β-hemoglobinopathies

β - 血红蛋白病
  • 文章类型: Journal Article
    基因治疗在治疗各种疾病方面显示出巨大的潜力,特别是遗传性血液疾病,如血友病,镰状细胞病,和地中海贫血。在理解疾病相关基因的调控网络方面的进展已经导致了对其他治疗靶点的识别。尤其是β-血红蛋白病。红系调节因子BCL11A为β-血红蛋白病提供了最有希望的治疗靶标,并且使用商业化的基因治疗产品Casgevy在2023年被批准在英国和美国使用。值得注意的是,创新基因编辑技术的出现进一步拓宽了基因治疗领域,为治疗提供新的可能性。深入研究表明,基础编辑和主要编辑,基于CRISPR技术,能够在造血干细胞中进行精确的单碱基修饰,以解决离体和体内遗传性血液疾病。在这次审查中,我们概述了基因疗法的现状,专注于遗传性血液疾病的临床研究和基因治疗产品,潜在基因靶标的评估,以及目前基因治疗实践中使用的基因编辑工具,这为未来针对更广泛的疾病建立更安全、更有效的基因治疗方法提供了启示。
    Gene therapy has shown significant potential in treating various diseases, particularly inherited blood disorders such as hemophilia, sickle cell disease, and thalassemia. Advances in understanding the regulatory network of disease-associated genes have led to the identification of additional therapeutic targets for treatment, especially for β-hemoglobinopathies. Erythroid regulatory factor BCL11A offers the most promising therapeutic target for β-hemoglobinopathies and reduction of its expression using the commercialized gene therapy product Casgevy was approved for use in the UK and USA in 2023. Notably, the emergence of innovative gene editing technologies has further broadened the gene therapy landscape, presenting new possibilities for treatment. Intensive studies indicate that base editing and prime editing, built upon CRISPR technology, enable precise single-base modification in hematopoietic stem cells for addressing inherited blood disorders ex vivo and in vivo. In this review, we present an overview of the current landscape of gene therapies, focusing on clinical research and gene therapy products for inherited blood disorders, evaluation of potential gene targets, and the gene editing tools employed in current gene therapy practices, which provides an insight for the establishment of safer and more effective gene therapy methods for a wider range of diseases in the future.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    近年来,成簇的规则间隔短回文重复序列(CRISPR)和CRISPR相关(Cas)蛋白已成为一种革命性的基因编辑工具,用于治疗影响不同器官系统的遗传性疾病。比如血液和肌肉。血液和神经肌肉遗传疾病都受益于基因组编辑方法,但在临床翻译中面临不同的挑战。CRISPR/Cas9技术离体修饰造血干细胞的能力极大地加速了血液疾病遗传疗法的发展。在过去的十年里,许多临床试验已经启动,目前正在取得令人鼓舞的结果.最近FDA批准了Casgevy,第一个基于CRISPR/Cas9的药物用于治疗严重镰状细胞病和输血依赖性β-地中海贫血,代表了该领域的重要里程碑,并突出了该技术的巨大潜力。目前,类似的临床前努力正在将CRISPR疗法扩展到其他血液系统疾病,例如原发性免疫缺陷。在神经肌肉领域,CRISPR/Cas9的多功能性有助于产生杜氏肌营养不良症(DMD)的新细胞和动物模型,提供创新平台,以加快治疗解决方案的临床前开发。已经提出了使用CRISPR工具箱遗传恢复肌营养不良蛋白产生的几种纠正性干预措施,并在不同的DMD动物模型中显示了有希望的结果。尽管这些进展代表了CRISPR/Cas9疗法临床转化为DMD的重要一步,还有许多障碍需要克服,例如与高病毒载体剂量相关的体内递送方法,以及安全性和免疫学问题。总的来说,在血液和神经肌肉领域获得的结果强调了CRISPR/Cas9对受这些衰弱状况影响的患者的转化作用.由于每个领域都面临着不同和具体的挑战,CRISPR疗法的临床转化可能因遗传疾病的不同而有差异.正在进行的调查和临床试验将解决这些疗法的风险和局限性,包括长期疗效,潜在的遗传毒性,和不良免疫反应。这篇综述提供了基于CRISPR的技术在临床前和临床环境中对单基因血液病和肌营养不良的不同应用的见解,并比较了这两个领域的进展,同时突出了当前的趋势。困难,和需要克服的挑战。
    In recent years, clustered regularly interspaced short palindromic repeats (CRISPRs) and CRISPR-associated (Cas) protein have emerged as a revolutionary gene editing tool to treat inherited disorders affecting different organ systems, such as blood and muscles. Both hematological and neuromuscular genetic disorders benefit from genome editing approaches but face different challenges in their clinical translation. The ability of CRISPR/Cas9 technologies to modify hematopoietic stem cells ex vivo has greatly accelerated the development of genetic therapies for blood disorders. In the last decade, many clinical trials were initiated and are now delivering encouraging results. The recent FDA approval of Casgevy, the first CRISPR/Cas9-based drug for severe sickle cell disease and transfusion-dependent β-thalassemia, represents a significant milestone in the field and highlights the great potential of this technology. Similar preclinical efforts are currently expanding CRISPR therapies to other hematologic disorders such as primary immunodeficiencies. In the neuromuscular field, the versatility of CRISPR/Cas9 has been instrumental for the generation of new cellular and animal models of Duchenne muscular dystrophy (DMD), offering innovative platforms to speed up preclinical development of therapeutic solutions. Several corrective interventions have been proposed to genetically restore dystrophin production using the CRISPR toolbox and have demonstrated promising results in different DMD animal models. Although these advances represent a significant step forward to the clinical translation of CRISPR/Cas9 therapies to DMD, there are still many hurdles to overcome, such as in vivo delivery methods associated with high viral vector doses, together with safety and immunological concerns. Collectively, the results obtained in the hematological and neuromuscular fields emphasize the transformative impact of CRISPR/Cas9 for patients affected by these debilitating conditions. As each field suffers from different and specific challenges, the clinical translation of CRISPR therapies may progress differentially depending on the genetic disorder. Ongoing investigations and clinical trials will address risks and limitations of these therapies, including long-term efficacy, potential genotoxicity, and adverse immune reactions. This review provides insights into the diverse applications of CRISPR-based technologies in both preclinical and clinical settings for monogenic blood disorders and muscular dystrophy and compare advances in both fields while highlighting current trends, difficulties, and challenges to overcome.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    β-血红蛋白病如β-地中海贫血(BT)和镰状细胞病(SCD)是遗传性单基因血液病,具有显著的全球负担。因此,由于缺乏有效的治疗方法,早期和负担得起的诊断可以减轻发病率并降低死亡率。目前,Sanger测序被认为是BT和SCD的黄金标准基因测试,但它的通量非常低,需要多个扩增子和更多的测序反应来覆盖整个HBB基因。为了解决这个问题,我们已经证明了一种基于无提取单扩增子的方法,该方法使用可扩展的非侵入性基于扩增子的精确测序(SNAPseq)测定法和下一代测序(NGS)催化,用临床样本筛选整个β-珠蛋白基因.我们使用非侵入性颊拭子样品和简单的手指刺血优化了测定,以直接扩增粗裂解物。SNAPseq显示出高灵敏度和特异性,与Sanger测序有100%的一致性.此外,为了促进无缝报告,我们根据ACMG和AMP指南对变异进行系统分类后,通过数据整合,创建了一个更简单的自动化流程,为BT和SCD的致病性突变提供了全面的资源.据我们所知,这是基于NGS的高通量SNAPseq方法的第一份报告,该方法用于在自动化流水线中以高灵敏度在单一检测中同时检测BT和SCD.
    β-hemoglobinopathies such as β-thalassemia (BT) and Sickle cell disease (SCD) are inherited monogenic blood disorders with significant global burden. Hence, early and affordable diagnosis can alleviate morbidity and reduce mortality given the lack of effective cure. Currently, Sanger sequencing is considered to be the gold standard genetic test for BT and SCD, but it has a very low throughput requiring multiple amplicons and more sequencing reactions to cover the entire HBB gene. To address this, we have demonstrated an extraction-free single amplicon-based approach for screening the entire β-globin gene with clinical samples using Scalable noninvasive amplicon-based precision sequencing (SNAPseq) assay catalyzing with next-generation sequencing (NGS). We optimized the assay using noninvasive buccal swab samples and simple finger prick blood for direct amplification with crude lysates. SNAPseq demonstrates high sensitivity and specificity, having a 100% agreement with Sanger sequencing. Furthermore, to facilitate seamless reporting, we have created a much simpler automated pipeline with comprehensive resources for pathogenic mutations in BT and SCD through data integration after systematic classification of variants according to ACMG and AMP guidelines. To the best of our knowledge, this is the first report of the NGS-based high throughput SNAPseq approach for the detection of both BT and SCD in a single assay with high sensitivity in an automated pipeline.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    通过抑制调节结构域的破坏重新激活沉默的γ-珠蛋白表达为治疗β-血红蛋白病提供了治疗策略。这里,我们使用了变压器基础编辑器(TBE),最近开发的胞嘧啶碱基编辑器,没有可检测到的脱靶突变,破坏造血干细胞中的转录因子结合基序。通过使用tBE对六个基序进行功能筛选,我们发现,直接破坏HBG1/2启动子中的BCL11A结合基序触发了最高的γ-珠蛋白表达。通过与使用Cas9核酸酶或常规BE(ABE8e和hA3A-BE3)的其他临床和临床前策略的并排比较,我们发现,在HBG1/2启动子处,tBE介导的BCL11A结合基序的破坏触发了健康和β-地中海贫血患者造血干/祖细胞中最高的胎儿血红蛋白,而没有可检测到的DNA或RNA脱靶突变.通过tBE持久的治疗编辑持续在造血干细胞的繁殖中,证明在HBG1/2启动子中tBE介导的编辑是治疗β-血红蛋白病的安全有效的策略。
    Reactivating silenced γ-globin expression through the disruption of repressive regulatory domains offers a therapeutic strategy for treating β-hemoglobinopathies. Here, we used transformer base editor (tBE), a recently developed cytosine base editor with no detectable off-target mutations, to disrupt transcription-factor-binding motifs in hematopoietic stem cells. By performing functional screening of six motifs with tBE, we found that directly disrupting the BCL11A-binding motif in HBG1/2 promoters triggered the highest γ-globin expression. Via a side-by-side comparison with other clinical and preclinical strategies using Cas9 nuclease or conventional BEs (ABE8e and hA3A-BE3), we found that tBE-mediated disruption of the BCL11A-binding motif at the HBG1/2 promoters triggered the highest fetal hemoglobin in healthy and β-thalassemia patient hematopoietic stem/progenitor cells while exhibiting no detectable DNA or RNA off-target mutations. Durable therapeutic editing by tBE persisted in repopulating hematopoietic stem cells, demonstrating that tBE-mediated editing in HBG1/2 promoters is a safe and effective strategy for treating β-hemoglobinopathies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    血红蛋白转换是尚未完全阐明的复杂生物过程。由于HbF对β-地中海贫血和镰状细胞病等疾病进程的积极影响,因此调节胎儿血红蛋白(HbF)表达抑制的机制尤其令人感兴趣。影响全世界无数人健康的遗传性血红蛋白疾病。几种转录因子与HbF的控制有关,其中BCL11A已成为HbF沉默的主要参与者。SOX6也涉及HbF的沉默,并且对于小鼠胚胎血红蛋白的沉默至关重要。BCL11A和SOX6在分化期间共表达并在红细胞区室中物理相互作用。在这项研究中,我们观察到BCL11A敲除通过激活microRNA(miR)-365-3p导致SOX6的转录后下调。通过miR-365-3p的瞬时异位表达或基因编辑下调SOX6激活红系细胞中的胚胎和胎儿β样珠蛋白基因表达。BCL11A和SOX6的同步表达对于血红蛋白转换至关重要。在这项研究中,我们确定了一个BCL11A/miR-365-3p/SOX6进化保守的途径,提供对胚胎和胎儿珠蛋白基因调控的见解,为治疗β-血红蛋白病提供新的靶点。
    Hemoglobin switching is a complex biological process not yet fully elucidated. The mechanism regulating the suppression of fetal hemoglobin (HbF) expression is of particular interest because of the positive impact of HbF on the course of diseases such as β-thalassemia and sickle cell disease, hereditary hemoglobin disorders that affect the health of countless individuals worldwide. Several transcription factors have been implicated in the control of HbF, of which BCL11A has emerged as a major player in HbF silencing. SOX6 has also been implicated in silencing HbF and is critical to the silencing of the mouse embryonic hemoglobins. BCL11A and SOX6 are co-expressed and physically interact in the erythroid compartment during differentiation. In this study, we observe that BCL11A knockout leads to post-transcriptional downregulation of SOX6 through activation of microRNA (miR)-365-3p. Downregulating SOX6 by transient ectopic expression of miR-365-3p or gene editing activates embryonic and fetal β-like globin gene expression in erythroid cells. The synchronized expression of BCL11A and SOX6 is crucial for hemoglobin switching. In this study, we identified a BCL11A/miR-365-3p/SOX6 evolutionarily conserved pathway, providing insights into the regulation of the embryonic and fetal globin genes suggesting new targets for treating β-hemoglobinopathies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    β0-地中海贫血缺失去除5'β-珠蛋白启动子通常呈现具有高血红蛋白(Hb)A2和HbF水平的表型。我们报告了大量具有3.4kb缺失的β0地中海贫血的分子特征和表型基因型相关性。
    总共148个科目,包括127个杂合子,20例HbE-β-地中海贫血患者,和一个α-珠蛋白基因三重复的双杂合子,被招募。进行Hb和DNA分析以鉴定地中海贫血突变和四个高HbF单核苷酸多态性(SNP),包括γ-珠蛋白启动子处的四个碱基对缺失(-AGCA),OR51B6基因的rs5006884,-158Gγ-XmnI,3'Aγ-珠蛋白基因和5'δ-珠蛋白基因之间的BCL11A结合基序(TGGTCA)。
    发现具有3.4kb缺失的杂合β0-地中海贫血和HbE-β0-地中海贫血具有显着更高的Hb,血细胞比容,平均红细胞体积,与其他突变相比,平均红细胞血红蛋白和HbF值。具有3.4kb缺失的杂合β0-地中海贫血和α-地中海贫血的共同遗传与更高的MCV和MCH值相关。HbE-β0-地中海贫血患者具有非输血依赖性地中海贫血表型,无输血时平均Hb约为10g/dL。迄今为止尚未描述的双杂合β0-地中海贫血,具有3.4kb缺失和α-珠蛋白基因三联,表现为普通的β-地中海贫血性状。大多数受试者具有所检查的四个高HbFSNP的野生型序列。在具有和不具有这些SNP的受试者之间没有观察到HbF的显著差异。5'β-珠蛋白启动子的去除可能是这种异常表型的原因。
    结果表明,具有3.4kb缺失的β0-地中海贫血是轻度的β-地中海贫血等位基因。此信息应在遗传咨询和产前地中海贫血诊断中提供。
    β 0-thalassemia deletion removing 5´β-globin promoter usually presents phenotype with high hemoglobin (Hb) A2 and Hb F levels. We report the molecular characteristics and phenotype-genotype correlation in a large cohort of the β 0-thalassemia with 3.4 kb deletion.
    A total of 148 subjects, including 127 heterozygotes, 20 Hb E-β-thalassemia patients, and a double heterozygote with α-globin gene triplication, were recruited. Hb and DNA analysis were performed to identify thalassemia mutations and four high Hb F single nucleotide polymorphisms (SNPs) including four base pair deletion (-AGCA) at A γ-globin promoter, rs5006884 on OR51B6 gene, -158 G γ-XmnI, BCL11A binding motifs (TGGTCA) between 3´A γ-globin gene and 5´δ-globin gene.
    It was found that heterozygous β 0-thalassemia and Hb E-β 0-thalassemia with 3.4 kb deletion had significantly higher Hb, hematocrit, mean corpuscular volume, mean corpuscular hemoglobin and Hb F values as compared with those with other mutations. Co-inheritance of heterozygous β 0-thalassemia with 3.4 kb deletion and α-thalassemia was associated with even higher MCV and MCH values. The Hb E-β 0-thalassemia patients carried a non-transfusion-dependent thalassemia phenotype with an average Hb of around 10 g/dL without blood transfusion. A hitherto undescribed double heterozygous β 0-thalassemia with 3.4 kb deletion and α-globin gene triplication presented as a plain β-thalassemia trait. Most of the subjects had wild-type sequences for the four high Hb F SNPs examined. No significant difference in Hb F was observed between those of subjects with and without these SNPs. Removal of the 5´β-globin promoter may likely be responsible for this unusual phenotype.
    The results indicate that β 0-thalassemia with 3.4 kb deletion is a mild β-thalassemia allele. This information should be provided at genetic counseling and prenatal thalassemia diagnosis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    了解珠蛋白基因的功能和控制的研究导致了20世纪和21世纪一些最令人兴奋的分子发现和生物医学突破。珠蛋白基因基因座的广泛表征,伴随着在人类造血干细胞和祖细胞(HPSC)中利用病毒作为人类基因递送工具的开创性工作,通过自体造血干细胞移植和基因治疗(HSCT-GT)导致了转化和成功的治疗。由于对β-珠蛋白基因簇的深入理解,首先考虑自体HSCT-GT的疾病是两种流行的β-血红蛋白病:镰状细胞病和β-地中海贫血,既影响功能性β-珠蛋白链,又导致大量发病率。两种条件都适用于同种异体HSCT;然而,这种疗法具有严重的风险,并且使用HLA匹配的家庭供体(大多数患者无法使用)最有效,以获得最佳的治疗和安全获益.来自无关或单倍体相同的供体的移植具有更高的风险,尽管它们正在逐步改善。相反,HSCT-GT利用患者自己的HSPC,扩大接触更多患者。据报道,一些基因治疗临床试验已经取得了显著的疾病改善,更多的正在进行中。基于自体HSCT-GT的安全性和治疗成功,2022年,美国食品和药物管理局(FDA)批准了用于β-地中海贫血的HSCT-GT(Zynteglo™)。这篇综述阐明了β-珠蛋白基因的研究历程,面对逆境,和取得的成就;它强调了β-珠蛋白基因座的重要分子和遗传发现,描述了主要的珠蛋白载体,最后描述了镰状细胞病和β-地中海贫血的临床试验的有希望的结果。
    Investigations to understand the function and control of the globin genes have led to some of the most exciting molecular discoveries and biomedical breakthroughs of the 20th and 21st centuries. Extensive characterization of the globin gene locus, accompanied by pioneering work on the utilization of viruses as human gene delivery tools in human hematopoietic stem and progenitor cells (HPSCs), has led to transformative and successful therapies via autologous hematopoietic stem-cell transplant with gene therapy (HSCT-GT). Due to the advanced understanding of the β-globin gene cluster, the first diseases considered for autologous HSCT-GT were two prevalent β-hemoglobinopathies: sickle cell disease and β-thalassemia, both affecting functional β-globin chains and leading to substantial morbidity. Both conditions are suitable for allogeneic HSCT; however, this therapy comes with serious risks and is most effective using an HLA-matched family donor (which is not available for most patients) to obtain optimal therapeutic and safe benefits. Transplants from unrelated or haplo-identical donors carry higher risks, although they are progressively improving. Conversely, HSCT-GT utilizes the patient\'s own HSPCs, broadening access to more patients. Several gene therapy clinical trials have been reported to have achieved significant disease improvement, and more are underway. Based on the safety and the therapeutic success of autologous HSCT-GT, the U.S. Food and Drug Administration (FDA) in 2022 approved an HSCT-GT for β-thalassemia (Zynteglo™). This review illuminates the β-globin gene research journey, adversities faced, and achievements reached; it highlights important molecular and genetic findings of the β-globin locus, describes the predominant globin vectors, and concludes by describing promising results from clinical trials for both sickle cell disease and β-thalassemia.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    β样珠蛋白基因的表达在生命过程中受到转录因子的发育调节,β-珠蛋白基因座的染色质循环和表观基因组修饰。表观基因组修饰,如组蛋白甲基化/去甲基化和乙酰化/去乙酰化和DNA甲基化,与基因表达的上调或下调有关。对这些机制及其基因表达结果的理解为开发治疗各种疾病的新治疗策略铺平了道路。如β-血红蛋白病。组蛋白脱乙酰酶和DNA甲基转移酶抑制剂目前正在血红蛋白病患者的临床试验中进行测试。然而,这些方法往往是不确定的,非特异性及其全球效应带来了严重的安全问题。表观基因组编辑是最近开发的有前途的工具,它由DNA识别域(锌指,转录激活因子样效应子或死簇规则间隔短回文重复序列Cas9)与染色质修饰酶的催化结构域融合。它提供了更具体的疾病相关基因靶向(例如,重新激活胎儿γ-珠蛋白基因并改善血红蛋白病表型的能力),并促进了无疤痕基因治疗方法的发展。这里,我们总结了β-珠蛋白基因座的表观基因组调控机制,我们讨论了表观基因组编辑在血红蛋白病治疗中的应用。
    Beta-like globin gene expression is developmentally regulated during life by transcription factors, chromatin looping and epigenome modifications of the β-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 β-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 β-globin locus, and we discuss the application of epigenome editing for the treatment of hemoglobinopathies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    血红素调节抑制剂(HRI)是调节红系细胞中mRNA翻译的血红素感应激酶。血红素缺乏,HRI被激活以磷酸化真核起始因子2α并停止球蛋白的产生,从而避免无血红素珠蛋白链的积累。HRI通过与HRIN末端和激酶结构域内的一个或两个血红素结合结构域结合而被血红素抑制。最近发现HRI抑制成人红系细胞中胎儿血红蛋白(HbF)的产生。HRI的耗尽增加了HbF的产量,为镰状细胞病或地中海贫血患者提供治疗可利用的靶标,这得益于升高的HbF水平。已知HRI是通过自磷酸化激活的寡聚酶,尽管HRI低聚物的确切性质,它与自磷酸化的关系,其血红素调节模式尚不清楚。这里,我们采用生物化学和生物物理研究来证明HRI形成了一种不依赖于自磷酸化的二聚体,HRI中的C端卷曲螺旋结构域对于二聚体形成是必不可少的,和二聚体的形成促进了HRI的有效自磷酸化和活化。我们还采用动力学研究来证明血红素抑制HRI的主要途径是通过激酶域内的血红素结合位点。并且这种抑制相对独立于ATP和真核起始因子2α底物的结合。一起,这些研究强调了血红素抑制的模式和二聚化在人类HRI血红素感应活性中的重要性。
    The heme-regulated inhibitor (HRI) is a heme-sensing kinase that regulates mRNA translation in erythroid cells. In heme deficiency, HRI is activated to phosphorylate eukaryotic initiation factor 2α and halt production of globins, thus avoiding accumulation of heme-free globin chains. HRI is inhibited by heme via binding to one or two heme-binding domains within the HRI N-terminal and kinase domains. HRI has recently been found to inhibit fetal hemoglobin (HbF) production in adult erythroid cells. Depletion of HRI increases HbF production, presenting a therapeutically exploitable target for the treatment of patients with sickle cell disease or thalassemia, which benefit from elevated HbF levels. HRI is known to be an oligomeric enzyme that is activated through autophosphorylation, although the exact nature of the HRI oligomer, its relation to autophosphorylation, and its mode of heme regulation remain unclear. Here, we employ biochemical and biophysical studies to demonstrate that HRI forms a dimeric species that is not dependent on autophosphorylation, the C-terminal coiled-coil domain in HRI is essential for dimer formation, and dimer formation facilitates efficient autophosphorylation and activation of HRI. We also employ kinetic studies to demonstrate that the primary avenue by which heme inhibits HRI is through the heme-binding site within the kinase domain, and that this inhibition is relatively independent of binding of ATP and eukaryotic initiation factor 2α substrates. Together, these studies highlight the mode of heme inhibition and the importance of dimerization in human HRI heme-sensing activity.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    镰状细胞病(SCD)是与急性和慢性疼痛相关的常见遗传性血液疾病,进行性多器官损伤,早期死亡。操纵人类基因组技术的最新进展,一个世纪的研究和技术的发展,使隔离,高效的基因改造,和自体患者造血干细胞(HSC)的重新植入,意味着治愈大多数SCD患者可能很快在富裕国家成为现实。并行,正在进行的研究正在寻求更容易的治疗方法,例如,体内提供的基因疗法和新药,最终可以在大多数SCD患者居住的低收入和中等收入国家使用。
    Sickle cell disease (SCD) is a common genetic blood disorder associated with acute and chronic pain, progressive multiorgan damage, and early mortality. Recent advances in technologies to manipulate the human genome, a century of research and the development of techniques enabling the isolation, efficient genetic modification, and reimplantation of autologous patient hematopoietic stem cells (HSCs), mean that curing most patients with SCD could soon be a reality in wealthy countries. In parallel, ongoing research is pursuing more facile treatments, such as in-vivo-delivered genetic therapies and new drugs that can eventually be administered in low- and middle-income countries where most SCD patients reside.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号