Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.745
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 180(5): 819, 2020 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-32142671

RESUMO

Sickle cell disease (SCD) is caused by a point mutation in the ß-globin gene that creates hemoglobin S (HbS). Upon deoxygenation, HbS forms long polymers that distort the shape of red blood cells, causing hemolysis and vaso-occlusion. Voxelotor inhibits HbS polymerization, the root cause of SCD complications. To view this Bench to Bedside, open or download the PDF.


Assuntos
Anemia Falciforme/genética , Benzaldeídos/uso terapêutico , Hemoglobina Falciforme/antagonistas & inibidores , Pirazinas/uso terapêutico , Pirazóis/uso terapêutico , Globinas beta/genética , Anemia Falciforme/epidemiologia , Hemoglobina Falciforme/genética , Humanos , Mutação Puntual/genética , Polimerização/efeitos dos fármacos
2.
Cell ; 173(2): 430-442.e17, 2018 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-29606353

RESUMO

Fetal hemoglobin (HbF, α2γ2) level is genetically controlled and modifies severity of adult hemoglobin (HbA, α2ß2) disorders, sickle cell disease, and ß-thalassemia. Common genetic variation affects expression of BCL11A, a regulator of HbF silencing. To uncover how BCL11A supports the developmental switch from γ- to ß- globin, we use a functional assay and protein binding microarray to establish a requirement for a zinc-finger cluster in BCL11A in repression and identify a preferred DNA recognition sequence. This motif appears in embryonic and fetal-expressed globin promoters and is duplicated in γ-globin promoters. The more distal of the duplicated motifs is mutated in individuals with hereditary persistence of HbF. Using the CUT&RUN approach to map protein binding sites in erythroid cells, we demonstrate BCL11A occupancy preferentially at the distal motif, which can be disrupted by editing the promoter. Our findings reveal that direct γ-globin gene promoter repression by BCL11A underlies hemoglobin switching.


Assuntos
Proteínas de Transporte/metabolismo , Hemoglobina Fetal/genética , Proteínas Nucleares/metabolismo , Sequência de Bases , Sítios de Ligação , Proteínas de Transporte/genética , Linhagem Celular , Cromatina/metabolismo , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Células Eritroides/citologia , Células Eritroides/metabolismo , Edição de Genes , Humanos , Proteínas Nucleares/genética , Regiões Promotoras Genéticas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Repressoras , Dedos de Zinco/genética , Globinas beta/genética , Talassemia beta/genética , Talassemia beta/patologia , gama-Globinas/genética
3.
Annu Rev Biochem ; 86: 461-484, 2017 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-28654322

RESUMO

Self-catalyzed DNA depurination is a sequence-specific physiological mechanism mediated by spontaneous extrusion of a stem-loop catalytic intermediate. Hydrolysis of the 5'G residue of the 5'GA/TGG loop and of the first 5'A residue of the 5'GAGA loop, together with particular first stem base pairs, specifies their hydrolysis without involving protein, cofactor, or cation. As such, this mechanism is the only known DNA catalytic activity exploited by nature. The consensus sequences for self-depurination of such G- and A-loop residues occur in all genomes examined across the phyla, averaging one site every 2,000-4,000 base pairs. Because apurinic sites are subject to error-prone repair, leading to substitution and short frameshift mutations, they are both a source of genome damage and a means for creating sequence diversity. Their marked overrepresentation in genomes, and largely unchanging density from the lowest to the highest organisms, indicate their selection over the course of evolution. The mutagenicity at such sites in many human genes is associated with loss of function of key proteins responsible for diverse diseases.


Assuntos
Adenina/metabolismo , Síndrome de Bloom/genética , DNA Catalítico/genética , Guanina/metabolismo , Polimorfismo Genético , Síndrome de Werner/genética , Evolução Biológica , Síndrome de Bloom/metabolismo , Síndrome de Bloom/patologia , Catálise , Reparo do DNA , DNA Catalítico/metabolismo , DNA Cruciforme/genética , DNA Cruciforme/metabolismo , DNA de Cadeia Simples/genética , DNA de Cadeia Simples/metabolismo , Humanos , Hidrólise , Sequências Repetidas Invertidas , Mutação , Síndrome de Werner/metabolismo , Síndrome de Werner/patologia , Globinas beta/genética , Globinas beta/metabolismo
4.
Cell ; 170(5): 1028-1043.e19, 2017 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-28841410

RESUMO

Cis-regulatory elements (CREs) are commonly recognized by correlative chromatin features, yet the molecular composition of the vast majority of CREs in chromatin remains unknown. Here, we describe a CRISPR affinity purification in situ of regulatory elements (CAPTURE) approach to unbiasedly identify locus-specific chromatin-regulating protein complexes and long-range DNA interactions. Using an in vivo biotinylated nuclease-deficient Cas9 protein and sequence-specific guide RNAs, we show high-resolution and selective isolation of chromatin interactions at a single-copy genomic locus. Purification of human telomeres using CAPTURE identifies known and new telomeric factors. In situ capture of individual constituents of the enhancer cluster controlling human ß-globin genes establishes evidence for composition-based hierarchical organization. Furthermore, unbiased analysis of chromatin interactions at disease-associated cis-elements and developmentally regulated super-enhancers reveals spatial features that causally control gene transcription. Thus, comprehensive and unbiased analysis of locus-specific regulatory composition provides mechanistic insight into genome structure and function in development and disease.


Assuntos
Sistemas CRISPR-Cas , Endonucleases/metabolismo , Técnicas Genéticas , Elementos Reguladores de Transcrição , Animais , Biotinilação , Células Cultivadas , Células-Tronco Embrionárias/metabolismo , Endonucleases/genética , Elementos Facilitadores Genéticos , Humanos , Células K562 , Camundongos , RNA Guia de Cinetoplastídeos/metabolismo , Telômero/metabolismo , Globinas beta/genética
5.
Cell ; 163(5): 1124-1137, 2015 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-26582132

RESUMO

In activated B lymphocytes, AID initiates antibody variable (V) exon somatic hypermutation (SHM) for affinity maturation in germinal centers (GCs) and IgH switch (S) region DNA breaks (DSBs) for class-switch recombination (CSR). To resolve long-standing questions, we have developed an in vivo assay to study AID targeting of passenger sequences replacing a V exon. First, we find AID targets SHM hotspots within V exon and S region passengers at similar frequencies and that the normal SHM process frequently generates deletions, indicating that SHM and CSR employ the same mechanism. Second, AID mutates targets in diverse non-Ig passengers in GC B cells at levels similar to those of V exons, definitively establishing the V exon location as "privileged" for SHM. Finally, Peyer's patch GC B cells generate a reservoir of V exons that are highly mutated before selection for affinity maturation. We discuss the implications of these findings for harnessing antibody diversification mechanisms.


Assuntos
Linfócitos B/metabolismo , Citidina Desaminase/genética , Switching de Imunoglobulina , Hipermutação Somática de Imunoglobulina , Recombinação V(D)J , Animais , Humanos , Camundongos , Mutação , Globinas beta/genética
6.
Cell ; 162(4): 900-10, 2015 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-26276636

RESUMO

CTCF and the associated cohesin complex play a central role in insulator function and higher-order chromatin organization of mammalian genomes. Recent studies identified a correlation between the orientation of CTCF-binding sites (CBSs) and chromatin loops. To test the functional significance of this observation, we combined CRISPR/Cas9-based genomic-DNA-fragment editing with chromosome-conformation-capture experiments to show that the location and relative orientations of CBSs determine the specificity of long-range chromatin looping in mammalian genomes, using protocadherin (Pcdh) and ß-globin as model genes. Inversion of CBS elements within the Pcdh enhancer reconfigures the topology of chromatin loops between the distal enhancer and target promoters and alters gene-expression patterns. Thus, although enhancers can function in an orientation-independent manner in reporter assays, in the native chromosome context, the orientation of at least some enhancers carrying CBSs can determine both the architecture of topological chromatin domains and enhancer/promoter specificity. These findings reveal how 3D chromosome architecture can be encoded by linear genome sequences.


Assuntos
Cromossomos/metabolismo , Técnicas Genéticas , Proteínas Repressoras/metabolismo , Animais , Sítios de Ligação , Fator de Ligação a CCCTC , Caderinas/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Cromossomos/química , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , DNA/química , Elementos Facilitadores Genéticos , Expressão Gênica , Genoma Humano , Humanos , Células K562 , Camundongos , Regiões Promotoras Genéticas , Globinas beta/genética , Coesinas
7.
Cell ; 158(4): 849-860, 2014 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-25126789

RESUMO

Distal enhancers commonly contact target promoters via chromatin looping. In erythroid cells, the locus control region (LCR) contacts ß-type globin genes in a developmental stage-specific manner to stimulate transcription. Previously, we induced LCR-promoter looping by tethering the self-association domain (SA) of Ldb1 to the ß-globin promoter via artificial zinc fingers. Here, we show that targeting the SA to a developmentally silenced embryonic globin gene in adult murine erythroblasts triggers its transcriptional reactivation. This activity depends on the LCR, consistent with an LCR-promoter looping mechanism. Strikingly, targeting the SA to the fetal γ-globin promoter in primary adult human erythroblasts increases γ-globin promoter-LCR contacts, stimulating transcription to approximately 85% of total ß-globin synthesis, with a reciprocal reduction in adult ß-globin expression. Our findings demonstrate that forced chromatin looping can override a stringent developmental gene expression program and suggest a novel approach to control the balance of globin gene transcription for therapeutic applications.


Assuntos
Cromatina/metabolismo , Hemoglobina Fetal/genética , Técnicas Genéticas , Região de Controle de Locus Gênico , Ativação Transcricional , Globinas beta/genética , Animais , Antígenos CD34/metabolismo , Cromatina/química , Embrião de Mamíferos/metabolismo , Eritroblastos/metabolismo , Hemoglobinopatias/genética , Hemoglobinopatias/terapia , Humanos , Camundongos , Cultura Primária de Células
8.
Mol Cell ; 81(5): 998-1012.e7, 2021 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-33440169

RESUMO

Pre-mRNA processing steps are tightly coordinated with transcription in many organisms. To determine how co-transcriptional splicing is integrated with transcription elongation and 3' end formation in mammalian cells, we performed long-read sequencing of individual nascent RNAs and precision run-on sequencing (PRO-seq) during mouse erythropoiesis. Splicing was not accompanied by transcriptional pausing and was detected when RNA polymerase II (Pol II) was within 75-300 nucleotides of 3' splice sites (3'SSs), often during transcription of the downstream exon. Interestingly, several hundred introns displayed abundant splicing intermediates, suggesting that splicing delays can take place between the two catalytic steps. Overall, splicing efficiencies were correlated among introns within the same transcript, and intron retention was associated with inefficient 3' end cleavage. Remarkably, a thalassemia patient-derived mutation introducing a cryptic 3'SS improved both splicing and 3' end cleavage of individual ß-globin transcripts, demonstrating functional coupling between the two co-transcriptional processes as a determinant of productive gene output.


Assuntos
Células Eritroides/metabolismo , Eritropoese/genética , RNA Polimerase II/genética , Splicing de RNA , Elongação da Transcrição Genética , Globinas beta/genética , Animais , Sequência de Bases , Diferenciação Celular , Linhagem Celular Tumoral , Células Eritroides/citologia , Éxons , Humanos , Íntrons , Leucócitos/citologia , Leucócitos/metabolismo , Camundongos , Mutação , Clivagem do RNA , RNA Polimerase II/metabolismo , Sítios de Splice de RNA , Spliceossomos/genética , Spliceossomos/metabolismo , Globinas beta/deficiência , Talassemia beta/genética , Talassemia beta/metabolismo , Talassemia beta/patologia
9.
Mol Cell ; 81(5): 983-997.e7, 2021 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-33539786

RESUMO

Gene transcription occurs via a cycle of linked events, including initiation, promoter-proximal pausing, and elongation of RNA polymerase II (Pol II). A key question is how transcriptional enhancers influence these events to control gene expression. Here, we present an approach that evaluates the level and change in promoter-proximal transcription (initiation and pausing) in the context of differential gene expression, genome-wide. This combinatorial approach shows that in primary cells, control of gene expression during differentiation is achieved predominantly via changes in transcription initiation rather than via release of Pol II pausing. Using genetically engineered mouse models, deleted for functionally validated enhancers of the α- and ß-globin loci, we confirm that these elements regulate Pol II recruitment and/or initiation to modulate gene expression. Together, our data show that gene expression during differentiation is regulated predominantly at the level of initiation and that enhancers are key effectors of this process.


Assuntos
Elementos Facilitadores Genéticos , Regiões Promotoras Genéticas , RNA Polimerase II/genética , Iniciação da Transcrição Genética , alfa-Globinas/genética , Globinas beta/genética , Animais , Diferenciação Celular , Éxons , Feto , Regulação da Expressão Gênica , Biblioteca Gênica , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Íntrons , Células K562 , Fígado/citologia , Fígado/metabolismo , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , RNA Polimerase II/metabolismo , Transdução de Sinais , alfa-Globinas/deficiência , Globinas beta/deficiência
10.
Annu Rev Cell Dev Biol ; 30: 1-21, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25288111

RESUMO

In this perspective I look back on the twists and turns that influenced the direction of my scientific career over the past 40 years. From my early ambition to be a chemist to my training in Philadelphia and Bethesda as a molecular biologist, I benefited enormously from generous and valuable mentoring. In my independent career in Philadelphia and Princeton, I was motivated by a keen interest in the changes in gene expression that direct the development of the mammalian embryo and inspired by the creativity and energy of my students, fellows, and research staff. After twelve years as President of Princeton University, I have happily returned to the faculty of the Department of Molecular Biology.


Assuntos
Biologia Molecular/história , Universidades/história , Sequência de Aminoácidos , Animais , Canadá , Passeio de Cromossomo , Desenvolvimento Embrionário/genética , Proteínas do Olho/genética , Proteínas do Olho/história , Regulação da Expressão Gênica no Desenvolvimento , Impressão Genômica , História do Século XX , História do Século XXI , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/história , Humanos , Camundongos , Dados de Sequência Molecular , National Institutes of Health (U.S.) , New Jersey , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/genética , Fatores de Transcrição Box Pareados/história , Splicing de RNA , RNA Longo não Codificante/genética , RNA Longo não Codificante/história , Proteínas Repressoras/genética , Proteínas Repressoras/história , Estados Unidos , alfa-Fetoproteínas/genética , alfa-Fetoproteínas/história , Globinas beta/genética , Globinas beta/história
11.
Cell ; 149(6): 1233-44, 2012 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-22682246

RESUMO

Chromatin loops juxtapose distal enhancers with active promoters, but their molecular architecture and relationship with transcription remain unclear. In erythroid cells, the locus control region (LCR) and ß-globin promoter form a chromatin loop that requires transcription factor GATA1 and the associated molecule Ldb1. We employed artificial zinc fingers (ZF) to tether Ldb1 to the ß-globin promoter in GATA1 null erythroblasts, in which the ß-globin locus is relaxed and inactive. Remarkably, targeting Ldb1 or only its self-association domain to the ß-globin promoter substantially activated ß-globin transcription in the absence of GATA1. Promoter-tethered Ldb1 interacted with endogenous Ldb1 complexes at the LCR to form a chromatin loop, causing recruitment and phosphorylation of RNA polymerase II. ZF-Ldb1 proteins were inactive at alleles lacking the LCR, demonstrating that their activities depend on long-range interactions. Our findings establish Ldb1 as a critical effector of GATA1-mediated loop formation and indicate that chromatin looping causally underlies gene regulation.


Assuntos
Cromatina/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas com Domínio LIM/metabolismo , Transcrição Gênica , Globinas beta/genética , Animais , Linhagem Celular , Separação Celular , Proteínas de Ligação a DNA/química , Embrião de Mamíferos/citologia , Eritroblastos/metabolismo , Feminino , Fator de Transcrição GATA1/metabolismo , Regulação da Expressão Gênica , Proteínas com Domínio LIM/química , Masculino , Camundongos , Regiões Promotoras Genéticas , Estrutura Terciária de Proteína , Dedos de Zinco
12.
Nature ; 595(7866): 295-302, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34079130

RESUMO

Sickle cell disease (SCD) is caused by a mutation in the ß-globin gene HBB1. We used a custom adenine base editor (ABE8e-NRCH)2,3 to convert the SCD allele (HBBS) into Makassar ß-globin (HBBG), a non-pathogenic variant4,5. Ex vivo delivery of mRNA encoding the base editor with a targeting guide RNA into haematopoietic stem and progenitor cells (HSPCs) from patients with SCD resulted in 80% conversion of HBBS to HBBG. Sixteen weeks after transplantation of edited human HSPCs into immunodeficient mice, the frequency of HBBG was 68% and hypoxia-induced sickling of bone marrow reticulocytes had decreased fivefold, indicating durable gene editing. To assess the physiological effects of HBBS base editing, we delivered ABE8e-NRCH and guide RNA into HSPCs from a humanized SCD mouse6 and then transplanted these cells into irradiated mice. After sixteen weeks, Makassar ß-globin represented 79% of ß-globin protein in blood, and hypoxia-induced sickling was reduced threefold. Mice that received base-edited HSPCs showed near-normal haematological parameters and reduced splenic pathology compared to mice that received unedited cells. Secondary transplantation of edited bone marrow confirmed that the gene editing was durable in long-term haematopoietic stem cells and showed that HBBS-to-HBBG editing of 20% or more is sufficient for phenotypic rescue. Base editing of human HSPCs avoided the p53 activation and larger deletions that have been observed following Cas9 nuclease treatment. These findings point towards a one-time autologous treatment for SCD that eliminates pathogenic HBBS, generates benign HBBG, and minimizes the undesired consequences of double-strand DNA breaks.


Assuntos
Adenina/metabolismo , Anemia Falciforme/genética , Anemia Falciforme/terapia , Edição de Genes , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Globinas beta/genética , Animais , Antígenos CD34/metabolismo , Proteína 9 Associada à CRISPR/metabolismo , Modelos Animais de Doenças , Feminino , Terapia Genética , Genoma Humano/genética , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/patologia , Humanos , Masculino , Camundongos
13.
Blood ; 143(19): 1980-1991, 2024 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-38364109

RESUMO

ABSTRACT: The switch from fetal hemoglobin (γ-globin, HBG) to adult hemoglobin (ß-globin, HBB) gene transcription in erythroid cells serves as a paradigm for a complex and clinically relevant developmental gene regulatory program. We previously identified HIC2 as a regulator of the switch by inhibiting the transcription of BCL11A, a key repressor of HBG production. HIC2 is highly expressed in fetal cells, but the mechanism of its regulation is unclear. Here we report that HIC2 developmental expression is controlled by microRNAs (miRNAs), as loss of global miRNA biogenesis through DICER1 depletion leads to upregulation of HIC2 and HBG messenger RNA. We identified the adult-expressed let-7 miRNA family as a direct posttranscriptional regulator of HIC2. Ectopic expression of let-7 in fetal cells lowered HIC2 levels, whereas inhibition of let-7 in adult erythroblasts increased HIC2 production, culminating in decommissioning of a BCL11A erythroid enhancer and reduced BCL11A transcription. HIC2 depletion in let-7-inhibited cells restored BCL11A-mediated repression of HBG. Together, these data establish that fetal hemoglobin silencing in adult erythroid cells is under the control of a miRNA-mediated inhibitory pathway (let-7 ⊣ HIC2 ⊣ BCL11A ⊣ HBG).


Assuntos
Hemoglobina Fetal , Fatores de Transcrição Kruppel-Like , MicroRNAs , Proteínas Repressoras , Humanos , Globinas beta/genética , Globinas beta/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/metabolismo , Eritroblastos/metabolismo , Eritroblastos/citologia , Hemoglobina Fetal/genética , Hemoglobina Fetal/metabolismo , gama-Globinas/genética , gama-Globinas/metabolismo , Regulação da Expressão Gênica , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Ribonuclease III/genética , Ribonuclease III/metabolismo , Transcrição Gênica
14.
Nature ; 578(7794): 229-236, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32051598

RESUMO

Genome editing, which involves the precise manipulation of cellular DNA sequences to alter cell fates and organism traits, has the potential to both improve our understanding of human genetics and cure genetic disease. Here I discuss the scientific, technical and ethical aspects of using CRISPR (clustered regularly interspaced short palindromic repeats) technology for therapeutic applications in humans, focusing on specific examples that highlight both opportunities and challenges. Genome editing is-or will soon be-in the clinic for several diseases, with more applications under development. The rapid pace of the field demands active efforts to ensure that this breakthrough technology is used responsibly to treat, cure and prevent genetic disease.


Assuntos
Anemia Falciforme/genética , Anemia Falciforme/terapia , Edição de Genes/métodos , Edição de Genes/tendências , Genoma Humano/genética , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Sistemas CRISPR-Cas/genética , Edição de Genes/ética , Edição de Genes/normas , Mutação em Linhagem Germinativa/genética , Humanos , Especificidade de Órgãos/genética , Segurança do Paciente , Globinas beta/genética
15.
N Engl J Med ; 386(7): 617-628, 2022 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-34898139

RESUMO

BACKGROUND: Sickle cell disease is characterized by the painful recurrence of vaso-occlusive events. Gene therapy with the use of LentiGlobin for sickle cell disease (bb1111; lovotibeglogene autotemcel) consists of autologous transplantation of hematopoietic stem and progenitor cells transduced with the BB305 lentiviral vector encoding a modified ß-globin gene, which produces an antisickling hemoglobin, HbAT87Q. METHODS: In this ongoing phase 1-2 study, we optimized the treatment process in the initial 7 patients in Group A and 2 patients in Group B with sickle cell disease. Group C was established for the pivotal evaluation of LentiGlobin for sickle cell disease, and we adopted a more stringent inclusion criterion that required a minimum of four severe vaso-occlusive events in the 24 months before enrollment. In this unprespecified interim analysis, we evaluated the safety and efficacy of LentiGlobin in 35 patients enrolled in Group C. Included in this analysis was the number of severe vaso-occlusive events after LentiGlobin infusion among patients with at least four vaso-occlusive events in the 24 months before enrollment and with at least 6 months of follow-up. RESULTS: As of February 2021, cell collection had been initiated in 43 patients in Group C; 35 received a LentiGlobin infusion, with a median follow-up of 17.3 months (range, 3.7 to 37.6). Engraftment occurred in all 35 patients. The median total hemoglobin level increased from 8.5 g per deciliter at baseline to 11 g or more per deciliter from 6 months through 36 months after infusion. HbAT87Q contributed at least 40% of total hemoglobin and was distributed across a mean (±SD) of 85±8% of red cells. Hemolysis markers were reduced. Among the 25 patients who could be evaluated, all had resolution of severe vaso-occlusive events, as compared with a median of 3.5 events per year (range, 2.0 to 13.5) in the 24 months before enrollment. Three patients had a nonserious adverse event related or possibly related to LentiGlobin that resolved within 1 week after onset. No cases of hematologic cancer were observed during up to 37.6 months of follow-up. CONCLUSIONS: One-time treatment with LentiGlobin resulted in sustained production of HbAT87Q in most red cells, leading to reduced hemolysis and complete resolution of severe vaso-occlusive events. (Funded by Bluebird Bio; HGB-206 ClinicalTrials.gov number, NCT02140554.).


Assuntos
Anemia Falciforme/terapia , Terapia Genética , Vetores Genéticos , Transplante de Células-Tronco Hematopoéticas , Hemoglobinas/genética , Lentivirus , Transplante de Células-Tronco , Globinas beta/genética , Adolescente , Adulto , Anemia Falciforme/sangue , Anemia Falciforme/complicações , Criança , Feminino , Hemoglobina Fetal , Hemoglobinas/análise , Hemoglobinas/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Grau de Desobstrução Vascular , Adulto Jovem
16.
N Engl J Med ; 386(2): 138-147, 2022 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-34898140

RESUMO

Gene therapy with LentiGlobin for sickle cell disease (bb1111, lovotibeglogene autotemcel) consists of autologous transplantation of a patient's hematopoietic stem cells transduced with the BB305 lentiviral vector that encodes the ßA-T87Q-globin gene. Acute myeloid leukemia developed in a woman approximately 5.5 years after she had received LentiGlobin for sickle cell disease as part of the initial cohort (Group A) of the HGB-206 study. An analysis of peripheral-blood samples revealed that blast cells contained a BB305 lentiviral vector insertion site. The results of an investigation of causality indicated that the leukemia was unlikely to be related to vector insertion, given the location of the insertion site, the very low transgene expression in blast cells, and the lack of an effect on expression of surrounding genes. Several somatic mutations predisposing to acute myeloid leukemia were present after diagnosis, which suggests that patients with sickle cell disease are at increased risk for hematologic malignant conditions after transplantation, most likely because of a combination of risks associated with underlying sickle cell disease, transplantation procedure, and inadequate disease control after treatment. (Funded by Bluebird Bio.).


Assuntos
Anemia Falciforme/terapia , Expressão Gênica , Terapia Genética/efeitos adversos , Transplante de Células-Tronco Hematopoéticas , Leucemia Mieloide Aguda/etiologia , Globinas beta/genética , Adulto , Anemia Falciforme/complicações , Anemia Falciforme/genética , Carcinogênese , Feminino , Vetores Genéticos , Humanos , Lentivirus , Fatores de Risco , Análise de Sequência de RNA , Transgenes , Transplante Autólogo
17.
N Engl J Med ; 386(5): 415-427, 2022 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-34891223

RESUMO

BACKGROUND: Betibeglogene autotemcel (beti-cel) gene therapy for transfusion-dependent ß-thalassemia contains autologous CD34+ hematopoietic stem cells and progenitor cells transduced with the BB305 lentiviral vector encoding the ß-globin (ßA-T87Q) gene. METHODS: In this open-label, phase 3 study, we evaluated the efficacy and safety of beti-cel in adult and pediatric patients with transfusion-dependent ß-thalassemia and a non-ß0/ß0 genotype. Patients underwent myeloablation with busulfan (with doses adjusted on the basis of pharmacokinetic analysis) and received beti-cel intravenously. The primary end point was transfusion independence (i.e., a weighted average hemoglobin level of ≥9 g per deciliter without red-cell transfusions for ≥12 months). RESULTS: A total of 23 patients were enrolled and received treatment, with a median follow-up of 29.5 months (range, 13.0 to 48.2). Transfusion independence occurred in 20 of 22 patients who could be evaluated (91%), including 6 of 7 patients (86%) who were younger than 12 years of age. The average hemoglobin level during transfusion independence was 11.7 g per deciliter (range, 9.5 to 12.8). Twelve months after beti-cel infusion, the median level of gene therapy-derived adult hemoglobin (HbA) with a T87Q amino acid substitution (HbAT87Q) was 8.7 g per deciliter (range, 5.2 to 10.6) in patients who had transfusion independence. The safety profile of beti-cel was consistent with that of busulfan-based myeloablation. Four patients had at least one adverse event that was considered by the investigators to be related or possibly related to beti-cel; all events were nonserious except for thrombocytopenia (in 1 patient). No cases of cancer were observed. CONCLUSIONS: Treatment with beti-cel resulted in a sustained HbAT87Q level and a total hemoglobin level that was high enough to enable transfusion independence in most patients with a non-ß0/ß0 genotype, including those younger than 12 years of age. (Funded by Bluebird Bio; HGB-207 ClinicalTrials.gov number, NCT02906202.).


Assuntos
Produtos Biológicos/uso terapêutico , Terapia Genética/métodos , Globinas beta/genética , Talassemia beta/terapia , Adolescente , Adulto , Produtos Biológicos/efeitos adversos , Bussulfano/uso terapêutico , Criança , Transfusão de Eritrócitos/efeitos adversos , Eritropoese , Feminino , Vetores Genéticos , Genótipo , Hemoglobinas/análise , Humanos , Sobrecarga de Ferro/prevenção & controle , Lentivirus/genética , Masculino , Pessoa de Meia-Idade , Agonistas Mieloablativos/uso terapêutico , Talassemia beta/sangue , Talassemia beta/genética
18.
Genome Res ; 32(3): 488-498, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35031571

RESUMO

Although it is known that the mutation rate varies across the genome, previous estimates were based on averaging across various numbers of positions. Here, we describe a method to measure the origination rates of target mutations at target base positions and apply it to a 6-bp region in the human hemoglobin subunit beta (HBB) gene and to the identical, paralogous hemoglobin subunit delta (HBD) region in sperm cells from both African and European donors. The HBB region of interest (ROI) includes the site of the hemoglobin S (HbS) mutation, which protects against malaria, is common in Africa, and has served as a classic example of adaptation by random mutation and natural selection. We found a significant correspondence between de novo mutation rates and past observations of alleles in carriers, showing that mutation rates vary substantially in a mutation-specific manner that contributes to the site frequency spectrum. We also found that the overall point mutation rate is significantly higher in Africans than in Europeans in the HBB region studied. Finally, the rate of the 20A→T mutation, called the "HbS mutation" when it appears in HBB, is significantly higher than expected from the genome-wide average for this mutation type. Nine instances were observed in the African HBB ROI, where it is of adaptive significance, representing at least three independent originations; no instances were observed elsewhere. Further studies will be needed to examine mutation rates at the single-mutation resolution across these and other loci and organisms and to uncover the molecular mechanisms responsible.


Assuntos
Globinas beta , Talassemia beta , Heterozigoto , Humanos , Mutação , Taxa de Mutação , Globinas beta/genética , Talassemia beta/genética
19.
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
20.
Blood ; 142(25): 2198-2215, 2023 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-37738561

RESUMO

ABSTRACT: Regulation of RNA polymerase II (RNAPII) activity is an essential process that governs gene expression; however, its contribution to the fundamental process of erythropoiesis remains unclear. hexamethylene bis-acetamide inducible 1 (HEXIM1) regulates RNAPII activity by controlling the location and activity of positive transcription factor ß. We identified a key role for HEXIM1 in controlling erythroid gene expression and function, with overexpression of HEXIM1 promoting erythroid proliferation and fetal globin expression. HEXIM1 regulated erythroid proliferation by enforcing RNAPII pausing at cell cycle check point genes and increasing RNAPII occupancy at genes that promote cycle progression. Genome-wide profiling of HEXIM1 revealed that it was increased at both repressed and activated genes. Surprisingly, there were also genome-wide changes in the distribution of GATA-binding factor 1 (GATA1) and RNAPII. The most dramatic changes occurred at the ß-globin loci, where there was loss of RNAPII and GATA1 at ß-globin and gain of these factors at γ-globin. This resulted in increased expression of fetal globin, and BGLT3, a long noncoding RNA in the ß-globin locus that regulates fetal globin expression. GATA1 was a key determinant of the ability of HEXIM1 to repress or activate gene expression. Genes that gained both HEXIM1 and GATA1 had increased RNAPII and increased gene expression, whereas genes that gained HEXIM1 but lost GATA1 had an increase in RNAPII pausing and decreased expression. Together, our findings reveal a central role for universal transcription machinery in regulating key aspects of erythropoiesis, including cell cycle progression and fetal gene expression, which could be exploited for therapeutic benefit.


Assuntos
Eritropoese , Fatores de Transcrição , Humanos , Eritropoese/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Regulação da Expressão Gênica , Transcrição Gênica , Globinas beta/genética , Globinas beta/metabolismo , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , Fator de Transcrição GATA1/genética , Fator de Transcrição GATA1/metabolismo , Proteínas de Ligação a RNA/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA