Fetal expression of a human Aγ globin transgene rescues globin chain imbalance but not hemolysis in EKLF null mouse embryos

Blood ◽  
2000 ◽  
Vol 95 (5) ◽  
pp. 1827-1833 ◽  
Author(s):  
A. C. Perkins ◽  
K. R. Peterson ◽  
G. Stamatoyannopoulos ◽  
H. E. Witkowska ◽  
S. H. Orkin

Mice lacking the erythroid Kruppel-like factor (EKLF) die in utero at embryonic day 15 (E15) from severe anemia. EKLF−/− embryos display a marked deficit in β-globin gene expression. To test whether β-globin deficiency was solely responsible for the anemia and intrauterine death, we corrected the globin chain imbalance in EKLF−/− embryos by breeding with a strain of mice that express high levels of human γ-globin. Despite efficient production of hybrid m2-hγ2 hemoglobin in the fetal livers of EKLF−/− animals, hemolysis was not corrected and survival was not prolonged. We concluded that deficiency of nonglobin EKLF target genes is a major contributor to the definitive red blood cell abnormalities and prenatal death in EKLF−/−embryos. These results suggest that strategies designed to antagonize EKLF function in adults with hemoglobinopathy, in an attempt to reactivate γ-globin gene expression, may adversely affect other essential aspects of red blood cell physiology.

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2128-2128
Author(s):  
Hernan Sabio ◽  
Natalia Dixon ◽  
Ferdane Kutlar ◽  
Niren Patel ◽  
Hanfang Zhang ◽  
...  

Abstract Abstract 2128 Clinical phenotype in β-thalassemia syndromes is determined by the degree of chain imbalance. An increase in γ-globin production will compensate for the absent or deficient β-globin synthesis and will result in the amelioration of the chain imbalance, and hence an improvement in clinical features. The known genotypes of δβ-thalassemia are associated with an increase in Hb F production, which results in the amelioration of the clinical presentation. Most δβ-thalassemias result from deletions that remove the δ- and β-globin genes, (δβ)0 with a compensatory increase in γ-globin (Hb F) expression. We report an unusual case of homozygous δ0β+ thalassemia that provides interesting insights into increased γ-globin expression and the regulation of β-globin gene expression. An 8-year old boy of African ancestry presented with lifelong jaundice and pallor. He also experienced episodes of worsening symptoms. He exhibited frontal bossing, pale mucosa, scleral icterus, and moderate splenomegaly. He was known to have G6PD deficiency and was suspected of having additional erythrocyte pathology. The CBC revealed a Hb of 8.7, Hct 26.4, MCV 64.7, WBC 10,700, platelets 283,000, reticulocytes 2.2%, and total bilirubin 5.3. Hemoglobin analysis by HPLC and IEF revealed HbA 13.4%, Hb F 86.6%, and no additional components. Alpha thalassemia −3.7kb deletion was not detected. Globin chain analysis revealed α, β, Gγ and AγI chains. DNA analysis revealed a novel Senegalese-type deletion of the beta and delta genes, resulting in a delta0 beta+ thalassemia. The subject's parents who were both from the same small village in Niger had normal hematology values. Their hemoglobin analyses revealed Hb A 94. 8%, Hb A2 2.0%, Hb F 3.2% and Hb A 93.5%, Hb A2 2.1%, Hb F 4.5% in the father and mother, respectively. They were both heterozygous for the delta-beta deletion identified in their son. DNA analysis revealed a breakpoint in the delta gene at nucleotides 54755–54760 and a breakpoint in the beta gene at nucleotides 62153– 62158 [GenBank Ref ID: HUMHBB] with a 5 nucleotide “CAACA” bp region overlapping area. The subject, who is homozygous for the identified deletions, has a clinical phenotype of thalassemia intermedia. He has not yet required red cell transfusions. This is the first instance of a Senegalese-type deletion occurring in the homozygous state. The genotype provides insights into regulation of globin gene expression. While the ∼7 Kb deletion in the δβ-intergenic region may be responsible for the increased expression of the γ-globin gene similar to Hb Lepore deletions, the continued low level expression of the β-globin gene is most probably the result of the juxtaposition of the inefficient δ-globin promoter brought in the vicinity of the β-globin gene. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2261-2261
Author(s):  
Adlette Inati ◽  
Mario Kahale ◽  
Susan P Perrine ◽  
Ali T Taher ◽  
Suzanne Koussa ◽  
...  

Abstract Treatment of β-thalassemia is still largely dependent on supportive care, including blood transfusions and iron chelation. No oral drugs have been shown to consistently improve the severe anemia in thalassemia. Hematopoietic stem cell transplantation is potentially curative, but only a fraction of patients are eligible. Reactivation of fetal globin gene expression and increased γ-globin chain production is an attractive therapeutic approach, because it can compensate for defective β-globin chain production and the consequent α/β-chain imbalance that is a hallmark of β-thalassaemia. HQK-1001 (2,2-dimethylbutyrate, sodium salt), a new butyrate derivative with moderate potency but greater bioavailability, no cytotoxicity, and no HDAC-inhibitory activity compared to earlier butyrates, was shown to stimulate γ-globin production and erythropoiesis in animal models and in vitro. HQK-1001 reactivates γ-globin gene expression by inducing targeted dissociation of the inhibitory complex HDAC3 and its adaptor protein NCoR from the γ-globin gene promoter. HQK-1001 also enhances erythropoiesis by prolonging phosphorylation and activation of STAT-5, and enhancing Bcl-xL expression. In a multicenter dose-escalation study, HQK-1001 administered orally at 10, 20, 30 and 40 mg/kg once daily for 8 weeks in 21 subjects with non-transfusion-dependent β-thalassemia (NTDT) was well-tolerated and increased fetal hemoglobin (Hb F) in most subjects, with best results observed at 20 mg/kg. This single-center study (NCT01642758) evaluated HQK-1001 administered at 20 mg/kg/day for a longer duration than previously studied. Patients 18-50 years old with NTDT of Mediterranean phenotypes were eligible if their hemoglobin (Hb) was between 6.0 and 9.0 g/dL within the prior 30 days and had no transfusions within the prior 3 months. HQK-1001 was administered at 20 mg/kg once daily for 24 weeks. Folic acid was administered daily and oral iron supplementation was prescribed if serum ferritin was <700 ng/mL and stopped if ferritin exceeded 1000 ng/mL. Clinical and laboratory assessments were performed every 4 weeks. Ten subjects were enrolled, 7 male and 3 female, with a mean age of 29.4 years (range: 18-52 years). Eight subjects were splenectomized and 2 had palpable splenomegaly. Mean laboratory values at baseline were Hb F = 26.6% (range: 7.9-73.8%), Hb = 7.7 g/dL (range: 6.2-9.6 g/L) and reticulocytes = 10.9% (range: 7.1-15.7%). β-thalassemia mutations included homozygous IVS I-6 (C-T) in 7 of 10 subjects; and single subjects with homozygous cd29, homozygous IVS I-110 (G-A), and heterozygous IVSI-110 (G-A) / IVS II-I (G-A). One subject discontinued participation at Week 16 because of worsening anemia requiring a transfusion. Hb F increased in all subjects (range: 2.3-9.8%), with a mean increase of 4.8% (p < 0.0001). Total Hb increased in 7 subjects, with a mean increase of 0.6 g/dL (range: 0.1-1.5 g/dL) in those with increased Hb F. Adverse events were mild or moderate and reversible; fatigue, in 3 subjects, was the most common, followed by nausea, epigastric pain, dyspepsia, and fever, reported in 2 subjects each. There were mild and reversible increases in AST in 5 subjects and ALT in 4. Analyses of 3 major genetic modifier loci influencing baseline HbF levels identified favorable SNPs in the HMIP locus in 4 subjects, in BCL-11A in 3 subjects, and Xmn-I in one. No association was identified between specific modifiers and pharmacodynamic responses in this small group. In conclusion, HQK-1001 was well-tolerated and resulted in increased Hb F in all subjects and a modest increase in total Hb in 70% of subjects. The findings suggest that further HQK-1001 studies enrolling larger numbers of genetically characterized patients for longer periods appear warranted. Funding for this study was provided by a research grant from HemaQuest Pharmaceuticals and by the Georges N. Khoriaty Foundation. Disclosures: Inati: HemaQuest Pharmaceuticals: Research Funding. Perrine:Phoenicia BioSciences, Inc.: Employment, Equity Ownership, Membership on an entity’s Board of Directors or advisory committees, Research Funding; HemaQuest Pharmaceuticals: Equity Ownership. Ghalie:HemaQuest Pharmaceuticals: Employment.


Blood ◽  
2004 ◽  
Vol 103 (4) ◽  
pp. 1518-1520 ◽  
Author(s):  
David P. Steensma ◽  
Vip Viprakasit ◽  
Alex Hendrick ◽  
David K. Goff ◽  
Joanne Leach ◽  
...  

Abstract Rarely, myelodysplastic syndrome (MDS) is complicated by an acquired form of α-thalassemia (α-thalassemia in myelodysplastic syndrome [ATMDS]) characterized by hypochromic, microcytic, anisopoikilocytic red blood cells with hemoglobin H (HbH) inclusions. Acquired mutations in ATRX, a chromatin remodeling gene, have recently been found in 12 patients with typical features of ATMDS, though they have not been detected in MDS patients with similar red blood cell findings but little HbH. The α-globin genes themselves have appeared normal in all ATMDS patients studied to date. Here we characterize the molecular defect in a unique MDS patient with rare HbH inclusions in which an abnormal clone lost a greater than 1.9-Mb segment of the telomeric region of the short arm of one allele of chromosome 16, including both α-globin genes. Red blood cell changes associated with this acquired somatic genotype (––/αα) are surprisingly severe, demonstrating that a minor globin chain imbalance may be unexpectedly deleterious during the abnormal erythropoiesis that occurs in the context of MDS.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 186-186 ◽  
Author(s):  
Thais Arouca Fornari ◽  
Carolina Lanaro ◽  
Regiane Ferreira ◽  
Paula Barbim Donate ◽  
Geraldo Passos ◽  
...  

Abstract Introduction Hereditary Persistence of Fetal Hemoglobin (HPFH) and δβ-thalassemia are genetic disorders characterized by elevated levels of fetal hemoglobin (HbF) in adulthood. Deletions of variable sizes, and at different positions, that involve the β-globin gene complex on chromosome 11 are associated with these disorders. The distinction between these conditions is made based on clinical and hematological data. Deletional HPFH includes a wide range of conditions, but typically it is characterized in heterozygotes by levels of HbF of 15% to 30% with normal red blood cell indices, while heterozygotes for δβ-thalassemia tend to have elevated levels of HbF that are lower (5% to 20%) and accompanied by mild anemia with hypochromic, microcytic red blood cell indices. MicroRNAs (miRNAs) have been described to have a possible role in globin switching and can modulate transcriptional erythroid-specific regulators. To the best of our knowledge miRNAs have not been analyzed in HPFH and δβ-thalassemia. The aim of this study was to investigate the miRNAs expression profile and possible post-transcriptional role of these molecules in relation to the lack of normal suppression of γ genes in these genetic disorders. Methods CD34(+)-derived erythroid cells from two HPFH-2 individuals and two δβ-thalassaemia Sicilian type patients (DB) and healthy controls (CTRL) were cultured for 13 days to examine the expression profile of miRNAs. The miRNAs were hybridized using an Agilent miRNA microarray platform and the profiles were obtained through bioinformatics data analysis using GeneSpring software. qPCR analysis was used to validate the miRNA expression (TaqMan® miRNAs assays) and to quantify gene expression of 19 transcription factors. Different databases, such as miRBase, TargetScan, microRNA.org and BioGPS were used to determine the predicted targets of miRNAs data found. Results Six miRNAs were up-regulated in HPFH and in DB compared to CTRL: miR-146b-5p, miR-181a, miR-342-3p, miR-362-3p, miR-362-5p and miR-365. Five miRNAs were up-regulated in HPFH, compared to DB: miR-223, miR-630, miR-638, miR-1246 and miR-1290. Nine miRNAs were up-regulated in DB compared to HPFH: miR-10a, miR-21*, miR-33b*, miR-96, miR-128, miR-194, miR-210, miR-424 and miR-1275. The ALK4 and the GATA2 mRNAs were significantly up-regulated in HPFH, compared to DB. The BCL11A and the SH3BGRL2 mRNAs were significantly down-regulated in HPFH compared to DB. In silico analysis and the literature show that several miRNAs have targets related to HbF production or erythropoiesis. These include miR-10a, miR-33b*, miR-96, miR-128, miR-210, miR-223, miR-342-3p, miR-362-3p, miR-424 and miR-630. Conclusion The comparison of miRNA and transcription factor profiles suggests differences in the expressions of several miRNAs that may influence γ-globin gene expression. These data may contribute to understanding the phenotypic differences found between deletional HPFH and δβ-thalassemia. Financial support by FAPESP and CNPq/INCTS. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3479-3479
Author(s):  
Jianqiong Zhu ◽  
Hongzhen Li ◽  
Wulin Aerbajinai ◽  
Chutima Kumkhaek ◽  
Kyung Chin ◽  
...  

Abstract β-hemoglobinopathies are inherited disorders caused by mutations/deletions in the β-globin chain that lead to structurally defective β-globin chains or reduced (or absent) β-globin chain production. These diseases affect multiple organs and are associated with considerable morbidity and mortality, representing a major public health challenge. Current gene therapy approaches for the treatment of hemoglobinopathies involve viral transduction of hematopoietic stem cells with antisickling globin genes. Hemoglobin A2 (HA2, α2δ2), expressed at a low level due to the lack of Eklf binding motif in its promoter region, is fully functional and could be a valid anti-sickling agent in sickle cell disease, as well as a substitute of hemoglobin A in β-thalassemia. We had previously demonstrated that two Eklf-GATA1 fusion proteins could significantly activate δ-globin expression in human CD34+ cells. Here we report the effects of Eklf-GATA1 on hemoglobin expression and phenotypic correction using erythrocytes cultured from mouse hematopoietic progenitor cells with sickle cell disease. We found that enforced expression of Eklf-GATA1 fusion protein enhanced globin gene expression in the erythrocytesas compared with vector control. The long-form Eklf-GATA1 up-regulated β-globin gene expression 1.8-fold, δ-globin gene expression 3.3-fold, and γ-globin gene expression 1.7-fold. The medium-form EKLF-GATA1 up-regulated δ-globin gene expression 2.6-fold and γ-globin 1.3-fold, but had no significant effect on β-globin gene expression. HPLC revealed a percentage of HA2 was increased from 2.1 % in vector-transduced cells to 8.9% in medium-form Eklf-GATA-transduced-cells (p<0.01) and 6.3% in long-form Eklf-GATA-transduced-cells (p<0.01). Upon deoxygenation, the percentage of sickling erythrocyte was lower to 30.6% in medium-form Eklf-GATA-transduced cells as compared with 40.7% in vector-transduced-cells (p<0.05). Flow cytometry analyses of CD71/GPA and thiazole orange staining indicated that erythroid cell differentiation and enucleation were not affected by Eklf-GATA1. ChIP-sequencing analysis has demonstrated that Eklf-GATA1 fusion proteins and GATA1 having a similar protein-DNA binding pattern at a global level. Our results have found that the long form Eklf-GATA1 fusion protein has a major effect on δ-globin induction than β-globin; the medium form Eklf-GATA1 is able to elevate δ-globin expression without having an effect on β-globin expression. The above findings indicate that these fusion constructs could be a valuable genetic therapeutic tool for hemoglobinopathies, and warrant further preclinical study and evaluation. Disclosures No relevant conflicts of interest to declare.


Biomolecules ◽  
2021 ◽  
Vol 11 (5) ◽  
pp. 755
Author(s):  
Nur Atikah Zakaria ◽  
Md Asiful Islam ◽  
Wan Zaidah Abdullah ◽  
Rosnah Bahar ◽  
Abdul Aziz Mohamed Yusoff ◽  
...  

Thalassemia, an inherited quantitative globin disorder, consists of two types, α– and β–thalassemia. β–thalassemia is a heterogeneous disease that can be asymptomatic, mild, or even severe. Considerable research has focused on investigating its underlying etiology. These studies found that DNA hypomethylation in the β–globin gene cluster is significantly related to fetal hemoglobin (HbF) elevation. Histone modification reactivates γ-globin gene expression in adults and increases β–globin expression. Down-regulation of γ–globin suppressor genes, i.e., BCL11A, KLF1, HBG-XMN1, HBS1L-MYB, and SOX6, elevates the HbF level. β–thalassemia severity is predictable through FLT1, ARG2, NOS2A, and MAP3K5 gene expression. NOS2A and MAP3K5 may predict the β–thalassemia patient’s response to hydroxyurea, a HbF-inducing drug. The transcription factors NRF2 and BACH1 work with antioxidant enzymes, i.e., PRDX1, PRDX2, TRX1, and SOD1, to protect erythrocytes from oxidative damage, thus increasing their lifespan. A single β–thalassemia-causing mutation can result in different phenotypes, and these are predictable by IGSF4 and LARP2 methylation as well as long non-coding RNA expression levels. Finally, the coinheritance of β–thalassemia with α–thalassemia ameliorates the β–thalassemia clinical presentation. In conclusion, the management of β–thalassemia is currently limited to genetic and epigenetic approaches, and numerous factors should be further explored in the future.


1982 ◽  
Vol 8 (2) ◽  
pp. 163-178 ◽  
Author(s):  
Devi Vembu ◽  
Neal S. Young ◽  
Marcia Willing ◽  
Eve Church ◽  
Linda Sanders-Haigh ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document