Bone Marrow Failure Disorders

骨髓衰竭疾病
  • 文章类型: Journal Article
    细胞遗传学研究对于骨髓衰竭综合征(BMFSs)患者的诊断和随访至关重要。但是由于细胞不足,获得高质量的结果通常是具有挑战性的。光学基因组作图(OGM),一种能够以高分辨率检测大多数类型染色体结构变异(SV)的新技术,越来越多地在许多环境中使用,包括恶性血液病.在这里,我们比较了20例不同BMFSs患者的常规细胞遗传学技术和OGM。仅在三名受试者(15%)中获得了20个核型中期,并且在任何样品中均未发现SV。一名培养失败的患者通过荧光原位杂交显示染色体1q增加,OGM证实了这一点。相比之下,OGM在所有科目中都提供了良好的质量结果,在其中14个(70%)中检测到SV,主要对应于标准技术未观察到的隐秘亚显微改变。因此,OGM成为一种强大的工具,可在低细胞BMFSs中提供完整且可评估的结果,减少多个测试到一个单一的测定和克服一些传统技术的主要限制。此外,除了确认常规技术检测到的异常之外,OGM发现了超出检测极限的新变化。
    Cytogenetic studies are essential in the diagnosis and follow up of patients with bone marrow failure syndromes (BMFSs), but obtaining good quality results is often challenging due to hypocellularity. Optical Genome Mapping (OGM), a novel technology capable of detecting most types chromosomal structural variants (SVs) at high resolution, is being increasingly used in many settings, including hematologic malignancies. Herein, we compared conventional cytogenetic techniques to OGM in 20 patients with diverse BMFSs. Twenty metaphases for the karyotype were only obtained in three subjects (15%), and no SVs were found in any of the samples. One patient with culture failure showed a gain in chromosome 1q by fluorescence in situ hybridization, which was confirmed by OGM. In contrast, OGM provided good quality results in all subjects, and SVs were detected in 14 of them (70%), mostly corresponding to cryptic submicroscopic alterations not observed by standard techniques. Therefore, OGM emerges as a powerful tool that provides complete and evaluable results in hypocellular BMFSs, reducing multiple tests into a single assay and overcoming some of the main limitations of conventional techniques. Furthermore, in addition to confirming the abnormalities detected by conventional techniques, OGM found new alterations beyond their detection limits.
    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
    骨髓衰竭(BMF)已成为研究最多的自身免疫性疾病之一,特别是由于它作为一种遗传性疾病的流行,也是化疗的结果。BMF与严重症状相关,如出血发作和感染易感性,并且通常具有潜在的特征,比如贫血,血小板减少症,和中性粒细胞减少症.目前BMF的治疗需要干细胞移植或化疗来诱导免疫抑制。然而,与这些治疗相关的供体细胞可用性或剂量相关的毒性有限.优化这些治疗已经成为一种需要。聚合物基材料越来越受欢迎,由于目前的研究工作集中在合成用于干细胞扩增的新型细胞基质,以解决有限的供体细胞可用性,以及应用聚合物递送载体以在胞内递送有助于免疫抑制的货物。这里,我们讨论了聚合物材料在BMF背景下增强治疗的重要性和影响。
    Bone marrow failure (BMF) has become one of the most studied autoimmune disorders, particularly due to its prevalence both as an inherited disease, but also as a result of chemotherapies. BMF is associated with severe symptoms such as bleeding episodes and susceptibility to infections, and often has underlying characteristics, such as anemia, thrombocytopenia, and neutropenia. The current treatment landscape for BMF requires stem cell transplantation or chemotherapies to induce immune suppression. However, there is limited donor cell availability or dose related toxicity associated with these treatments. Optimizing these treatments has become a necessity. Polymer-based materials have become increasingly popular, as current research efforts are focused on synthesizing novel cell matrices for stem cell expansion to solve limited donor cell availability, as well as applying polymer delivery vehicles to intracellularly deliver cargo that can aid in immunosuppression. Here, we discuss the importance and impact of polymer materials to enhance therapeutics in the context of BMF.
    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)是一组异质性的白血病前造血系统疾病,其特征是由于无效的造血作用而导致外周血中的血细胞减少以及骨髓(BM)中的正常或高细胞和形态发育异常。炎性BM微环境和造血干/祖细胞(HSPC)的程序性细胞死亡被认为是MDS中无效造血的主要原因。焦亡,凋亡和坏死(统称,在MDS患者的BM组织中观察到PANoptosis),提示PANoptosis在MDS发病机制中的重要作用。半胱天冬酶8(Casp8)是PANoptosis的主要调节因子,在大多数MDS患者的HSPCs中下调,在具有SRSF2突变的MDS患者的HSPCs中异常剪接。为了研究PANoptosis在造血中的作用,我们产生了诱导型Casp8敲除小鼠(Casp8-/-)。Mx1-Cre-Casp8-/-小鼠在polyI:C注射的10天内死于BM失败,这是由于HSPC的消耗。Rosa-ERT2Cre-Casp8-/-小鼠是健康的,在Casp8缺失后的前1.5个月内BM造血没有显著变化。由于Casp8-/-HSPC对感染或炎症诱导的坏死的超敏反应,这类小鼠在感染或低剂量polyI:C/LPS注射时出现BM衰竭,这可以通过Ripk3缺失来预防。然而,由于Ripk1-Tbk1信号的激活,无法通过Ripk3删除来挽救Casp8-/-HSPC的自我更新能力受损。最重要的是,用Casp8-/-BM细胞移植的小鼠在移植后4个月内出现MDS样疾病,如贫血所示,血小板减少和骨髓增生异常。我们的研究表明,Casp8,Ripk3-Mlkl和Ripk1-Tbk1活性的平衡在调节HSPCs的生存和自我更新中起着至关重要的作用。其破坏会导致炎症和BM衰竭,导致MDS样疾病。
    Myelodysplastic syndromes (MDS) are a heterogeneous group of pre-leukemic hematopoietic disorders characterized by cytopenia in peripheral blood due to ineffective hematopoiesis and normo- or hypercellularity and morphologic dysplasia in bone marrow (BM). An inflammatory BM microenvironment and programmed cell death of hematopoietic stem/progenitor cells (HSPCs) are thought to be the major causes of ineffective hematopoiesis in MDS. Pyroptosis, apoptosis and necroptosis (collectively, PANoptosis) are observed in BM tissues of MDS patients, suggesting an important role of PANoptosis in MDS pathogenesis. Caspase 8 (Casp8) is a master regulator of PANoptosis, which is downregulated in HSPCs from most MDS patients and abnormally spliced in HSPCs from MDS patients with SRSF2 mutation. To study the role of PANoptosis in hematopoiesis, we generated inducible Casp8 knockout mice (Casp8-/-). Mx1-Cre-Casp8-/- mice died of BM failure within 10 days of polyI:C injections due to depletion of HSPCs. Rosa-ERT2Cre-Casp8-/- mice are healthy without significant changes in BM hematopoiesis within the first 1.5 months after Casp8 deletion. Such mice developed BM failure upon infection or low dose polyI:C/LPS injections due to the hypersensitivity of Casp8-/- HSPCs to infection or inflammation-induced necroptosis which can be prevented by Ripk3 deletion. However, impaired self-renewal capacity of Casp8-/- HSPCs cannot be rescued by Ripk3 deletion due to activation of Ripk1-Tbk1 signaling. Most importantly, mice transplanted with Casp8-/- BM cells developed MDS-like disease within 4 months of transplantation as demonstrated by anemia, thrombocytopenia and myelodysplasia. Our study suggests an essential role for a balance in Casp8, Ripk3-Mlkl and Ripk1-Tbk1 activities in the regulation of survival and self-renewal of HSPCs, the disruption of which induces inflammation and BM failure, resulting in MDS-like disease.
    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
    先天性角化病(DC)是一种与端粒功能障碍内在相关的多系统疾病,导致骨髓衰竭(BMF)。尽管DC的病理主要是由端粒相关基因突变驱动的,基因融合的含义,由于端粒诱导的基因组不稳定性而出现,仍未探索。我们精心分析了来自DC患者的RNA-Seq数据中的基因融合,以提供对DC进展的更深入了解。随后将最重要的DC特异性基因融合体进行计算机评估,以确定生物物理和结构属性。包括电荷图案化,固有的紊乱,和自我联想的倾向。然后使用深度学习驱动的结构预测和分子动力学模拟来分析选定的候选物,以评估它们形成高阶低聚物的潜力。我们的探索表明,参与融合事件的基因在维护基因组稳定性中起着至关重要的作用。促进造血,抑制肿瘤。值得注意的是,我们的分析突出了一个特别无序的聚两性电解质融合蛋白,表现出强大的高阶寡聚动力学。最后,这项研究强调了几种高置信度基因融合在DC中BMF进展中的潜在意义,特别是通过基因组稳定性的失调,造血,和肿瘤抑制。此外,我们认为这些融合蛋白可能具有有害作用,特别是在诱导蛋白毒性驱动的造血破坏中。
    Dyskeratosis Congenita (DC) is a multisystem disorder intrinsically associated with telomere dysfunction, leading to bone marrow failure (BMF). Although the pathology of DC is largely driven by mutations in telomere-associated genes, the implications of gene fusions, which emerge due to telomere-induced genomic instability, remain unexplored. We meticulously analyzed gene fusions in RNA-Seq data from DC patients to provide deeper insights into DC\'s progression. The most significant DC-specific gene fusions were subsequently put through in silico assessments to ascertain biophysical and structural attributes, including charge patterning, inherent disorder, and propensity for self-association. Selected candidates were then analyzed using deep learning-powered structural predictions and molecular dynamics simulations to gauge their potential for forming higher-order oligomers. Our exploration revealed that genes participating in fusion events play crucial roles in upholding genomic stability, facilitating hematopoiesis, and suppressing tumors. Notably, our analysis spotlighted a particularly disordered polyampholyte fusion protein that exhibits robust higher-order oligomerization dynamics. To conclude, this research underscores the potential significance of several high-confidence gene fusions in the progression of BMF in DC, particularly through the dysregulation of genomic stability, hematopoiesis, and tumor suppression. Additionally, we propose that these fusion proteins might hold a detrimental role, specifically in inducing proteotoxicity-driven hematopoietic disruptions.
    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
    人类骨髓衰竭(BMF)综合征是由于造血干细胞和祖细胞(HSPC)的丧失,这种损失归因于细胞死亡;然而,细胞死亡的触发因素,和机制仍然未知。在BMF期间,肿瘤坏死因子-α(TNFα)和干扰素-γ(IFNγ)增加。已知这些配体诱导坏死,由RIPK1、RIPK3和MLKL介导的细胞死亡的炎症形式。我们以前发现,患有造血RIPK1缺乏症(Ripk1HEMKO)的小鼠表现出炎症,HSPC损失,还有BMF,RIPK3缺乏症部分改善了;然而,RIPK3是否通过其介导坏死性凋亡或其他形式的细胞死亡的功能发挥其作用尚不清楚.这里,我们证明了类似于RIPK3缺乏症,MLKL缺乏症可显着延长生存期,而Ripk3缺乏症可部分恢复Ripk1HEMKO小鼠的造血功能,这表明这些小鼠的坏死和凋亡均有助于BMF。使用鼠标模型,我们表明,核酸传感器Z-DNA结合蛋白1(ZBP1)在小鼠RIPK1缺陷的骨髓细胞中上调,并且ZBP1在内源性核酸传感中的功能是HSPC死亡所必需的,并有助于BMF。我们还提供了IFNγ介导Ripk1HEMKO小鼠HSPC死亡的证据,因为IFNγ而不是TNFα受体信号的消融显着延长了这些小鼠的存活。一起,这些数据表明RIPK1通过阻止ZBP1激活和诱导HSPC死亡来维持造血稳态.
    Human bone marrow failure (BMF) syndromes result from the loss of hematopoietic stem and progenitor cells (HSPC), and this loss has been attributed to cell death; however, the cell death triggers, and mechanisms remain unknown. During BMF, tumor necrosis factor-α (TNFα) and interferon-γ (IFNγ) increase. These ligands are known to induce necroptosis, an inflammatory form of cell death mediated by RIPK1, RIPK3, and MLKL. We previously discovered that mice with a hematopoietic RIPK1 deficiency (Ripk1HEM KO) exhibit inflammation, HSPC loss, and BMF, which is partially ameliorated by a RIPK3 deficiency; however, whether RIPK3 exerts its effects through its function in mediating necroptosis or other forms of cell death remains unclear. Here, we demonstrate that similar to a RIPK3 deficiency, an MLKL deficiency significantly extends survival and like Ripk3 deficiency partially restores hematopoiesis in Ripk1HEM KO mice revealing that both necroptosis and apoptosis contribute to BMF in these mice. Using mouse models, we show that the nucleic acid sensor Z-DNA binding protein 1 (ZBP1) is up-regulated in mouse RIPK1-deficient bone marrow cells and that ZBP1\'s function in endogenous nucleic acid sensing is necessary for HSPC death and contributes to BMF. We also provide evidence that IFNγ mediates HSPC death in Ripk1HEM KO mice, as ablation of IFNγ but not TNFα receptor signaling significantly extends survival of these mice. Together, these data suggest that RIPK1 maintains hematopoietic homeostasis by preventing ZBP1 activation and induction of HSPC death.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Letter
    暂无摘要。
    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
    遗传性骨髓衰竭综合征(IBMFS)是由相关基因的种系突变引起的一组异质性疾病,以骨髓衰竭(BMF)为特征。疾病特异性器官受累,and,在大多数情况下,易患恶性肿瘤.它们与免疫骨髓衰竭的区别通常具有挑战性,特别是当出现在成年期或非典型时。在评估所有新的BMF患者的遗传性病因时,功能(疾病特异性测定)和遗传测试的组合是最佳的。然而,基因检测费用昂贵,由于资源限制,可能无法在全球范围内使用;在这种情况下,临床病史,标准实验室测试,算法的使用可以指导诊断。遗传结果的解释可能具有挑战性,必须反映致病性的评估,继承模式,临床表型,和使用的标本类型。由于基因组学的逐步使用,新的IBMFS继续被识别,扩大这些疾病的范围。
    The inherited bone marrow failure syndromes (IBMFS) are a heterogenous group of disorders caused by germline mutations in related genes and characterized by bone marrow failure (BMF), disease specific organ involvement, and, in most cases, predisposition to malignancy. Their distinction from immune marrow failure can often be challenging, particularly when presentations occur in adulthood or are atypical. A combination of functional (disease specific assays) and genetic testing is optimal in assessing all new BMF patients for an inherited etiology. However, genetic testing is costly and may not be available worldwide due to resource constraints; in such cases, clinical history, standard laboratory testing, and the use of algorithms can guide diagnosis. Interpretation of genetic results can be challenging and must reflect assessment of pathogenicity, inheritance pattern, clinical phenotype, and specimen type used. Due to the progressive use of genomics, new IBMFS continue to be identified, widening the spectrum of these disorders.
    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
    造血细胞移植(HCT)可以治愈遗传性骨髓衰竭综合征(IBMFS)患者的血液异常并降低血液系统癌症的风险。然而,因为它的高死亡率,通常保留HCT,直到IBMFS患者出现危及生命的血细胞减少或髓系恶性肿瘤。在这一点上,结果很差。缺乏能够准确预测转化并能够及时干预的筛查测试。这些未知因素和风险限制了HCT在IBMFS患者中的使用,有时直到发生明显的疾病相关后遗症。IBMFS的主要目标是减少与细胞治疗相关的并发症,以便在大量输血暴露之前考虑早期干预。合并症的发生,或恶性转化。近几十年来,疾病特异性同种异体HCT试验在IBMFS条件下取得了显著改善,包括范可尼贫血和先天性角化障碍。这在很大程度上是由于调节强度的显著降低,以解决这些患者对细胞毒性化疗和放射的敏感性增加。这些方法的成功还可能表明在IBMFS病症中利用造血干细胞和祖细胞的内在适应性缺陷的能力。现在,随着造血中追踪体细胞遗传进化的进展和量身定制的最小强度调节方案,这个问题出现了:是时候对IBMFS进行预防性HCT了吗?
    Hematopoietic cell transplantation (HCT) can cure blood dyscrasias and reduce the risk of hematologic cancers in patients with inherited bone marrow failure syndromes (IBMFS). However, because of its high mortality rate, HCT is generally reserved until patients with IBMFS manifest life-threatening cytopenias or myeloid malignancy, at which point outcomes are poor. Screening tests that accurately predict transformation and enable timely intervention are lacking. These unknowns and risks limit the use of HCT in patients with IBMFS, sometimes until significant disease-related sequelae have occurred. A major goal for IBMFS is to reduce cellular therapy-related complications to the point that earlier intervention can be considered before significant transfusion exposure, occurrence of comorbidities, or malignant transformation. In recent decades, disease-specific allogeneic HCT trials have yielded significant improvements in outcomes in IBMFS conditions, including Fanconi anemia and dyskeratosis congenita. This is in large part due to marked reductions in conditioning intensity to address the increased sensitivity of these patients to cytotoxic chemotherapy and radiation. The success of these approaches may also indicate an ability to leverage intrinsic fitness defects of hematopoietic stem and progenitor cells across IBMFS disorders. Now with advances in tracking somatic genetic evolution in hematopoiesis and tailored minimal intensity conditioning regimens, this question arises: is it time for preventative HCT for IBMFS?
    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
    范可尼贫血(FA)一直被认为是儿童早期严重的遗传性骨髓衰竭(BMF)疾病。因此,这种多系统疾病的管理以前对于许多专门从事青少年和年轻人护理(AYA)的血液学家来说并不熟悉.AYA患者中FA的诊断增加,通过广泛可用的种系基因组测试,在为BMF进行匹配的同胞和替代供者造血干细胞移植(HSCT)后,FA儿童的长期生存率得到改善,而且在不久的将来,需要在体外基因治疗后实现血液学稳定的患者中进行长期监测,这都是AYA人群中FA管理值得特别考虑的原因.在这次审查中,我们解决了AYA合并FA患者的独特挑战和循证实践建议.讨论的具体主题包括尚未接受HSCT的AYA患者的血液学监测,发生在FA的骨髓性恶性肿瘤的管理,AYAFA患者非血液系统恶性肿瘤和器官功能障碍的诊断和治疗,以及对接受基因治疗的FA患者进行长期监测的考虑因素。
    Fanconi anemia (FA) has long been considered a severe inherited bone marrow failure (BMF) disorder of early childhood. Thus, management of this multisystem disorder has previously been unfamiliar to many hematologists specializing in the care of adolescents and young adults (AYA). The increased diagnosis of FA in AYA patients, facilitated by widely available germline genomic testing, improved long-term survival of children with FA following matched sibling and alternative donor hematopoietic stem cell transplantation (HSCT) performed for BMF, and expanding need in the near future for long-term monitoring in patients achieving hematologic stabilization following ex vivo gene therapy are all reasons why management of FA in AYA populations deserves specific consideration. In this review, we address the unique challenges and evidence-based practice recommendations for the management of AYA patients with FA. Specific topics addressed include hematologic monitoring in AYA patients yet to undergo HSCT, management of myeloid malignancies occurring in FA, diagnosis and management of nonhematologic malignances and organ dysfunction in AYA patients with FA, and evolving considerations for the long-term monitoring of patients with FA undergoing gene therapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号