Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters








Publication year range
1.
PLoS One ; 18(12): e0289860, 2023.
Article in English | MEDLINE | ID: mdl-38134183

ABSTRACT

Elevated levels of Fetal Hemoglobin interfere with polymerization of sickle hemoglobin thereby reducing anemia, lessening the severity of symptoms, and increasing life span of patients with sickle cell disease. An affordable, small molecule drug that stimulates HbF expression in vivo would be ideally suited to treat the large numbers of SCD patients that exist worldwide. Our previous work showed that administration of the LSD1 (KDM1A) inhibitor RN-1 to normal baboons increased Fetal Hemoglobin (HbF) and was tolerated over a prolonged treatment period. HbF elevations were associated with changes in epigenetic modifications that included increased levels of H3K4 di-and tri-methyl lysine at the γ-globin promoter. While dramatic effects of the loss of LSD1 on hematopoietic differentiation have been observed in murine LSD1 gene deletion and silencing models, the effect of pharmacological inhibition of LSD1 in vivo on hematopoietic differentiation is unknown. The goal of these experiments was to investigate the in vivo mechanism of action of the LSD1 inhibitor RN-1 by determining its effect on γ-globin expression in highly purified subpopulations of bone marrow erythroid cells enriched for varying stages of erythroid differentiation isolated directly from baboons treated with RN-1 and also by investigating the effect of RN1 on the global transcriptome in a highly purified population of proerythroblasts. Our results show that RN-1 administered to baboons targets an early event during erythroid differentiation responsible for γ-globin repression and increases the expression of a limited number of genes including genes involved in erythroid differentiation such as GATA2, GFi-1B, and LYN.


Subject(s)
Anemia, Sickle Cell , Histone Demethylases , Animals , Humans , Mice , Anemia, Sickle Cell/genetics , Fetal Hemoglobin/genetics , gamma-Globins/genetics , Gene Expression , Histone Demethylases/antagonists & inhibitors , Papio anubis/genetics
2.
Blood Adv ; 7(15): 3891-3902, 2023 08 08.
Article in English | MEDLINE | ID: mdl-36884303

ABSTRACT

Increased fetal hemoglobin (HbF) levels reduce the symptoms of sickle cell disease (SCD) and increase the lifespan of patients. Because curative strategies for bone marrow transplantation and gene therapy technologies remain unavailable to a large number of patients, the development of a safe and effective pharmacological therapy that increases HbF offers the greatest potential for disease intervention. Although hydroxyurea increases HbF, a substantial proportion of patients fail to demonstrate an adequate response. Pharmacological inhibitors of DNA methyltransferase (DNMT1) and lysine-specific demethylase 1A (LSD1), 2 epigenome-modifying enzymes associated with the multiprotein corepressor complex recruited to the repressed γ-globin gene, are powerful in vivo inducers of HbF. The hematological side effects of these inhibitors limit feasible clinical exposures. We evaluated whether administering these drugs in combination could reduce the dose and/or time of exposure to any single agent to minimize adverse effects, while achieving additive or synergistic increases in HbF. The DNMT1 inhibitor decitabine (0.5 mg/kg per day) and the LSD1 inhibitor RN-1 (0.25 mg/kg per day) administered in combination 2 days per week produced synergistic increases in F-cells, F-reticulocytes, and γ-globin messenger RNA in healthy baboons. Large increases in HbF and F-cells were observed in healthy, nonanemic, and anemic (phlebotomized) baboons. Combinatorial therapy targeting epigenome-modifying enzymes could thus be a useful strategy for producing larger increases in HbF to modify the clinical course of SCD.


Subject(s)
Anemia, Sickle Cell , gamma-Globins , Humans , Animals , Decitabine/pharmacology , Decitabine/therapeutic use , gamma-Globins/genetics , Epigenome , Fetal Hemoglobin/genetics , Anemia, Sickle Cell/genetics , Papio/genetics , Histone Demethylases/genetics , Histone Demethylases/therapeutic use
3.
Exp Hematol ; 67: 60-64.e2, 2018 11.
Article in English | MEDLINE | ID: mdl-30125603

ABSTRACT

Increased levels of fetal hemoglobin (HbF) lessen the severity of symptoms and increase the life span of patients with sickle cell disease (SCD). More effective strategies to increase HbF are needed because the current standard of care, hydroxyurea, is not effective in a significant proportion of patients. Treatment of the millions of patients projected worldwide would best be accomplished with an orally administered drug therapy that increased HbF. LSD1 is a component of corepressor complexes that repress γ-globin gene expression and are a therapeutic target for HbF reactivation. We have shown that subcutaneous administration of RN-1, a pharmacological LSD1 inhibitor, increased γ-globin expression in SCD mice and baboons, which are widely acknowledged as the best animal model in which to test the activity of HbF-inducing drugs. The objective of this investigation was to test the effect of oral administration of a new LSD1 inhibitor, ORY-3001. Oral administration of ORY-3001 to SCD mice (n = 3 groups) increased γ-globin expression, Fetal Hemoglobin (HbF)-containing (F) cells, and F reticulocytes (retics). In normal baboons (n = 7 experiments) treated with ORY-3001, increased F retics, γ-globin chain synthesis, and γ-globin mRNA were observed. Experiments in anemic baboons (n = 2) showed that ORY-3001 increased F retics (PA8695, predose = 24%, postdose = 66.8%; PA8698: predose = 13%, postdose = 93.6%), γ-globin chain synthesis (PA8695: predose = 0.07 γ/γ+ß, postdose = 0.20 γ/γ+ß; PA8698: predose = 0.02 γ/γ+ß, postdose = 0.44 γ/γ+ß), and γ-globin mRNA (PA8695: predose = 0.06 γ/γ+ß, postdose = 0.18 γ/γ+ß; PA8698: predose = 0.03 γ/γ+ß, postdose = 0.33 γ/γ+ß). We conclude that oral administration of ORY-3001 increases F retics, γ-globin chain synthesis, and γ-globin mRNA in baboons and SCD mice, supporting further efforts toward the development of this drug for SCD therapy.


Subject(s)
Anemia, Sickle Cell/drug therapy , Enzyme Inhibitors/therapeutic use , Fetal Hemoglobin/biosynthesis , Histone Demethylases/antagonists & inhibitors , gamma-Globins/biosynthesis , Administration, Oral , Anemia/blood , Anemia/drug therapy , Anemia, Sickle Cell/blood , Animals , Blood Cell Count , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Enzyme Inhibitors/administration & dosage , Female , Fetal Hemoglobin/genetics , Gene Expression Regulation/drug effects , Mice , Papio , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reticulocytes/metabolism , gamma-Globins/genetics
5.
Haematologica ; 101(6): 688-97, 2016 06.
Article in English | MEDLINE | ID: mdl-26858356

ABSTRACT

Increased fetal hemoglobin levels lessen the severity of symptoms and increase the lifespan of patients with sickle cell disease. Hydroxyurea, the only drug currently approved for the treatment of sickle cell disease, is not effective in a large proportion of patients and therefore new pharmacological agents that increase fetal hemoglobin levels have long been sought. Recent studies identifying LSD-1 as a repressor of γ-globin expression led to experiments demonstrating that the LSD-1 inhibitor RN-1 increased γ-globin expression in the sickle cell mouse model. Because the arrangement and developmental stage-specific expression pattern of the ß-like globin genes is highly conserved between man and baboon, the baboon model remains the best predictor of activity of fetal hemoglobin-inducing agents in man. In this report, we demonstrate that RN-1 increases γ-globin synthesis, fetal hemoglobin, and F cells to high levels in both anemic and non-anemic baboons with activity comparable to decitabine, the most potent fetal hemoglobin-inducing agent known. RN-1 not only restores high levels of fetal hemoglobin but causes the individual 5' Iγ- and 3' Vγ-globin chains to be synthesized in the ratio characteristic of fetal development. Increased fetal hemoglobin was associated with increased levels of acetylated Histone H3, H3K4Me2, H3K4Me3, and RNA polymerase II at the γ-globin gene, and diminished γ-globin promoter DNA methylation. RN-1 is likely to induce clinically relevant levels of fetal hemoglobin in patients with sickle cell disease, although careful titration of the dose may be required to minimize myelotoxicity.


Subject(s)
Enzyme Inhibitors/pharmacology , Fetal Hemoglobin/biosynthesis , Fetal Hemoglobin/genetics , Histone Demethylases/antagonists & inhibitors , Anemia/blood , Anemia/drug therapy , Anemia/etiology , Animals , Blood Cell Count , DNA Methylation/drug effects , Disease Models, Animal , Gene Expression Regulation/drug effects , Histones/metabolism , Papio , Reticulocytes/drug effects , Reticulocytes/metabolism , gamma-Globins/biosynthesis , gamma-Globins/genetics
6.
Epigenetics ; 10(5): 397-407, 2015.
Article in English | MEDLINE | ID: mdl-25932923

ABSTRACT

The mechanism responsible for developmental stage-specific regulation of γ-globin gene expression involves DNA methylation. Previous results have shown that the γ-globin promoter is nearly fully demethylated during fetal liver erythroid differentiation and partially demethylated during adult bone marrow erythroid differentiation. The hypothesis that 5-hydroxymethylcytosine (5 hmC), a known intermediate in DNA demethylation pathways, is involved in demethylation of the γ-globin gene promoter during erythroid differentiation was investigated by analyzing levels of 5-methylcytosine (5 mC) and 5 hmC at a CCGG site within the 5' γ-globin gene promoter region in FACS-purified cells from baboon bone marrow and fetal liver enriched for different stages of erythroid differentiation. Our results show that 5 mC and 5 hmC levels at the γ-globin promoter are dynamically modulated during erythroid differentiation with peak levels of 5 hmC preceding and/or coinciding with demethylation. The Tet2 and Tet3 dioxygenases that catalyze formation of 5 hmC are expressed during early stages of erythroid differentiation and Tet3 expression increases as differentiation proceeds. In baboon CD34+ bone marrow-derived erythroid progenitor cell cultures, γ-globin expression was positively correlated with 5 hmC and negatively correlated with 5 mC at the γ-globin promoter. Supplementation of culture media with Vitamin C, a cofactor of the Tet dioxygenases, reduced γ-globin promoter DNA methylation and increased γ-globin expression when added alone and in an additive manner in combination with either DNA methyltransferase or LSD1 inhibitors. These results strongly support the hypothesis that the Tet-mediated 5 hmC pathway is involved in developmental stage-specific regulation of γ-globin expression by mediating demethylation of the γ-globin promoter.


Subject(s)
Cell Differentiation/drug effects , Cytosine/analogs & derivatives , DNA Methylation/drug effects , Erythroid Cells/cytology , Promoter Regions, Genetic/drug effects , gamma-Globins/metabolism , 5-Methylcytosine/metabolism , Animals , Animals, Newborn , Antineoplastic Agents/pharmacology , Ascorbic Acid/pharmacology , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Bone Marrow Cells , Cells, Cultured , Cytosine/metabolism , Cytosine/pharmacology , Decitabine , Dioxygenases/metabolism , Erythroid Cells/drug effects , Erythroid Cells/metabolism , Histone Demethylases/metabolism , Humans , Hydroxyurea/pharmacology , Liver/cytology , Liver/drug effects , Papio anubis , Tranylcypromine/pharmacology
7.
Exp Hematol ; 43(7): 546-53.e1-3, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25931013

ABSTRACT

Increased levels of fetal hemoglobin are associated with decreased symptoms and increased lifespan in patients with sickle cell disease (SCD). Hydroxyurea, the only drug currently approved for SCD, is not effective in a large fraction of patients, and therefore, new agents are urgently needed. Recently it was found that lysine demethylase 1, an enzyme that removes monomethyl and dimethyl residues from the lysine 4 residue of histone H3, is a repressor of γ-globin gene expression. In this article, we have compared the ability of tranylcypromine (TCP) and a more potent TCP derivative, RN-1, to increase γ-globin expression in cultured baboon erythroid progenitor cells and in the SCD mouse model. The results indicate that the ability of RN-1 to induce F cells and γ-globin mRNA in SCD mice is similar to that of decitabine, the most powerful fetal hemoglobin-inducing drug known, and greater than that of either TCP or hydroxyurea. We conclude that RN-1 and other lysine demethylase 1 inhibitors may be promising new γ-globin-inducing agents for the treatment of SCD that warrant further studies in other preclinical models, such as nonhuman primates.


Subject(s)
Anemia, Sickle Cell/genetics , Fetal Hemoglobin/biosynthesis , Histone Demethylases/antagonists & inhibitors , Reticulocytes/drug effects , Tranylcypromine/pharmacology , gamma-Globins/biosynthesis , Anemia, Sickle Cell/drug therapy , Anemia, Sickle Cell/enzymology , Animals , Disease Models, Animal , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacology , Erythroid Precursor Cells/drug effects , Erythroid Precursor Cells/metabolism , Erythropoiesis/drug effects , Fetal Hemoglobin/genetics , Gene Expression Regulation/drug effects , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Histones/metabolism , Humans , Hydroxyurea/pharmacology , Methylation , Mice , Mice, Transgenic , Papio , Protein Processing, Post-Translational/drug effects , Transgenes/drug effects , Tranylcypromine/analogs & derivatives , Tretinoin/pharmacology , U937 Cells , gamma-Globins/genetics
8.
Blood ; 119(5): 1240-7, 2012 Feb 02.
Article in English | MEDLINE | ID: mdl-22160381

ABSTRACT

The deoxycytidine analog decitabine (DAC) can deplete DNA methyl-transferase 1 (DNMT1) and thereby modify cellular epigenetics, gene expression, and differentiation. However, a barrier to efficacious and accessible DNMT1-targeted therapy is cytidine deaminase, an enzyme highly expressed in the intestine and liver that rapidly metabolizes DAC into inactive uridine counterparts, severely limiting exposure time and oral bioavailability. In the present study, the effects of tetrahydrouridine (THU), a competitive inhibitor of cytidine deaminase, on the pharmacokinetics and pharmacodynamics of oral DAC were evaluated in mice and nonhuman primates. Oral administration of THU before oral DAC extended DAC absorption time and widened the concentration-time profile, increasing the exposure time for S-phase-specific depletion of DNMT1 without the high peak DAC levels that can cause DNA damage and cytotoxicity. THU also decreased interindividual variability in pharmacokinetics seen with DAC alone. One potential clinical application of DNMT1-targeted therapy is to increase fetal hemoglobin and treat hemoglobinopathy. Oral THU-DAC at a dose that would produce peak DAC concentrations of less than 0.2µM administered 2×/wk for 8 weeks to nonhuman primates was not myelotoxic, hypomethylated DNA in the γ-globin gene promoter, and produced large cumulative increases in fetal hemoglobin. Combining oral THU with oral DAC changes DAC pharmacology in a manner that may facilitate accessible noncytotoxic DNMT1-targeted therapy.


Subject(s)
Azacitidine/analogs & derivatives , Tetrahydrouridine/pharmacology , Administration, Oral , Animals , Antimetabolites/pharmacology , Antimetabolites, Antineoplastic/adverse effects , Antimetabolites, Antineoplastic/metabolism , Antimetabolites, Antineoplastic/pharmacokinetics , Area Under Curve , Azacitidine/administration & dosage , Azacitidine/adverse effects , Azacitidine/metabolism , Azacitidine/pharmacokinetics , Biological Availability , DNA Damage/drug effects , DNA Methylation/drug effects , Decitabine , Drug Interactions , Female , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Inactivation, Metabolic , Injections, Intravenous , Injections, Subcutaneous , Mice , Papio anubis
9.
Exp Hematol ; 39(1): 26-36.e1, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20974210

ABSTRACT

OBJECTIVE: These studies were performed to test the hypothesis that DNMT1 is required for maintenance of DNA methylation and repression of the γ-globin gene in adult-stage erythroid cells. MATERIALS AND METHODS: DNMT1 levels were reduced by nucleofection of small interfering RNA targeting DNMT1 in chemical inducer of dimerization-dependent multipotential mouse bone marrow cells containing the human ß-globin gene locus in the context of a yeast artificial chromosome and in primary cultures of erythroid progenitor cells derived from CD34(+) baboon bone marrow cells. The effect of reduced DNMT1 levels on globin gene expression was measured by real-time polymerase chain reaction and the effect on globin chain synthesis in primary erythroid progenitor cell cultures was determined by biosynthetic radiolabeling of globin chains followed by high-performance liquid chromatography analysis. The effect on DNA methylation was determined by bisulfite sequence analysis. RESULTS: Reduced DNMT1 levels in cells treated with siDNMT1 were associated with increased expression of γ-globin messenger RNA, an increased γ/γ+ß chain ratio in cultured erythroid progenitors, and decreased DNA methylation of the γ-globin promoter. Similar effects were observed in cells treated with decitabine, a pharmacological inhibitor of DNA methyltransferase inhibitor. CONCLUSIONS: DNMT1 is required to maintain DNA methylation of the γ-globin gene promoter and repress γ-globin gene expression in adult-stage erythroid cells.


Subject(s)
Bone Marrow Cells/metabolism , DNA (Cytosine-5-)-Methyltransferases/physiology , Erythroid Precursor Cells/metabolism , gamma-Globins/genetics , Animals , Antigens, CD34/immunology , Blotting, Western , Cells, Cultured , Chromosomes, Artificial, Yeast , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation , Dimerization , Erythroid Precursor Cells/immunology , Humans , Mice , Papio , RNA, Small Interfering/genetics , Transfection
10.
J Transl Med ; 8: 92, 2010 Oct 08.
Article in English | MEDLINE | ID: mdl-20932295

ABSTRACT

BACKGROUND: S110 is a novel dinucleoside analog that could have advantages over existing DNA methyltransferase (DNMT) inhibitors such as decitabine. A potential therapeutic role for S110 is to increase fetal hemoglobin (HbF) levels to treat ß-hemoglobinopathies. In these experiments the effect of S110 on HbF levels in baboons and its ability to reduce DNA methylation of the γ-globin gene promoter in vivo were evaluated. METHODS: The effect of S110 on HbF and γ-globin promoter DNA methylation was examined in cultured human erythroid progenitors and in vivo in the baboon pre-clinical model. S110 pharmacokinetics was also examined in the baboon model. RESULTS: S110 increased HbF and reduced DNA methylation of the γ-globin promoter in human erythroid progenitors and in baboons when administered subcutaneously. Pharmacokinetic analysis was consistent with rapid conversion of S110 into the deoxycytosine analog decitabine that binds and depletes DNA. CONCLUSION: S110 is rapidly converted into decitabine, hypomethylates DNA, and induces HbF in cultured human erythroid progenitors and the baboon pre-clinical model.


Subject(s)
Azacitidine/analogs & derivatives , Fetal Hemoglobin/metabolism , Oligonucleotides/pharmacology , Animals , Azacitidine/pharmacokinetics , Azacitidine/pharmacology , DNA Methylation , Fetal Hemoglobin/genetics , Oligonucleotides/pharmacokinetics , Papio , Promoter Regions, Genetic
11.
Exp Hematol ; 38(11): 989-993.e1, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20713129

ABSTRACT

OBJECTIVE: The mechanism responsible for increased fetal hemoglobin levels following decitabine treatment remains controversial. These experiments were performed to evaluate the role of transcriptional vs. translational mechanisms in the ability of decitabine to increase fetal hemoglobin levels in vivo. MATERIALS AND METHODS: Three normal, nonanemic baboons were treated with decitabine subcutaneously (0.5 mg/kg/d) for 10 days. The effect of decitabine on globin chain synthesis and globin messenger RNA levels was measured in pre- and posttreatment bone marrow aspirates by biosynthetic radiolabeling with [(3)H] leucine followed by separation of globin chains by high-performance liquid chromatography, and real-time polymerase chain reaction, respectively. The effect on DNA methylation of the ɛ- and γ-globin gene promoters was determined by bisulfite sequence analysis. RESULTS: Decitabine treatment of normal, nonanemic baboons induced similar increases in the γ/γ+ß chain synthetic ratio and the γ/total ß-like globin RNA ratio and also increased expression of ɛ-globin transcripts. Increased expression of ɛ- and γ-globin was associated with decreased DNA methylation of the ɛ- and γ-globin gene promoters. CONCLUSIONS: Decitabine increases fetal hemoglobin in vivo by transcriptional activation of the γ-globin gene.


Subject(s)
Azacitidine/analogs & derivatives , Fetal Hemoglobin/genetics , gamma-Globins/genetics , Animals , Azacitidine/administration & dosage , Azacitidine/pharmacology , Chromatography, High Pressure Liquid , DNA Methylation/drug effects , Decitabine , Fetal Hemoglobin/metabolism , Papio , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transcription, Genetic/drug effects , beta-Globins/genetics , beta-Globins/metabolism , epsilon-Globins/genetics , epsilon-Globins/metabolism , gamma-Globins/metabolism
12.
Exp Hematol ; 37(10): 1131-42, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19576949

ABSTRACT

OBJECTIVE: To investigate the mechanism(s) responsible for increased gamma-globin expression in vivo in decitabine-treated baboons and in vitro in cultured erythroid progenitor cells (EPC) from adult baboon bone marrow (BM). MATERIALS AND METHODS: Fetal liver, adult BM erythroid cells pre- and post-decitabine, and cultured EPCs were analyzed for distribution of RNA polymerase II, histone acetylation, and histone H3 (lys4) trimethyl throughout the gamma-globin gene complex by chromatin immunoprecipitation. DNA methylation of the gamma-globin promoter was determined by bisulfite sequencing. Expression of the baboon Igamma- and Vgamma-globin chains was determined by high performance liquid chromatography (HPLC). Expression of BCL11A, a recently identified repressor of gamma-globin expression, was analyzed by Western blot. RESULTS: Increased gamma-globin expression in decitabine-treated baboons and cultured EPC correlated with increased levels of RNA polymerase II, histone acetylation, and histone H3 (lys4) trimethyl associated with the gamma-globin gene consistent with a transcriptional activation mechanism. Cultured EPC expressed the Igamma- and Vgamma-globin chains in a pattern characteristic of fetal development. The level of DNA methylation of the gamma-globin gene promoter in EPC cultures was similar to BM erythroid cells from normal adult baboons. Different BCL11A isoforms were observed in BM erythroid cells and cultured EPC. CONCLUSION: The mechanism responsible for increased gamma-globin expression in cultured EPC was unexpectedly not associated with increased DNA hypomethylation of the gamma-globin gene promoter compared to normal BM erythroid cells, in contrast to BM erythroid cells of decitabine-treated baboons. Rather, increased fetal hemoglobin in EPC cultures was associated with a fetal Igamma/Vgamma chain ratio and a difference in the size of the BCL11A protein compared to normal BM erythroid cells.


Subject(s)
Azacitidine/analogs & derivatives , Erythroid Precursor Cells/metabolism , Papio anubis/genetics , Transcription, Genetic/drug effects , gamma-Globins/genetics , Age Factors , Animals , Azacitidine/pharmacology , Carrier Proteins/physiology , Cells, Cultured/metabolism , Chromatin Immunoprecipitation , DNA Methylation/drug effects , Decitabine , Fetal Blood/metabolism , Fetal Hemoglobin/biosynthesis , Fetal Hemoglobin/genetics , Gestational Age , Nuclear Proteins/physiology , Phlebotomy , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , Protein Isoforms/physiology , Protein Processing, Post-Translational , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , SUMO-1 Protein/metabolism , gamma-Globins/biosynthesis
13.
Am J Hematol ; 82(11): 981-5, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17696208

ABSTRACT

The silencing of tumor suppressor genes associated with increased DNA methylation of the promoter regions is a frequent observation in many forms of cancer. Reactivation of these genes using pharmacological inhibitors of DNA methyltransferase such as 5-aza-2'-deoxycytidine (decitabine) is a worthwhile therapeutic goal. The effectiveness and tolerability of low-dose intravenous and subcutaneous decitabine regimens to demethylate and reactivate expression of the methylated gamma-globin gene in baboons and in patients with sickle cell disease led to successful trials of low-dose regimens of this drug in patients with myelodysplastic syndrome. Since these low-dose regimens are well-tolerated with minimal toxicity, they are suitable for chronic dosing to maintain promoter hypomethylation and expression of target genes. The development of an orally administered therapy using DNA methyltransferase inhibitors would facilitate such chronic approaches to therapy. We tested the ability of decitabine and a new salt derivative, decitabine mesylate, to reactivate the methylated gamma-globin gene in baboons when administered orally. Our results demonstrate that oral administration of these drugs at doses 17-34 times optimal subcutaneous doses of decitabine reactivates fetal hemoglobin, demethylates the epsilon- and gamma-globin gene promoters, and increases histone acetylation of these promoters in baboons (Papio anubis).


Subject(s)
Azacitidine/analogs & derivatives , DNA Methylation/drug effects , DNA Modification Methylases/antagonists & inhibitors , Fetal Hemoglobin , Gene Silencing/drug effects , Administration, Oral , Animals , Azacitidine/administration & dosage , Azacitidine/pharmacokinetics , Decitabine , Fetal Hemoglobin/drug effects , Fetal Hemoglobin/genetics , Gene Expression Regulation/drug effects , Globins/drug effects , Globins/genetics , Globins/metabolism , Papio anubis
14.
Exp Hematol ; 35(1): 48-55, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17198873

ABSTRACT

OBJECTIVE: To determine whether the difference in gamma-globin gene promoter methylation in terminal erythroblasts at the fetal and adult stages of development is a result of fetal stage-specific demethylation or adult stage-specific de novo methylation during erythropoiesis. MATERIALS AND METHODS: Fetal liver- (FL, n = 2) and adult bone marrow- (ABM, n = 3) derived hematopoietic stem/progenitor cells and mature erythroblasts were purified by passage through a Miltenyi Magnetic Column followed by fluorescein-activated cell sorting (FACS) into subpopulations, defined by expression of CD34 and CD36 antigens. CD34(+)CD36(-), CD34(+)CD36(+), and CD34(-)CD36(+) subpopulations were purified by FACS and their degree of differentiation verified using the colony-forming cell assay. The methylation pattern of 5 CpG sites in the gamma-globin promoter region of these purified cell populations was determined using bisulfite sequencing. RESULTS: The gamma-globin promoter was highly methylated in the earliest stage of hematopoietic stem progenitor cells (CD34(+)CD36(-)) and methylation progressively decreased as erythroid differentiation progressed in FL and appears so in ABM as well. CONCLUSIONS: These data support a model in which differences in the methylation pattern of the gamma-globin gene in differentiating erythroblasts at different stages of development is the result of fetal stage-specific demethylation associated with transcriptional activation, rather than de novo methylation in the adults. The difference in the extent of gamma-globin gene demethylation in FL and ABM is correlated with the difference in gamma-globin expression at these developmental stages.


Subject(s)
Cell Differentiation/genetics , Cell Lineage/genetics , DNA Methylation , Erythroid Cells/cytology , Globins/genetics , Hematopoietic Stem Cells/cytology , Promoter Regions, Genetic/genetics , Animals , Bone Marrow Cells , Cells, Cultured , Erythroblasts , Erythropoiesis/genetics , Fetus/cytology , Liver/cytology , Papio
15.
Blood Cells Mol Dis ; 36(2): 269-78, 2006.
Article in English | MEDLINE | ID: mdl-16527500

ABSTRACT

The baboon is a suitable and relevant animal model to study the mechanism of human globin gene switching. This investigation addresses the role of DNA methylation and histone coding in globin gene switching in the baboon, Papio anubis. Bisulfite sequencing and chromatin immunoprecipitation studies were performed in erythroid cells purified from fetuses of varying gestational ages and from adult bone marrow to analyze the manner that changes in DNA methylation of the epsilon-, gamma-, and beta-globin promoters and association of ac-H3, ac-H4, H3-dimeK4, H3-dimeK36, and H3-dimeK79 with the epsilon-, gamma-, and beta-globin promoters occur during development. Changes in DNA methylation of the epsilon- and gamma-globin gene promoters during transitional stages of globin gene switching were consistent with the stochastic model of methylation and a role of DNA methylation in gene silencing. Enrichment of ac-H3, ac-H4, and pol II at the promoters of developmentally active genes was observed, while the pattern of distribution of H3-dimeK4 and H3-dimeK79 suggests that these modifications are found near both currently and formerly active promoters. Enrichment of H3-dimeK36 at the silenced epsilon-globin gene promoter was observed. These studies demonstrate that coordinated epigenetic modifications in the chromatin structure of the beta-like globin gene promoters accompany the highly regulated changes in expression patterns of these genes during development.


Subject(s)
Chromatin/chemistry , DNA Methylation , Globins/genetics , Histones , Promoter Regions, Genetic/genetics , Animals , Fetus , Gene Expression Regulation, Developmental/genetics , Papio anubis
16.
Exp Hematol ; 34(3): 339-47, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16543068

ABSTRACT

OBJECTIVE: Treatment with the DNA demethylating drug 5-aza-2'-deoxycytidine (Dacogen; DAC) increased fetal hemoglobin and F cells to therapeutically significant levels in patients with sickle cell disease. To gain more insight into the mechanism of action of this drug and to increase our understanding of the relationship between DNA methylation and chromatin structure, we have determined the effect of DAC on covalent histone modifications of chromatin associated with the epsilon, gamma-, and beta-globin promoters in purified bone marrow erythroid cells of four baboons (P. anubis) pre- and posttreatment. RESULTS: Fetal hemoglobin increased from 6.45%+/-1.75% in pretreatment samples to 62.1%+/-7.94% following DAC. DNA methylation of three CpG sites within the epsilon-globin promoter and 5 CpG sites within the gamma-globin promoter decreased more than 50% following DAC treatment. Levels of RNA polymerase II, acetyl-histone H3, acetyl-histone H4, dimethyl-histone H3 (lys4), dimethyl-histone H3 (lys36), and dimethyl-histone H3 (lys79) associated with the epsilon-, gamma-, and beta-globin promoters were determined by chromatin immunoprecipitation of formaldehyde-fixed chromatin followed by real-time PCR. Dacogen treatment increased the association of RNA polymerase II, acetyl-histone H3, and acetyl-histone H4 with the gamma-globin promoter but did not significantly affect the association of dimethyl-histone H3 (lys4), dimethyl-histone H3 (lys36), and dimethyl-histone H3 (lys79) with the epsilon-, gamma-, and beta-globin gene promoters. CONCLUSION: These experiments illustrate the usefulness of the baboon model to investigate the mechanism of pharmacologic reactivation of fetal hemoglobin synthesis at the molecular level.


Subject(s)
Azacitidine/analogs & derivatives , Chromatin/drug effects , Globins/genetics , Histones/drug effects , Promoter Regions, Genetic , Animals , Azacitidine/pharmacology , Base Sequence , Chromatin/chemistry , Chromatin Immunoprecipitation , DNA Primers , Decitabine , Histones/chemistry , Papio
SELECTION OF CITATIONS
SEARCH DETAIL