Transdifferentiation

转分化
  • 文章类型: Journal Article
    使用生长因子和化学物质将间充质干细胞(MSC)转化为神经细胞。尽管这些神经细胞能有效调节疾病症状,它们在替代丢失的神经细胞方面效果较差。直接转分化似乎是产生再生医学应用所需细胞的有希望的方法。Sox2是神经祖细胞(NP)命运决定中的关键转录因子,经常用于将不同细胞类型转分化为NP。这里,我们证明了单个转录因子的过表达,Sox2在人脂肪组织来源的间充质干细胞(hAT-MSCs)中导致产生诱导的NPs样细胞,增殖性和传代性,并显示出分化为三个神经谱系的潜力。NP已知为具有分化为少突胶质细胞的潜能的祖细胞。在体内,移植到脱髓鞘的成年小鼠脑后,他们幸存下来,分化并整合到成人大脑中,同时参与髓鞘再生过程和行为改善。本报告介绍了一个有益的,从可获得的成人来源产生NPs的低成本和有效的方法,用于治疗神经退行性疾病的自体应用,包括多发性硬化症和其他脱髓鞘疾病的髓鞘再生疗法。
    Mesenchymal stem cells (MSCs) are converted to neural cells using growth factors and chemicals. Although these neural cells are effective at modulating disease symptoms, they are less effective at replacing lost neural cells. Direct transdifferentiation seems to be a promising method for generating the required cells for regenerative medicine applications. Sox2 is a key transcription factor in neural progenitor (NP) fate determination and has been frequently used for transdifferentiating different cell types to NPs. Here, we demonstrated that the overexpression of a single transcription factor, Sox2, in human adipose tissue-derived mesenchymal stem cells (hAT-MSCs) led to the generation of induced NPs-like cells that were clonogenic, proliferative and passageable, and showed the potential to differentiate into three neural lineages. NPs are known as progenitors with the potential to differentiate into oligodendrocytes. In vivo, following transplantation into demyelinated adult mouse brains, they survived, differentiated and integrated into the adult brain while participating in the remyelination process and behavioral improvement. This report introduces a beneficial, low-cost and effective approach for generating NPs from an accessible adult source for autologous applications in treating neurodegenerative diseases, including remyelination therapies for multiple sclerosis and other demyelinating diseases.
    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
    背景:怀孕期间胰岛β细胞团扩张,但是潜在的机制还没有完全理解。这项研究检查了怀孕和自助餐厅饮食对胰岛形态的影响,相关的细胞增殖/凋亡率以及β细胞谱系。
    方法:非妊娠和妊娠Ins1Cre/+;Rosa26-eYFP转基因小鼠维持正常或高脂肪自助餐,妊娠18天获得的胰腺组织。胰岛形态的免疫组织化学改变,β-/α-细胞增殖和凋亡,以及胰岛细胞身份,评估新生和导管细胞转分化。
    结果:妊娠正常饮食小鼠显示体重和血糖增加。自助餐厅喂养减轻了这种体重增加,同时引起明显的高血糖。维持正常饮食的妊娠小鼠表现出胰岛和β细胞区域的典型扩张,由于增加的β细胞增殖和存活以及导管向β细胞转分化和β细胞新生,伴随着减少的β细胞去分化。这种妊娠诱导的胰岛适应受到自助餐厅饮食的严重限制。因此,这些小鼠的胰岛表现出高水平的β细胞凋亡和去分化,伴随着β细胞增殖的减少和缺乏妊娠诱导的β细胞新生和转分化,在正常饮食的怀孕小鼠中观察到完全相反的胰岛细胞修饰。
    结论:妊娠期间β细胞群的增加是通过各种机制产生的,包括现有β细胞的增殖和存活,导管细胞的转分化以及β细胞新生。值得注意的是,自助餐厅喂养几乎完全取消了妊娠诱导的胰岛适应,这可能有助于在饮食引起的代谢应激的情况下妊娠糖尿病的发展。
    BACKGROUND: Pancreatic islet β-cell mass expands during pregnancy, but underlying mechanisms are not fully understood. This study examines the impact of pregnancy and cafeteria diet on islet morphology, associated cellular proliferation/apoptosis rates as well as β-cell lineage.
    METHODS: Non-pregnant and pregnant Ins1Cre/+;Rosa26-eYFP transgenic mice were maintained on either normal or high-fat cafeteria diet, with pancreatic tissue obtained at 18 days gestation. Immunohistochemical changes in islet morphology, β-/α-cell proliferation and apoptosis, as well as islet cell identity, neogenesis and ductal cell transdifferentiation were assessed.
    RESULTS: Pregnant normal diet mice displayed an increase in body weight and glycaemia. Cafeteria feeding attenuated this weight gain while causing overt hyperglycaemia. Pregnant mice maintained on a normal diet exhibited typical expansion in islet and β-cell area, owing to increased β-cell proliferation and survival as well as ductal to β-cell transdifferentiation and β-cell neogenesis, alongside decreased β-cell dedifferentiation. Such pregnancy-induced islet adaptations were severely restricted by cafeteria diet. Accordingly, islets from these mice displayed high levels of β-cell apoptosis and dedifferentiation, together with diminished β-cell proliferation and lack of pregnancy-induced β-cell neogenesis and transdifferentiation, entirely opposing islet cell modifications observed in pregnant mice maintained on a normal diet.
    CONCLUSIONS: Augmentation of β-cell mass during gestation arises through various mechanisms that include proliferation and survival of existing β-cells, transdifferentiation of ductal cells as well as β-cell neogenesis. Remarkably, cafeteria feeding almost entirely annuls pregnancy-induced islet adaptations, which may contribute to the development of gestational diabetes in the setting of dietary provoked metabolic 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
    在开发的第一周,人类胚胎形成由内部细胞团和滋养外胚层(TE)细胞组成的胚泡,后者是胎盘滋养层的祖细胞。这里,我们研究了从胚泡早期到晚期的人TE中转录本的表达。我们鉴定了转录因子GATA2,GATA3,TFAP2C和KLF5的富集,并表征了它们在TE发育过程中的蛋白质表达动力学。通过诱导型过表达和mRNA转染,我们确定这些因素,和MYC一起,足以从引发的人胚胎干细胞建立诱导的滋养层干细胞(iTSC)。这些iTSCs自我更新并概括了形态学特征,基因表达谱,和定向分化潜力,与现有的人类TSC相似。每个系统的遗漏,或多种因素的组合,揭示了GATA2和GATA3对iTSC转分化的重要性。总之,这些发现提供了对可能在人类TE中起作用的转录因子网络的见解,并拓宽了建立早期人类胎盘祖细胞细胞模型的方法,这可能在未来对胎盘相关疾病的模型是有用的。
    During the first week of development, human embryos form a blastocyst composed of an inner cell mass and trophectoderm (TE) cells, the latter of which are progenitors of placental trophoblast. Here, we investigated the expression of transcripts in the human TE from early to late blastocyst stages. We identified enrichment of the transcription factors GATA2, GATA3, TFAP2C and KLF5 and characterised their protein expression dynamics across TE development. By inducible overexpression and mRNA transfection, we determined that these factors, together with MYC, are sufficient to establish induced trophoblast stem cells (iTSCs) from primed human embryonic stem cells. These iTSCs self-renew and recapitulate morphological characteristics, gene expression profiles, and directed differentiation potential, similar to existing human TSCs. Systematic omission of each, or combinations of factors, revealed the crucial importance of GATA2 and GATA3 for iTSC transdifferentiation. Altogether, these findings provide insights into the transcription factor network that may be operational in the human TE and broaden the methods for establishing cellular models of early human placental progenitor cells, which may be useful in the future to model placental-associated diseases.
    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
    恶性黑色素瘤是一种强大的肿瘤,起源于神经c起源的黑素细胞,在不同的解剖位置发现,主要在皮肤上,其次是眼睛和粘膜。这种肿瘤由于其显著的表型多样性而突出。转分化,分化为不同于肿瘤起源的细胞系的过程,和表型可塑性,以行为变化为特征,形态学,和生理响应不同的环境条件,可能会使黑色素瘤成为不小心的病理学家的诊断难题。在这个案例报告中,我们提出了一个具有挑战性的黑色素瘤软骨转分化的案例,以阐明其临床,病态,和分子方面。
    Malignant melanoma is a formidable tumor originating from melanocytes of neural crest origin, found in various anatomical locations, primarily in the skin, followed by the eyes and mucosal membranes. This tumor stands out due to its remarkable phenotypic diversity. Transdifferentiation, the process of differentiation into cell lineages other than the one from which the tumor originated, and phenotypic plasticity, characterized by changes in behavior, morphology, and physiology in response to different environmental conditions, can make melanoma a diagnostic conundrum for unwary pathologists. In this case report, we present a challenging case of melanoma with cartilaginous transdifferentiation to shed light on its clinical, pathological, and molecular aspects.
    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
    亨廷顿病(HD)是一种无法治愈的遗传性疾病,由编码突变亨廷顿蛋白(mHTT)的HTT基因中CAG重复序列的扩展引起。尽管在细胞和动物模型中进行了大量研究,mHTT的生物学作用及其对纹状体神经元的毒性的潜在机制尚未建立,迄今为止,尚无针对HD患者的有效治疗方法.我们产生并表征了一种新的真皮成纤维细胞(HDDF,亨廷顿病皮肤成纤维细胞)来自确诊的HD患者。我们还研究了HDDF细胞的生长特性,为规范标记染色,对这些细胞进行核型分析,并调查了他们的表型。HDDF细胞通过转分化成功地重编程为诱导的纹状体神经元。新的成纤维细胞系可用作细胞模型,以研究mHTT的生物学作用以及通过重编程技术从其获得的成纤维细胞和诱导的神经元细胞中HD发病机理的表现。
    Huntington\'s disease (HD) is an incurable hereditary disease caused by expansion of the CAG repeats in the HTT gene encoding the mutant huntingtin protein (mHTT). Despite numerous studies in cellular and animal models, the mechanisms underlying the biological role of mHTT and its toxicity to striatal neurons have not yet been established and no effective therapy for HD patients has been developed so far. We produced and characterized a new line of dermal fibroblasts (HDDF, Huntington\'s disease dermal fibroblasts) from a patient with a confirmed HD diagnosis. We also studied the growth characteristics of HDDF cells, stained them for canonical markers, karyotyped these cells, and investigated their phenotype. HDDF cells was successfully reprogrammed into induced striatal neurons via transdifferentiation. The new fibroblast line can be used as a cell model to study the biological role of mHTT and manifestations of HD pathogenesis in both fibroblasts and induced neuronal cells obtained from them by reprogramming techniques.
    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
    含CKLF样MARVEL跨膜结构域3(CMTM3),CMTM家族的一个成员,与肿瘤的发生和进展密切相关,在免疫系统中起着至关重要的作用,心血管系统,男性生殖系统。最近,CMTM3已成为治疗与骨形成相关的疾病的潜在靶标。然而,需要更多的研究来了解CMTM3调节成骨分化过程的机制.在这项研究中,我们观察到在BMP4诱导的C2C12成肌细胞向成骨细胞转分化过程中Cmtm3表达的显著下调。Cmtm3过表达抑制BMP4诱导的C2C12细胞的增殖和成骨分化,而它的击倒反过来促进了这一过程。机械上,Cmtm3过表达上调p53和p21的蛋白质和mRNA水平。相反,Cmtm3敲除产生相反的效果。此外,我们发现Cmtm3与p53相互作用,并通过抑制蛋白酶体介导的泛素化和降解增加蛋白质稳定性.值得注意的是,Trp53下调消除了Cmtm3对BMP4诱导的C2C12成肌细胞增殖和成骨分化的抑制作用。总的来说,我们的发现为CMTM3在调节成肌细胞增殖和转分化为成骨细胞中的作用提供了关键见解,强调其在成骨研究中的意义。
    CKLF-like MARVEL transmembrane domain-containing 3 (CMTM3), a member of the CMTM family that is closely related to tumor occurrence and progression, plays crucial roles in the immune system, cardiovascular system, and male reproductive system. Recently, CMTM3 has emerged as a potential target for treating diseases related to bone formation. However, additional studies are needed to understand the mechanisms by which CMTM3 regulates the process of osteogenic differentiation. In this study, we observed a significant downregulation of Cmtm3 expression during the transdifferentiation of C2C12 myoblasts into osteoblasts induced by BMP4. Cmtm3 overexpression suppressed proliferation and osteogenic differentiation in BMP4-induced C2C12 cells, whereas its knockdown conversely facilitated the process. Mechanistically, Cmtm3 overexpression upregulated both the protein and mRNA levels of p53 and p21. Conversely, Cmtm3 knockdown exerted the opposite effects. Additionally, we found that Cmtm3 interacts with p53 and increases protein stability by inhibiting proteasome-mediated ubiquitination and degradation. Notably, Trp53 downregulation abrogated the inhibitory effect of Cmtm3 on BMP4-induced proliferation and osteogenic differentiation of C2C12 myoblasts. Collectively, our findings provide key insights into the role of CMTM3 in regulating myoblast proliferation and transdifferentiation into osteoblasts, highlighting its significance in osteogenesis research.
    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
    帕金森病(PD)是一种神经退行性疾病,由大脑中产生多巴胺的神经元丢失引起,引起运动症状,如震颤和僵硬。虽然目前的治疗方法如药物治疗和脑深部刺激可以缓解症状,他们没有解决神经元丢失的根本原因。因此,细胞替代疗法成为一种有前途的治疗策略。然而,临床相关数量的可植入多巴胺能(DA)细胞的产生仍然是一个挑战。细胞重编程技术的最新进展为产生治疗量的所需类型的患者特异性细胞开辟了广阔的可能性。主要的细胞重编程策略涉及通过病毒转导或转染强制表达单个或一组基因。或者通过小分子,被称为化学方法,这是一个更容易和更安全的方法。在我们之前的研究中,使用小分子方法(表观遗传修饰剂和SMAD抑制剂如Dorsomorphin和SB431542的组合),我们已经能够从人类间充质干细胞(hMSCs)产生DA祖细胞。这项研究的目的是进一步改进DA祖细胞的生成方法,并在帕金森病动物模型中测试其治疗效果。结果表明,向我们的DA细胞诱导方案中添加自噬增强剂(AE)进一步提高了DA祖细胞的产量。结果还表明,将DA祖细胞移植到PD小鼠模型中存活,集成,改善PD运动症状。这些数据表明化学产生的DA细胞可以是非常有前途和安全的PD细胞治疗剂。
    Parkinson\'s disease (PD) is a neurodegenerative disorder resulting from the loss of dopamine-producing neurons in the brain, causing motor symptoms like tremors and stiffness. Although current treatments like medication and deep brain stimulation can alleviate symptoms, they don\'t address the root cause of neuron loss. Therefore, cell replacement therapy emerges as a promising treatment strategy. However, the generation of engraftable dopaminergic (DA) cells in clinically relevant quantities is still a challenge. Recent advances in cell reprogramming technologies open up vast possibilities to produce patient-specific cells of a desired type in therapeutic quantities. The main cell reprogramming strategies involve the enforced expression of individual or sets of genes through viral transduction or transfection, or through small molecules, known as the chemical approach, which is a much easier and safer method. In our previous studies, using a small molecule approach (combinations of epigenetic modifiers and SMAD inhibitors such asDorsomorphin and SB431542), we have been able to generate DA progenitors from human mesenchymal stem cells (hMSCs). The aim of this study was to further improve the method for the generation of DA progenitors and to test their therapeutic effect in an animal model of Parkinson\'s. The results showed that the addition of an autophagy enhancer (AE) to our DA cell induction protocol further increased the yield of DA progenitor cells. The results also showed that DA progenitors transplanted into the mouse model of PD survived, integrated, and improved PD motor symptoms. These data suggest that chemically-produced DA cells can be very promising and safe cellular therapeutics for PD.
    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
    目的:II型糖尿病(T2D)源于胰岛素抵抗,β细胞功能障碍是其进展的标志。研究表明,β细胞在T2D发育过程中发生凋亡或去分化。转录因子PAX4对β分化和存活至关重要,因此可能是T2D胰岛β细胞功能的潜在增强剂。材料和方法:将人PAX4cDNA与腺病毒载体一起递送到T2D人胰岛中,并检查了其对β细胞的影响。结果:PAX4基因递送显著提高β细胞存活率,T2D人胰岛中的β细胞组成增加。表达PAX4的胰岛中的基础胰岛素和葡萄糖刺激的胰岛素分泌明显高于未处理或对照处理的T2D人胰岛。结论:在T2D人胰岛中引入PAX4表达可改善β细胞功能,因此可以为T2D治疗提供治疗益处。
    II型糖尿病(T2D)由胰岛素抵抗引起,β细胞功能障碍在其进展中起关键作用。β细胞质量和功能的缺陷主要归因于通过凋亡导致的β细胞死亡;然而,最近的研究表明,β细胞衰竭也可能是由β细胞去分化引起的-也就是说,β细胞经历成熟身份的丧失,在T2D发育过程中采用祖细胞样或产生胰高血糖素的α细胞状态。因此,防止β细胞去分化同时促进其存活的策略对于T2D治疗是有益的。在这项研究中,我们探索了PAX4,β分化和存活的关键转录因子,可以减轻T2D患者胰岛β细胞功能障碍。要做到这一点,通过基于腺病毒载体的载体将人PAX4cDNA递送到从T2D供体分离的人胰岛中,Ad5.评价Pax4及其对β细胞功能的影响。结果表明,PAX4的表达显着提高了T2D胰岛中的β细胞存活并增加了β细胞组成。值得注意的是,PAX4处理的T2D胰岛的基础胰岛素分泌和葡萄糖刺激的胰岛素分泌明显高于对照处理的胰岛。数据表明,将PAX4基因递送到T2D人胰岛中可增强β细胞质量和功能,因此可以在T2D的治疗中提供治疗益处。
    Aim: Type II diabetes (T2D) stems from insulin resistance, with β-cell dysfunction as a hallmark in its progression. Studies reveal that β cells undergo apoptosis or dedifferentiation during T2D development. The transcription factor PAX4 is vital for β differentiation and survival, thus may be a potential enhancer of β-cell function in T2D islets. Materials & methods: Human PAX4 cDNA was delivered into T2D human islets with an adenoviral vector, and its effects on β cells were examined. Results: PAX4 gene delivery significantly improved β-cell survival, and increased β-cell composition in the T2D human islets. Basal insulin and glucose-stimulated insulin secretion in PAX4-expressing islets were substantially higher than untreated or control-treated T2D human islets. Conclusion: Introduced PAX4 expression in T2D human islets improves β-cell function, thus could provide therapeutic benefits for T2D treatment.
    Type II diabetes (T2D) results from insulin resistance, with β-cell dysfunction playing a pivotal role in its progression. Deficits in β-cell mass and function have been attributed primarily to β-cell death through apoptosis; however, recent studies suggest β-cell failure can also arise from β-cell dedifferentiation – that is, β cells undergo a loss of mature identity, adopting either progenitor-like or glucagon-producing α cell states during T2D development. Therefore, a strategy preventing β-cell dedifferentiation while promoting its survival is beneficial for T2D treatment. In this study, we explored whether PAX4, a critical transcription factor for β differentiation and survival, could alleviate β-cell dysfunction in human islets derived from T2D patients. To accomplish that, human PAX4 cDNA was delivered into human islets isolated from T2D donors by an adenoviral vector-based vector, Ad5.Pax4 and its effects on β-cell function were evaluated. The results showed PAX4 expression significantly improved β-cell survival and increased β-cell composition in the T2D islets. Notably, PAX4-treated T2D islets exhibited significantly higher basal insulin secretion and glucose-stimulated insulin secretion than control-treated islets. The data demonstrate that PAX4 gene delivery into T2D human islets enhances β-cell mass and function, and thus may offer therapeutic benefits in the treatment of T2D.
    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
    直接重编程为产生功能性内皮细胞(ECs)提供了新的突破,而无需中间干细胞或祖细胞状态。为心血管研究和治疗提供有前途的资源。ETV2是一种关键的转录因子,已被确定为指定内皮谱系的先驱因子。实现精确的ETV2诱导对于有效的内皮重编程至关重要,而维持重编程的细胞表型依赖于生长因子和小分子的特定组合。因此,我们在此提供了一个简单而全面的方案,用于从人真皮成纤维细胞(HDFs)产生两种不同类型的重编程ECs(rECs).早期的rECs表现出强大的新生血管形成特性,但缺乏成熟的EC表型,而晚期rECs表现出与人类出生后ECs的表型相似性,并且具有与早期rECs相似的新血管形成能力。两种细胞类型都可以来自人类体细胞,使它们适合个性化的疾病调查,药物发现,和疾病治疗。
    Direct reprogramming provides a novel breakthrough for generating functional endothelial cells (ECs) without the need for intermediate stem or progenitor states, offering a promising resource for cardiovascular research and treatment. ETV2 is a key transcription factor that has been identified as a pioneering factor for specifying endothelial lineage. Achieving precise ETV2 induction is essential for effective endothelial reprogramming, and maintaining the reprogrammed cellular phenotype relies on a specific combination of growth factors and small molecules. Thus, we hereby provide a straightforward and comprehensive protocol for generating two distinct types of reprogrammed ECs (rECs) from human dermal fibroblasts (HDFs). Early rECs demonstrate a robust neovascularization property but lack the mature EC phenotype, while late rECs exhibit phenotypical similarity to human postnatal ECs and have a neovascularization capacity similar to early rECs. Both cell types can be derived from human somatic source cells, making them suitable for personalized disease investigations, drug discovery, and disease therapy.
    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
    胰腺内的β细胞在胰岛素的产生和分泌中起着关键作用。对血糖水平波动的反应。然而,肥胖等因素,饮食习惯,长期的胰岛素抵抗会损害β细胞功能,有助于2型糖尿病(T2D)的发展。这种功能障碍的一个关键方面涉及β细胞去分化和转分化,其中这些细胞失去了它们的专门特征并采用不同的身份,特别是向祖细胞或其他胰腺细胞类型如α细胞的转变。这个过程显着导致β细胞功能障碍和T2D的进展,通常超过β细胞完全丢失的影响。β细胞特有的特定基因和转录因子表达的改变,随着表观遗传修饰和环境因素如炎症,氧化应激,和线粒体功能障碍,支持β细胞去分化和T2D的发生。最近的研究强调了靶向β细胞去分化以有效管理T2D的潜在治疗价值。在这次审查中,我们的目标是剖析控制β细胞去分化的复杂机制,并探索源于这些见解的治疗途径。
    The β-cells within the pancreas play a pivotal role in insulin production and secretion, responding to fluctuations in blood glucose levels. However, factors like obesity, dietary habits, and prolonged insulin resistance can compromise β-cell function, contributing to the development of Type 2 Diabetes (T2D). A critical aspect of this dysfunction involves β-cell dedifferentiation and transdifferentiation, wherein these cells lose their specialized characteristics and adopt different identities, notably transitioning towards progenitor or other pancreatic cell types like α-cells. This process significantly contributes to β-cell malfunction and the progression of T2D, often surpassing the impact of outright β-cell loss. Alterations in the expressions of specific genes and transcription factors unique to β-cells, along with epigenetic modifications and environmental factors such as inflammation, oxidative stress, and mitochondrial dysfunction, underpin the occurrence of β-cell dedifferentiation and the onset of T2D. Recent research underscores the potential therapeutic value for targeting β-cell dedifferentiation to manage T2D effectively. In this review, we aim to dissect the intricate mechanisms governing β-cell dedifferentiation and explore the therapeutic avenues stemming from these insights.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号