RNA Splicing

RNA 剪接
  • 文章类型: Journal Article
    前mRNA剪接是转录后修饰的重要步骤,并在植物的各种生理过程中发挥作用。人NHP2L在剪接体组装过程中与U4snRNA结合;它参与RNA剪接并介导人类肿瘤的发展。然而,在植物中还没有发现直系同源物。因此,我们报道了At4g12600编码直系同源NHP2L蛋白,和AtSNU13与剪接体复合物的成分相关;atsnu13突变体在抗病性方面表现出受损的抗性,表明AtSNU13是植物免疫的正调节因子。与野生型植物相比,atsnu13突变导致防御相关基因的剪接模式改变和防御相关基因的表达减少,例如RBOHD和ALD1。进一步研究表明,AtSNU13促进U4/U6之间的相互作用。U5三-snRNP特异性27K和目标mRNA中的基序调节RNA剪接。我们的研究强调了AtSNU13通过影响防御相关基因的前mRNA剪接在调节植物免疫中的作用。
    Pre-mRNA splicing is a significant step for post-transcriptional modifications and functions in a wide range of physiological processes in plants. Human NHP2L binds to U4 snRNA during spliceosome assembly; it is involved in RNA splicing and mediates the development of human tumors. However, no ortholog has yet been identified in plants. Therefore, we report At4g12600 encoding the ortholog NHP2L protein, and AtSNU13 associates with the component of the spliceosome complex; the atsnu13 mutant showed compromised resistance in disease resistance, indicating that AtSNU13 is a positive regulator of plant immunity. Compared to wild-type plants, the atsnu13 mutation resulted in altered splicing patterns for defense-related genes and decreased expression of defense-related genes, such as RBOHD and ALD1. Further investigation shows that AtSNU13 promotes the interaction between U4/U6.U5 tri-snRNP-specific 27 K and the motif in target mRNAs to regulate the RNA splicing. Our study highlights the role of AtSNU13 in regulating plant immunity by affecting the pre-mRNA splicing of defense-related genes.
    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
    LncRNA在癌症进展和靶向中起着至关重要的作用,但很难确定参与结直肠癌(CRC)进展的关键lncRNAs.我们使用21对IV期CRC组织和邻近正常组织将FAM83H-AS1鉴定为肿瘤促进相关lncRNA。体外和体内实验表明,在CRC细胞中敲低FAM83H-AS1抑制肿瘤的增殖和转移,反之亦然。m6A修饰对于通过作者METTL3和读者IGF2BP2/IGFBP3的FAM83H-AS1RNA稳定性至关重要。PTBP1-一种RNA结合蛋白-负责CRC中的FAM83H-AS1功能。FAM83H-AS1的外显子4上的T4(1770-2440nt)和T5(2440-2743nt)提供了PTBP1RRM2相互作用的平台。我们的结果表明,m6A修饰通过磷酸化PTBP1对其RNA剪接作用失调FAM83H-AS1致癌作用。在患者来源的异种移植模型中,ASO-FAM83H-AS1显著抑制胃肠道(GI)肿瘤的生长,不仅是CRC,还有GC和ESCC。ASO-FAM83H-AS1和奥沙利铂/顺铂的组合与单独使用任一种药剂的治疗相比显著抑制肿瘤生长。值得注意的是,所有这三种胃肠道癌均有病理完全缓解。我们的研究结果表明,FAM83H-AS1靶向治疗将使主要接受铂类药物治疗的胃肠道肿瘤患者受益。
    LncRNA plays a crucial role in cancer progression and targeting, but it has been difficult to identify the critical lncRNAs involved in colorectal cancer (CRC) progression. We identified FAM83H-AS1 as a tumor-promoting associated lncRNA using 21 pairs of stage IV CRC tissues and adjacent normal tissues. In vitro and in vivo experiments revealed that knockdown of FAM83H-AS1 in CRC cells inhibited tumor proliferation and metastasis, and vice versa. M6A modification is critical for FAM83H-AS1 RNA stability through the writer METTL3 and the readers IGF2BP2/IGFBP3. PTBP1-an RNA binding protein-is responsible for the FAM83H-AS1 function in CRC. T4 (1770-2440 nt) and T5 (2440-2743 nt) on exon 4 of FAM83H-AS1 provide a platform for PTBP1 RRM2 interactions. Our results demonstrated that m6A modification dysregulated the FAM83H-AS1 oncogenic role by phosphorylated PTBP1 on its RNA splicing effect. In patient-derived xenograft models, ASO-FAM83H-AS1 significantly suppressed the growth of gastrointestinal (GI) tumors, not only CRC but also GC and ESCC. The combination of ASO-FAM83H-AS1 and oxaliplatin/cisplatin significantly suppressed tumor growth compared with treatment with either agent alone. Notably, there was pathological complete response in all these three GI cancers. Our findings suggest that FAM83H-AS1 targeted therapy would benefit patients primarily receiving platinum-based therapy in GI cancers.
    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
    这项研究分析了临床数据,全外显子组测序结果,以及发育迟缓和智力障碍儿童的体外小基因功能实验。男病人,4岁,出生后3个月开始出现癫痫发作,并表现出发育迟缓。在1至2岁之间进行康复训练。没有其他重要的家庭病史。通过全面的家族外显子组基因检测,在受影响的儿童中鉴定出OPHN1基因第11外显子的半合子变异:c.1025+1G>A.家庭隔离分析证实了患者母亲中存在这种变异,以前没有报道过。根据ACMG指南,该变异被归类为可能的致病变异.作为对这种变体的回应,设计并进行了体外小基因功能实验,证实突变影响基因mRNA的正常剪接,导致在Intron11的左侧保留56bp。已证实OPHN1:c.1025+1G>A是儿童X连锁智力障碍的致病原因,临床表型包括发育迟缓和癫痫发作。
    This research analyzes the clinical data, whole-exome sequencing results, and in vitro minigene functional experiments of a child with developmental delay and intellectual disability. The male patient, aged 4, began experiencing epileptic seizures at 3 months post-birth and has shown developmental delay. Rehabilitation training was administered between the ages of one and two. There were no other significant family medical histories. Through comprehensive family exome genetic testing, a hemizygous variant in the 11th exon of the OPHN1 gene was identified in the affected child: c.1025 + 1G > A. Family segregation analysis confirmed the presence of this variant in the patient\'s mother, which had not been previously reported. According to the ACMG guidelines, this variant was classified as a likely pathogenic variant. In response to this variant, an in vitro minigene functional experiment was designed and conducted, confirming that the mutation affects the normal splicing of the gene\'s mRNA, resulting in a 56 bp retention on the left side of Intron 11. It was confirmed that OPHN1: c.1025 + 1G > A is the pathogenic cause of X-linked intellectual disabilities in the child, with clinical phenotypes including developmental delay and seizures.
    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
    白细胞介素-17(IL-17)家族细胞因子促进保护性炎症抵抗病原体,而且还促进自身免疫和肿瘤的发展。IL-17对调节性T细胞(Tregs)的直接信号尚未报道,可能有助于解释这些二分法反应。
    我们通过将Foxp3-YFP-Cre小鼠与Il17ra-flox小鼠(Il17raΔTreg小鼠)杂交,在Treg中产生了Il17ra的条件性敲除。随后,我们将Il17raΔTreg小鼠的骨髓细胞过继转移到散发性结直肠癌的小鼠模型(Cdx2-Cre/ApcF/),在结直肠癌中选择性消融IL-17对Tregs的直接信号传导。对来自小鼠结肠直肠肿瘤的纯化Treg进行单细胞RNA测序和批量RNA测序,并与人类肿瘤浸润的Treg细胞进行比较。
    IL-17受体A(IL-17RA)在存在于小鼠肠系膜淋巴结和结肠肿瘤中的Treg中表达。IL-17RA消融,特别是在Tregs中,导致Th17细胞增加,并加剧了肿瘤的发展。机械上,肿瘤浸润性Tregs表现出与其激活相关的独特基因特征,成熟,和抑制函数,并且该签名部分由IL-17向Tregs的直接信号传导支持。为了研究Treg编程的途径,我们发现肿瘤Tregs中IL-17RA的缺失导致RNA剪接减少,和下调几种RNA结合蛋白,已知这些蛋白可以调节可变剪接并促进Treg功能。
    IL-17直接向Tregs发出信号,并促进其成熟和功能。该信号传导途径构成了一个负反馈回路,可控制CRC中的促癌炎症。
    UNASSIGNED: Interleukin-17 (IL-17) family cytokines promote protective inflammation for pathogen resistance, but also facilitate autoimmunity and tumor development. A direct signal of IL-17 to regulatory T cells (Tregs) has not been reported and may help explain these dichotomous responses.
    UNASSIGNED: We generated a conditional knockout of Il17ra in Tregs by crossing Foxp3-YFP-Cre mice to Il17ra-flox mice (Il17ra ΔTreg mice). Subsequently, we adoptively transferred bone marrow cells from Il17ra ΔTreg mice to a mouse model of sporadic colorectal cancer (Cdx2-Cre +/Apc F/+), to selectively ablate IL-17 direct signaling on Tregs in colorectal cancer. Single cell RNA sequencing and bulk RNA sequencing were performed on purified Tregs from mouse colorectal tumors, and compared to those of human tumor infiltrating Treg cells.
    UNASSIGNED: IL-17 Receptor A (IL-17RA) is expressed in Tregs that reside in mouse mesenteric lymph nodes and colon tumors. Ablation of IL-17RA, specifically in Tregs, resulted in increased Th17 cells, and exacerbated tumor development. Mechanistically, tumor-infiltrating Tregs exhibit a unique gene signature that is linked to their activation, maturation, and suppression function, and this signature is in part supported by the direct signaling of IL-17 to Tregs. To study pathways of Treg programming, we found that loss of IL-17RA in tumor Tregs resulted in reduced RNA splicing, and downregulation of several RNA binding proteins that are known to regulate alternative splicing and promote Treg function.
    UNASSIGNED: IL-17 directly signals to Tregs and promotes their maturation and function. This signaling pathway constitutes a negative feedback loop that controls cancer-promoting inflammation in CRC.
    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
    目的:集落刺激因子1受体(CSF1R)相关性白质脑病是由CSF1R基因突变引起的一种进展迅速的神经退行性疾病。本研究旨在鉴定和研究CSF1R的一个新的内含子突变(c.1754-3C>G)对剪接的影响。
    方法:使用全外显子组测序鉴定了一种新的内含子突变。为了研究这种突变的影响,我们使用各种生物信息学工具来分析CSF1R基因的转录及其编码蛋白的三维结构。此外,进行逆转录聚合酶链反应(RT-PCR)以验证研究结果.
    结果:在CSF1R中发现了一个新的突变(c.1754-3C>G),由于3'剪接位点共有序列NYAG/G的破坏,导致外显子13跳跃。通过RT-PCR和Sanger测序在突变携带者的外周血中进一步验证了该外显子跳跃事件。蛋白质结构预测表明酪氨酸激酶结构域的破坏,截短的蛋白质显示出显著的结构改变。
    结论:我们的发现强调了内含子错误剪接突变在CSF1R相关白质脑病诊断和治疗中的重要性。
    OBJECTIVE: Colony stimulating factor 1 receptor (CSF1R)-related leukoencephalopathy is a rapidly progressing neurodegenerative disease caused by CSF1R gene mutations. This study aimed to identify and investigate the effect of a novel intronic mutation (c.1754-3C>G) of CSF1R on splicing.
    METHODS: A novel intronic mutation was identified using whole-exome sequencing. To investigate the impact of this mutation, we employed various bioinformatics tools to analyze the transcription of the CSF1R gene and the three-dimensional structure of its encoded protein. Furthermore, reverse transcription polymerase chain reaction (RT-PCR) was performed to validate the findings.
    RESULTS: A novel mutation (c.1754-3C>G) in CSF1R was identified, which results in exon 13 skipping due to the disruption of the 3\' splice site consensus sequence NYAG/G. This exon skipping event was further validated in the peripheral blood of the mutation carrier through RT-PCR and Sanger sequencing. Protein structure prediction indicated a disruption in the tyrosine kinase domain, with the truncated protein showing significant structural alterations.
    CONCLUSIONS: Our findings underscore the importance of intronic mis-splicing mutations in the diagnosis and management of CSF1R-related leukoencephalopathy.
    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
    U4snRNP在前信使RNA(mRNA)剪接中的重要作用已被充分确立。在这项研究中,我们利用特异性靶向U4snRNA的反义吗啉代寡核苷酸(AMO)在HeLa细胞中实现U4snRNP的功能性敲低。我们的结果表明,这种敲除导致全球内含子过早裂解和聚腺苷酸化(PCPA)事件,与U1AMO治疗观察到的效果相当,如mRNA3'-seq分析所示。此外,我们的研究表明,这可能是人类和小鼠细胞系中的常见现象。此外,我们表明U4AMO处理破坏了转录延伸,通过RNAPII的染色质免疫沉淀测序(ChIP-seq)分析证明。总的来说,我们的结果确定了U4snRNP在抑制PCPA中的独特作用,并表明了一个模型,其中剪接在共转录mRNA加工的背景下固有地抑制内含子切割和聚腺苷酸化.
    The essential role of U4 snRNP in pre-messenger RNA (mRNA) splicing has been well established. In this study, we utilized an antisense morpholino oligonucleotide (AMO) specifically targeting U4 snRNA to achieve functional knockdown of U4 snRNP in HeLa cells. Our results showed that this knockdown resulted in global intronic premature cleavage and polyadenylation (PCPA) events, comparable to the effects observed with U1 AMO treatment, as demonstrated by mRNA 3\'-seq analysis. Furthermore, our study suggested that this may be a common phenomenon in both human and mouse cell lines. Additionally, we showed that U4 AMO treatment disrupted transcription elongation, as evidenced by chromatin immunoprecipitation sequencing (ChIP-seq) analysis for RNAPII. Collectively, our results identified a unique role for U4 snRNP in the inhibition of PCPA and indicated a model wherein splicing intrinsically inhibits intronic cleavage and polyadenylation in the context of cotranscriptional mRNA processing.
    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
    背景:对Euglenida中核内含子的研究不足。本研究旨在通过识别Euglenagracilis中的大量内含子来研究Euglenida中的核内含子(E.gracilis),包括顺式剪接的常规和非常规内含子,以及交叉剪接的外突。我们还检查了这些内含子的序列特征。
    结果:共鉴定出28,337个内含子和11,921个外端。常规和非常规内含子具有不同的剪接位点特征;前者是典型的GT/C-AG剪接位点,而后者能够与其末端序列形成结构化基序。我们观察到短内含子对规范的GT-AG内含子具有偏好。值得注意的是,普通E.gracilis中的常规内含子和反离子表现出明显的富含胞苷的聚嘧啶束,与在其他生物体中观察到的富含胸苷的区域相反。此外,E.gracilis中的SL-RNA,以及其他反式剪接物种,可以与相应的U6形成一个最近发现的主题,称为扩展的U6/5\'ss双工。我们还描述了一种新型的可变剪接模式。确定了该原生生物中内含子的串联重复序列,它们的含量与人类相当。
    结论:我们的发现突出了E.gracilis内含子的独特特征,并提供了对这些内含子剪接机制的见解,以及Euglenida的基因组学和进化。
    BACKGROUND: Nuclear introns in Euglenida have been understudied. This study aimed to investigate nuclear introns in Euglenida by identifying a large number of introns in Euglena gracilis (E. gracilis), including cis-spliced conventional and nonconventional introns, as well as trans-spliced outrons. We also examined the sequence characteristics of these introns.
    RESULTS: A total of 28,337 introns and 11,921 outrons were identified. Conventional and nonconventional introns have distinct splice site features; the former harbour canonical GT/C-AG splice sites, whereas the latter are capable of forming structured motifs with their terminal sequences. We observed that short introns had a preference for canonical GT-AG introns. Notably, conventional introns and outrons in E. gracilis exhibited a distinct cytidine-rich polypyrimidine tract, in contrast to the thymidine-rich tracts observed in other organisms. Furthermore, the SL-RNAs in E. gracilis, as well as in other trans-splicing species, can form a recently discovered motif called the extended U6/5\' ss duplex with the respective U6s. We also describe a novel type of alternative splicing pattern in E. gracilis. The tandem repeat sequences of introns in this protist were determined, and their contents were comparable to those in humans.
    CONCLUSIONS: Our findings highlight the unique features of E. gracilis introns and provide insights into the splicing mechanism of these introns, as well as the genomics and evolution of Euglenida.
    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
    转移是晚期结直肠癌(CRC)治疗失败的关键因素之一。转移性CRC经常对化学治疗剂产生抗性。这项研究旨在从涉及肿瘤转移和化学抗性的长链非编码RNA(lncRNAs)编码的“隐藏”蛋白中鉴定新的调节因子。方法:采用CRISPR/Cas9文库功能筛查来鉴定高侵袭性CRC模型中癌症转移的关键抑制剂。西方印迹,免疫荧光染色,入侵,迁移,伤口愈合,WST-1,集落形成,功能增益和丧失实验,体内实验转移模型,多重免疫组织化学染色,免疫组织化学,qRT-PCR,和RT-PCR用于评估FOXP3,PRDM16-DT,HNRNPA2B1和L-CHEK2。RNA测序,免疫共沉淀,qRT-PCR,RT-PCR,RNA亲和纯化,RNA免疫沉淀,MeRIP-定量PCR,荧光原位杂交,进行染色质免疫沉淀和荧光素酶报告基因测定以获得对PRDM16-DT在癌症转移和化学耐药性中的作用的机制见解。通过体内选择建立了耐奥沙利铂的CRC细胞系。WST-1,集落形成,入侵,迁移,Biacore技术,使用功能增益和功能丧失实验以及体内实验转移模型来确定升麻苷H-1在CRC中的功能和机制。结果:新蛋白PRDM16-DT,由LINC00982编码,被鉴定为癌症转移和化学耐药抑制因子。PRDM16-DT水平下调与CRC患者的恶性表型和不良预后呈正相关。在FOXP3的转录调控下,PRDM16-DT直接与HNRNPA2B1相互作用,并竞争性降低HNRNPA2B1与CHEK2外显子9的结合,从而形成长CHEK2(L-CHEK2),随后促进E-cadherin分泌。PRDM16-DT诱导的E-cadherin分泌抑制成纤维细胞活化,进而通过减少MMP9分泌抑制CRC转移。菊苣苷H-1,一种天然化合物,可以结合FOXP3的LEU89,HIS91和LEU92,并显着上调PRDM16-DT表达以抑制CRC转移并逆转奥沙利铂耐药。结论:lncRNALINC00982可表达一种新的蛋白PRDM16-DT,在CRC的肿瘤转移和耐药中起新的调控作用。升麻苷H-1可作用于PRDM16-DT信号通路的上游,减轻肿瘤化疗耐药。
    Metastasis is one of the key factors of treatment failure in late-stage colorectal cancer (CRC). Metastatic CRC frequently develops resistance to chemotherapeutic agents. This study aimed to identify the novel regulators from \"hidden\" proteins encoded by long noncoding RNAs (lncRNAs) involved in tumor metastasis and chemoresistance. Methods: CRISPR/Cas9 library functional screening was employed to identify the critical suppressor of cancer metastasis in highly invasive CRC models. Western blotting, immunofluorescence staining, invasion, migration, wound healing, WST-1, colony formation, gain- and loss-of-function experiments, in vivo experimental metastasis models, multiplex immunohistochemical staining, immunohistochemistry, qRT-PCR, and RT-PCR were used to assess the functional and clinical significance of FOXP3, PRDM16-DT, HNRNPA2B1, and L-CHEK2. RNA-sequencing, co-immunoprecipitation, qRT-PCR, RT-PCR, RNA affinity purification, RNA immunoprecipitation, MeRIP-quantitative PCR, fluorescence in situ hybridization, chromatin immunoprecipitation and luciferase reporter assay were performed to gain mechanistic insights into the role of PRDM16-DT in cancer metastasis and chemoresistance. An oxaliplatin-resistant CRC cell line was established by in vivo selection. WST-1, colony formation, invasion, migration, Biacore technology, gain- and loss-of-function experiments and an in vivo experimental metastasis model were used to determine the function and mechanism of cimicifugoside H-1 in CRC. Results: The novel protein PRDM16-DT, encoded by LINC00982, was identified as a cancer metastasis and chemoresistance suppressor. The down-regulated level of PRDM16-DT was positively associated with malignant phenotypes and poor prognosis of CRC patients. Transcriptionally regulated by FOXP3, PRDM16-DT directly interacted with HNRNPA2B1 and competitively decreased HNRNPA2B1 binding to exon 9 of CHEK2, resulting in the formation of long CHEK2 (L-CHEK2), subsequently promoting E-cadherin secretion. PRDM16-DT-induced E-cadherin secretion inhibited fibroblast activation, which in turn suppressed CRC metastasis by decreasing MMP9 secretion. Cimicifugoside H-1, a natural compound, can bind to LEU89, HIS91, and LEU92 of FOXP3 and significantly upregulated PRDM16-DT expression to repress CRC metastasis and reverse oxaliplatin resistance. Conclusions: lncRNA LINC00982 can express a new protein PRDM16-DT to function as a novel regulator in cancer metastasis and drug resistance of CRC. Cimicifugoside H-1 can act on the upstream of the PRDM16-DT signaling pathway to alleviate cancer chemoresistance.
    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
    胚乳,谷物的主要贮藏器官,决定了粮食的产量和品质。线粒体为干物质积累提供能量,在胚乳发育中。尽管线粒体单链DNA结合蛋白(mtSSB)在维持单链线粒体DNA中起着规范作用,它们在RNA加工和胚乳发育中的分子功能仍然不清楚。这里,我们报告了一个有缺陷的水稻胚乳突变体,粉质胚乳26(flo26),在胚乳中形成异常的淀粉粒。基于图的克隆和互补实验表明,FLO26等位基因编码线粒体单链DNA结合蛋白,命名为mtSSB1.1。mtSSB1.1功能的丧失会影响许多线粒体编码基因的转录水平和nad1的RNA剪接,nad1是线粒体中呼吸链复合物I的核心成分。因此,功能失调的成熟nad1导致复合物I活性急剧下降,从而减少ATP的产生。我们的结果表明,mtSSB1.1通过稳定水稻线粒体RNA的剪接,在维持线粒体功能和胚乳发育中起重要作用。
    Endosperm, the major storage organ in cereal grains, determines the grain yield and quality. Mitochondria provide the energy for dry matter accumulation, in the endosperm development. Although mitochondrial single-stranded DNA-binding proteins (mtSSBs) play a canonical role in the maintenance of single-stranded mitochondrial DNA, their molecular functions in RNA processing and endosperm development remain obscure. Here, we report a defective rice endosperm mutant, floury endosperm26 (flo26), which develops abnormal starch grains in the endosperm. Map-based cloning and complementation experiments showed that FLO26 allele encodes a mitochondrial single-stranded DNA-binding protein, named as mtSSB1.1. Loss of function of mtSSB1.1 affects the transcriptional level of many mitochondrially-encoded genes and RNA splicing of nad1, a core component of respiratory chain complex I in mitochondria. As a result, dysfunctional mature nad1 led to dramatically decreased complex I activity, thereby reducing ATP production. Our results reveal that mtSSB1.1 plays an important role in the maintenance of mitochondrial function and endosperm development by stabilizing the splicing of mitochondrial RNA in rice.
    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
    受精卵的分裂产生全能卵裂球。在人类8细胞卵裂球中,发生合子基因组激活(ZGA)以启动个体发育程序。然而,在人体细胞中捕获和维持全能性构成了重大挑战。这里,我们实现了培养人类全能卵裂球样细胞(hTBLC)。我们发现剪接抑制可以将人类多能干细胞瞬时重编程为ZGA样细胞(ZLCs),其随后在长期传代后转变为稳定的hTBLC。与报道的8细胞样细胞(8CLC)不同,ZLC和hTBLC都广泛沉默多能基因。有趣的是,ZLCs激活一组特定的ZGA特异性基因,和hTBLC富含前ZGA特异性基因。在自发分化过程中,hTBLC重新进入中间ZLC阶段,并进一步产生外爆炸(EPI)-,原始内胚层(PrE)-,和类似滋养外胚层(TE)的谱系,有效地概括了人类植入前的发育。具有胚胎和胚胎外发育能力,hTBLC可以在体外自主产生胚泡样结构而没有外部细胞信号传导。总之,我们的研究提供了人类细胞全能性的关键标准和见解.
    The cleavage of zygotes generates totipotent blastomeres. In human 8-cell blastomeres, zygotic genome activation (ZGA) occurs to initiate the ontogenesis program. However, capturing and maintaining totipotency in human cells pose significant challenges. Here, we realize culturing human totipotent blastomere-like cells (hTBLCs). We find that splicing inhibition can transiently reprogram human pluripotent stem cells into ZGA-like cells (ZLCs), which subsequently transition into stable hTBLCs after long-term passaging. Distinct from reported 8-cell-like cells (8CLCs), both ZLCs and hTBLCs widely silence pluripotent genes. Interestingly, ZLCs activate a particular group of ZGA-specific genes, and hTBLCs are enriched with pre-ZGA-specific genes. During spontaneous differentiation, hTBLCs re-enter the intermediate ZLC stage and further generate epiblast (EPI)-, primitive endoderm (PrE)-, and trophectoderm (TE)-like lineages, effectively recapitulating human pre-implantation development. Possessing both embryonic and extraembryonic developmental potency, hTBLCs can autonomously generate blastocyst-like structures in vitro without external cell signaling. In summary, our study provides key criteria and insights into human cell totipotency.
    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