MECOM

MECOM
  • 文章类型: Journal Article
    目的:探讨MDS1和EVI1复合物位点(MECOM)在肺腺癌(LUAD)中的临床价值。方法:生物信息学和实验验证证实了LUAD中MECOM的表达水平。通过受试者工作特征(ROC)曲线和Cox回归分析分析MECOM值。结果:LUAD患者血清MECOM水平较低,且与性别相关,TNM阶段,肿瘤大小,淋巴结转移和远处转移。ROC曲线显示,LUAD的MECOM曲线下面积为0.804,值得注意的是,晚期LUAD可达到0.889;特异性高达90%。结论:MECOM可能有助于LUAD患者的独立识别,特别是在高级阶段。
    肺腺癌是肺癌的一种常见类型,具有很高的发病率和死亡率。然而,目前仍缺乏有效鉴别肺腺癌患者的临床指标。MECOM基因编码的蛋白质是调节基因表达的DNA结合蛋白,已发现在许多癌症中发挥促癌作用,但是我们发现它可能在肺腺癌中起抑癌作用。本研究旨在证实MECOM是否可以作为肺腺癌的预测因子。我们的结果表明,肺腺癌患者的血清MECOM水平低于健康人,MECOM水平较低的患者生存率较短。也就是说,血清MECOM水平较低的患者可能提示发生肺腺癌和死亡的风险较高.因此,MECOM基因有望成为与肺腺癌和死亡风险相关的预测因子.
    Objective: We aimed to explore the clinical value of MDS1 and EVI1 complex locus (MECOM) in lung adenocarcinoma (LUAD). Methods: Bioinformatics and experimental validation confirmed MECOM expression levels in LUAD. The value of MECOM was analyzed by receiver operating characteristic (ROC) curve and Cox regression analysis. Results: Serum MECOM levels were lower in LUAD and correlated with gender, TNM stage, tumor size, lymph node metastasis and distant metastasis. The ROC curve showed that the area under the curve of MECOM was 0.804 for LUAD and, of note, could reach 0.889 for advanced LUAD; specificity was up to 90%. Conclusion: MECOM may contribute to independently identifying LUAD patients, particularly in advanced stages.
    Lung adenocarcinoma is a common type of lung cancer with a high incidence and death rate. However, clinical indicators that effectively identify lung adenocarcinoma patients are still lacking. The protein encoded by the MECOM gene is a DNA-binding protein regulating gene expression, which has been found to play a cancer-promoting role in many cancers, but we found that it may play a cancer-suppressing role in lung adenocarcinoma. This study aimed to confirm whether MECOM can be a predictor for lung adenocarcinoma. Our results showed that lung adenocarcinoma patients had lower serum MECOM levels than healthy people, and patients with lower MECOM levels had a shorter survival rate. That is, patients with lower serum MECOM levels may indicate a high risk of developing lung adenocarcinoma and death. Thus, the MECOM gene is expected to be a predictor associated with the risk of developing lung adenocarcinoma and death.
    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: English Abstract
    在急性髓细胞性白血病(AML)中,inv(3)(q21q26)或t(3;3)(q21;q26)代表的EVI1重排通过增强子的结构重排引起EVI1过表达,并导致预后不良。我和我的同事对EVI1重排的骨髓性肿瘤进行了突变分析,并鉴定了SF3B1,一种核心RNA剪接因子,作为最常见的共同突变基因。的确,携带人源化inv(3)(q21q26)等位基因的转基因小鼠中潜伏性白血病的发展通过Sf3b1突变的共同发生显着加速。有趣的是,我们发现这个SF3B1突变体诱导了EVI1本身的错误剪接,其产生了在EVI1的DNA结合结构域附近具有6个氨基酸的框内插入的异常EVI1同种型。该异常EVI1同种型表现出不同于野生型EVI1的DNA结合活性,并且显著增强小鼠造血干细胞的自我更新能力。我们还鉴定了SF3B1突变体诱导的这种EVI1错误剪接所需的隐蔽分支点和外显子剪接增强子。这些数据为进一步阐明EVI1重排AML的分子机制和潜在治疗候选物提供了基础。
    In acute myeloid leukemia (AML), EVI1 rearrangement represented by inv(3)(q21q26) or t(3;3)(q21;q26) causes EVI1 overexpression via structural rearrangement of an enhancer, and confers poor prognosis. My colleagues and I performed a mutational analysis of EVI1-rearranged myeloid neoplasms and identified SF3B1, a core RNA splicing factor, as the most commonly co-mutated gene. Indeed, latent leukemia development in transgenic mice bearing the humanized inv(3)(q21q26) allele was significantly accelerated by co-occurrence of Sf3b1 mutation. Intriguingly, we found that this SF3B1 mutant induced mis-splicing of EVI1 itself, which generated an aberrant EVI1 isoform with in-frame insertion of 6 amino acids near the DNA-binding domain of EVI1. This aberrant EVI1 isoform exhibited DNA-binding activity different from wild-type EVI1 and significantly enhanced the self-renewal capacity of murine hematopoietic stem cells. We also identified the cryptic branch point and exonic splicing enhancer required for this EVI1 mis-splicing induced by the SF3B1 mutant. These data provide a basis for further elucidation of the molecular mechanism and potential therapeutic candidates for EVI1-rearranged AML.
    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
    在本研究中,目的探讨热休克蛋白B7(HSPB7)对肺腺癌(LUAD)的作用及可能机制。进行生物信息学分析以探讨HSPB7表达与LUAD患者之间的关系。MTT,菌落形成,进行伤口愈合和Transwell测定以检查增殖,H1975和A549细胞的迁移和侵袭能力。进行蛋白质印迹分析以确定相应的蛋白质表达。进行了共免疫沉淀和染色质免疫沉淀测定,以揭示HSPB7与骨髓增生异常综合征1和生态病毒整合位点1复杂基因座(MECOM)之间的相互作用。此外,BALB/c裸鼠皮下注射A549细胞,并测量肿瘤的重量和大小。HSPB7在LUAD组织和细胞中下调,其表达水平与患者预后相关。细胞功能数据显示HSPB7沉默促进肺癌细胞增殖,迁移,侵袭和上皮间质转化(EMT);而HSPB7的过表达导致相反的结果。此外,生物信息学分析显示HSPB7抑制糖酵解。HSPB7降低葡萄糖消耗,乳酸生产,和乳酸脱氢酶A,己糖激酶2和丙酮酸激酶肌肉同工型2蛋白水平。结果表明MECOM是HSPB7的转录因子。总的来说,这些结果表明HSPB7受MECOM调控,HSPB7减弱LUAD细胞增殖,迁移,通过抑制糖酵解进行侵袭和EMT。
    In the present study, it was aimed to investigate the effects and potential mechanisms of heat shock protein B7 (HSPB7) on lung adenocarcinoma (LUAD). Bioinformatic analysis was performed to explore the association between HSPB7 expression and patients with LUAD. MTT, colony formation, wound healing and Transwell assays were performed to examine the proliferative, migratory and invasive abilities of H1975 and A549 cells. Western blot analysis was conducted to determine the corresponding protein expression. Co‑Immunoprecipitation and Chromatin immunoprecipitation assays were carried out to reveal the interaction between HSPB7 and myelodysplastic syndrome 1 and ecotropic viral integration site 1 complex locus (MECOM). In addition, an animal model was conducted by the subcutaneous injection of A549 cells into BALB/c nude mice, and tumor weight and size were measured. HSPB7 was downregulated in LUAD tissues and cells, and its expression level correlated with patient prognosis. Cell functional data revealed that silencing of HSPB7 promoted lung cancer cell proliferation, migration, invasion and epithelial mesenchymal transition (EMT); whereas overexpression of HSPB7 led to the opposite results. Furthermore, bioinformatics analysis showed that HSPB7 inhibited glycolysis. HSPB7 decreased glucose consumption, lactic acid production, and lactate dehydrogenase A, hexokinase 2 and pyruvate kinase muscle isoform 2 protein levels. The results demonstrated that MECOM was a transcription factor of HSPB7. Collectively, these results suggested that HSPB7 is regulated by MECOM, and that HSPB7 attenuates LUAD cell proliferation, migration, invasion and EMT by inhibiting glycolysis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Case Reports
    2014年,对一名27岁的X连锁慢性肉芽肿病(X-CGD)患者进行了使用MFGS-gp91phox逆转录病毒载体的干细胞基因治疗。患者的难治性感染得到解决,而氧化酶阳性中性粒细胞在6个月内消失。基因治疗32个月后,患者出现骨髓增生异常综合征(MDS),并且在胚细胞中鉴定了载体整合到MECOM基因座中。在基因治疗后12个月,在大多数骨髓细胞中可检测到载体整合到MECOM中。然而,患者表现出正常的造血,直到MDS发作,表明MECOM反式激活有助于克隆造血,并且爆炸转化可能在获得额外的遗传损伤后出现。在全基因组测序中,WT1抑癌基因的双等位基因缺失,发生在肿瘤发生之前,被确定为潜在的候选遗传改变。原始细胞中的前病毒CYBBcDNA仅在编码链中包含108个G到A突变,提示在CD34阳性细胞转导过程中发生APOBEC3介导的超突变。超突变介导的氧化酶活性丧失可能促进了具有MECOM反式激活的克隆的存活和增殖。我们的数据为X-CGD基因治疗中白血病发展的复杂机制提供了有价值的见解。
    Stem cell gene therapy using the MFGS-gp91phox retroviral vector was performed on a 27-year-old patient with X-linked chronic granulomatous disease (X-CGD) in 2014. The patient\'s refractory infections were resolved, whereas the oxidase-positive neutrophils disappeared within 6 months. Thirty-two months after gene therapy, the patient developed myelodysplastic syndrome (MDS), and vector integration into the MECOM locus was identified in blast cells. The vector integration into MECOM was detectable in most myeloid cells at 12 months after gene therapy. However, the patient exhibited normal hematopoiesis until the onset of MDS, suggesting that MECOM transactivation contributed to clonal hematopoiesis, and the blast transformation likely arose after the acquisition of additional genetic lesions. In whole-genome sequencing, the biallelic loss of the WT1 tumor suppressor gene, which occurred immediately before tumorigenesis, was identified as a potential candidate genetic alteration. The provirus CYBB cDNA in the blasts contained 108 G-to-A mutations exclusively in the coding strand, suggesting the occurrence of APOBEC3-mediated hypermutations during the transduction of CD34-positive cells. A hypermutation-mediated loss of oxidase activity may have facilitated the survival and proliferation of the clone with MECOM transactivation. Our data provide valuable insights into the complex mechanisms underlying the development of leukemia in X-CGD gene therapy.
    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
    MECOM相关综合征(MECOM-AS)是一种罕见疾病,其特征是变核细胞性血小板减少症,进行性骨髓衰竭,高外显率的全血细胞减少症和尺桡骨滑膜。临床表型还可能包括手指畸形,心脏和肾脏改变,听力损失,B细胞缺乏和感染倾向。综合症,通常在新生儿期被诊断为严重的血小板减少症,是由MECOM基因突变引起的,编码转录因子EVI1。EVI1功能改变与血小板减少症的相关机制尚不清楚。在患有严重血小板减少症的儿科患者中,我们确定了MECOM基因的新变体(p。P634L),在pAP-1增强子元件和靶基因的启动子上测试了其作用,表明突变损害了转录因子的抑制活性。此外,我们证明了EVI1控制MPL的转录调控,一种基因,其突变负责先天性阿米迦勒血小板减少症(CAMT),可能解释了MECOM-AS和CAMT之间的部分重叠。
    MECOM-associated syndrome (MECOM-AS) is a rare disease characterized by amegakaryocytic thrombocytopenia, progressive bone marrow failure, pancytopenia and radioulnar synostosis with high penetrance. The clinical phenotype may also include finger malformations, cardiac and renal alterations, hearing loss, B-cell deficiency and predisposition to infections. The syndrome, usually diagnosed in the neonatal period because of severe thrombocytopenia, is caused by mutations in the MECOM gene, encoding for the transcription factor EVI1. The mechanism linking the alteration of EVI1 function and thrombocytopenia is poorly understood. In a paediatric patient affected by severe thrombocytopenia, we identified a novel variant of the MECOM gene (p.P634L), whose effect was tested on pAP-1 enhancer element and promoters of targeted genes showing that the mutation impairs the repressive activity of the transcription factor. Moreover, we demonstrated that EVI1 controls the transcriptional regulation of MPL, a gene whose mutations are responsible for congenital amegakaryocytic thrombocytopenia (CAMT), potentially explaining the partial overlap between MECOM-AS and CAMT.
    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
    MECOM缺乏症是最近发现的由造血转录因子MECOM单倍体不足引起的先天性免疫错误和遗传性骨髓衰竭综合征。它在遗传性骨髓衰竭综合征中是独一无二的,其中许多存在于童年或青春期后期,由于全血细胞减少症的发病年龄和严重程度,强调MECOM在造血过程中的重要性和基因剂量依赖性。已经在MECOM缺乏症患者的一部分中描述了B细胞淋巴细胞减少和低丙种球蛋白血症。虽然目前尚不清楚B细胞缺乏的潜在机制,最近的工作为MECOM在造血干细胞(HSC)维持中的功能提供了机械见解。MECOM与调控增强子结合,所述调控增强子控制HSC维持和自我更新所必需的基因网络的表达。杂合突变,如MECOM缺乏的骨髓衰竭所示,导致MECOM网络表达失调。MECOM缺乏的造血外表现,包括肾脏和心脏异常,径向骨滑膜,clindactyly,听力损失,已被报道。具有特定基因型的个体有一些全身性表现,有孤立的轻度血小板减少症或无血液学异常。突出了一些MECOM结构域突变的组织特异性。那些患有MECOM相关骨髓衰竭的婴儿需要HSC移植才能存活。这里,我们回顾了导致MECOM缺陷的患者表型和伴随基因型的扩展队列,以及MECOM调控人类HSC维持和B细胞发育的潜在机制。
    MECOM deficiency is a recently identified inborn error of immunity and inherited bone marrow failure syndrome caused by haploinsufficiency of the hematopoietic transcription factor MECOM. It is unique among inherited bone marrow failure syndromes, many of which present during later childhood or adolescence, because of the early age of onset and severity of the pancytopenia, emphasizing the importance and gene dose dependency of MECOM during hematopoiesis. B-cell lymphopenia and hypogammaglobulinemia have been described in a subset of patients with MECOM deficiency. While the mechanisms underlying the B-cell deficiency are currently unknown, recent work has provided mechanistic insights into the function of MECOM in hematopoietic stem cell (HSC) maintenance. MECOM binds to regulatory enhancers that control the expression of a network of genes essential for HSC maintenance and self-renewal. Heterozygous mutations, as seen in MECOM-deficient bone marrow failure, lead to dysregulated MECOM network expression. Extra-hematopoietic manifestations of MECOM deficiency, including renal and cardiac anomalies, radioulnar synostosis, clinodactyly, and hearing loss, have been reported. Individuals with specific genotypes have some of the systemic manifestations with isolated mild thrombocytopenia or without hematologic abnormalities, highlighting the tissue specificity of mutations in some MECOM domains. Those infants with MECOM-associated bone marrow failure require HSC transplantation for survival. Here, we review the expanding cohort of patient phenotypes and accompanying genotypes resulting in MECOM deficiency, and the proposed mechanisms underlying MECOM regulation of human HSC maintenance and B-cell development.
    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
    遗传性骨髓衰竭(BMF)综合征在遗传上是多样化的-超过100个基因与这些综合征相关,并且列表正在迅速扩大。最近发现的BMF综合征患者的风险评估和遗传咨询本质上是困难的,因为疾病机制,外显率,基因型-表型关联,表型异质性,血液系统恶性肿瘤的风险和疾病进展的克隆标志物未知或不清楚.这篇综述旨在以稀疏简洁的数据阐明最近描述的BMF综合征,并强调与ADH5/ALDH2,DNAJC21,ERCC6L2和MECOM中的种系变异相关的那些。这将提供重要的数据,可能有助于个性化和改善这些患者的护理。
    Inherited bone marrow failure (BMF) syndromes are genetically diverse - more than 100 genes have been associated with those syndromes and the list is rapidly expanding. Risk assessment and genetic counseling of patients with recently discovered BMF syndromes is inherently difficult as disease mechanisms, penetrance, genotype-phenotype associations, phenotypic heterogeneity, risk of hematologic malignancies and clonal markers of disease progression are unknown or unclear. This review aims to shed light on recently described BMF syndromes with sparse concise data and with an emphasis on those associated with germline variants in ADH5/ALDH2, DNAJC21, ERCC6L2 and MECOM. This will provide important data that may help to individualize and improve care for these patients.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Preprint
    转录因子MECOM,PAX8,SOX17和WT1是高级浆液性卵巢癌(HGSC)的候选主调节因子,然而它们在假设的起源组织中的合作作用,输卵管分泌上皮(FTSEC)未知。我们产生了26个表观基因组(CUT&TAG,CUT&RUN,ATAC-seq和HiC)数据集和24个RNA-seq转录因子敲低谱,然后在FTSEC和HGSC模型中进行RNA测序,以定义顺式和反式受这些因子调节的结合位点和基因集。这表明MECOM,PAX8、SOX17和WT1是谱系丰富的,超级增强子相关的主调节因子,其协同DNA结合模式和靶基因在肿瘤发展过程中重新连接。所有四种TFs对于HGSC克隆形成和存活是不可缺少的,但仅PAX8和WT1的耗竭损害FTSEC细胞存活。这四种TF仅在HGSC中而在FTSEC中被转录抑制剂药理学抑制。总的来说,我们的数据表明,肿瘤特异性表观遗传重塑与MECOM密切相关,PAX8、SOX17和WT1活性以及这些转录因子可通过转录抑制剂以肿瘤特异性方式靶向。
    The transcription factors MECOM, PAX8, SOX17 and WT1 are candidate master regulators of high-grade serous \'ovarian\' cancer (HGSC), yet their cooperative role in the hypothesized tissue of origin, the fallopian tube secretory epithelium (FTSEC) is unknown. We generated 26 epigenome (CUT&TAG, CUT&RUN, ATAC-seq and HiC) data sets and 24 profiles of RNA-seq transcription factor knock-down followed by RNA sequencing in FTSEC and HGSC models to define binding sites and gene sets regulated by these factors in cis and trans. This revealed that MECOM, PAX8, SOX17 and WT1 are lineage-enriched, super-enhancer associated master regulators whose cooperative DNA-binding patterns and target genes are re-wired during tumor development. All four TFs were indispensable for HGSC clonogenicity and survival but only depletion of PAX8 and WT1 impaired FTSEC cell survival. These four TFs were pharmacologically inhibited by transcriptional inhibitors only in HGSCs but not in FTSECs. Collectively, our data highlights that tumor-specific epigenetic remodeling is tightly related to MECOM, PAX8, SOX17 and WT1 activity and these transcription factors are targetable in a tumor-specific manner through transcriptional inhibitors.
    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
    MECOM基因编码造血干细胞自我更新和维持所必需的多种蛋白质同种型。种系MECOM变异与先天性血小板减少症有关,尺桡骨融合和骨髓衰竭;然而,MECOM相关综合征的表型谱不断扩大,新的致病变异体也在不断被发现.我们描述了八名无关患者,他们增加了MECOM相关综合征的先前已知表型和遗传缺陷。由于每个受试者都有独特的MECOM变体,该系列未能证明基因型与表型之间有明确的相关性,但可能提示了影响基因表达和随后表型的其他修饰因子的作用.对扩大的血液学和非血液学临床特征的识别允许快速的分子诊断。早期发现危及生命的并发症,和改善家庭遗传咨询。一个中央的国际公开可访问的数据库,以共享带注释的MECOM变体将促进其临床解释,并为执行功能性MECOM研究提供基础。
    The MECOM gene encodes multiple protein isoforms that are essential for hematopoietic stem cell self-renewal and maintenance. Germline MECOM variants have been associated with congenital thrombocytopenia, radioulnar synostosis and bone marrow failure; however, the phenotypic spectrum of MECOM-associated syndromes continues to expand and novel pathogenic variants continue to be identified. We describe eight unrelated patients who add to the previously known phenotypes and genetic defects of MECOM-associated syndromes. As each subject presented with unique MECOM variants, the series failed to demonstrate clear genotype-to-phenotype correlation but may suggest a role for additional modifiers that affect gene expression and subsequent phenotype. Recognition of the expanded hematologic and non-hematologic clinical features allows for rapid molecular diagnosis, early identification of life-threatening complications, and improved genetic counseling for families. A centralized international publicly accessible database to share annotated MECOM variants would advance their clinical interpretation and provide a foundation to perform functional MECOM studies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Case Reports
    我们医院出现了一名62岁的女性视力障碍。眼部检查发现双侧罗斯斑点。实验室数据显示白细胞增多(236,200μl)与过量的blast(11%)。体格检查和计算机断层扫描(CT)显示全身性淋巴结肿大。骨髓检查显示其成分为9.2%。骨髓细胞染色体分析显示46,XX,t(3;12)(q26.2;p13),t(9;22)(q34.1;q11.2)在80%的中期(16/20)。腹股沟淋巴结活检显示髓过氧化物酶(MPO)阳性异常细胞弥漫性增殖。荧光原位杂交分析用于检测BCR-ABL1融合基因并分裂MECOM和ETV6的信号。她被诊断出患有新发慢性粒细胞白血病(CML)髓外爆炸危机。她接受了酪氨酸激酶抑制剂(TKI)联合化疗和异基因造血干细胞移植,并获得了主要的分子反应。在这项研究中,我们报道了一例CML在爆炸期,最初表现为髓外,其中细胞遗传学和分子分析在分期方法中很有用。
    A 62-year-old woman was presented at our hospital with visual disturbance. An ocular examination revealed bilateral Roth spots. Laboratory data revealed leukocytosis (236,200 µl) with an excess blast (11%). Physical examination and computed tomography (CT) showed systemic lymphadenopathy. A bone marrow examination revealed a composition of 9.2% blast. Chromosomal analysis on bone marrow cells revealed 46,XX,t (3;12)(q26.2;p13),t (9;22)(q34.1;q11.2) in 80% of metaphases (16/20). Inguinal lymph node biopsy revealed diffuse proliferation of myeloperoxidase (MPO)-positive abnormal cells. Fluorescence in situ hybridization analysis was used to detect the BCR-ABL1 fusion gene and split the signals of MECOM and ETV6. She was diagnosed with de-novo chronic myeloid leukemia (CML) extramedullary blast crisis. She received tyrosine kinase inhibitor (TKI) combination chemotherapy and allogeneic hematopoietic stem cell transplantation and achieved a major molecular response. In this study, we reported a case of CML in blast-phase initially presenting as extramedullary, in which cytogenetic and molecular analyses were useful in the staging method.
    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