Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
1.
Annu Rev Genomics Hum Genet ; 24: 255-275, 2023 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-37624668

RESUMO

Sickle cell disease (SCD) is a monogenic blood disease caused by a point mutation in the gene coding for ß-globin. The abnormal hemoglobin [sickle hemoglobin (HbS)] polymerizes under low-oxygen conditions and causes red blood cells to sickle. The clinical presentation varies from very severe (with acute pain, chronic pain, and early mortality) to normal (few complications and a normal life span). The variability of SCD might be due (in part) to various genetic modulators. First, we review the main genetic factors, polymorphisms, and modifier genes that influence the expression of globin or otherwise modulate the severity of SCD. Considering SCD as a complex, multifactorial disorder is important for the development of appropriate pharmacological and genetic treatments. Second, we review the characteristics, advantages, and disadvantages of the latest advances in gene therapy for SCD, from lentiviral-vector-based approaches to gene-editing strategies.


Assuntos
Dor Aguda , Anemia Falciforme , Dor Crônica , Hemoglobinas Anormais , Humanos , Anemia Falciforme/genética , Anemia Falciforme/terapia , Eritrócitos
2.
Blood ; 141(10): 1169-1179, 2023 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-36508706

RESUMO

ß-Thalassemia (BT) is one of the most common genetic diseases worldwide and is caused by mutations affecting ß-globin production. The only curative treatment is allogenic hematopoietic stem/progenitor cells (HSPCs) transplantation, an approach limited by compatible donor availability and immunological complications. Therefore, transplantation of autologous, genetically-modified HSPCs is an attractive therapeutic option. However, current gene therapy strategies based on the use of lentiviral vectors are not equally effective in all patients and CRISPR/Cas9 nuclease-based strategies raise safety concerns. Thus, base editing strategies aiming to correct the genetic defect in patients' HSPCs could provide safe and effective treatment. Here, we developed a strategy to correct one of the most prevalent BT mutations (IVS1-110 [G>A]) using the SpRY-ABE8e base editor. RNA delivery of the base editing system was safe and led to ∼80% of gene correction in the HSPCs of patients with BT without causing dangerous double-strand DNA breaks. In HSPC-derived erythroid populations, this strategy was able to restore ß-globin production and correct inefficient erythropoiesis typically observed in BT both in vitro and in vivo. In conclusion, this proof-of-concept study paves the way for the development of a safe and effective autologous gene therapy approach for BT.


Assuntos
Talassemia beta , Humanos , Talassemia beta/genética , Talassemia beta/terapia , Edição de Genes , Sistemas CRISPR-Cas , Mutação , Globinas beta/genética
3.
Mol Ther ; 31(7): 2257-2265, 2023 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-36905119

RESUMO

Electroporation of the Cas9 ribonucleoprotein (RNP) complex offers the advantage of preventing off-target cleavages and potential immune responses produced by long-term expression of the nuclease. Nevertheless, the majority of engineered high-fidelity Streptococcus pyogenes Cas9 (SpCas9) variants are less active than the wild-type enzyme and are not compatible with RNP delivery. Building on our previous studies on evoCas9, we developed a high-fidelity SpCas9 variant suitable for RNP delivery. The editing efficacy and precision of the recombinant high-fidelity Cas9 (rCas9HF), characterized by the K526D substitution, was compared with the R691A mutant (HiFi Cas9), which is currently the only available high-fidelity Cas9 that can be used as an RNP. The comparative analysis was extended to gene substitution experiments where the two high fidelities were used in combination with a DNA donor template, generating different ratios of non-homologous end joining (NHEJ) versus homology-directed repair (HDR) for precise editing. The analyses revealed a heterogeneous efficacy and precision indicating different targeting capabilities between the two variants throughout the genome. The development of rCas9HF, characterized by an editing profile diverse from the currently used HiFi Cas9 in RNP electroporation, increases the genome editing solutions for the highest precision and efficient applications.


Assuntos
Sistemas CRISPR-Cas , Streptococcus pyogenes , Streptococcus pyogenes/genética , Edição de Genes , Proteína 9 Associada à CRISPR/genética , Eletroporação
4.
Mol Ther ; 30(1): 47-53, 2022 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-33823304

RESUMO

Motor neuron diseases are untreatable with common pharmacological approaches. Spinal muscular atrophy (SMA) is caused by SMN1 gene mutations leading to lowered SMN expression. Symptoms are alleviated in infants with a higher copy number of the SMN2 gene, which, however, displays a splicing defect resulting in low SMN levels. Amyotrophic lateral sclerosis (ALS) is caused by a number of mutations, with C9orf72 repeat expansions the most common genetic cause and SOD1 gain-of-function mutations the first genetic cause identified for this disease. Genetic therapies based on oligonucleotides that enhance SMN2 splicing and SMN production or lower SOD1 expression have shown promise in initial clinical trials for individuals with SMA and ALS harboring SOD1 mutations, respectively. Gene addition/silencing approaches using adeno-associated viruses (AAVs) are also currently under clinical investigation in trials for SMA and ALS. Here we provide a brief overview of these efforts and their advantages and challenges. We also review genome editing approaches aimed at correcting the disease-causing mutations or modulating the expression of genetic modifiers, e.g., by repairing SOD1 mutations or the SMN2 splicing defect or deleting C9orf72 expanded repeats. These studies have shown promising results to approach therapeutic trials that should significantly lower the progression of these deadly disorders.


Assuntos
Esclerose Lateral Amiotrófica , Atrofia Muscular Espinal , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/terapia , Edição de Genes , Humanos , Lactente , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/metabolismo , Atrofia Muscular Espinal/terapia , Oligonucleotídeos/metabolismo , Splicing de RNA , Proteína 1 de Sobrevivência do Neurônio Motor/genética , Proteína 1 de Sobrevivência do Neurônio Motor/metabolismo
5.
Mol Ther ; 30(1): 145-163, 2022 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-34418541

RESUMO

Sickle cell disease (SCD) is caused by a mutation in the ß-globin gene leading to polymerization of the sickle hemoglobin (HbS) and deformation of red blood cells. Autologous transplantation of hematopoietic stem/progenitor cells (HSPCs) genetically modified using lentiviral vectors (LVs) to express an anti-sickling ß-globin leads to some clinical benefit in SCD patients, but it requires high-level transgene expression (i.e., high vector copy number [VCN]) to counteract HbS polymerization. Here, we developed therapeutic approaches combining LV-based gene addition and CRISPR-Cas9 strategies aimed to either knock down the sickle ß-globin and increase the incorporation of an anti-sickling globin (AS3) in hemoglobin tetramers, or to induce the expression of anti-sickling fetal γ-globins. HSPCs from SCD patients were transduced with LVs expressing AS3 and a guide RNA either targeting the endogenous ß-globin gene or regions involved in fetal hemoglobin silencing. Transfection of transduced cells with Cas9 protein resulted in high editing efficiency, elevated levels of anti-sickling hemoglobins, and rescue of the SCD phenotype at a significantly lower VCN compared to the conventional LV-based approach. This versatile platform can improve the efficacy of current gene addition approaches by combining different therapeutic strategies, thus reducing the vector amount required to achieve a therapeutic VCN and the associated genotoxicity risk.


Assuntos
Anemia Falciforme , Edição de Genes , Anemia Falciforme/genética , Anemia Falciforme/terapia , Proteína 9 Associada à CRISPR/genética , Hemoglobina Fetal/genética , Edição de Genes/métodos , Humanos , Globinas beta/genética
6.
Hum Mol Genet ; 28(R1): R24-R30, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31322165

RESUMO

Recently, gene therapy clinical trials have been successfully applied to hemoglobinopathies, such as sickle cell disease (SCD) and ß-thalassemia. Among the great discoveries that led to the design of genetic approaches to cure these disorders is the discovery of the ß-globin locus control region and several associated transcription factors, which determine hemoglobin switching as well as high-level, erythroid-specific expression of genes at the ß-globin locus. Moreover, increasing evidence shows that lentiviral vectors are efficient tools to insert large DNA elements into nondividing hematopoietic stem cells, showing reassuring safe integration profiles. Alternatively, genome editing could restore expression of fetal hemoglobin or target specific mutations to restore expression of the wild-type ß-globin gene. The most recent clinical trials for ß-thalassemia and SCD are showing promising outcomes: patients were able to discontinue transfusions or had reduced transfusion requirements. However, toxic myeloablation and the high cost of current ex vivo hematopoietic stem cell gene therapy platforms represent a barrier to a widespread application of these approaches. In this review, we summarize these gene therapy strategies and ongoing clinical trials. Finally, we discuss possible strategies to improve outcomes, reduce myeloablative regimens and future challenges to reduce the cost of gene therapy platform.


Assuntos
Terapia Genética , Hemoglobinopatias/genética , Hemoglobinopatias/terapia , Animais , Ensaios Clínicos como Assunto , Regulação da Expressão Gênica , Predisposição Genética para Doença , Terapia Genética/efeitos adversos , Terapia Genética/economia , Terapia Genética/métodos , Terapia Genética/tendências , Vetores Genéticos/genética , Transplante de Células-Tronco Hematopoéticas , Hemoglobinas/genética , Humanos , Mutação , Transdução Genética , Resultado do Tratamento
7.
Blood ; 134(15): 1203-1213, 2019 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-31467062

RESUMO

ß-Thalassemia and sickle cell disease (SCD) are the most prevalent monogenic diseases. These disorders are caused by quantitative or qualitative defects in the production of adult hemoglobin. Gene therapy is a potential treatment option for patients lacking an allogenic compatible hematopoietic stem cell (HSC) donor. New-generation lentiviral vectors (LVs) carrying a ß-globin-like gene have revolutionized this field by allowing effective HSC transduction, with no evidence of genotoxicity to date. Several clinical trials with different types of vector are underway worldwide; the initial results are encouraging with regard to the sustained production of therapeutic hemoglobin, improved biological parameters, a lower transfusion requirement, and better quality of life. Long-term follow-up studies will confirm the safety of LV-based gene therapy. The optimization of patient conditioning, HSC harvesting, and HSC transduction has further improved the therapeutic potential of this approach. Novel LV-based strategies for reactivating endogenous fetal hemoglobin (HbF) are also promising, because elevated HbF levels can reduce the severity of both ß-thalassemia and SCD. Lastly, genome-editing approaches designed to correct the disease-causing mutation or reactivate HbF are currently under investigation. Here, we discuss the clinical outcomes of current LV-based gene addition trials and the promising advantages of novel alternative therapeutic strategies.


Assuntos
Hemoglobina Fetal/genética , Edição de Genes/métodos , Terapia Genética/métodos , Hemoglobinopatias/terapia , Globinas beta/genética , Anemia Falciforme/genética , Anemia Falciforme/terapia , Animais , Ensaios Clínicos como Assunto , Terapia Genética/efeitos adversos , Vetores Genéticos/genética , Vetores Genéticos/uso terapêutico , Hemoglobinopatias/genética , Humanos , Lentivirus/genética , Talassemia beta/genética , Talassemia beta/terapia
8.
Haematologica ; 106(10): 2707-2719, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32855279

RESUMO

While ineffective erythropoiesis has long been recognized as a key contributor to anemia in thalassemia, its role in anemia of sickle cell disease (SCD) has not been critically explored. Using in vitro and in vivo derived human erythroblasts we assessed the extent of ineffective erythropoiesis in SCD. Modeling the bone marrow hypoxic environment, we found that hypoxia induces death of sickle erythroblasts starting at the polychromatic stage, positively selecting cells with high levels of fetal hemoglobin (HbF). Cell death was associated with cytoplasmic sequestration of heat shock protein 70 and was rescued by induction of HbF synthesis. Importantly, we document that in the bone marrow of SCD patients similar cell loss occurs during the final stages of terminal differentiation. Our study provides evidence for ineffective erythropoiesis in SCD and highlights an anti-apoptotic role for HbF during the terminal stages of erythroid differentiation. These findings imply that the beneficial effect on anemia of increased HbF levels is not only due to the increased life span of red cells but also a consequence of decreased ineffective erythropoiesis.


Assuntos
Anemia Falciforme , Hemoglobina Fetal , Eritroblastos , Eritrócitos , Eritropoese , Humanos
9.
IUBMB Life ; 72(1): 10-26, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31574210

RESUMO

The members of the GATA family of transcription factors have homologous zinc fingers and bind to similar sequence motifs. Recent advances in genome-wide technologies and the integration of bioinformatics data have led to a better understanding of how GATA factors regulate gene expression; GATA-factor-induced transcriptional and epigenetic changes have now been analyzed at unprecedented levels of detail. Here, we review the results of genome-wide studies of GATA factor occupancy in human and murine cell lines and primary cells (as determined by chromatin immunoprecipitation sequencing), and then discuss the molecular mechanisms underlying the mediation of transcriptional and epigenetic regulation by GATA factors.


Assuntos
Epigênese Genética , Fatores de Transcrição GATA/metabolismo , Regulação Neoplásica da Expressão Gênica , Genoma , Neoplasias/patologia , Animais , Biologia Computacional , Fatores de Transcrição GATA/genética , Humanos , Neoplasias/genética , Neoplasias/metabolismo
10.
Blood ; 131(17): 1960-1973, 2018 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-29519807

RESUMO

Naturally occurring, large deletions in the ß-globin locus result in hereditary persistence of fetal hemoglobin, a condition that mitigates the clinical severity of sickle cell disease (SCD) and ß-thalassemia. We designed a clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated protein 9 (Cas9) (CRISPR/Cas9) strategy to disrupt a 13.6-kb genomic region encompassing the δ- and ß-globin genes and a putative γ-δ intergenic fetal hemoglobin (HbF) silencer. Disruption of just the putative HbF silencer results in a mild increase in γ-globin expression, whereas deletion or inversion of a 13.6-kb region causes a robust reactivation of HbF synthesis in adult erythroblasts that is associated with epigenetic modifications and changes in chromatin contacts within the ß-globin locus. In primary SCD patient-derived hematopoietic stem/progenitor cells, targeting the 13.6-kb region results in a high proportion of γ-globin expression in erythroblasts, increased HbF synthesis, and amelioration of the sickling cell phenotype. Overall, this study provides clues for a potential CRISPR/Cas9 genome editing approach to the therapy of ß-hemoglobinopathies.


Assuntos
Anemia Falciforme , Sistemas CRISPR-Cas , Hemoglobina Fetal , Edição de Genes , Loci Gênicos , Células-Tronco Hematopoéticas/metabolismo , Globinas beta/genética , Anemia Falciforme/genética , Anemia Falciforme/metabolismo , Anemia Falciforme/patologia , Anemia Falciforme/terapia , Linhagem Celular , Hemoglobina Fetal/biossíntese , Hemoglobina Fetal/genética , Células-Tronco Hematopoéticas/patologia , Humanos , Globinas beta/metabolismo
11.
Haematologica ; 105(5): 1240-1247, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31537695

RESUMO

Although studies of mixed chimerism following hematopoietic stem cell transplantation in patients with sickle cell disease (SCD) may provide insights into the engraftment needed to correct the disease and into immunological reconstitution, an extensive multilineage analysis is lacking. We analyzed chimerism simultaneously in peripheral erythroid and granulomonocytic precursors/progenitors, highly purified B and T lymphocytes, monocytes, granulocytes and red blood cells (RBC). Thirty-four patients with mixed chimerism and ≥12 months of follow-up were included. A selective advantage of donor RBC and their progenitors/precursors led to full chimerism in mature RBC (despite partial engraftment of other lineages), and resulted in the clinical control of the disease. Six patients with donor chimerism <50% had hemolysis (reticulocytosis) and higher HbS than their donor. Four of them had donor chimerism <30%, including a patient with AA donor (hemoglobin >10 g/dL) and three with AS donors (hemoglobin <10 g/dL). However, only one vaso-occlusive crisis occurred with 68.7% HbS. Except in the patients with the lowest chimerism, the donor engraftment was lower for T cells than for the other lineages. In a context of mixed chimerism after hematopoietic stem cell transplantation for SCD, myeloid (rather than T cell) engraftment was the key efficacy criterion. Results show that myeloid chimerism as low as 30% was sufficient to prevent a vaso-occlusive crisis in transplants from an AA donor but not constantly from an AS donor. However, the correction of hemolysis requires higher donor chimerism levels (i.e ≥50%) in both AA and AS recipients. In the future, this group of patients may need a different therapeutic approach.


Assuntos
Anemia Falciforme , Transplante de Células-Tronco Hematopoéticas , Anemia Falciforme/terapia , Quimerismo , Terapia Genética , Hematopoese , Humanos , Quimeras de Transplante , Transplante Homólogo
12.
Mol Ther ; 27(1): 137-150, 2019 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-30424953

RESUMO

Editing the ß-globin locus in hematopoietic stem cells is an alternative therapeutic approach for gene therapy of ß-thalassemia and sickle cell disease. Using the CRISPR/Cas9 system, we genetically modified human hematopoietic stem and progenitor cells (HSPCs) to mimic the large rearrangements in the ß-globin locus associated with hereditary persistence of fetal hemoglobin (HPFH), a condition that mitigates the clinical phenotype of patients with ß-hemoglobinopathies. We optimized and compared the efficiency of plasmid-, lentiviral vector (LV)-, RNA-, and ribonucleoprotein complex (RNP)-based methods to deliver the CRISPR/Cas9 system into HSPCs. Plasmid delivery of Cas9 and gRNA pairs targeting two HPFH-like regions led to high frequency of genomic rearrangements and HbF reactivation in erythroblasts derived from sorted, Cas9+ HSPCs but was associated with significant cell toxicity. RNA-mediated delivery of CRISPR/Cas9 was similarly toxic but much less efficient in editing the ß-globin locus. Transduction of HSPCs by LVs expressing Cas9 and gRNA pairs was robust and minimally toxic but resulted in poor genome-editing efficiency. Ribonucleoprotein (RNP)-based delivery of CRISPR/Cas9 exhibited a good balance between cytotoxicity and efficiency of genomic rearrangements as compared to the other delivery systems and resulted in HbF upregulation in erythroblasts derived from unselected edited HSPCs.


Assuntos
Proteína 9 Associada à CRISPR/metabolismo , Sistemas CRISPR-Cas/fisiologia , Terapia Genética/métodos , Células-Tronco Hematopoéticas/metabolismo , Anemia Falciforme/genética , Anemia Falciforme/metabolismo , Anemia Falciforme/terapia , Proteína 9 Associada à CRISPR/genética , Sistemas CRISPR-Cas/genética , Edição de Genes/métodos , Células-Tronco Hematopoéticas/citologia , Hemoglobinopatias/genética , Hemoglobinopatias/metabolismo , Hemoglobinopatias/terapia , Ribonucleoproteínas Nucleares Heterogêneas/genética , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Humanos , Plasmídeos/genética , RNA Guia de Cinetoplastídeos/genética , RNA Guia de Cinetoplastídeos/metabolismo , Talassemia beta/genética , Talassemia beta/metabolismo , Talassemia beta/terapia
13.
Haematologica ; 103(5): 778-786, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29472357

RESUMO

Sickle cell disease is characterized by chronic anemia and vaso-occlusive crises, which eventually lead to multi-organ damage and premature death. Hematopoietic stem cell transplantation is the only curative treatment but it is limited by toxicity and poor availability of HLA-compatible donors. A gene therapy approach based on the autologous transplantation of lentiviral-corrected hematopoietic stem and progenitor cells was shown to be efficacious in one patient. However, alterations of the bone marrow environment and properties of the red blood cells hamper the harvesting and immunoselection of patients' stem cells from bone marrow. The use of Filgrastim to mobilize large numbers of hematopoietic stem and progenitor cells into the circulation has been associated with severe adverse events in sickle cell patients. Thus, broader application of the gene therapy approach requires the development of alternative mobilization methods. We set up a phase I/II clinical trial whose primary objective was to assess the safety of a single injection of Plerixafor in sickle cell patients undergoing red blood cell exchange to decrease the hemoglobin S level to below 30%. The secondary objective was to measure the efficiency of mobilization and isolation of hematopoietic stem and progenitor cells. No adverse events were observed. Large numbers of CD34+ cells were mobilized extremely quickly. Importantly, the mobilized cells contained high numbers of hematopoietic stem cells, expressed high levels of stemness genes, and engrafted very efficiently in immunodeficient mice. Thus, Plerixafor can be safely used to mobilize hematopoietic stem cells in sickle cell patients; this finding opens up new avenues for treatment approaches based on gene addition and genome editing. Clinicaltrials.gov identifier: NCT02212535.


Assuntos
Anemia Falciforme/terapia , Transfusão de Sangue , Mobilização de Células-Tronco Hematopoéticas , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Compostos Heterocíclicos/administração & dosagem , Anemia Falciforme/metabolismo , Anemia Falciforme/patologia , Fármacos Anti-HIV/administração & dosagem , Antígenos CD34/metabolismo , Antidrepanocíticos/administração & dosagem , Benzilaminas , Estudos de Casos e Controles , Células Cultivadas , Estudos de Coortes , Ciclamos , Células-Tronco Hematopoéticas/citologia , Humanos , Hidroxiureia/administração & dosagem
14.
Mol Ther ; 25(5): 1142-1154, 2017 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-28377044

RESUMO

ß-Thalassemia and sickle cell disease (SCD) are the world's two most widely disseminated hereditary hemoglobinopathies. ß-Thalassemia originated in the Mediterranean, Middle Eastern, and Asian regions, and SCD originated in central Africa. However, subsequent population migration means that these two diseases are now global and thus constitute a growing health problem in many countries. Despite remarkable improvements in medical care for patients with ß-hemoglobinopathies, there is still only one definitive treatment option: allogeneic hematopoietic stem cell (HSC) transplantation. The development of gene therapy for ß-hemoglobinopathies has been justified by (1) the limited availability of human leukocyte antigen (HLA)-identical donors, (2) the narrow window of application of HSC transplantation to the youngest patients, and (3) recent advances in HSC-based gene therapy. The huge ongoing efforts in translational medicine and the high number of related publications show that gene therapy has the potential to become the treatment of choice for patients who lack either an HLA genoidentical sibling or an alternative, medically acceptable donor. In this dynamic scientific context, we first summarize the main steps toward clinical translation of this therapeutic approach and then discuss novel lentiviral- and genome editing-based treatment strategies for ß-hemoglobinopathies.


Assuntos
Anemia Falciforme/terapia , Edição de Genes/métodos , Terapia Genética/métodos , Transplante de Células-Tronco Hematopoéticas , Globinas beta/genética , Talassemia beta/terapia , Anemia Falciforme/genética , Anemia Falciforme/metabolismo , Anemia Falciforme/patologia , Gammaretrovirus/genética , Gammaretrovirus/imunologia , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/imunologia , Antígenos HLA , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Lentivirus/genética , Lentivirus/imunologia , Mutação , Doadores de Tecidos , Transplante Homólogo , Globinas beta/metabolismo , Talassemia beta/genética , Talassemia beta/metabolismo , Talassemia beta/patologia
15.
EMBO J ; 29(2): 442-56, 2010 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-19927129

RESUMO

GATA transcription factors interact with FOG proteins to regulate tissue development by activating and repressing transcription. FOG-1 (ZFPM1), a co-factor for the haematopoietic factor GATA-1, binds to the NuRD co-repressor complex through a conserved N-terminal motif. Surprisingly, we detected NuRD components at both repressed and active GATA-1/FOG-1 target genes in vivo. In addition, while NuRD is required for transcriptional repression in certain contexts, we show a direct requirement of NuRD also for FOG-1-dependent transcriptional activation. Mice in which the FOG-1/NuRD interaction is disrupted display defects similar to germline mutations in the Gata1 and Fog1 genes, including anaemia and macrothrombocytopaenia. Gene expression analysis in primary mutant erythroid cells and megakaryocytes (MKs) revealed an essential function for NuRD during both the repression and activation of select GATA-1/FOG-1 target genes. These results show that NuRD is a critical co-factor for FOG-1 and underscore the versatile use of NuRD by lineage-specific transcription factors to activate and repress gene transcription in the appropriate cellular and genetic context.


Assuntos
Fator de Transcrição GATA1/metabolismo , Hematopoese , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/metabolismo , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Animais , Células Cultivadas , Células Eritroides/citologia , Células Eritroides/metabolismo , Fator de Transcrição GATA1/genética , Megacariócitos/citologia , Megacariócitos/metabolismo , Camundongos , Proteínas Nucleares/genética , Fatores de Transcrição/genética , Transcrição Gênica , Ativação Transcricional
16.
Cell Stem Cell ; 31(4): 435-436, 2024 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-38579680

RESUMO

Lee et al.1 analyzed the impacts of lentiviral vector transduction and CRISPR-Cas9/homology-directed repair editing on hematopoietic stem and progenitor cell (HSPC) engraftment and clonal dynamics. The study suggests that relative to lentiviral-vector-mediated gene addition, homology-directed repair editing is inefficient in vivo and might impair the engraftment and differentiation of HSPCs.


Assuntos
Edição de Genes , Transplante de Células-Tronco Hematopoéticas , Animais , Humanos , Células-Tronco Hematopoéticas/metabolismo , Modelos Animais , Sistemas CRISPR-Cas/genética
17.
Blood Rev ; 65: 101185, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38493007

RESUMO

Recent advancements in gene editing illuminate new potential therapeutic approaches for Sickle Cell Disease (SCD), a debilitating monogenic disorder caused by a point mutation in the ß-globin gene. Despite the availability of several FDA-approved medications for symptomatic relief, allogeneic hematopoietic stem cell transplantation (HSCT) remains the sole curative option, underscoring a persistent need for novel treatments. This review delves into the growing field of gene editing, particularly the extensive research focused on curing haemoglobinopathies like SCD. We examine the use of techniques such as CRISPR-Cas9 and homology-directed repair, base editing, and prime editing to either correct the pathogenic variant into a non-pathogenic or wild-type one or augment fetal haemoglobin (HbF) production. The article elucidates ways to optimize these tools for efficacious gene editing with minimal off-target effects and offers insights into their effective delivery into cells. Furthermore, we explore clinical trials involving alternative SCD treatment strategies, such as LentiGlobin therapy and autologous HSCT, distilling the current findings. This review consolidates vital information for the clinical translation of gene editing for SCD, providing strategic insights for investigators eager to further the development of gene editing for SCD.


Assuntos
Anemia Falciforme , Hemoglobinopatias , Humanos , Edição de Genes/métodos , Sistemas CRISPR-Cas , Anemia Falciforme/genética , Anemia Falciforme/terapia , Hemoglobinopatias/genética , Hemoglobina Fetal/genética
18.
Nat Commun ; 15(1): 3478, 2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38658578

RESUMO

The expansion of the CRISPR-Cas toolbox is highly needed to accelerate the development of therapies for genetic diseases. Here, through the interrogation of a massively expanded repository of metagenome-assembled genomes, mostly from human microbiomes, we uncover a large variety (n = 17,173) of type II CRISPR-Cas loci. Among these we identify CoCas9, a strongly active and high-fidelity nuclease with reduced molecular size (1004 amino acids) isolated from an uncultivated Collinsella species. CoCas9 is efficiently co-delivered with its sgRNA through adeno associated viral (AAV) vectors, obtaining efficient in vivo editing in the mouse retina. With this study we uncover a collection of previously uncharacterized Cas9 nucleases, including CoCas9, which enriches the genome editing toolbox.


Assuntos
Sistemas CRISPR-Cas , Edição de Genes , Microbiota , Edição de Genes/métodos , Humanos , Animais , Camundongos , Microbiota/genética , Dependovirus/genética , Proteína 9 Associada à CRISPR/metabolismo , Proteína 9 Associada à CRISPR/genética , RNA Guia de Sistemas CRISPR-Cas/genética , RNA Guia de Sistemas CRISPR-Cas/metabolismo , Retina/metabolismo , Clostridiales/genética , Clostridiales/enzimologia , Células HEK293 , Vetores Genéticos/metabolismo , Vetores Genéticos/genética
19.
Nat Commun ; 15(1): 4965, 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38862518

RESUMO

Sickle cell disease is a devastating blood disorder that originates from a single point mutation in the HBB gene coding for hemoglobin. Here, we develop a GMP-compatible TALEN-mediated gene editing process enabling efficient HBB correction via a DNA repair template while minimizing risks associated with HBB inactivation. Comparing viral versus non-viral DNA repair template delivery in hematopoietic stem and progenitor cells in vitro, both strategies achieve comparable HBB correction and result in over 50% expression of normal adult hemoglobin in red blood cells without inducing ß-thalassemic phenotype. In an immunodeficient female mouse model, transplanted cells edited with the non-viral strategy exhibit higher engraftment and gene correction levels compared to those edited with the viral strategy. Transcriptomic analysis reveals that non-viral DNA repair template delivery mitigates P53-mediated toxicity and preserves high levels of long-term hematopoietic stem cells. This work paves the way for TALEN-based autologous gene therapy for sickle cell disease.


Assuntos
Anemia Falciforme , Edição de Genes , Terapia Genética , Células-Tronco Hematopoéticas , Nucleases dos Efetores Semelhantes a Ativadores de Transcrição , Anemia Falciforme/terapia , Anemia Falciforme/genética , Edição de Genes/métodos , Animais , Células-Tronco Hematopoéticas/metabolismo , Humanos , Feminino , Camundongos , Terapia Genética/métodos , Nucleases dos Efetores Semelhantes a Ativadores de Transcrição/metabolismo , Nucleases dos Efetores Semelhantes a Ativadores de Transcrição/genética , Transplante de Células-Tronco Hematopoéticas , Globinas beta/genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Reparo do DNA , Mutação , Talassemia beta/terapia , Talassemia beta/genética , Modelos Animais de Doenças , Técnicas de Transferência de Genes
20.
Genes (Basel) ; 14(3)2023 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-36980849

RESUMO

Beta-like globin gene expression is developmentally regulated during life by transcription factors, chromatin looping and epigenome modifications of the ß-globin locus. Epigenome modifications, such as histone methylation/demethylation and acetylation/deacetylation and DNA methylation, are associated with up- or down-regulation of gene expression. The understanding of these mechanisms and their outcome in gene expression has paved the way to the development of new therapeutic strategies for treating various diseases, such as ß-hemoglobinopathies. Histone deacetylase and DNA methyl-transferase inhibitors are currently being tested in clinical trials for hemoglobinopathies patients. However, these approaches are often uncertain, non-specific and their global effect poses serious safety concerns. Epigenome editing is a recently developed and promising tool that consists of a DNA recognition domain (zinc finger, transcription activator-like effector or dead clustered regularly interspaced short palindromic repeats Cas9) fused to the catalytic domain of a chromatin-modifying enzyme. It offers a more specific targeting of disease-related genes (e.g., the ability to reactivate the fetal γ-globin genes and improve the hemoglobinopathy phenotype) and it facilitates the development of scarless gene therapy approaches. Here, we summarize the mechanisms of epigenome regulation of the ß-globin locus, and we discuss the application of epigenome editing for the treatment of hemoglobinopathies.


Assuntos
Epigênese Genética , Hemoglobinopatias , Humanos , Epigênese Genética/genética , Epigenoma , Hemoglobinopatias/genética , Hemoglobinopatias/terapia , Globinas beta/genética , Globinas beta/metabolismo , Cromatina , DNA/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA