Anemia, Diamond-Blackfan

贫血,Diamond - Blackfan
  • 文章类型: Journal Article
    Diamond-Blackfan贫血(DBA)是一种罕见的遗传性疾病,影响骨髓产生红细胞的能力,导致严重贫血和各种身体异常。大约75%的DBA病例涉及核糖体蛋白(RP)基因的杂合突变,将其归类为核糖体病,RPS19是最常见的突变基因。非RP突变,例如在GATA1中,也已确定。目前的治疗包括糖皮质激素,输血,和造血干细胞移植(HSCT),HSCT是唯一的治疗选择,尽管存在供体可用性和免疫并发症等挑战。基因治疗,特别是使用慢病毒载体和CRISPR/Cas9技术,作为一个有希望的替代方案出现。这篇综述探讨了基因治疗的潜力,重点关注慢病毒载体和CRISPR/Cas9技术与非整合慢病毒载体的结合,作为DBA的治愈解决方案。它突出了DBA治疗领域的变革性进步,为受这种情况影响的个人提供希望。
    Diamond-Blackfan anemia (DBA) is a rare genetic disorder affecting the bone marrow\'s ability to produce red blood cells, leading to severe anemia and various physical abnormalities. Approximately 75% of DBA cases involve heterozygous mutations in ribosomal protein (RP) genes, classifying it as a ribosomopathy, with RPS19 being the most frequently mutated gene. Non-RP mutations, such as in GATA1, have also been identified. Current treatments include glucocorticosteroids, blood transfusions, and hematopoietic stem cell transplantation (HSCT), with HSCT being the only curative option, albeit with challenges like donor availability and immunological complications. Gene therapy, particularly using lentiviral vectors and CRISPR/Cas9 technology, emerges as a promising alternative. This review explores the potential of gene therapy, focusing on lentiviral vectors and CRISPR/Cas9 technology in combination with non-integrating lentiviral vectors, as a curative solution for DBA. It highlights the transformative advancements in the treatment landscape of DBA, offering hope for individuals affected by this condition.
    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
    这项研究为未来慢病毒介导的基因疗法在核糖体蛋白S19(RPS19)突变引起的DiamondBlackfan贫血(DBA)患者中奠定了基础。显示新的安全有效疗法的证据。数据显示,与范可尼贫血(FA)患者不同,DBA患者的造血干细胞(HSC)储库没有明显减少,提示这些细胞的收集不应构成对DBA基因治疗的显著限制。随后,开发了2种临床适用的慢病毒载体。在以前的慢病毒载体中,PGK.CoRPS19LV,密码子优化版本的RPS19由磷酸甘油酸激酶启动子(PGK)驱动,已经用于不同的基因治疗试验,包括FA基因治疗。在后一种情况下,EF1α。CoRPS19LV,RPS19表达由延伸因子α短启动子驱动,EF1α(s)。临床前实验显示,用PGK转导DBA患者CD34+细胞。CoRPS19LV恢复红系分化,并证明了校正后的DBACD34+细胞的长期繁殖特性,提供改善红细胞成熟的证据。同时,使用一种适用于患者血细胞的潜在新方法验证了核糖体生物发生的长期恢复,基于核糖体RNA甲基化分析。最后,体内安全性研究和前病毒插入位点分析显示,慢病毒介导的基因治疗是无毒的.
    This study lays the groundwork for future lentivirus-mediated gene therapy in patients with Diamond Blackfan anemia (DBA) caused by mutations in ribosomal protein S19 (RPS19), showing evidence of a new safe and effective therapy. The data show that, unlike patients with Fanconi anemia (FA), the hematopoietic stem cell (HSC) reservoir of patients with DBA was not significantly reduced, suggesting that collection of these cells should not constitute a remarkable restriction for DBA gene therapy. Subsequently, 2 clinically applicable lentiviral vectors were developed. In the former lentiviral vector, PGK.CoRPS19 LV, a codon-optimized version of RPS19 was driven by the phosphoglycerate kinase promoter (PGK) already used in different gene therapy trials, including FA gene therapy. In the latter one, EF1α.CoRPS19 LV, RPS19 expression was driven by the elongation factor alpha short promoter, EF1α(s). Preclinical experiments showed that transduction of DBA patient CD34+ cells with the PGK.CoRPS19 LV restored erythroid differentiation, and demonstrated the long-term repopulating properties of corrected DBA CD34+ cells, providing evidence of improved erythroid maturation. Concomitantly, long-term restoration of ribosomal biogenesis was verified using a potentially novel method applicable to patients\' blood cells, based on ribosomal RNA methylation analyses. Finally, in vivo safety studies and proviral insertion site analyses showed that lentivirus-mediated gene therapy was nontoxic.
    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
    在深度独立于数据的采集蛋白质组分析方面的最新进展已经实现了对>10,000种蛋白质的全面定量分析。在这里,进行了遗传性骨髓衰竭综合征(IBMFS)的综合蛋白质基因组分析,以揭示其生物学特征,并在发现队列中开发基于蛋白质组学的诊断测定;先天性角化异常(n=12),范可尼贫血(n=11),Diamond-Blackfan贫血(DBA,n=9),Shwachman-Diamond综合征(SDS,n=6),ADH5/ALDH2缺乏症(n=4),和其他IBMFS(n=18)。无监督蛋白质组聚类确定了八个独立簇(C1-C8),核糖体通路在C1和C2中特异性下调,富集DBA和SDS,分别。6例SDS患者SBDS蛋白表达明显下降,其中两个不是通过单独的DNA测序来诊断的。4例ADH5/ALDH2缺乏患者显示ADH5蛋白表达显著降低。为了进行大规模的快速IBMFS筛查,对来自IBMFS相关血液病患者(n=390)和健康对照(n=27)的417个样本进行靶向蛋白质组学分析.在SDS和ADH5/ALDH2缺乏症中,SBDS和ADH5蛋白表达显著降低,分别。首次整合的蛋白质基因组分析的临床应用将有助于IBMFS的诊断和筛选。在缺乏适当的临床筛查测试的地方。
    Recent advances in in-depth data-independent acquisition proteomic analysis have enabled comprehensive quantitative analysis of >10,000 proteins. Herein, an integrated proteogenomic analysis for inherited bone marrow failure syndrome (IBMFS) was performed to reveal their biological features and to develop a proteomic-based diagnostic assay in the discovery cohort; dyskeratosis congenita (n = 12), Fanconi anemia (n = 11), Diamond-Blackfan anemia (DBA, n = 9), Shwachman-Diamond syndrome (SDS, n = 6), ADH5/ALDH2 deficiency (n = 4), and other IBMFS (n = 18). Unsupervised proteomic clustering identified eight independent clusters (C1-C8), with the ribosomal pathway specifically downregulated in C1 and C2, enriched for DBA and SDS, respectively. Six patients with SDS had significantly decreased SBDS protein expression, with two of these not diagnosed by DNA sequencing alone. Four patients with ADH5/ALDH2 deficiency showed significantly reduced ADH5 protein expression. To perform a large-scale rapid IBMFS screening, targeted proteomic analysis was performed on 417 samples from patients with IBMFS-related hematological disorders (n = 390) and healthy controls (n = 27). SBDS and ADH5 protein expressions were significantly reduced in SDS and ADH5/ALDH2 deficiency, respectively. The clinical application of this first integrated proteogenomic analysis would be useful for the diagnosis and screening of IBMFS, where appropriate clinical screening tests are lacking.
    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
    在过去的十年中,多种单细胞技术的出现导致对正常造血功能及其在各种血液系统疾病中的扰动的认识得到了改善。Diamond-Blackfan贫血(DBA)就是这样一种疾病,其中单细胞分析有助于描绘该疾病背后的细胞和分子缺陷。DBA是由编码核糖体蛋白(RP)的基因中功能种系变体的杂合缺失引起的。尽管核糖体在造血中具有广泛的作用,DBA中最常见和最严重的血细胞减少是贫血.在这篇综述中,我们将讨论单细胞研究-包括克隆细胞培养试验,荧光激活细胞分选(FACS)和单细胞RNA测序(scRNAseq)-已导致对DBA发病机理的见解。主要的治疗方法是定期输血,糖皮质激素或造血干细胞移植(HSCT),但都与显着的发病率和死亡率有关。因此,我们将概述单细胞研究如何为DBA提供新疗法。此外,我们将讨论DBA如何作为一个有用的模型来理解正常的红细胞生成,分子调节过程中的稳态和对压力的反应。
    The emergence of multiomic single-cell technologies over the last decade has led to improved insights into both normal hematopoiesis and its perturbation in a variety of hematological disorders. Diamond-Blackfan anemia (DBA) syndrome is one such disorder where single-cell assays have helped to delineate the cellular and molecular defects underlying the disease. DBA is caused by heterozygous loss-of-function germline variants in genes encoding ribosomal proteins (RPs). Despite the widespread role of ribosomes in hematopoiesis, the most frequent and severe cytopenia in DBA is anemia. In this review we discussed how single-cell studies, including clonogenic cell culture assays, fluorescence-activated cell sorting (FACS) and single-cell RNA sequencing (scRNA-seq), have led to insights into the pathogenesis of DBA. The main therapies are regular blood transfusions, glucocorticoids, or hematopoietic stem cell transplantation (HSCT) but all are associated with significant morbidity and mortality. We will therefore outline how single-cell studies can inform new therapies for DBA. Furthermore, we discussed how DBA serves as a useful model for understanding normal erythropoiesis in terms of its cellular hierarchy, molecular regulation during homeostasis, and response to \"stress.\"
    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
    Diamond-Blackfan贫血(DBA),80多年前首次描述,是一种先天性红细胞生成障碍,有先天缺陷和癌症的倾向。尽管科学进步,这种慢性,衰弱,限制生命的疾病继续导致大量的身体,心理,以及对患者及其家人的经济损失。受影响患者的高度复杂的医疗需求需要专业知识和多学科护理。然而,在有效地将科学发现与临床实践联系起来以及向提供者传播最新知识和最佳实践方面仍然存在差距。继2008年第一个国际共识发表后,我们对遗传学的理解有了进展,自然史,和DBA的临床管理强烈支持需要新的共识建议.2014年在弗莱堡,德国,由包括临床医生在内的53名专家组成的小组,诊断医生,来自27个国家的研究人员召集。在患者倡导者的支持下,小组在随后的几年中多次开会,参与正在进行的讨论。这些会议导致在2024年制定了新的共识建议,取代了以前的准则。考虑到不同的表型,包括没有贫血的表现,专家组同意采用DBA综合征这一术语.我们提出了新的简化诊断标准,描述DBA综合征的遗传学及其表型,并引入治疗标准的重大变化。这些变化包括将泼尼松维持剂量降低至每天最大0·3mg/kg,将输血前血红蛋白提高到9-10g/dL,与年龄无关,建议早期积极螯合,扩大造血干细胞移植的适应症,并建议进行系统的临床监测,包括早期结直肠癌筛查。总之,当前的实践指南标准化了诊断,治疗,并对全世界所有年龄段的DBA综合征患者进行长期监测。
    Diamond-Blackfan anaemia (DBA), first described over 80 years ago, is a congenital disorder of erythropoiesis with a predilection for birth defects and cancer. Despite scientific advances, this chronic, debilitating, and life-limiting disorder continues to cause a substantial physical, psychological, and financial toll on patients and their families. The highly complex medical needs of affected patients require specialised expertise and multidisciplinary care. However, gaps remain in effectively bridging scientific discoveries to clinical practice and disseminating the latest knowledge and best practices to providers. Following the publication of the first international consensus in 2008, advances in our understanding of the genetics, natural history, and clinical management of DBA have strongly supported the need for new consensus recommendations. In 2014 in Freiburg, Germany, a panel of 53 experts including clinicians, diagnosticians, and researchers from 27 countries convened. With support from patient advocates, the panel met repeatedly over subsequent years, engaging in ongoing discussions. These meetings led to the development of new consensus recommendations in 2024, replacing the previous guidelines. To account for the diverse phenotypes including presentation without anaemia, the panel agreed to adopt the term DBA syndrome. We propose new simplified diagnostic criteria, describe the genetics of DBA syndrome and its phenocopies, and introduce major changes in therapeutic standards. These changes include lowering the prednisone maintenance dose to maximum 0·3 mg/kg per day, raising the pre-transfusion haemoglobin to 9-10 g/dL independent of age, recommending early aggressive chelation, broadening indications for haematopoietic stem-cell transplantation, and recommending systematic clinical surveillance including early colorectal cancer screening. In summary, the current practice guidelines standardise the diagnostics, treatment, and long-term surveillance of patients with DBA syndrome of all ages worldwide.
    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: Case Reports
    儿童贫血很常见,找到根本原因通常并不复杂。然而,在某些情况下,基础诊断是罕见且难以诊断的。
    一名幼儿出现严重贫血,红细胞指数正常,网织红细胞计数低。其余血液学参数正常,有轻微的血小板增多.此时,诊断为儿童短暂性红细胞减少症(TEC)。该儿童在整个儿童期继续患有轻度贫血,间歇性大细胞增多症和网织红细胞减少症。生长发育正常,并且没有心脏或肾脏先天性异常的迹象,也没有任何颅面或指骨缺陷。反复骨髓检查未发现明显异常。十几岁时,该患者通过基于外显子组的基因组小组被诊断出患有Diamond-Blackfan贫血,该基因组显示RPL11基因发生突变。
    先天性骨髓衰竭综合征并不总是以经典方式存在,导致延迟诊断。对于具有持续性血液学实验室参数异常的患者,不同基因面板的可用性增加提供了更准确的诊断途径的可能性。这对于充分的随访和遗传咨询很重要。
    Anemia in children is common and finding the underlying cause is often uncomplicated. However, in some cases, the underlying diagnosis is rare and difficult to diagnose.
    A toddler presented with severe anemia with normal red cell indices and a low reticulocyte count. The remaining hematological parameters were normal, bar a slight thrombocytosis. At this point a diagnosis of transient erythroblastopenia of childhood (TEC) was made. The child continued to have slight anemia with intermittent macrocytosis and reticulocytopenia throughout childhood. Growth and development was normal, and there were no signs of congenital abnormalities in the heart or kidneys nor any craniofacial or phalangeal defects. Repeated bone marrow examinations showed no significant abnormal findings. As a teenager the patient was diagnosed with Diamond-Blackfan anemia through an exome-based gene panel which revealed a mutation in the RPL11 gene.
    Congenital bone marrow failure syndromes do not always present in the classical way, leading to a delayed diagnosis. The increasing availability of different gene panels for patients with persistent abnormal hematological laboratory parameters offers the possibility of a more accurate diagnostic pathway, which is important for adequate follow-up and genetic counselling.
    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
    钻石黑风扇贫血(DBA)的丰富历史,最初在1938年被描述为先天性发育不良2反映了儿科血液学的演变。在他们的论文中,作者1介绍了一项使用血小板生成素模拟剂eltrombopag治疗DBA中红细胞衰竭的临床试验结果.低回复率掩盖了这项工作的重要性。评论:邓肯等人。艾曲波帕治疗难治性/复发性Diamond-Blackfan贫血。BrJHaematol2024;204:2077-2085。
    The rich history surrounding Diamond-Blackfan anaemia (DBA), originally described in 1938 as congenital hypoplastic anaemia2 reflects the evolution of paediatric haematology. In their paper, the authors1 present the results of a clinical trial using the thrombopoietin-mimetic agent eltrombopag to treat red cell failure in DBA. A low response rate belies the importance of this work. Commentary on: Duncan et al. Treatment of refractory/relapsed Diamond-Blackfan anaemia with eltrombopag. Br J Haematol 2024;204:2077-2085.
    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
    Diamond-Blackfan贫血(DBA)是一种罕见的,遗传性骨髓衰竭综合征伴有核糖体缺陷,导致正常血红素合成的珠蛋白链产生减慢,导致过量的反应性铁/血红素和红细胞特异性细胞毒性。Eltrombopag,非肽类血小板生成素受体激动剂,是一种有效的细胞内铁螯合剂,在另一项试验中,在RPS19突变的DBA患者中诱导了强大的持久反应。我们假设eltrombopag可以改善DBA患者的RBC产生。我们进行了一个单中心,单臂试点研究(NCT04269889)评估每天6个月的安全性和红细胞反应,DBA患者的固定剂量艾曲波帕。15例依赖输血(每3-5周)的患者(中位年龄18[范围2-56])接受治疗。一名反应者血红蛋白持续改善,红细胞输血频率减少>50%。值得注意的是,7/15(41%)患者由于无症状的血小板增多而需要减少剂量或持续停药eltrombopag。尽管回复率低,艾曲波帕现在已经改善了一些DBA患者的红细胞生成,具有良好的安全性。在大多数患者中,由于血小板增多导致的剂量限制可能会导致铁螯合不足,从而减少血红素产生并改善贫血。未来的工作将集中在患者的红细胞生成动力学和血红素合成抑制剂的使用上,而不会影响其他造血谱系。
    Diamond-Blackfan anaemia (DBA) is a rare, inherited bone marrow failure syndrome with a ribosomal defect causing slowed globin chain production with normal haem synthesis, causing an overabundance of reactive iron/haem and erythroid-specific cellular toxicity. Eltrombopag, a non-peptide thrombopoietin receptor agonist, is a potent intracellular iron chelator and induced a robust durable response in an RPS19-mutated DBA patient on another trial. We hypothesized eltrombopag would improve RBC production in DBA patients. We conducted a single-centre, single-arm pilot study (NCT04269889) assessing safety and erythroid response of 6 months of daily, fixed-dose eltrombopag for DBA patients. Fifteen transfusion-dependent (every 3-5 weeks) patients (median age 18 [range 2-56]) were treated. One responder had sustained haemoglobin improvement and >50% reduction in RBC transfusion frequency. Of note, 7/15 (41%) patients required dose reductions or sustained discontinuation of eltrombopag due to asymptomatic thrombocytosis. Despite the low response rate, eltrombopag has now improved erythropoiesis in several patients with DBA with a favourable safety profile. Dosing restrictions due to thrombocytosis may cause insufficient iron chelation to decrease haem production and improve anaemia in most patients. Future work will focus on erythropoiesis dynamics in patients and use of haem synthesis inhibitors without an impact on other haematopoietic lineages.
    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
    Diamond-Blackfan贫血(DBA)是人类中描述的第一个核糖体病。DBA是一种先天性再生障碍性贫血,以大细胞性再生性贫血为特征,表现为BFU-e/CFU-e发育红系祖细胞阶段之间的分化阻断。在50%的DBA案例中,注意到各种畸形。引人注目的是,对于具有相对红细胞样倾向的血液疾病,DBA是由于24种不同核糖体蛋白(RP)基因中的核糖体单倍体不足。在DBA样疾病中已经描述了一些其他基因,但它们不适合经典的DBA表型(Sankaran等人。,2012;vanDooijeweert等人。,2022年;Toki等人。,2018年;Kim等人。,2017年[1-4])。RP基因中的单倍体不足导致核糖体RNA(rRNA)成熟缺陷,这是DBA的标志。然而,DBA中类红细胞嗜性缺陷的机制理解仍有待完全定义。DBA中的红系缺陷最近以非排他性的方式与许多机制相关联,这些机制包括:1)翻译中的缺陷,特别是对于GATA1类红细胞基因;2)HSP70,GATA1伴侣,3)游离血红素毒性。此外,响应核糖体应激的p53激活涉及DBA病理生理学。因此,DBA表型可能来自各种参与者的综合贡献,这可以解释在DBA患者中观察到的异质性表型,即使在同一个家庭里。
    Diamond-Blackfan anemia (DBA) was the first ribosomopathy described in humans. DBA is a congenital hypoplastic anemia, characterized by macrocytic aregenerative anemia, manifesting by differentiation blockage between the BFU-e/CFU-e developmental erythroid progenitor stages. In 50 % of the DBA cases, various malformations are noted. Strikingly, for a hematological disease with a relative erythroid tropism, DBA is due to ribosomal haploinsufficiency in 24 different ribosomal protein (RP) genes. A few other genes have been described in DBA-like disorders, but they do not fit into the classical DBA phenotype (Sankaran et al., 2012; van Dooijeweert et al., 2022; Toki et al., 2018; Kim et al., 2017 [1-4]). Haploinsufficiency in a RP gene leads to defective ribosomal RNA (rRNA) maturation, which is a hallmark of DBA. However, the mechanistic understandings of the erythroid tropism defect in DBA are still to be fully defined. Erythroid defect in DBA has been recently been linked in a non-exclusive manner to a number of mechanisms that include: 1) a defect in translation, in particular for the GATA1 erythroid gene; 2) a deficit of HSP70, the GATA1 chaperone, and 3) free heme toxicity. In addition, p53 activation in response to ribosomal stress is involved in DBA pathophysiology. The DBA phenotype may thus result from the combined contributions of various actors, which may explain the heterogenous phenotypes observed in DBA patients, even within the same family.
    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: Case Reports
    Diamond-Blackfan贫血(DBA)是一种先天性贫血,伴有红细胞发育不全。大多数致病基因是核糖体蛋白。GATA1,红细胞生成所需的造血主转录因子,也是DBA的原因。GATA1位于Xp11.23;因此,DBA仅在X连锁遗传模式的雄性中发育。这里,我们报告了1例女性新生婴儿的短暂性红细胞减少症和中度贫血,该婴儿具有从头GATA1变异。在这个病人身上,在红系细胞中观察到GATA1野生型等位基因甲基化增加.GATA1的倾斜溶解可能会导致女性患者轻度短暂性红细胞减少症。
    Diamond-Blackfan anemia (DBA) is a congenital anemia with erythroid cell aplasia. Most of the causative genes are ribosomal proteins. GATA1, a hematopoietic master transcription factor required for erythropoiesis, also causes DBA. GATA1 is located on Xp11.23; therefore, DBA develops only in males in an X-linked inheritance pattern. Here, we report a case of transient erythroblastopenia and moderate anemia in a female newborn infant with a de novo GATA1 variant. In this patient, increased methylation of the GATA1 wild-type allele was observed in erythroid cells. Skewed lyonization of GATA1 may cause mild transient erythroblastopenia in a female patient.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号