentiation potential, and a normal karyotype, offering a valuable model for studying the mechanism of EYS-related RP and new therapeutic development.

2.
PLoS One ; 19(4): e0300958, 2024.
Article in English | MEDLINE | ID: mdl-38625890

ABSTRACT

Oxidative damage to erythroid cells plays a key role in the pathogenesis of thalassemia. The oxidative stress in thalassemia is potentiated by heme, nonheme iron, and free iron produced by the Fenton reaction, due to degradation of the unstable hemoglobin and iron overload. In addition, the levels of antioxidant enzymes and molecules are significantly decreased in erythrocytes in α- and ß-thalassemia. The control of oxidative stress in red blood cells (RBCs) is known to be mediated by microRNAs (miRNAs). In erythroid cells, microR-214 (miR-214) has been reported to respond to external oxidative stress. However, the molecular mechanisms underlying this phenomenon remain unclear, especially during thalassemic erythropoiesis. In the present study, to further understand how miR-214 aggravates oxidative stress in thalassemia erythroid cells, we investigated the molecular mechanism of miR-214 and its regulation of the oxidative status in thalassemia erythrocytes. We have reported a biphasic expression of miR-214 in ß- and α-thalassemia. In the present study the effect of miR-214 expression was investigated by using miR -inhibitor and -mimic transfection in erythroid cell lines induced by hemin. Our study showed a biphasic expression of miR-214 in ß- and α-thalassemia. Subsequently, we examined the effect of miR-214 on erythroid differentiation in thalassemia. Our study reveals the loss-of-function of miR-214 during translational activation of activating transcription factor 4 mRNA, leading to decreased reactive oxygen species levels and increased glutathione levels in thalassemia erythroid cell. Our results suggest that the expression of activating transcription factor 4 regulated by miR-214 is important for oxidative stress modulation in thalassemic erythroid cells. Our findings can help to better understand the molecular mechanism of miRNA and transcription factors in regulation of oxidative status in erythroid cells, particularly in thalassemia, and could be useful for managing and relieving severe anemia symptoms in patients in the future.


Subject(s)
MicroRNAs , alpha-Thalassemia , beta-Thalassemia , Humans , Activating Transcription Factor 4/metabolism , Oxidative Stress/genetics , Erythroid Cells/metabolism , beta-Thalassemia/pathology , MicroRNAs/metabolism , Iron
3.
Protein Expr Purif ; 210: 106313, 2023 10.
Article in English | MEDLINE | ID: mdl-37276914

ABSTRACT

Many therapeutic proteins are expressed in Escherichia coli bacteria for the low cost and high yield obtained. However, these gram-negative bacteria also generate undesirable endotoxin byproducts such as lipopolysaccharides (LPS). These endotoxins can induce a human immune response and cause severe inflammation. To mitigate this problem, we have employed the ClearColi BL21 (DE3) endotoxin-free cells as an expression host for Cas9 protein production. Cas9 is an endonuclease enzyme that plays a key role in the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) and CRISPR associated protein 9 (CRISPR/Cas9) genome editing technique. This technology is very promising for use in diagnostics as well as treatment of diseases, especially for genetic diseases such as thalassemia. The potential uses for this technology thus generate a considerable interest for Cas9 utilization as a therapeutic protein in clinical treatment. Therefore, special care in protein production should be a major concern. Accordingly, we expressed the Cas9 protein in endotoxin-free bacterial cells achieving 99% purity with activity comparable to commercially available Cas9. Our protocol therefore yields a cost-effective product suitable for invitro experiments with stem cells.


Subject(s)
CRISPR-Associated Protein 9 , CRISPR-Cas Systems , Humans , Endotoxins/genetics , Gene Editing/methods , Repressor Proteins
4.
PLoS One ; 18(3): e0281059, 2023.
Article in English | MEDLINE | ID: mdl-36888630

ABSTRACT

Imbalanced globin chain output contributes to thalassemia pathophysiology. Hence, induction of fetal hemoglobin in ß-thalassemia and other ß-hemoglobinopathies are of continuing interest for therapeutic approaches. Genome-wide association studies have identified three common genetic loci: namely ß-globin (HBB), an intergenic region between MYB and HBS1L, and BCL11A underlying quantitative fetal hemoglobin production. Here, we report that knockdown of HBS1L (all known variants) using shRNA in early erythroblast obtained from ß0-thalassemia/HbE patients triggers an upregulation of γ-globin mRNA 1.69 folds. There is modest perturbation of red cell differentiation assessed by flow cytometry and morphology studies. The levels of α- and ß-globin mRNAs are relatively unaltered. Knockdown of HBS1L also increases the percentage of fetal hemoglobin around 16.7 folds when compared to non-targeting shRNA. Targeting HBS1L is attractive because of the potent induction of fetal hemoglobin and the modest effect on cell differentiation.


Subject(s)
Thalassemia , beta-Thalassemia , Humans , beta-Thalassemia/genetics , Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , RNA, Small Interfering/genetics , Genome-Wide Association Study , Erythroid Cells/metabolism , Carrier Proteins/genetics , beta-Globins/genetics
5.
Stem Cell Res ; 65: 102964, 2022 12.
Article in English | MEDLINE | ID: mdl-36395688

ABSTRACT

Choroideremia (CHM) is a monogenic, X-linked inherited retinal disease caused by mutations in the CHM gene. CHM patients develop progressive loss of vision due to degeneration of cell layers in the retina. In this report, the human-induced pluripotent stem cell, MUi032-A, was generated from CD34+ hematopoietic stem/progenitor cells of a male CHM patient by co-electroporation of non-integration episomal vectors containing OCT4/shp53, Sox-2/KLF4, and L-MYC/LIN-28. The MUi032-A showed normal karyotype and a hemizygous c.715C > T mutation. They expressed pluripotency markers and differentiated into cells derived from three germ layers. This cell line may be useful for disease mechanisms and gene therapy studies.


Subject(s)
Adaptor Proteins, Signal Transducing , Choroideremia , Hemizygote , Induced Pluripotent Stem Cells , Humans , Male , Adaptor Proteins, Signal Transducing/genetics , Mutation/genetics , Choroideremia/genetics , Choroideremia/pathology , Cell Line
6.
Blood Adv ; 6(11): 3280-3285, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35240686

ABSTRACT

Human γ-globin is predominantly expressed in fetal liver erythroid cells during gestation from 2 nearly identical genes, HBG1 and HBG2, that are both perinatally silenced. Reactivation of these fetal genes in adult red blood cells can ameliorate many symptoms associated with the inherited ß-globinopathies, sickle cell disease, and Cooley anemia. Although promising genetic strategies to reactivate the γ-globin genes to treat these diseases have been explored, there are significant barriers to their effective implementation worldwide; alternatively, pharmacological induction of γ-globin synthesis could readily reach the majority of affected individuals. In this study, we generated a CRISPR knockout library that targeted all erythroid genes for which prospective or actual therapeutic compounds already exist. By probing this library for genes that repress fetal hemoglobin (HbF), we identified several novel, potentially druggable, γ-globin repressors, including VHL and PTEN. We demonstrate that deletion of VHL induces HbF through activation of the HIF1α pathway and that deletion of PTEN induces HbF through AKT pathway stimulation. Finally, we show that small-molecule inhibitors of PTEN and EZH induce HbF in both healthy and ß-thalassemic human primary erythroid cells.


Subject(s)
beta-Thalassemia , gamma-Globins , Adult , Erythroid Cells/metabolism , Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , Humans , Prospective Studies , beta-Thalassemia/genetics , beta-Thalassemia/therapy , gamma-Globins/genetics , gamma-Globins/metabolism
7.
Exp Ther Med ; 22(4): 1105, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34504559

ABSTRACT

The Krüppel-like factor (KLF) family dominates highly conserved three zinc finger DNA binding domains at the C-terminus and variable transactivation domains at the N-terminus. Humans possess 18 KLF genes that are differentially expressed in various tissues. Several KLFs recognize a specific CACCC DNA motif that is commonly found within hematopoietic-specific promoters. To investigate those KLFs that are involved in human hemoglobin (Hb) switching, the present study analyzed a previous microarray data set from fetal and adult erythroid cells and validated the mRNA expression levels of 18 KLFs by reverse transcription-quantitative PCR (RT-qPCR). KLF with a decreased expression level in the fetuses was selected for a functional study in human erythroid progenitor cells using lentiviral-based short hairpin RNA knockdown. The fetuses demonstrated a lower level of KLF4 mRNA expression when compared with the adults. Downregulation of KLF4 in erythroid progenitor cells from healthy individuals and individuals with ß0-thalassemia/HbE evidenced the increasing embryonic and fetal globin mRNA expression with neither significant cytotoxicity nor gene expression alteration of the examined globin regulators, KLF1, B-cell lymphoma/leukemia 11A and lymphoma/leukemia-related factor. These findings demonstrate that the downregulation of KLF4 is associated with increased embryonic and fetal globin gene expression in human erythroid progenitor cells. Moreover, identifying putative compounds or molecular approaches that effectively downregulate KLF4 and further induce embryonic globin expression may provide an alternative therapeutic strategy for α-globin substitution in severe α-thalassemia.

8.
Blood ; 138(18): 1691-1704, 2021 11 04.
Article in English | MEDLINE | ID: mdl-34324630

ABSTRACT

Histone H3 lysine 4 methylation (H3K4Me) is most often associated with chromatin activation, and removing H3K4 methyl groups has been shown to be coincident with gene repression. H3K4Me demethylase KDM1a/LSD1 is a therapeutic target for multiple diseases, including for the potential treatment of ß-globinopathies (sickle cell disease and ß-thalassemia), because it is a component of γ-globin repressor complexes, and LSD1 inactivation leads to robust induction of the fetal globin genes. The effects of LSD1 inhibition in definitive erythropoiesis are not well characterized, so we examined the consequences of conditional inactivation of Lsd1 in adult red blood cells using a new Gata1creERT2 bacterial artificial chromosome transgene. Erythroid-specific loss of Lsd1 activity in mice led to a block in erythroid progenitor differentiation and to the expansion of granulocyte-monocyte progenitor-like cells, converting hematopoietic differentiation potential from an erythroid fate to a myeloid fate. The analogous phenotype was also observed in human hematopoietic stem and progenitor cells, coincident with the induction of myeloid transcription factors (eg, PU.1 and CEBPα). Finally, blocking the activity of the transcription factor PU.1 or RUNX1 at the same time as LSD1 inhibition rescued myeloid lineage conversion to an erythroid phenotype. These data show that LSD1 promotes erythropoiesis by repressing myeloid cell fate in adult erythroid progenitors and that inhibition of the myeloid-differentiation pathway reverses the lineage switch induced by LSD1 inactivation.


Subject(s)
Erythroid Cells/cytology , Erythropoiesis , Histone Demethylases/metabolism , Myeloid Cells/cytology , Animals , Cell Line , Cells, Cultured , Erythroid Cells/metabolism , Gene Deletion , Histone Demethylases/genetics , Humans , Mice , Myeloid Cells/metabolism
9.
Hematol Rep ; 13(4): 9215, 2021 Nov 26.
Article in English | MEDLINE | ID: mdl-35003571

ABSTRACT

Induction of fetal hemoglobin (HbF) ameliorates the clinical severity of ß-thalassemias. Histone methyltransferase LSD1 enzyme removes methyl groups from the activating chromatin mark histone 3 lysine 4 at silenced genes, including the γ-globin genes. LSD1 inhibitor RN-1 induces HbF levels in cultured human erythroid cells. Here, the HbF-inducing activity of RN-1 was investigated in erythroid progenitor cells derived from ß0-thalassemia/ hemoglobin E (HbE) patients. The significant and reproducible increases in γ-globin transcript and HbF expression upon RN-1 treatment were demonstrated in erythroid cells with divergent HbF baseline levels, the average of HbF induction was 17.7±0.8%. RN-1 at low concentration did not affect viability and proliferation of erythroid cells, but decreases in cell number were observed in cells treated with RN-1 at high concentration. Delayed terminal erythroid differentiation was revealed in ß0-thalassemia/HbE erythroid cells treated with RN-1 as similar to other compounds that target LSD1 activity. Downregulation of repressors of γ- globin expression; NCOR1 and SOX6, was observed in RN-1 treatment. These findings provide proof of the concept that LSD1 epigenetic enzyme is a potential therapeutic target for ß0-thalassemia/HbE patients.

10.
Hum Gene Ther ; 32(9-10): 473-480, 2021 05.
Article in English | MEDLINE | ID: mdl-32977730

ABSTRACT

Restoration of correct splicing of ßIVS2-654-globin pre-mRNA was previously accomplished in erythroid cells from ß-thalassemia/HbE patients by an engineered U7 small nuclear RNA (snRNA) that carried a sequence targeted to the cryptic branch point and an exonic splicing enhancer, U7.BP+623 snRNA. In this study, this approach was tested in thalassemic mice carrying the ßIVS2-654 mutation. While correction of ßIVS2-654 pre-mRNA splicing was achieved in erythroid progenitors transduced with a lentiviral vector carrying the U7.BP+623 snRNA, a high level of truncated U7.BP+623 snRNA was also observed. The discrepancy of processing of the modified U7 snRNA in human and mouse constructs hamper the evaluation of pathologic improvement in mouse model.


Subject(s)
RNA Precursors , beta-Globins , Animals , Erythroid Precursor Cells/metabolism , Humans , Mice , RNA Precursors/genetics , RNA Precursors/metabolism , RNA Splicing/genetics , RNA, Small Nuclear/genetics , RNA, Small Nuclear/metabolism , beta-Globins/genetics
11.
Int J Lab Hematol ; 42(6): 727-733, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32706939

ABSTRACT

INTRODUCTION: Several DNA-based approaches including a reverse dot-blot hybridization (RDB) have been established for detection of ß-thalassemia genotypes to provide accurate genetic counseling and prenatal diagnosis for prevention and control of severe ß-thalassemia. However, one of major concerns of these techniques is a risk of misdiagnosis due to a lack of DNA controls. Here, we constructed positive DNA controls for ß-thalassemia genotyping in order to ensure that all steps in the analysis are performed properly. METHODS: Four recombinant ß-globin plasmids, including a normal sequence and three different mutant panels covering 10 common ß-thalassemia mutations in Asia, were constructed by a conventional cloning method followed by sequential rounds of site-directed mutagenesis. These positive DNA controls were further validated by RDB analysis. RESULTS: We demonstrated the applicability of established positive DNA controls for ß-thalassemia genotyping in terms of accuracy and reproducibility by RDB analysis. We further combined three mutant ß-globin plasmids into a single positive control, which showed positive signals for both normal and mutant probes of all tested mutations. Therefore, only two positive DNA controls, normal and combined mutant ß-globin plasmids, are required for detecting 10 common ß-thalassemia mutations by RDB, reducing the cost, time, and efforts in the routine diagnosis. CONCLUSION: The ß-globin DNA controls established here provide efficient alternatives to a conventional DNA source from peripheral blood, which is more difficult to obtain. They also provide a platform for future development of ß-globin plasmid controls with other mutations, which can also be suitable for other DNA-based approaches.


Subject(s)
Asian People/genetics , DNA Probes/genetics , Genotype , Mutation , beta-Globins/genetics , beta-Thalassemia/genetics , Female , Humans , Male , Nucleic Acid Hybridization , beta-Thalassemia/diagnosis
12.
Ann Hematol ; 99(9): 2027-2036, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32567028

ABSTRACT

Increased expression of fetal hemoglobin (HbF) improves the clinical severity of ß-thalassemia patients. EHMT1/2 histone methyltransferases are epigenetic modifying enzymes that are responsible for catalyzing addition of the repressive histone mark H3K9me2 at silenced genes, including the γ-globin genes. UNC0638, a chemical inhibitor of EHMT1/2, has been shown to induce HbF expression in human erythroid progenitor cell cultures. Here, we report the HbF-inducing activity of UNC0638 in erythroid progenitor cells from ß-thalassemia/HbE patients. UNC0638 treatment led to significant increases in γ-globin mRNA, HbF expression, and HbF-containing cells in the absence of significant cytotoxicity. Moreover, UNC0638 showed additive effects on HbF induction in combination with the immunomodulatory drug pomalidomide and the DNMT1 inhibitor decitabine. These studies provide a scientific proof of concept that a small molecule targeting EHMT1/2 epigenetic enzymes, used alone or in combination with pomalidomide or decitabine, is a potential therapeutic approach for HbF induction. Further development of structural analogs of UNC0638 with similar biological effects but improved pharmacokinetic properties may lead to promising therapies and possible clinical application for the treatment of ß-thalassemia.


Subject(s)
Erythroid Precursor Cells/metabolism , Fetal Hemoglobin/biosynthesis , Hemoglobin E/metabolism , Quinazolines/pharmacology , beta-Thalassemia/metabolism , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Erythroid Precursor Cells/drug effects , Fetal Hemoglobin/genetics , Gene Expression , Humans , beta-Thalassemia/genetics
14.
Sci Rep ; 9(1): 7672, 2019 05 21.
Article in English | MEDLINE | ID: mdl-31113996

ABSTRACT

A cytosine to thymine mutation at nucleotide 654 of human ß-globin intron 2 (ßIVS2-654) is one of the most common mutations causing ß-thalassaemia in Chinese and Southeast Asians. This mutation results in aberrant ß-globin pre-mRNA splicing and prevents synthesis of ß-globin protein. Splicing correction using synthetic splice-switching oligonucleotides (SSOs) has been shown to restore expression of the ß-globin protein, but to maintain therapeutically relevant levels of ß-globin it would require lifelong administration. Here, we demonstrate long-term splicing correction using U7 snRNA lentiviral vectors engineered to target several pre-mRNA splicing elements on the ßIVS2-654-globin pre-mRNA such as cryptic 3' splice site, aberrant 5' splice site, cryptic branch point and an exonic splicing enhancer. A double-target engineered U7 snRNAs targeted to the cryptic branch point and an exonic splicing enhancer, U7.BP + 623, was the most effective in a model cell line, HeLa IVS2-654. Moreover, the therapeutic potential of the vector was demonstrated in erythroid progenitor cells derived from ßIVS2-654-thalassaemia/HbE patients, which showed restoration of correctly spliced ß-globin mRNA and led to haemoglobin A synthesis, and consequently improved thalassaemic erythroid cell pathology. These results demonstrate proof of concept of using the engineered U7 snRNA lentiviral vector for treatment of ß-thalassaemia.


Subject(s)
RNA Splicing , RNA, Small Nuclear/genetics , RNAi Therapeutics/methods , beta-Globins/genetics , beta-Thalassemia/therapy , Animals , Cells, Cultured , Erythroid Precursor Cells/metabolism , Genetic Vectors/genetics , HeLa Cells , Hemoglobin E/genetics , Hemoglobin E/metabolism , Humans , Mice , RNA, Small Nuclear/metabolism , beta-Globins/metabolism , beta-Thalassemia/genetics
16.
Genes Dev ; 32(23-24): 1537-1549, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30463901

ABSTRACT

Human globin gene production transcriptionally "switches" from fetal to adult synthesis shortly after birth and is controlled by macromolecular complexes that enhance or suppress transcription by cis elements scattered throughout the locus. The DRED (direct repeat erythroid-definitive) repressor is recruited to the ε-globin and γ-globin promoters by the orphan nuclear receptors TR2 (NR2C1) and TR4 (NR2C2) to engender their silencing in adult erythroid cells. Here we found that nuclear receptor corepressor-1 (NCoR1) is a critical component of DRED that acts as a scaffold to unite the DNA-binding and epigenetic enzyme components (e.g., DNA methyltransferase 1 [DNMT1] and lysine-specific demethylase 1 [LSD1]) that elicit DRED function. We also describe a potent new regulator of γ-globin repression: The deubiquitinase BRCA1-associated protein-1 (BAP1) is a component of the repressor complex whose activity maintains NCoR1 at sites in the ß-globin locus, and BAP1 inhibition in erythroid cells massively induces γ-globin synthesis. These data provide new mechanistic insights through the discovery of novel epigenetic enzymes that mediate γ-globin gene repression.


Subject(s)
Gene Expression Regulation/genetics , Nuclear Receptor Co-Repressor 1/genetics , Nuclear Receptor Co-Repressor 1/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism , gamma-Globins/genetics , Binding Sites , Cell Line , Enzyme Activation/genetics , Epigenesis, Genetic/genetics , Erythroid Cells/metabolism , Gene Silencing , HEK293 Cells , Humans , K562 Cells , Nuclear Receptor Subfamily 2, Group C, Member 1/metabolism , Protein Domains , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/metabolism
17.
Biochem Biophys Res Commun ; 499(1): 86-92, 2018 04 30.
Article in English | MEDLINE | ID: mdl-29550480

ABSTRACT

Repair of a splicing defect of ß-globin pre-mRNA harboring hemoglobin E (HbE) mutation was successfully accomplished in erythroid cells from patients with ß-thalassemia/HbE disorder by a synthetic splice-switching oligonucleotide (SSO). However, its application is limited by short-term effectiveness and requirement of lifelong periodic administration of SSO, especially for chronic diseases like thalassemias. Here, we engineered lentiviral vectors that stably express U7 small nuclear RNA (U7 snRNA) carrying the splice-switching sequence of the SSO that restores correct splicing of ßE-globin pre-mRNA and achieves a long-term therapeutic effect. Using a two-step tiling approach, we systematically screened U7 snRNAs carrying splice-switching SSO sequences targeted to the cryptic 5' splice site created by HbE mutation. We tested this approach and identified the most responsive element for mediating splicing correction in engineered U7 snRNAs in HeLa-ßE cell model cell line. Remarkably, the U7 snRNA lentiviral vector (U7 ßE4+1) targeted to this region effectively restored the correctly-spliced ßE-globin mRNA for at least 5 months. Moreover, the effects of the U7 ßE4+1 snRNA lentiviral vector were also evident as upregulation of the correctly-spliced ßE-globin mRNA in erythroid progenitor cells from ß-thalassemia/HbE patients treated with the vector, which led to improvements of pathologies in erythroid progenitor cells from thalassemia patients. These results suggest that the splicing correction of ßE-globin pre-mRNA by the engineered U7 snRNA lentiviral vector provides a promising, long-term treatment for ß-thalassemia/HbE.


Subject(s)
Erythroid Precursor Cells/metabolism , Genetic Engineering/methods , Genetic Therapy/methods , RNA Precursors/genetics , RNA Splicing , RNA, Small Nuclear/genetics , beta-Globins/genetics , Base Sequence , Erythroid Precursor Cells/pathology , Exons , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HeLa Cells , Hemoglobin E/genetics , Hemoglobin E/metabolism , Humans , Lentivirus/genetics , Lentivirus/metabolism , Mutation , Primary Cell Culture , RNA Precursors/metabolism , RNA Splice Sites , RNA, Small Nuclear/metabolism , beta-Globins/metabolism , beta-Thalassemia/genetics , beta-Thalassemia/metabolism , beta-Thalassemia/pathology , beta-Thalassemia/therapy
18.
Blood ; 130(23): 2537-2547, 2017 12 07.
Article in English | MEDLINE | ID: mdl-29018082

ABSTRACT

The orphan nuclear receptors TR4 (NR2C2) and TR2 (NR2C1) are the DNA-binding subunits of the macromolecular complex, direct repeat erythroid-definitive, which has been shown to repress ε- and γ-globin transcription during adult definitive erythropoiesis. Previous studies implied that TR2 and TR4 act largely in a redundant manner during erythroid differentiation; however, during the course of routine genetic studies, we observed multiple variably penetrant phenotypes in the Tr4 mutants, suggesting that indirect effects of the mutation might be masked by multiple modifying genes. To test this hypothesis, Tr4+/- mutant mice were bred into a congenic C57BL/6 background and their phenotypes were reexamined. Surprisingly, we found that homozygous Tr4 null mutant mice expired early during embryogenesis, around embryonic day 7.0, and well before erythropoiesis commences. We further found that Tr4+/- erythroid cells failed to fully differentiate and exhibited diminished proliferative capacity. Analysis of Tr4+/- mutant erythroid cells revealed that reduced TR4 abundance resulted in decreased expression of genes required for heme biosynthesis and erythroid differentiation (Alad and Alas2), but led to significantly increased expression of the proliferation inhibitory factor, cyclin dependent kinase inhibitor (Cdkn1c) These studies support a vital role for TR4 in promoting erythroid maturation and proliferation, and demonstrate that TR4 and TR2 execute distinct, individual functions during embryogenesis and erythroid differentiation.


Subject(s)
Cell Differentiation/genetics , Erythroid Cells/cytology , Erythroid Cells/metabolism , Nuclear Receptor Subfamily 2, Group C, Member 2/genetics , Anemia/blood , Anemia/genetics , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Proliferation/genetics , Erythropoiesis/genetics , Gene Expression Regulation, Developmental , Genes, Lethal , Heterozygote , Homozygote , Lymphopoiesis/genetics , Mice , Mice, Knockout , Mutation , Myelopoiesis/genetics , Nuclear Receptor Subfamily 2, Group C, Member 2/metabolism
19.
Stem Cells Transl Med ; 6(4): 1059-1069, 2017 04.
Article in English | MEDLINE | ID: mdl-28213976

ABSTRACT

The therapeutic use of patient-specific induced pluripotent stem cells (iPSCs) is emerging as a potential treatment of ß-thalassemia. Ideally, patient-specific iPSCs would be genetically corrected by various approaches to treat ß-thalassemia including lentiviral gene transfer, lentivirus-delivered shRNA, and gene editing. These corrected iPSCs would be subsequently differentiated into hematopoietic stem cells and transplanted back into the same patient. In this article, we present a proof of principle study for disease modeling and screening using iPSCs to test the potential use of the modified U7 small nuclear (sn) RNA to correct a splice defect in IVS2-654 ß-thalassemia. In this case, the aberration results from a mutation in the human ß-globin intron 2 causing an aberrant splicing of ß-globin pre-mRNA and preventing synthesis of functional ß-globin protein. The iPSCs (derived from mesenchymal stromal cells from a patient with IVS2-654 ß-thalassemia/hemoglobin (Hb) E) were transduced with a lentivirus carrying a modified U7 snRNA targeting an IVS2-654 ß-globin pre-mRNA in order to restore the correct splicing. Erythroblasts differentiated from the transduced iPSCs expressed high level of correctly spliced ß-globin mRNA suggesting that the modified U7 snRNA was expressed and mediated splicing correction of IVS2-654 ß-globin pre-mRNA in these cells. Moreover, a less active apoptosis cascade process was observed in the corrected cells at transcription level. This study demonstrated the potential use of a genetically modified U7 snRNA with patient-specific iPSCs for the partial restoration of the aberrant splicing process of ß-thalassemia. Stem Cells Translational Medicine 2017;6:1059-1069.


Subject(s)
Erythroid Cells/cytology , Erythroid Cells/metabolism , Gene Expression/genetics , Induced Pluripotent Stem Cells/cytology , RNA, Small Nuclear/genetics , beta-Globins/genetics , Animals , Humans , Induced Pluripotent Stem Cells/metabolism , RNA Splicing/genetics , RNA Splicing/physiology , Transcriptome/genetics , beta-Thalassemia/genetics , beta-Thalassemia/metabolism
20.
Blood ; 126(3): 386-96, 2015 Jul 16.
Article in English | MEDLINE | ID: mdl-26031919

ABSTRACT

Inhibition of lysine-specific demethylase 1 (LSD1) has been shown to induce fetal hemoglobin (HbF) levels in cultured human erythroid cells in vitro. Here we report the in vivo effects of LSD1 inactivation by a selective and more potent inhibitor, RN-1, in a sickle cell disease (SCD) mouse model. Compared with untreated animals, RN-1 administration leads to induced HbF synthesis and to increased frequencies of HbF-positive cells and mature erythrocytes, as well as fewer reticulocytes and sickle cells, in the peripheral blood of treated SCD mice. In keeping with these observations, histologic analyses of the liver and spleen of treated SCD mice verified that they do not exhibit the necrotic lesions that are usually associated with SCD. These data indicate that RN-1 can effectively induce HbF levels in red blood cells and reduce disease pathology in SCD mice, and may therefore offer new therapeutic possibilities for treating SCD.


Subject(s)
Anemia, Sickle Cell/prevention & control , Fetal Hemoglobin/biosynthesis , Histone Demethylases/antagonists & inhibitors , Rhodamines/pharmacology , Spiro Compounds/pharmacology , Splenomegaly/prevention & control , Thiophenes/pharmacology , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/pathology , Animals , Blotting, Western , Cells, Cultured , Disease Models, Animal , Fetal Hemoglobin/drug effects , Flow Cytometry , Humans , Immunoenzyme Techniques , Mice , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Splenomegaly/blood , Splenomegaly/pathology , beta-Globins/genetics , beta-Globins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...