Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Nature ; 610(7933): 783-790, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36224385

RESUMO

Around birth, globin expression in human red blood cells (RBCs) shifts from γ-globin to ß-globin, which results in fetal haemoglobin (HbF, α2γ2) being gradually replaced by adult haemoglobin (HbA, α2ß2)1. This process has motivated the development of innovative approaches to treat sickle cell disease and ß-thalassaemia by increasing HbF levels in postnatal RBCs2. Here we provide therapeutically relevant insights into globin gene switching obtained through a CRISPR-Cas9 screen for ubiquitin-proteasome components that regulate HbF expression. In RBC precursors, depletion of the von Hippel-Lindau (VHL) E3 ubiquitin ligase stabilized its ubiquitination target, hypoxia-inducible factor 1α (HIF1α)3,4, to induce γ-globin gene transcription. Mechanistically, HIF1α-HIF1ß heterodimers bound cognate DNA elements in BGLT3, a long noncoding RNA gene located 2.7 kb downstream of the tandem γ-globin genes HBG1 and HBG2. This was followed by the recruitment of transcriptional activators, chromatin opening and increased long-range interactions between the γ-globin genes and their upstream enhancer. Similar induction of HbF occurred with hypoxia or with inhibition of prolyl hydroxylase domain enzymes that target HIF1α for ubiquitination by the VHL E3 ubiquitin ligase. Our findings link globin gene regulation with canonical hypoxia adaptation, provide a mechanism for HbF induction during stress erythropoiesis and suggest a new therapeutic approach for ß-haemoglobinopathies.


Assuntos
gama-Globinas , Humanos , Cromatina , Hemoglobina Fetal/biossíntese , Hemoglobina Fetal/genética , gama-Globinas/biossíntese , gama-Globinas/genética , Hipóxia/genética , Prolil Hidroxilases/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , RNA Longo não Codificante , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/genética , Eritropoese
2.
Nucleic Acids Res ; 49(17): 9711-9723, 2021 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-34379783

RESUMO

Human fetal globin (γ-globin) genes are developmentally silenced after birth, and reactivation of γ-globin expression in adulthood ameliorates symptoms of hemoglobin disorders, such as sickle cell disease (SCD) and ß-thalassemia. However, the mechanisms by which γ-globin expression is precisely regulated are still incompletely understood. Here, we found that NonO (non-POU domain-containing octamer-binding protein) interacted directly with SOX6, and repressed the expression of γ-globin gene in human erythroid cells. We showed that NonO bound to the octamer binding motif, ATGCAAAT, of the γ-globin proximal promoter, resulting in inhibition of γ-globin transcription. Depletion of NonO resulted in significant activation of γ-globin expression in K562, HUDEP-2, and primary human erythroid progenitor cells. To confirm the role of NonO in vivo, we further generated a conditional knockout of NonO by using IFN-inducible Mx1-Cre transgenic mice. We found that induced NonO deletion reactivated murine embryonic globin and human γ-globin gene expression in adult ß-YAC mice, suggesting a conserved role for NonO during mammalian evolution. Thus, our data indicate that NonO acts as a novel transcriptional repressor of γ-globin gene expression through direct promoter binding, and is essential for γ-globin gene silencing.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Hemoglobina Fetal/genética , Inativação Gênica , Proteínas de Ligação a RNA/metabolismo , gama-Globinas/genética , Animais , Células Cultivadas , Células Precursoras Eritroides/metabolismo , Hemoglobina Fetal/biossíntese , Humanos , Células K562 , Camundongos Knockout , Camundongos Transgênicos , Regiões Promotoras Genéticas , Fatores de Transcrição SOXD/metabolismo , gama-Globinas/biossíntese
3.
Am J Med Sci ; 362(1): 5-12, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33600783

RESUMO

ß-thalassemia is a lethal inherited disease resulting from ß-globin gene mutations. Severe ß-thalassemia requires regular blood transfusions. Other active interventions, including iron chelating, stem cell transplantation and gene therapy, have remarkably improved the quality of life and prolonged the survival of patients with transfusion-dependent ß-thalassemia, but all with significant limitations and complications. MicroRNAs (miRNAs), encoded by a class of endogenous genes, are found to play important roles in regulating globin expression. Among the miRNAs of particular interest related to ß-thalassemia, miR-15a/16-1, miR-486-3p, miR-26b, miR-199b-5p, miR-210, miR-34a, miR-138, miR-326, let-7, and miR-17/92 cluster elevate γ-globin expression, while miR-96, miR-146a, miR-223-3p, and miR-144 inhibit γ-globin expression. A couple of miRNAs, miR-144 and miR-150, repress α-globin expression, whereas miR-451 induces α-, ß- and γ-globin expression. Single nucleotide polymorphism in miRNA genes or their targeted genes might also contribute to the abnormal expression of hemoglobin. Moreover, changes in the expression of miR-125b, miR-210, miR-451, and miR-609 reflect the severity of anemia and hemolysis in ß-thalassemia patients. These results suggest that miRNAs are potential biomarkers for the diagnosis and prognosis of ß-thalassemia, and miRNA-based therapeutic strategy might be used as a coordinated approach for effectively treating ß-thalassemia.


Assuntos
MicroRNAs/genética , Talassemia beta/diagnóstico , Talassemia beta/genética , Animais , Humanos , MicroRNAs/biossíntese , Polimorfismo de Nucleotídeo Único/genética , alfa-Globinas/biossíntese , alfa-Globinas/genética , Talassemia beta/metabolismo , gama-Globinas/biossíntese , gama-Globinas/genética
4.
Exp Cell Res ; 394(2): 112168, 2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32653411

RESUMO

Pharmacologic induction of fetal hemoglobin (HbF) is an effective strategy for treating ß-hemoglobinopathies like ß-thalassemia and sickle cell anemia by ameliorating disease severity. Hydroxyurea is the only FDA-approved agent that induces HbF, but significant nonresponders and toxicity limit its clinical usefulness. This study relates preclinical investigation of Tenofovir disoproxil fumarate (TDF) as a potential HbF inducing agent, using human erythroleukemia cell line and a ß-YAC mouse model. Erythroid induction of K562 cells was studied by the benzidine/H2O2 reaction, total hemoglobin production was estimated by plasma hemoglobin assay kit, and γ-globin gene expression by RT-qPCR, whereas, fetal hemoglobin production was estimated by flow cytometry and immunofluorescence microscopy. We observed significantly increased γ- globin gene transcription and HbF expression mediated by TDF in K562 cells. Subsequent treatment of ß-YAC transgenic mice with TDF confirmed HbF induction in vivo through an increase in γ-globin gene expression and in the percentage of HbF positive red blood cells. Moreover, TDF showed no cytotoxic effect at HbF inducing concentrations. These data support the potential development of TDF for the treatment of hematological disorders, including ß-thalassemia and sickle cell anemia.


Assuntos
Hemoglobina Fetal/biossíntese , Tenofovir/farmacologia , gama-Globinas/biossíntese , Animais , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Células Eritroides/citologia , Células Eritroides/efeitos dos fármacos , Células Eritroides/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Células K562 , Camundongos Transgênicos , Tenofovir/química , Transcrição Gênica/efeitos dos fármacos , gama-Globinas/genética
5.
Blood ; 135(24): 2121-2132, 2020 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-32299090

RESUMO

Reactivation of fetal hemoglobin remains a critical goal in the treatment of patients with sickle cell disease and ß-thalassemia. Previously, we discovered that silencing of the fetal γ-globin gene requires the erythroid-specific eIF2α kinase heme-regulated inhibitor (HRI), suggesting that HRI might present a pharmacologic target for raising fetal hemoglobin levels. Here, via a CRISPR-Cas9-guided loss-of-function screen in human erythroblasts, we identify transcription factor ATF4, a known HRI-regulated protein, as a novel γ-globin regulator. ATF4 directly stimulates transcription of BCL11A, a repressor of γ-globin transcription, by binding to its enhancer and fostering enhancer-promoter contacts. Notably, HRI-deficient mice display normal Bcl11a levels, suggesting species-selective regulation, which we explain here by demonstrating that the analogous ATF4 motif at the murine Bcl11a enhancer is largely dispensable. Our studies uncover a linear signaling pathway from HRI to ATF4 to BCL11A to γ-globin and illustrate potential limits of murine models of globin gene regulation.


Assuntos
Fator 4 Ativador da Transcrição/genética , Hemoglobina Fetal/genética , Proteínas Repressoras/genética , eIF-2 Quinase/genética , Anemia Falciforme/sangue , Anemia Falciforme/genética , Anemia Falciforme/terapia , Animais , Sistemas CRISPR-Cas , Células Cultivadas , Elementos Facilitadores Genéticos , Eritroblastos/metabolismo , Regulação da Expressão Gênica , Inativação Gênica , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Especificidade da Espécie , Talassemia beta/sangue , Talassemia beta/genética , Talassemia beta/terapia , gama-Globinas/biossíntese , gama-Globinas/genética
6.
Curr Opin Hematol ; 27(3): 141-148, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32167946

RESUMO

PURPOSE OF REVIEW: ß-hemoglobinopathies, such as ß-Thalassemias (ß-Thal) and sickle cell disease (SCD) are among the most common inherited genetic disorders in humans worldwide. These disorders are characterized by a quantitative (ß-Thal) or qualitative (SCD) defects in adult hemoglobin production, leading to anemia, ineffective erythropoiesis and severe secondary complications. Reactivation of the fetal globin genes (γ-globin), making-up fetal hemoglobin (HbF), which are normally silenced in adults, represents a major strategy to ameliorate anemia and disease severity. RECENT FINDINGS: Following the identification of the first 'switching factors' for the reactivation of fetal globin gene expression more than 10 years ago, a multitude of novel leads have recently been uncovered. SUMMARY: Recent findings provided invaluable functional insights into the genetic and molecular networks controlling globin genes expression, revealing that complex repression systems evolved in erythroid cells to maintain HbF silencing in adults. This review summarizes these unique and exciting discoveries of the regulatory factors controlling the globin switch. New insights and novel leads for therapeutic strategies based on the pharmacological induction of HbF are discussed. This represents a major breakthrough for rational drug design in the treatment of ß-Thal and SCD.


Assuntos
Anemia Falciforme , Desenho de Fármacos , Hemoglobina Fetal , Regulação da Expressão Gênica , Talassemia beta , gama-Globinas , Anemia Falciforme/tratamento farmacológico , Anemia Falciforme/genética , Anemia Falciforme/metabolismo , Anemia Falciforme/patologia , Células Eritroides/metabolismo , Células Eritroides/patologia , Hemoglobina Fetal/biossíntese , Hemoglobina Fetal/genética , Humanos , Talassemia beta/tratamento farmacológico , Talassemia beta/genética , Talassemia beta/metabolismo , Talassemia beta/patologia , gama-Globinas/biossíntese , gama-Globinas/genética
7.
Curr Opin Hematol ; 27(3): 129-140, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32167945

RESUMO

PURPOSE OF REVIEW: The current review focuses on recent insights into the development of small molecule therapeutics to treat the ß-globinopathies. RECENT FINDINGS: Recent studies of fetal γ-globin gene regulation reveal multiple insights into how γ-globin gene reactivation may lead to novel treatment for ß-globinopathies. SUMMARY: We summarize current information regarding the binding of transcription factors that appear to be impeded or augmented by different hereditary persistence of fetal hemoglobin (HPFH) mutations. As transcription factors have historically proven to be difficult to target for therapeutic purposes, we next address the contributions of protein complexes associated with these HPFH mutation-affected transcription factors with the aim of defining proteins that might provide additional targets for chemical molecules to inactivate the corepressors. Among the enzymes associated with the transcription factor complexes, a group of corepressors with currently available inhibitors were initially thought to be good candidates for potential therapeutic purposes. We discuss possibilities for pharmacological inhibition of these corepressor enzymes that might significantly reactivate fetal γ-globin gene expression. Finally, we summarize the current clinical trial data regarding the inhibition of select corepressor proteins for the treatment of sickle cell disease and ß-thalassemia.


Assuntos
Anemia Falciforme , Mutação , Fatores de Transcrição , Talassemia beta , gama-Globinas , Anemia Falciforme/tratamento farmacológico , Anemia Falciforme/genética , Anemia Falciforme/metabolismo , Hemoglobina Fetal/biossíntese , Hemoglobina Fetal/genética , Humanos , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Talassemia beta/tratamento farmacológico , Talassemia beta/genética , Talassemia beta/metabolismo , gama-Globinas/biossíntese , gama-Globinas/genética
8.
Sci Rep ; 9(1): 11649, 2019 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-31406232

RESUMO

ß-Thalassaemia is one of the most common monogenic diseases with no effective cure in the majority of patients. Unbalanced production of α-globin in the presence of defective synthesis of ß-globin is the primary mechanism for anaemia in ß-thalassaemia. Clinical genetic data accumulated over three decades have clearly demonstrated that direct suppression of α-globin and induction of γ-globin are effective in reducing the globin chain imbalance in erythroid cells hence improving the clinical outcome of patients with ß-thalassaemia. Here, we show that the histone deacetylase inhibitor drug, vorinostat, in addition to its beneficial effects for patients with ß-thalassaemia through induction of γ-globin, has the potential to simultaneously suppress α-globin. We further show that vorinostat exhibits these synergistic beneficial effects in globin gene expression at nanomolar concentrations without perturbing erythroid expansion, viability, differentiation or the transcriptome. This new evidence will be helpful for the interpretation of existing clinical trials and future clinical studies that are directed towards finding a cure for ß-thalassaemia using vorinostat.


Assuntos
Inibidores de Histona Desacetilases/farmacologia , Vorinostat/farmacologia , alfa-Globinas/biossíntese , Talassemia beta/tratamento farmacológico , gama-Globinas/biossíntese , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Células Eritroides/efeitos dos fármacos , Sangue Fetal/citologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Inibidores de Histona Desacetilases/uso terapêutico , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Cultura Primária de Células , Vorinostat/uso terapêutico , alfa-Globinas/análise , Talassemia beta/sangue , gama-Globinas/análise
9.
Nat Med ; 25(5): 776-783, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30911135

RESUMO

Re-expression of the paralogous γ-globin genes (HBG1/2) could be a universal strategy to ameliorate the severe ß-globin disorders sickle cell disease (SCD) and ß-thalassemia by induction of fetal hemoglobin (HbF, α2γ2)1. Previously, we and others have shown that core sequences at the BCL11A erythroid enhancer are required for repression of HbF in adult-stage erythroid cells but are dispensable in non-erythroid cells2-6. CRISPR-Cas9-mediated gene modification has demonstrated variable efficiency, specificity, and persistence in hematopoietic stem cells (HSCs). Here, we demonstrate that Cas9:sgRNA ribonucleoprotein (RNP)-mediated cleavage within a GATA1 binding site at the +58 BCL11A erythroid enhancer results in highly penetrant disruption of this motif, reduction of BCL11A expression, and induction of fetal γ-globin. We optimize conditions for selection-free on-target editing in patient-derived HSCs as a nearly complete reaction lacking detectable genotoxicity or deleterious impact on stem cell function. HSCs preferentially undergo non-homologous compared with microhomology-mediated end joining repair. Erythroid progeny of edited engrafting SCD HSCs express therapeutic levels of HbF and resist sickling, while those from patients with ß-thalassemia show restored globin chain balance. Non-homologous end joining repair-based BCL11A enhancer editing approaching complete allelic disruption in HSCs is a practicable therapeutic strategy to produce durable HbF induction.


Assuntos
Edição de Genes/métodos , Células-Tronco Hematopoéticas/metabolismo , Sequência de Aminoácidos , Anemia Falciforme/sangue , Anemia Falciforme/genética , Anemia Falciforme/terapia , Sequência de Bases , Sistemas CRISPR-Cas , Proteínas de Transporte/genética , Elementos Facilitadores Genéticos , Células Precursoras Eritroides/metabolismo , Hemoglobina Fetal/biossíntese , Hemoglobina Fetal/genética , Transplante de Células-Tronco Hematopoéticas , Humanos , Mutação INDEL , Proteínas Nucleares/genética , RNA Guia de Cinetoplastídeos/genética , Proteínas Repressoras , Talassemia beta/sangue , Talassemia beta/genética , Talassemia beta/terapia , gama-Globinas/biossíntese , gama-Globinas/genética
10.
J Clin Invest ; 129(2): 598-615, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30422819

RESUMO

Current thalassemia gene therapy protocols require the collection of hematopoietic stem/progenitor cells (HSPCs), in vitro culture, lentivirus vector transduction, and retransplantation into myeloablated patients. Because of cost and technical complexity, it is unlikely that such protocols will be applicable in developing countries, where the greatest demand for a ß-thalassemia therapy lies. We have developed a simple in vivo HSPC gene therapy approach that involves HSPC mobilization and an intravenous injection of integrating HDAd5/35++ vectors. Transduced HSPCs homed back to the bone marrow, where they persisted long-term. HDAd5/35++ vectors for in vivo gene therapy of thalassemia had a unique capsid that targeted primitive HSPCs through human CD46, a relatively safe SB100X transposase-based integration machinery, a micro-LCR-driven γ-globin gene, and an MGMT(P140K) system that allowed for increasing the therapeutic effect by short-term treatment with low-dose O6-benzylguanine plus bis-chloroethylnitrosourea. We showed in "healthy" human CD46-transgenic mice and in a mouse model of thalassemia intermedia that our in vivo approach resulted in stable γ-globin expression in the majority of circulating red blood cells. The high marking frequency was maintained in secondary recipients. In the thalassemia model, a near-complete phenotypic correction was achieved. The treatment was well tolerated. This cost-efficient and "portable" approach could permit a broader clinical application of thalassemia gene therapy.


Assuntos
Eritrócitos , Regulação da Expressão Gênica , Terapia Genética , Células-Tronco Hematopoéticas/metabolismo , Talassemia beta , gama-Globinas , Adenoviridae , Animais , Linhagem Celular , Modelos Animais de Doenças , Eritrócitos/metabolismo , Eritrócitos/patologia , Vetores Genéticos , Células-Tronco Hematopoéticas/patologia , Humanos , Camundongos , Camundongos Transgênicos , Transdução Genética , Talassemia beta/genética , Talassemia beta/metabolismo , Talassemia beta/patologia , Talassemia beta/terapia , gama-Globinas/biossíntese , gama-Globinas/genética
11.
Genomics ; 111(6): 1771-1776, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-30529538

RESUMO

The master erythroid regulator KLF1,plays a pivotal role during erythroid lineage development by regulating the expression of many erythroid genes. Variations in the KLF1 gene are found to be associated with varied erythroid phenotypes. With the aim of determining the role of KLF1 gene variations in HbF induction and their genotype phenotype relationship, in this study, we screened 370 individuals with different hemoglobinopathy condition. Hematological analysis was carried out using automated blood cell counter and Variant II HPLC (Biorad). KLF1 gene mutations were screened using automated DNA sequencing. Expression analysis was carried out using q-RT PCR of KLF1, BCL11A and γ-globin after selective enrichment and culturing of CD 34 +ve cells into an erythroid lineage. Over all 14 KLF1 gene variations were identified, of which six variants were novel. The incidence of KLF1 gene mutations was found to be 8.1%. It was seen that KLF1 mutations contributed in borderline HbA2 levels as 7.6% of our borderline HbA2 cases showed presence of KLF1 variations. It also contributed in induction of HbF levels under stress erythropoietic conditions. Gene expression studies revealed inverse correlation of KLF1, BCL11A (reduced) with γ-globin gene expression (increased) in patients showing KLF1 gene mutations, thus indicating the role of KLF1 gene in regulating the γ-globin gene expression. The identification of genomic variants of the KLF1 may help in determining the functionally active domain of this protein and will facilitate in understanding the wide spectrum of phenotypes generated by these variants.


Assuntos
Eritropoese/genética , Doenças Genéticas Inatas/genética , Doenças Hematológicas/genética , Fatores de Transcrição Kruppel-Like/genética , Mutação , Eritrócitos/metabolismo , Eritrócitos/patologia , Feminino , Regulação da Expressão Gênica , Doenças Genéticas Inatas/metabolismo , Doenças Genéticas Inatas/patologia , Doenças Hematológicas/metabolismo , Doenças Hematológicas/patologia , Humanos , Fatores de Transcrição Kruppel-Like/metabolismo , Masculino , gama-Globinas/biossíntese , gama-Globinas/genética
12.
J Biol Chem ; 293(45): 17454-17463, 2018 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-30257864

RESUMO

Protein arginine methyltransferase 5 (PRMT5) is a member of the arginine methyltransferase protein family that critically mediates the symmetric dimethylation of Arg-3 at histone H4 (H4R3me2s) and is involved in many key cellular processes, including hematopoiesis. However, the post-translational modifications (PTMs) of PRMT5 that may affect its biological functions remain less well-understood. In this study, using MS analyses, we found that PRMT5 itself is methylated in human erythroleukemia Lys-562 cells. Biochemical assays revealed that coactivator-associated arginine methyltransferase 1 (CARM1) interacts directly with and methylates PRMT5 at Arg-505 both in vivo and in vitro. Substitutions at Arg-505 significantly reduced PRMT5's methyltransferase activity, decreased H4R3me2s enrichment at the γ-globin gene promoter, and increased the expression of the γ-globin gene in Lys-562 cells. Moreover, CARM1 knockdown consistently reduced PRMT5 activity and activated γ-globin gene expression. Importantly, we show that CARM1-mediated methylation of PRMT5 is essential for the intracellular homodimerization of PRMT5 to its active form. These results thus reveal a critical PTM of PRMT5 that represses human γ-globin gene expression. We conclude that CARM1-mediated asymmetric methylation of PRMT5 is critical for its dimerization and methyltransferase activity leading to the repression of γ-globin expression. Given PRMT5's crucial role in diverse cellular processes, these findings may inform strategies for manipulating its methyltransferase activity for managing hemoglobinopathy or cancer.


Assuntos
Regulação Neoplásica da Expressão Gênica , Leucemia Eritroblástica Aguda/metabolismo , Proteínas de Neoplasias/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo , gama-Globinas/biossíntese , Linhagem Celular Tumoral , Metilação de DNA/genética , DNA de Neoplasias/genética , DNA de Neoplasias/metabolismo , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/patologia , Proteínas de Neoplasias/genética , Proteína-Arginina N-Metiltransferases/genética , gama-Globinas/genética
13.
Exp Hematol ; 67: 60-64.e2, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30125603

RESUMO

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.


Assuntos
Anemia Falciforme/tratamento farmacológico , Inibidores Enzimáticos/uso terapêutico , Hemoglobina Fetal/biossíntese , Histona Desmetilases/antagonistas & inibidores , gama-Globinas/biossíntese , Administração Oral , Anemia/sangue , Anemia/tratamento farmacológico , Anemia Falciforme/sangue , Animais , Contagem de Células Sanguíneas , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/administração & dosagem , Feminino , Hemoglobina Fetal/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Papio , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Reticulócitos/metabolismo , gama-Globinas/genética
14.
Eur J Med Chem ; 154: 341-353, 2018 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-29852459

RESUMO

N-oxide derivatives 5(a-b), 8(a-b), and 11(a-c) were designed, synthesized and evaluated in vitro and in vivo as potential drugs that are able to ameliorate sickle cell disease (SCD) symptoms. All of the compounds demonstrated the capacity to releasing nitric oxide at different levels ranging from 0.8 to 30.1%, in vivo analgesic activity and ability to reduce TNF-α levels in the supernatants of monocyte cultures. The most active compound (8b) protected 50.1% against acetic acid-induced abdominal constrictions, while dipyrone, which was used as a control only protected 35%. Compounds 8a and 8b inhibited ADP-induced platelet aggregation by 84% and 76.1%, respectively. Both compounds increased γ-globin in K562 cells at 100 µM. The mechanisms involved in the γ-globin increase are related to the acetylation of histones H3 and H4 that is induced by these compounds. In vitro, the most promising compound (8b) was not cytotoxic, mutagenic and genotoxic.


Assuntos
Anemia Falciforme/tratamento farmacológico , Descoberta de Drogas , Histonas/metabolismo , Oxidiazóis/farmacologia , gama-Globinas/biossíntese , Ácido Acético/antagonistas & inibidores , Ácido Acético/farmacologia , Acetilação , Anemia Falciforme/metabolismo , Relação Dose-Resposta a Droga , Humanos , Células K562 , Estrutura Molecular , Óxido Nítrico/metabolismo , Oxidiazóis/síntese química , Oxidiazóis/química , Relação Estrutura-Atividade
16.
Exp Hematol ; 63: 33-40.e2, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29601850

RESUMO

Haploinsufficiency of erythroid Krüppel-like factor (EKLF/KLF1) has been shown recently to ameliorate the clinical severity of ß-thalassemia by increased expression levels of fetal hemoglobin (HbF). The underlying mechanisms for role of EKLF in regulating HbF are of great interest but remain incompletely understood. In this study, we used a combination of in silico, in vitro, and in vivo approaches to identify microRNAs (miRs) involved in EKLF regulation and to validate the role of miR-326 in HbF modification. We found that miR-326 suppresses EKLF expression directly by targeting its 3' untranslated region. miR-326 overexpression in K562 cells or CD34+ hematopoietic progenitor cells resulted in reduced EKLF protein levels and was associated with elevated expression of γ-globin, whereas inhibition of physiological miR-326 levels increased EKLF and thus reduced γ-globin expression. Moreover, miR-326 expression is positively correlated with HbF levels in ß-thalassemia patients. Our results suggest that miR-326 plays a key role in regulating EKLF expression and in modifying the HbF level, which may provide a new strategy for activating HbF in individuals with ß-thalassemia or sickle cell disease.


Assuntos
Células Eritroides/metabolismo , Fatores de Transcrição Kruppel-Like/biossíntese , MicroRNAs/fisiologia , gama-Globinas/biossíntese , Regiões 3' não Traduzidas , Adulto , Sítios de Ligação , Células Cultivadas , Regulação para Baixo , Células Eritroides/efeitos dos fármacos , Eritropoese , Hemoglobina Fetal/análise , Regulação da Expressão Gênica/efeitos dos fármacos , Genes Reporter , Células HEK293 , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Humanos , Células K562 , Fatores de Transcrição Kruppel-Like/genética , MicroRNAs/antagonistas & inibidores , MicroRNAs/farmacologia , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Recombinantes/biossíntese , Talassemia beta/sangue , gama-Globinas/genética
17.
J Cell Biochem ; 119(3): 2512-2519, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-28941328

RESUMO

ß-thalassemia is a common autosomal recessive disorder characterized by a deficiency in the synthesis of ß-chains. Evidences show that increased HbF levels improve the symptoms in patients with ß-thalassemia or sickle cell anemia. In this study, ZFN technology was applied to induce a mutation in the binding domain region of SOX6 to reactivate γ-globin expression. The sequences coding for ZFP arrays were designed and sub cloned in TDH plus as a transfer vector. The ZFN expression was confirmed using Western blot analysis. In the next step, using the site-directed mutagenesis strategy through the overlap PCR, a missense mutation (D64V) was induced in the catalytic domain of the integrase gene in the packaging plasmid and verified using DNA sequencing. Then, the integrase minus lentivirus containing ZFN cassette was packaged. Transduction of K562 cells with this virus was performed. Mutation detection assay was performed. The indel percentage of the cells transducted with lenti virus containing ZFN was 31%. After 5 days of erythroid differentiation with 15 µg/mL cisplatin, the levels of γ-globin mRNA were sixfold in the cells treated with ZFN compared to untreated cells. In the meantime, the measurement of HbF expression levels was carried out using hemoglobin electrophoresis and showed the same results. Integrase minus lentivirus can provide a useful tool for efficient transient gene expression and helps avoid disadvantages of gene targeting using the native virus. The ZFN strategy applied here to induce indel on SOX6 gene in adult erythroid progenitors may provide a method to activate fetal hemoglobin expression in individuals with ß-thalassemia.


Assuntos
Edição de Genes/métodos , Terapia Genética/métodos , Fatores de Transcrição SOXD/genética , Talassemia beta/genética , gama-Globinas/genética , Humanos , Células K562 , Mutação , Transdução Genética , Nucleases de Dedos de Zinco , gama-Globinas/biossíntese
18.
J Biol Chem ; 291(30): 15628-40, 2016 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-27231347

RESUMO

One mode of γ-globin gene silencing involves a GATA-1·FOG-1·Mi2ß repressor complex that binds to the -566 GATA site relative to the (A)γ-globin gene cap site. However, the mechanism of how this repressor complex is assembled at the -566 GATA site is unknown. In this study, we demonstrate that the O-linked N-acetylglucosamine (O-GlcNAc) processing enzymes, O-GlcNAc-transferase (OGT) and O-GlcNAcase (OGA), interact with the (A)γ-globin promoter at the -566 GATA repressor site; however, mutation of the GATA site to GAGA significantly reduces OGT and OGA promoter interactions in ß-globin locus yeast artificial chromosome (ß-YAC) bone marrow cells. When WT ß-YAC bone marrow cells are treated with the OGA inhibitor Thiamet-G, the occupancy of OGT, OGA, and Mi2ß at the (A)γ-globin promoter is increased. In addition, OGT and Mi2ß recruitment is increased at the (A)γ-globin promoter when γ-globin becomes repressed in postconception day E18 human ß-YAC transgenic mouse fetal liver. Furthermore, we show that Mi2ß is modified with O-GlcNAc, and both OGT and OGA interact with Mi2ß, GATA-1, and FOG-1. Taken together, our data suggest that O-GlcNAcylation is a novel mechanism of γ-globin gene regulation mediated by modulating the assembly of the GATA-1·FOG-1·Mi2ß repressor complex at the -566 GATA motif within the promoter.


Assuntos
Inativação Gênica/fisiologia , N-Acetilglucosaminiltransferases/metabolismo , Elementos de Resposta , beta-N-Acetil-Hexosaminidases/metabolismo , gama-Globinas/biossíntese , Animais , Fator de Transcrição GATA1/genética , Fator de Transcrição GATA1/metabolismo , Humanos , Células K562 , Camundongos , Camundongos Transgênicos , N-Acetilglucosaminiltransferases/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , beta-N-Acetil-Hexosaminidases/genética , gama-Globinas/genética
19.
Ann N Y Acad Sci ; 1368(1): 31-9, 2016 03.
Artigo em Inglês | MEDLINE | ID: mdl-26918894

RESUMO

The ß-hemoglobinopathies are the most common monogenic disorders in humans, with symptoms arising after birth when the fetal γ-globin genes are silenced and the adult ß-globin gene is activated. There is a growing appreciation that genome organization and the folding of chromosomes are key determinants of gene transcription. Underlying this function is the activity of transcriptional enhancers that increase the transcription of target genes over long linear distances. To accomplish this, enhancers engage in close physical contact with target promoters through chromosome folding or looping that is orchestrated by protein complexes that bind to both sites and stabilize their interaction. We find that enhancer activity can be redirected with concomitant changes in gene transcription. Both targeting the ß-globin locus control region (LCR) to the γ-globin gene in adult erythroid cells by tethering and epigenetic unmasking of a silenced γ-globin gene lead to increased frequency of LCR/γ-globin contacts and reduced LCR/ß-globin contacts. The outcome of these manipulations is robust, pancellular γ-globin transcription activation with a concomitant reduction in ß-globin transcription. These examples show that chromosome looping may be considered a therapeutic target for gene activation in ß-thalassemia and sickle cell disease.


Assuntos
Cromatina/genética , Proteínas de Ligação a DNA/genética , Células Eritroides , Hemoglobinopatias/genética , Proteínas com Domínio LIM/genética , Fatores de Transcrição/genética , Animais , Cromatina/metabolismo , Proteínas de Ligação a DNA/biossíntese , Células Eritroides/metabolismo , Terapia Genética/métodos , Terapia Genética/tendências , Hemoglobinopatias/metabolismo , Hemoglobinopatias/terapia , Histona-Lisina N-Metiltransferase/biossíntese , Histona-Lisina N-Metiltransferase/genética , Humanos , Proteínas com Domínio LIM/biossíntese , Quinazolinas/uso terapêutico , Fatores de Transcrição/biossíntese , Globinas beta/biossíntese , Globinas beta/genética , gama-Globinas/biossíntese , gama-Globinas/genética
20.
Haematologica ; 101(6): 688-97, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26858356

RESUMO

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.


Assuntos
Inibidores Enzimáticos/farmacologia , Hemoglobina Fetal/biossíntese , Hemoglobina Fetal/genética , Histona Desmetilases/antagonistas & inibidores , Anemia/sangue , Anemia/tratamento farmacológico , Anemia/etiologia , Animais , Contagem de Células Sanguíneas , Metilação de DNA/efeitos dos fármacos , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Histonas/metabolismo , Papio , Reticulócitos/efeitos dos fármacos , Reticulócitos/metabolismo , gama-Globinas/biossíntese , gama-Globinas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...