Pharmacological induction of fetal hemoglobin in sickle cell disease and [beta ]-thalassemia

2001 ◽  
Vol 38 (4) ◽  
pp. 367-373 ◽  
Author(s):  
George F. Atweh ◽  
Dimitris Loukopoulos
Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 488-488
Author(s):  
Karin E Wahlberg ◽  
Jie Jiang ◽  
Helen Rooks ◽  
Steve Best ◽  
Swee Lay Thein

Abstract A major ameliorating factor in sickle cell disease and beta-thalassemia is the inherent ability to produce fetal hemoglobin (HbF, a2g2), a highly heritable trait. We have previously mapped a major quantitative trait locus (QTL) controlling HbF levels to an array of single nucleotide polymorphisms (SNPs) within a 79 kb block between the gene for HBS1L (a G-protein/elongation factor) and the MYB proto-oncogene in Northern European Caucasians. These variants in chromosome 6q23.3, are referred to as HBS1L-MYB intergenic polymorphism (HMIP) and exist in three linkage disequilibrium (LD) blocks, HMIP-1, -2 and -3, of which HMIP-2 shows the strongest association. Association of HMIP with HbF has recently been replicated in two independent cohorts of patients with sickle cell disease, African-American and Brazilian, as well as in a cohort of Chinese patients with beta-thalassemia. Physiologically, MYB and HBS1L expression was simultaneously down-regulated in individuals with high HbF while over-expression of MYB inhibits gamma-globin expression. We also showed that HMIP influences erythrocyte, platelet and monocyte counts in humans. We hypothesised that the genetic variants in the MYB-HBS1L intergenic region are situated in a regulatory region and affect HbF levels by altering the expression of MYB and/or HBS1L. Initial DNase I hypersensitivity analysis of the HMIP-2 region in K562 cells identified a number of hypersensitive sites which encouraged further functional analysis of the region. To study activity and to map potential regulatory elements in the entire MYB-HBS1L region we used chromatin immunopreciptation on microarray (ChIP–chip) of acetylated histones H3 and 4, RNA polymerase II and GATA-1. ChIP was performed on chromatin from primary human erythroid precursor cells and enrichment for specific sequences was evaluated using a high resolution Nimblegen microarray covering a large part of chromosome 6q. Our ChIP-chip data show high levels of histone acetylation in the MYB-HBS1L intergenic region with a concentration in the HMIP-2 block, indicating that the region is transcriptionally active in erythroid precursors. Moreover, a number of potential cis-regulatory elements were identified in the same region as strong GATA-1 signals in coincidence with DNase I hypersensitive sites. These results suggest a regulatory region in the MYB-HBS1L intergenic region that could be important in erythroid development by controlling expression in the MYB locus.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1224-1224
Author(s):  
Hassana Fathallah ◽  
Ali Taher ◽  
Ali Bazarbachi ◽  
George F. Atweh

Abstract High levels of fetal hemoglobin (HbF) are known to ameliorate the pathophysiology of β-globin disorders. The objective of this study is twofold: the first is to evaluate the efficacy of hemin as an inducer of HbF in erythroid cells from patients with sickle cell disease (SCD) and β-thalassemia (β-thal); the second is to determine if the combination of butyrate and hemin can induce higher levels of expression of HbF than either agent alone. BFU-E derived cells from the peripheral blood of two patients with homozygous SCD, three patients with β-thal, one patient with sickle β-thalassemia (S/β-thal) and one normal individual (AA) were cultured in the absence (control) or presence of butyrate (B), hemin (H) or butyrate and hemin (B+H). As expected, the levels of γ-globin mRNA [expressed as % γ/(β+γ)] increased upon butyrate exposure in progenitor-derived erythroid cells from SS and S/β-thal patients, and to a lesser extent in patients with β-thal (P = 0.01). In contrast, butyrate did not increase γ-globin expression in BFU-E derived colonies from the AA individual. Moreover, hemin exposure increased the γ/(β+γ) ratio in all subjects (P = 0.02). These findings confirm that hemin can be an effective HbF inducing agent in SCD and β-thal. Although the mechanism of induction of HbF by hemin is not known, unlike butyrate, hemin is clearly not a direct inhibitor of histone deacetylases and is likely to induce HbF by a different mechanism of action. Thus, we investigated the effect of the combination of hemin and butyrate on γ-globin gene expression. Interestingly, the combination of butyrate and hemin resulted in additive increases in the γ/(β+γ) ratios in all patients compared to butyrate alone (P = 0.03) or hemin alone (P = 0.01) (Table I). Just as importantly, exposure to both drugs resulted in a decrease in the α/(β+γ) mRNA imbalance in β-thal, which is the predominant pathophysiological feature of this disorder. In conclusion, combination therapy consisting of butyrate and hemin, which are two agents with different mechanisms of action and different toxicity profiles, may provide a more effective way of inducing HbF in patients with SCD and β-thal. Table I mRNA SCD β-Thal S/β-Thal AA n 2 3 1 1 %γ/(β+γ) Control 36 42 26 7.1 B 45 50 41 6.9 H 55 55 52 15 B+H 60 61 59 13 α/(β+γ) Control 3.1 8.9 1.8 1.9 B 2.0 7.7 2.9 1.7 H 3.0 7.5 1.7 1.0 B+H 2.9 6.4 2.2 1.3


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3352-3352 ◽  
Author(s):  
Ling Lin ◽  
Adrian P. Rybak ◽  
Conrad Rinaldi ◽  
Jonathan Yen ◽  
Yanfang Fu ◽  
...  

Sickle cell disease (SCD) and Beta thalassemia are disorders of beta globin production and function that lead to severe anemia and significant disease complications across a multitude of organ systems. Autologous transplantation of hematopoietic stem cells engineered through the upregulation of fetal hemoglobin (HbF) or correction of the beta globin gene have the potential to reduce disease burden in patients with beta hemoglobinopathies. Base editing is a recently developed technology that enables precise modification of the genome without the introduction of double strand DNA breaks. Gamma globin gene promoters were comprehensively screened with cytosine and adenine base editors (ABE) for the identification of alterations that would derepress HbF. Three regions were identified that significantly upregulated HbF, and the most effective nucleotide residue conversions are supported by natural variation seen in patients with hereditary persistence of fetal hemoglobin (HPFH). ABEs have been developed that significantly increase the level of HbF following nucleotide conversion at key regulatory motifs within the HBG1 and HBG2 promoters. CD34+ hematopoietic stem and progenitor cells (HSPC) were purified at clinical scale and edited using a process designed to preserve self-renewal capacity. Editing at two independent sites with different ABEs reached 94 percent and resulted in up to 63 percent gamma globin by UPLC. The levels of HbF observed should afford protection to the majority of SCD and Beta thalassemia patients based on clinical observations of HPFH and non-interventional therapy that links higher HbF dosage with milder disease (Ngo et al, 2011 Brit J Hem; Musallam et al, 2012 Blood). Directly correcting the Glu6Val mutation of SCD has been a recent goal of genetic therapies designed for the SCD population. Current base editing technology cannot yet convert mutations like those that result from the A-T transversion in sickle beta globin; however, ABE variants have been designed to recognize and edit the opposite stranded adenine residue of valine. This results in the conversion of valine to alanine and the production of a naturally occurring variant known as Hb G-Makassar. Beta globin with alanine at this position does not contribute to polymer formation, and patients with Hb G-Makassar present with normal hematological parameters and red blood cell morphology. SCD patient fibroblasts edited with these ABE variants achieve up to 70 percent conversion of the target adenine. CD34 cells from healthy donors were then edited with a lead ABE variant, targeting a synonymous mutation in an adjacent proline that resides within the editing window and serves as a proxy for editing the SCD mutation. The average editing frequency was 40 percent. Donor myeloid chimerism documented at these levels in the allogeneic transplant setting exceeds the 20 percent that is required for reversing the sickle phenotype (Fitzhugh et al, 2017 Blood). These next generation editing approaches provide a promising new modality for treating patients with Beta thalassemia and SCD. Disclosures No relevant conflicts of interest to declare.


2020 ◽  
pp. 1-2
Author(s):  
Shriya Phadnis ◽  

Sickle cell disease is a common inherited, multisystem, monogenic disorder of red blood cells (erythrocytes) caused due to polymorphic changes in hemoglobin. The most commonly known condition; Sickle cell anemia wherein there aren’t enough healthy RBCs to carry adequate oxygen throughout the body is considered to be a common form if Sickle-cell disease. Other type of Sickle cell disease is Hemoglobin SC disease (HbSC) caused due to inheritance of beta s and beta c alleles. The third type of such disease is HbS thalassemia caused due to beta-thalassemia mutation in the beta-globin gene leading to Sickle hemoglobin (HbS). Polymerization of HbS due to presence of fetal hemoglobin in the erythrocytes that in turn reduces the concentration of HbS which becomes the prominent determinant to check the severity of the disease. Reduced concentrations of HbS also reduce hemolysis that prevents acute vaso- occlusion. This pain is caused as the irregular shaped RBCs and WBCs get entrapped in the small blood vessels causing vascular obstruction and tissue ischemia. HbS polymerization can also lead to hemolytic anemia which is a state where in rate of RBC destruction is faster than formation; such patients are likely to develop vasculopathy. During the process of hemolysis, hemoglobin is released into plasma that inhibits endothelial nitric oxide signaling causing endothelial cell dysfunction. Hemolysis is also associated with formation of erythrocyte microvesicles that acts as a activator of tissue factor. Malaria is considered to cause HbS. Sickle cell disease is found to be highest in the African continent mostly affecting the new borns. The cause of deaths is hug in Africa due to poor diagnostic facilities. Measures taken against H influenzae and S pneumoniae that is profoundly detected in African children with Sickle cell disease can help reduce the disease proximity. Implementation of early life screening can thereby be effective in this case.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 4848-4848
Author(s):  
Dalal H. Mulla-Ali ◽  
Kevin H.M. Kuo ◽  
Mathew Sermer ◽  
Anne Mcleod

Abstract Abstract 4848 Introduction: Patients with sickle cell disease (SCD) have worse maternal and fetal outcomes compared to the general population, and experience antepartum complications unique to SCD patients, including painful vasoocclusive crises (VOC), acute chest syndrome (ACS), stroke and symptomatic anemia. The relationship between pre-pregnancy SCD-specific complications and maternal/fetal outcomes and antepartum complications has not been explored. We hypothesize that increased rates of SCD-specific complications are associated with increased rates of antepartum SCD-specific complications and worsened maternal and fetal outcomes. We further hypothesize that elevation in fetal hemoglobin is associated with improved maternal/fetal outcomes. Materials and Methods: We conducted a retrospective review of patients with SCD (SS, SC, S/beta-thalassemia) whose pregnancies were managed at the Mount Sinai Hospital (MSH), a high risk obstetrics care institution in Ontario, Canada, between January 1st, 1999 and June 30th, 2009 based on the institution's electronic and paper-based medical records. Patients were jointly managed by a hematologist specialized in hemoglobinopathies and an obstetrician specialized in high risk obstetric care. We compared the pre-pregnancy and antepartum rates of SCD-specific complications (painful VOC, ACS, stroke, and on-demand transfusion requirements). Pre-pregnancy fetal hemoglobin level was analyzed according to the presence or absence of maternal/fetal complications (expressed as an aggregate of preterm delivery, placental insufficiency, low birth weight (<2500 g), the need of emergent Caesarian section, fetal anomalies and fetal death). The t-test was used to compare means of the two groups. Fisher's exact test was used to compare categorical frequency data. An alpha value of 0.05 was chosen as the level of significance. Results and Discussion: We identified 22 pregnancies in 22 patients with SCD, 4 patients had not delivered at the time of censor. Fourteen patients were HbSS, 7 were HbSC, 1 was HbS/beta-thalassemia. Mean maternal age was 31.1 years. Mean gestational age at delivery was 37 weeks (95% CI 36 to 38 weeks) and 5 (23%) were preterm (< 37 weeks). Eleven of the 18 deliveries (61%) were by Caesarian section and 7 were performed on an emergent basis (4 due to fetal distress and 3 due to failure to progress). Three (17%) were low birth weight (< 2500 g) and 2 (11%) were intrauterine growth restricted. Maternal and fetal outcomes and rates of antepartum complications were similar to the existing literature. There was no association between prior history of ACS and having an episode of ACS during pregnancy. A history of painful VOC was associated with having at least one episode of painful VOC during pregnancy (P = 0.0433). Pre-pregnancy history of on-demand red cell transfusion was also associated with the need of at least one unit of transfusion during pregnancy (P = 0.0048). However, the frequency and time of first painful VOC during pregnancy were not associated with worsened maternal/fetal outcome. There was no association between fetal hemoglobin level and antepartum rates of painful VOC (P = 0.4867), ACS (P = 0.3702), and maternal/fetal complications (P = 0.2489). The results suggest that patients with a history of painful VOC may be predisposed to having painful VOC during pregnancy. Similarly, patients with a history of on-demand transfusion may need transfusion during pregnancy. The present study is limited by small sample size and its single-centered and retrospective nature. Further observation studies with larger sample size are required to prospectively validate these results. Disclosures: Kuo: Novartis Canada: Research Funding.


2008 ◽  
Vol 105 (33) ◽  
pp. 11869-11874 ◽  
Author(s):  
G. Lettre ◽  
V. G. Sankaran ◽  
M. A. C. Bezerra ◽  
A. S. Araujo ◽  
M. Uda ◽  
...  

Hematology ◽  
2013 ◽  
Vol 2013 (1) ◽  
pp. 362-369 ◽  
Author(s):  
Deepa Manwani ◽  
Paul S. Frenette

Abstract Recurrent and unpredictable episodes of vaso-occlusion are the hallmark of sickle cell disease. Symptomatic management and prevention of these events using the fetal hemoglobin–reactivating agent hydroxyurea are currently the mainstay of treatment. Discoveries over the past 2 decades have highlighted the important contributions of various cellular and soluble participants in the vaso-occlusive cascade. The role of these elements and the opportunities for therapeutic intervention are summarized in this review.


2002 ◽  
Vol 2 ◽  
pp. 1706-1728 ◽  
Author(s):  
Martin H. Steinberg

High fetal hemoglobin (HbF) levels inhibit the polymerization of sickle hemoglobin (HbS) and reduce the complications of sickle cell disease. Pharmacologic agents that can reverse the switch from γ- to β-chain synthesis — γ-globin chains characterize HbF, and sickle β-globin chains are present in HbS — or selectively increase the proportion of adult erythroid precursors that maintain the ability to produce HbF are therapeutically useful. Hydroxyurea promotes HbF production by perturbing the maturation of erythroid precursors. This treatment increases the total hemoglobin concentration, reduces the vaso-occlusive complications of pain and acute chest syndrome, and attenuates mortality in adults. It is a promising beginning for pharmacologic therapy of sickle cell disease. Still, its effects are inconsistent, trials in infants and children are ongoing, and its ultimate value — and peril — when started early in life are still unknown.


Blood ◽  
1997 ◽  
Vol 90 (2) ◽  
pp. 891-892
Author(s):  
Rita Selby ◽  
Eric Nisbet-Brown ◽  
Raveen K. Basran ◽  
Lebe Chang ◽  
Nancy F. Olivieri

Sign in / Sign up

Export Citation Format

Share Document