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
    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: Review
    Diamond-Blackfan贫血(DBA)是一种与畸形相关的先天性骨髓衰竭综合征。DBA与核糖体生物发生缺陷有关,损害红细胞生成,导致再生性大细胞性贫血.该疾病具有常染色体显性遗传,通常在生命的第一年被诊断出,需要持续治疗。我们介绍一个年轻女子的案例,在21岁时,出现了严重的症状性贫血。虽然,由于畸形,从出生起就怀疑患有先天性综合症,直到患者被转诊至我们中心进行贫血评估后,才做出确诊.在她的新生儿病史中,她出现了贫血,需要输血,但后来保持了稳定,温和,在她的童年和青春期无症状性贫血。她的家族史在其他方面并不引人注目。为了解释有症状的贫血,维生素缺乏,自身免疫性疾病,出血原因,对髓样和淋巴样肿瘤进行了调查和排除。分子研究显示RPL5基因变异c.392dup,p.(Asn131Lysfs*6),确认DBA的诊断。所有家庭成员都有正常的血液值,没有人携带突变。这里,我们将讨论这个案例的不寻常演变,并重新审视文献。
    Diamond-Blackfan anemia (DBA) is a congenital bone marrow failure syndrome associated with malformations. DBA is related to defective ribosome biogenesis, which impairs erythropoiesis, causing hyporegenerative macrocytic anemia. The disease has an autosomal dominant inheritance and is commonly diagnosed in the first year of life, requiring continuous treatment. We present the case of a young woman who, at the age of 21, developed severe symptomatic anemia. Although, due to malformations, a congenital syndrome had been suspected since birth, a confirmation diagnosis was not made until the patient was referred to our center for an evaluation of her anemia. In her neonatal medical history, she presented with anemia that required red blood cell transfusions, but afterwards remained with a stable, mild, asymptomatic anemia throughout her childhood and adolescence. Her family history was otherwise unremarkable. To explain the symptomatic anemia, vitamin deficiencies, autoimmune diseases, bleeding causes, and myeloid and lymphoid neoplasms were investigated and ruled out. A molecular investigation showed the RPL5 gene variant c.392dup, p.(Asn131Lysfs*6), confirming the diagnosis of DBA. All family members have normal blood values and none harbored the mutation. Here, we will discuss the unusual evolution of this case and revisit the literature.
    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)是一种罕见的先天性骨髓衰竭疾病,其特征是红系发育不全。它主要影响婴儿,通常由核糖体蛋白(RP)基因的杂合等位基因变异引起。最近的研究还表明,非RP基因如GATA1,TSR2与DBA相关。P53激活,翻译功能障碍,炎症,不平衡的珠蛋白/血红素合成,和自噬失调被证明有助于破坏红细胞生成和受损的红细胞生成。DBA患者的主要治疗选择是皮质类固醇。然而,这些患者中有一半在长期治疗后对皮质类固醇治疗无反应,必须输血.造血干细胞移植是目前唯一的治疗选择,然而,治疗受到合适供体的可获得性和可能出现严重免疫并发症的限制.使用慢病毒载体的基因治疗的最新进展已显示出通过促进正常造血来治疗RPS19缺陷型DBA的希望。随着对DBA分子框架的深入了解,基因疗法等新兴疗法有望提供治愈性解决方案并促进对潜在疾病机制的理解。
    Diamond-Blackfan anemia (DBA) is a rare congenital bone marrow failure disorder characterized by erythroid hypoplasia. It primarily affects infants and is often caused by heterozygous allelic variations in ribosomal protein (RP) genes. Recent studies also indicated that non-RP genes like GATA1, TSR2, are associated with DBA. P53 activation, translational dysfunction, inflammation, imbalanced globin/heme synthesis, and autophagy dysregulation were shown to contribute to disrupted erythropoiesis and impaired red blood cell production. The main therapeutic option for DBA patients is corticosteroids. However, half of these patients become non-responsive to corticosteroid therapy over prolonged treatment and have to be given blood transfusions. Hematopoietic stem cell transplantation is currently the sole curative option, however, the treatment is limited by the availability of suitable donors and the potential for serious immunological complications. Recent advances in gene therapy using lentiviral vectors have shown promise in treating RPS19-deficient DBA by promoting normal hematopoiesis. With deepening insights into the molecular framework of DBA, emerging therapies like gene therapy hold promise for providing curative solutions and advancing comprehension of the underlying disease mechanisms.
    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
    p53活性组成型增加的小鼠表现出先天性角化异常(DC)的特征,由端粒维持缺陷引起的骨髓衰竭综合征(BMFS)。进一步的研究证实,在人类和老鼠身上,影响TP53或其调节因子MDM4的种系突变可能导致端粒短并改变造血,但也揭示了Diamond-Blackfan贫血(DBA)或Fanconi贫血(FA)的特征,两个BMFSs,分别,由核糖体功能或DNA修复缺陷引起。p53下调DC中突变的几个基因,通过与启动子序列(DKC1)结合或间接通过DREAM阻遏复合物(RTEL1,DCLRE1B),p53-DREAM通路抑制22个额外的端粒相关基因。有趣的是,任何DC致病基因的突变都会导致端粒功能障碍和随后的p53激活,从而进一步促进p53-DREAM靶标的抑制。同样,核糖体功能障碍和DNA损伤导致p53激活,p53-DREAM靶标包括DBA-因果基因TSR2,至少9个FA-因果基因,和影响核糖体或FA途径的38个其他基因。此外,BMFSs患者可能表现出大脑异常,p53-DREAM抑制小头症或小脑发育不全突变的16个基因。总之,正反馈回路和p53-DREAM靶标库可能导致不同分子来源的BMFS之间的部分表型重叠。
    Mice with a constitutive increase in p53 activity exhibited features of dyskeratosis congenita (DC), a bone marrow failure syndrome (BMFS) caused by defective telomere maintenance. Further studies confirmed, in humans and mice, that germline mutations affecting TP53 or its regulator MDM4 may cause short telomeres and alter hematopoiesis, but also revealed features of Diamond-Blackfan anemia (DBA) or Fanconi anemia (FA), two BMFSs, respectively, caused by defects in ribosomal function or DNA repair. p53 downregulates several genes mutated in DC, either by binding to promoter sequences (DKC1) or indirectly via the DREAM repressor complex (RTEL1, DCLRE1B), and the p53-DREAM pathway represses 22 additional telomere-related genes. Interestingly, mutations in any DC-causal gene will cause telomere dysfunction and subsequent p53 activation to further promote the repression of p53-DREAM targets. Similarly, ribosomal dysfunction and DNA lesions cause p53 activation, and p53-DREAM targets include the DBA-causal gene TSR2, at least 9 FA-causal genes, and 38 other genes affecting ribosomes or the FA pathway. Furthermore, patients with BMFSs may exhibit brain abnormalities, and p53-DREAM represses 16 genes mutated in microcephaly or cerebellar hypoplasia. In sum, positive feedback loops and the repertoire of p53-DREAM targets likely contribute to partial phenotypic overlaps between BMFSs of distinct molecular origins.
    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
    骨髓增生异常综合征(MDS)和DiamondBlackfan贫血(DBA)的贫血通常是大细胞性的,总是反映无效的红细胞生成,然而,不同的基因突变的结果。为了描绘导致细胞死亡的共同机制,我们研究了DBA和MDS-5q患者的单个红系骨髓细胞的命运。我们定义了一个不健康的(vs.健康)使用转录假时间和细胞表面蛋白的分化轨迹。细胞上调转铁蛋白受体(CD71)后,假时间轨迹立即发散,进口铁,启动血红素合成,尽管细胞死亡发生得更晚。注定死亡的细胞高度表达血红素反应基因,包括核糖体蛋白和珠蛋白基因,而存活的细胞下调血红素合成并上调DNA损伤反应,缺氧和HIF1途径。令人惊讶的是,来自对照组的24±12%的细胞遵循不健康的轨迹,这意味着血红素可能是一个变阻器,指导细胞生存或死亡。当血红素合成被琥珀酰丙酮抑制时,更多的DBA细胞遵循健康的轨迹并存活。我们还注意到,随着红系细胞成熟,保留内含子的信息数量很多,证实了CFU-E的快速循环,并证明细胞周期时间是分化阶段的不变属性。在伪时间测定中包括未剪接的RNA允许我们可靠地对齐独立的数据集并准确地查询阶段特异性转录组变化。MDS-5q(不同于DBA)来自体细胞突变,所以许多正常(未突变)的红系细胞持续存在。通过独立跟踪有和没有染色体5q缺失的细胞的红系分化,我们深入了解了为什么5q+细胞不能扩张以预防贫血。
    The anemias of myelodysplastic syndrome (MDS) and Diamond Blackfan anemia (DBA) are generally macrocytic and always reflect ineffective erythropoiesis yet result from diverse genetic mutations. To delineate shared mechanisms that lead to cell death, we studied the fate of single erythroid marrow cells from individuals with DBA or MDS-5q. We defined an unhealthy (vs healthy) differentiation trajectory using transcriptional pseudotime and cell surface proteins. The pseudotime trajectories diverge immediately after cells upregulate transferrin receptor (CD71), import iron, and initiate heme synthesis, although cell death occurs much later. Cells destined to die express high levels of heme-responsive genes, including ribosomal protein and globin genes, whereas surviving cells downregulate heme synthesis and upregulate DNA damage response, hypoxia, and HIF1 pathways. Surprisingly, 24% ± 12% of cells from control subjects follow the unhealthy trajectory, implying that heme might serve as a rheostat directing cells to live or die. When heme synthesis was inhibited with succinylacetone, more DBA cells followed the healthy trajectory and survived. We also noted high numbers of messages with retained introns that increased as erythroid cells matured, confirmed the rapid cycling of colony forming unit-erythroid, and demonstrated that cell cycle timing is an invariant property of differentiation stage. Including unspliced RNA in pseudotime determinations allowed us to reliably align independent data sets and accurately query stage-specific transcriptomic changes. MDS-5q (unlike DBA) results from somatic mutation, so many normal (unmutated) erythroid cells persist. By independently tracking erythroid differentiation of cells with and without chromosome 5q deletions, we gained insight into why 5q+ cells cannot expand to prevent anemia.
    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
    核糖体蛋白(Rp)基因单倍体不足可导致Diamond-Blackfan贫血(DBA),以红细胞生成缺陷和骨骼缺陷为特征。一些小鼠Rp突变概括了DBA表型,尽管其他人缺乏红细胞生成或骨骼缺陷。我们生成了一个条件敲除小鼠来部分删除Rps12。纯合Rps12缺失导致胚胎致死。遗传Rps12+/-基因型的小鼠有生长和形态缺陷,全血细胞减少和红细胞生成受损。骨髓中造血干细胞(HSC)和祖细胞(BM)的显着减少与竞争性和非竞争性BM移植后重新填充血液系统的能力降低有关。Rps12+/-突变体失去了HSC静止,在HSC和祖细胞中经历了ERK和MTOR激活并增加了整体翻译。使用Tal1-Cre-ERT在造血细胞中Rps12的出生后杂合子缺失也导致全血细胞减少,HSC数量减少。然而,出生后Cre-ERT诱导导致HSC和祖细胞翻译减少,这表明这是造血细胞中Rps12单倍体不足的最直接后果。因此,RpS12对HSC功能有很强的要求,除了红细胞生成。
    Ribosomal protein (Rp) gene haploinsufficiency can result in Diamond-Blackfan Anemia (DBA), characterized by defective erythropoiesis and skeletal defects. Some mouse Rp mutations recapitulate DBA phenotypes, although others lack erythropoietic or skeletal defects. We generated a conditional knockout mouse to partially delete Rps12. Homozygous Rps12 deletion resulted in embryonic lethality. Mice inheriting the Rps12KO/+ genotype had growth and morphological defects, pancytopenia, and impaired erythropoiesis. A striking reduction in hematopoietic stem cells (HSCs) and progenitors in the bone marrow (BM) was associated with decreased ability to repopulate the blood system after competitive and non-competitive BM transplantation. Rps12KO/+ lost HSC quiescence, experienced ERK and MTOR activation, and increased global translation in HSC and progenitors. Post-natal heterozygous deletion of Rps12 in hematopoietic cells using Tal1-Cre-ERT also resulted in pancytopenia with decreased HSC numbers. However, post-natal Cre-ERT induction led to reduced translation in HSCs and progenitors, suggesting that this is the most direct consequence of Rps12 haploinsufficiency in hematopoietic cells. Thus, RpS12 has a strong requirement in HSC function, in addition to erythropoiesis.
    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贫血是一种罕见的遗传性骨髓衰竭疾病,通常由核糖体蛋白基因突变引起。在本研究中,我们使用CRISPR-Cas9和同源定向修复建立了一个可追溯的RPS19缺陷型细胞模型,以研究临床适用的慢病毒载体在单细胞分辨率下的治疗效果.我们开发了一种温和的纳米吸管递送平台,以编辑原代人脐带血来源的CD34造血干细胞和祖细胞中的RPS19基因。编辑的细胞显示预期受损的红系分化表型,并通过单细胞RNA测序分析鉴定出具有异常细胞周期状态的特定红系祖细胞,并伴有TNFα/NF-κB和p53信号通路的富集。治疗载体可以通过激活细胞周期相关的信号通路来挽救异常的红细胞生成并促进红细胞生成。总的来说,这些结果确立了纳米吸管作为敏感的原发性造血干细胞和祖细胞中基于CRISPR-Cas9的基因编辑的温和选择,并为未来慢病毒基因治疗策略的临床研究提供支持。
    Diamond-Blackfan anemia is a rare genetic bone marrow failure disorder which is usually caused by mutations in ribosomal protein genes. In the present study, we generated a traceable RPS19-deficient cell model using CRISPR-Cas9 and homology-directed repair to investigate the therapeutic effects of a clinically applicable lentiviral vector at single-cell resolution. We developed a gentle nanostraw delivery platform to edit the RPS19 gene in primary human cord bloodderived CD34+ hematopoietic stem and progenitor cells. The edited cells showed expected impaired erythroid differentiation phenotype, and a specific erythroid progenitor with abnormal cell cycle status accompanied by enrichment of TNFα/NF-κB and p53 signaling pathways was identified by single-cell RNA sequencing analysis. The therapeutic vector could rescue the abnormal erythropoiesis by activating cell cycle-related signaling pathways and promoted red blood cell production. Overall, these results establish nanostraws as a gentle option for CRISPR-Cas9- based gene editing in sensitive primary hematopoietic stem and progenitor cells, and provide support for future clinical investigations of the lentiviral gene therapy strategy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号