Induced pluripotent stem cells (iPSCs)

诱导多能干细胞
  • 文章类型: Journal Article
    马凡氏综合征(MFS)是由FBN1基因突变引起的遗传性疾病。FBN1基因位点的基因突变影响编码蛋白的功能,纤溶蛋白1,一种在结缔组织中发现的形成微纤维的结构分子。MFS患者会出现严重的心血管并发症,包括胸主动脉瘤和主动脉夹层,这使得他们过早死亡的风险增加。这里,我们产生了两个在FBN1基因中具有突变的诱导多能干细胞(iPSC)系(p.C1942C>A和c.1954T>C),直接来源于MFS患者。我们已经表明,两个iPSC系都显示多能性标记的表达,正常核型和三系分化能力,代表了一种有价值的工具,用于确定干预这种疾病的新治疗策略。
    Marfan syndrome (MFS) is a hereditary condition caused by mutations in the FBN1 gene. Genetic mutations in the FBN1 locus impact the function of the encoded protein, Fibrillin 1, a structural molecule forming microfibrils found in the connective tissue. MFS patients develop severe cardiovascular complications including thoracic aortic aneurysm and aortic dissection, which predispose them to an enhanced risk of premature death. Here, we generated two induced pluripotent stem cell (iPSC) lines harboring mutations in the FBN1 gene (p.C1942C>A and c.1954 T>C), directly derived from MFS patients. We have shown that both iPSC lines displayed expression of pluripotency markers, normal karyotype and ability of trilineage differentiation, representing a valuable tool for the identification of new therapeutic strategies for intervening in this disease.
    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
    载脂蛋白E4(APOE4)等位基因是阿尔茨海默病(AD)的主要遗传危险因素。相比之下,已知APOE2可以降低AD风险,APOE3被定义为风险中性。APOE在大脑的生物能量稳态中起着重要作用,在AD患者的大脑中已经检测到早期代谢变化。尽管APOE主要由大脑中的星形胶质细胞表达,神经元也被证明是APOE的来源。然而,三种APOE亚型在神经元能量稳态中的不同作用仍然知之甚少。在这项研究中,我们从APOE等基因诱导的多能干细胞(iPSCs)中产生了纯的人神经元(iN细胞),表达APOE2,APOE3,APOE4或携带APOE敲除(KO)以研究APOE同工型对神经元能量代谢的特异性影响。我们表明,内源性产生的APOE4增强了APOE等基因iN细胞中的线粒体ATP产生,但在相应的iPS细胞系中却没有。这种效应既不与线粒体裂变或融合蛋白的表达水平相关,也不与APOE的细胞内或分泌水平相关。APOE2,APOE3和APOE4iN细胞相似。APOE-KOiN细胞中的ATP产生和基础呼吸与APOE4强烈不同,更类似于APOE2和APOE3iN细胞。表明APOE4的功能获得机制,而不是功能丧失。一起来看,我们在APOE同基因iN细胞中的发现揭示了APOE基因型依赖性和神经元特异性的氧化能量代谢调节。
    The apolipoprotein E4 (APOE4) allele represents the major genetic risk factor for Alzheimer\'s disease (AD). In contrast, APOE2 is known to lower the AD risk, while APOE3 is defined as risk neutral. APOE plays a prominent role in the bioenergetic homeostasis of the brain, and early-stage metabolic changes have been detected in the brains of AD patients. Although APOE is primarily expressed by astrocytes in the brain, neurons have also been shown as source for APOE. However, the distinct roles of the three APOE isoforms in neuronal energy homeostasis remain poorly understood. In this study, we generated pure human neurons (iN cells) from APOE-isogenic induced pluripotent stem cells (iPSCs), expressing either APOE2, APOE3, APOE4, or carrying an APOE knockout (KO) to investigate APOE isoform-specific effects on neuronal energy metabolism. We showed that endogenously produced APOE4 enhanced mitochondrial ATP production in APOE-isogenic iN cells but not in the corresponding iPS cell line. This effect neither correlated with the expression levels of mitochondrial fission or fusion proteins nor with the intracellular or secreted levels of APOE, which were similar for APOE2, APOE3, and APOE4 iN cells. ATP production and basal respiration in APOE-KO iN cells strongly differed from APOE4 and more closely resembled APOE2 and APOE3 iN cells, indicating a gain-of-function mechanism of APOE4 rather than a loss-of-function. Taken together, our findings in APOE isogenic iN cells reveal an APOE genotype-dependent and neuron-specific regulation of oxidative energy metabolism.
    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:细胞外囊泡(EV)具有阐明肌萎缩侧索硬化症(ALS)发病机理的潜力,并可作为生物标志物。值得注意的是,散发性ALS(SALS)患者的血清(sEVs)和脑脊液(CSF;cEVs)中EV蛋白谱的比较和纵向变化仍然未知。盐酸罗匹尼罗(ROPI;多巴胺D2受体[D2R]激动剂),通过基于诱导多能干细胞(iPSC)的药物发现确定了一种新的抗ALS药物候选物,在肌萎缩性侧索硬化(ROPALS)试验中,盐酸罗匹尼罗补救措施被建议抑制ALS疾病的进展,但是它的作用机制还不清楚。因此,我们试图揭示疾病进展的纵向变化以及ROPI对EV蛋白谱的影响.
    方法:我们以固定间隔从10名对照和20名参与ROPALS试验的SALS患者中收集血清和CSF。电动汽车的综合蛋白质组学分析,从这些样本中提取,使用液相色谱/质谱仪(LC/MS)进行。此外,我们产生了iPSC来源的星形胶质细胞(iPasts),并在有或没有ROPI治疗的情况下对星形胶质细胞进行了RNA测序.
    结果:研究结果揭示了与疾病状态有关的sEV和cEV蛋白谱的显著差异和高度一致性,时间和ROPI管理。在Sals,sEV和cEV均呈现炎症相关蛋白水平升高,但与未折叠蛋白反应(UPR)相关的水平降低.这些结果反映了疾病发作后的纵向变化,并与采样时修订的ALS功能评定量表(ALSFRS-R)相关。提示与SALS的发病和进展有关。ROPI似乎抵消了这些变化,在SALS中降低炎症相关蛋白水平并提高与UPR相关的蛋白水平,提出了抗ALS对EV蛋白谱的影响。使用iPast的反向翻译研究表明,这些变化可能部分反映了ROPI的DRD2依赖性神经炎症抑制作用。我们还确定了通过机器学习驱动的生物标志物搜索来预测诊断和疾病进展的生物标志物。
    结论:尽管样本量有限,这项研究是报告SALS患者血清和CSFEV时间序列蛋白质组学改变的先驱,提供对SALS发病机制的全面见解,ROPI引起的变化,以及潜在的预后和诊断生物标志物。
    BACKGROUND: Extracellular vesicles (EVs) hold the potential for elucidating the pathogenesis of amyotrophic lateral sclerosis (ALS) and serve as biomarkers. Notably, the comparative and longitudinal alterations in the protein profiles of EVs in serum (sEVs) and cerebrospinal fluid (CSF; cEVs) of sporadic ALS (SALS) patients remain uncharted. Ropinirole hydrochloride (ROPI; dopamine D2 receptor [D2R] agonist), a new anti-ALS drug candidate identified through induced pluripotent stem cell (iPSC)-based drug discovery, has been suggested to inhibit ALS disease progression in the Ropinirole Hydrochloride Remedy for Amyotrophic Lateral Sclerosis (ROPALS) trial, but its mechanism of action is not well understood. Therefore, we tried to reveal longitudinal changes with disease progression and the effects of ROPI on protein profiles of EVs.
    METHODS: We collected serum and CSF at fixed intervals from ten controls and from 20 SALS patients participating in the ROPALS trial. Comprehensive proteomic analysis of EVs, extracted from these samples, was conducted using liquid chromatography/mass spectrometer (LC/MS). Furthermore, we generated iPSC-derived astrocytes (iPasts) and performed RNA sequencing on astrocytes with or without ROPI treatment.
    RESULTS: The findings revealed notable disparities yet high congruity in sEVs and cEVs protein profiles concerning disease status, time and ROPI administration. In SALS, both sEVs and cEVs presented elevated levels of inflammation-related proteins but reduced levels associated with unfolded protein response (UPR). These results mirrored the longitudinal changes after disease onset and correlated with the revised ALS Functional Rating Scale (ALSFRS-R) at sampling time, suggesting a link to the onset and progression of SALS. ROPI appeared to counteract these changes, attenuating inflammation-related protein levels and boosting those tied to UPR in SALS, proposing an anti-ALS impact on EV protein profiles. Reverse translational research using iPasts indicated that these changes may partly reflect the DRD2-dependent neuroinflammatory inhibitory effects of ROPI. We have also identified biomarkers that predict diagnosis and disease progression by machine learning-driven biomarker search.
    CONCLUSIONS: Despite the limited sample size, this study pioneers in reporting time-series proteomic alterations in serum and CSF EVs from SALS patients, offering comprehensive insights into SALS pathogenesis, ROPI-induced changes, and potential prognostic and diagnostic biomarkers.
    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
    开发生产功能性内皮细胞(EC)的可靠方法对于血管生物学和再生医学的发展至关重要。在这项研究中,我们提出了一种简化和有效的方法,用于人诱导多能干细胞(iPSCs)分化为诱导ECs(iECs),该诱导ECs使用强力霉素诱导系统在体外和体内维持进行血管生成的能力,用于ETV2转录因子的瞬时表达.这种方法减轻了直接转染方法的局限性,如mRNA介导的分化,通过简化方案并增强不同干细胞系的可重复性。我们详细介绍了用于多西环素诱导的ETV2表达的iPSCs的产生及其随后分化为iECs。在四天内达到90%以上的效率。通过体外和体内测定,衍生iECs的功能性和表型稳定性得到严格验证.值得注意的是,这些细胞表现出关键的内皮标记和能力,包括体外微生理平台和皮下小鼠模型中血管网络的形成。此外,我们的结果揭示了通过我们的方法产生的iECs和初级ECs之间的紧密转录和蛋白质组排列,证实分化细胞的生物学相关性。我们的归纳方法的高效率和有效性为iPSC衍生的ECs在科学研究中的更广泛的应用和可访问性铺平了道路,为研究内皮生物学和开发基于EC的疗法提供了有价值的工具。
    The development of reliable methods for producing functional endothelial cells (ECs) is crucial for progress in vascular biology and regenerative medicine. In this study, we present a streamlined and efficient methodology for the differentiation of human induced pluripotent stem cells (iPSCs) into induced ECs (iECs) that maintain the ability to undergo vasculogenesis in vitro and in vivo using a doxycycline-inducible system for the transient expression of the ETV2 transcription factor. This approach mitigates the limitations of direct transfection methods, such as mRNA-mediated differentiation, by simplifying the protocol and enhancing reproducibility across different stem cell lines. We detail the generation of iPSCs engineered for doxycycline-induced ETV2 expression and their subsequent differentiation into iECs, achieving over 90% efficiency within four days. Through both in vitro and in vivo assays, the functionality and phenotypic stability of the derived iECs were rigorously validated. Notably, these cells exhibit key endothelial markers and capabilities, including the formation of vascular networks in a microphysiological platform in vitro and in a subcutaneous mouse model. Furthermore, our results reveal a close transcriptional and proteomic alignment between the iECs generated via our method and primary ECs, confirming the biological relevance of the differentiated cells. The high efficiency and effectiveness of our induction methodology pave the way for broader application and accessibility of iPSC-derived ECs in scientific research, offering a valuable tool for investigating endothelial biology and for the development of EC-based therapies.
    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
    干细胞分化为成骨细胞的承诺是一个高度调节和复杂的过程,涉及外在信号和内在转录机制的协调。虽然已经广泛研究了啮齿动物的成骨细胞分化,人类成骨的研究受到细胞来源和现有模型的限制。这里,我们系统地解剖了hPSC来源的成骨细胞,以鉴定参与人类成骨的功能性膜蛋白及其下游转录网络。我们的结果揭示了人类而不是啮齿动物成骨细胞中II型跨膜丝氨酸蛋白酶CORIN的富集。功能分析表明,CORIN耗竭显着损害成骨。全基因组ChIP富集和机制研究表明,p38MAPK介导的CEBPD上调是CORIN调节成骨所必需的。相反,在MSCs中富集的I型跨膜硫酸乙酰肝素蛋白聚糖SDC1通过类似的机制对成骨产生负调节作用。ChIP-seq,批量和单细胞转录组,和功能验证表明CEBPD在控制成骨中起关键作用。总之,我们的发现揭示了以前未被认可的CORIN介导的CEBPD转录组网络在驱动人类成骨细胞谱系承诺中的作用.
    The commitment of stem cells to differentiate into osteoblasts is a highly regulated and complex process that involves the coordination of extrinsic signals and intrinsic transcriptional machinery. While rodent osteoblastic differentiation has been extensively studied, research on human osteogenesis has been limited by cell sources and existing models. Here, we systematically dissect human pluripotent stem cell-derived osteoblasts to identify functional membrane proteins and their downstream transcriptional networks involved in human osteogenesis. Our results reveal an enrichment of type II transmembrane serine protease CORIN in humans but not rodent osteoblasts. Functional analyses demonstrated that CORIN depletion significantly impairs osteogenesis. Genome-wide chromatin immunoprecipitation enrichment and mechanistic studies show that p38 MAPK-mediated CCAAT enhancer binding protein delta (CEBPD) upregulation is required for CORIN-modulated osteogenesis. Contrastingly, the type I transmembrane heparan sulfate proteoglycan SDC1 enriched in mesenchymal stem cells exerts a negative regulatory effect on osteogenesis through a similar mechanism. Chromatin immunoprecipitation-seq, bulk and single-cell transcriptomes, and functional validations indicated that CEBPD plays a critical role in controlling osteogenesis. In summary, our findings uncover previously unrecognized CORIN-mediated CEBPD transcriptomic networks in driving human osteoblast lineage commitment.
    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
    我们的小组从具有广泛已知的红细胞抗原谱的献血者中产生了两个诱导多能干细胞(iPSC)系,用于体外红细胞(RBC)生产。一条生产线旨在在镰状细胞病(SCD)患者中产生红细胞用于输血,而另一种是用来制造红细胞面板试剂的。根据他们的红细胞表型选择了两名献血者,通过高通量DNA阵列分析进一步补充,以获得更全面的红细胞抗原谱。使用非整合质粒载体将来自供体外周血单核细胞的富集的成红细胞群体重编程为iPSC。对iPSC系进行表征并随后进行造血分化。iPSCPB02和iPSCPB12在体外和体内证明了iPSC特征,并保留了每个献血者的红细胞抗原谱的基因型。集落形成细胞测定证实iPSCPB02和iPSCPB12产生造血祖细胞。这两个iPSC系产生了确定的红细胞抗原谱,自我更新能力,和造血分化潜力。随着造血分化的改善,这些细胞可能在未来更有效地分化为红细胞。它们可以作为获得不依赖供体的红细胞和解决输血特定需求的补充方法。
    Our group generated two induced pluripotent stem cell (iPSC) lines for in vitro red blood cell (RBC) production from blood donors with extensively known erythrocyte antigen profiles. One line was intended to give rise to RBCs for transfusions in patients with sickle cell disease (SCD), while the other was developed to create RBC panel reagents. Two blood donors were selected based on their RBC phenotypes, further complemented by high-throughput DNA array analysis to obtain a more comprehensive erythrocyte antigen profile. Enriched erythroblast populations from the donors\' peripheral blood mononuclear cells were reprogrammed into iPSCs using nonintegrative plasmid vectors. The iPSC lines were characterized and subsequently subjected to hematopoietic differentiation. iPSC PB02 and iPSC PB12 demonstrated in vitro and in vivo iPSC features and retained the genotype of each blood donor\'s RBC antigen profile. Colony-forming cell assays confirmed that iPSC PB02 and iPSC PB12 generated hematopoietic progenitors. These two iPSC lines were generated with defined erythrocyte antigen profiles, self-renewal capacity, and hematopoietic differentiation potential. With improvements in hematopoietic differentiation, these cells could potentially be more efficiently differentiated into RBCs in the future. They could serve as a complementary approach for obtaining donor-independent RBCs and addressing specific demands for blood transfusions.
    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
    纤维化间质性肺疾病(FILDs),例如,由于特发性肺纤维化(IPF),是预后不良的慢性进行性疾病。这些疾病的管理具有挑战性,主要集中在抗纤维化药物的进展抑制上。因此,需要新的FILD治疗。近年来,已经针对FILD研究了使用各种干细胞的基于细胞的疗法,间充质干细胞(MSCs)的使用已被广泛报道,临床研究也正在进行中。诱导多能干细胞(iPSC)也被报道在FILD中具有抗纤维化作用;然而,在机制和副作用方面,这些研究不如MSCs。虽然MSCs显示出有效的抗纤维化作用,在供体短缺或细胞传代后增殖能力降低的情况下,需要考虑供体之间质量差异和稳定供应的可能性。iPSC来源的细胞的应用具有克服这些问题的潜力,并且可以导致细胞产品的一致质量和稳定的产品供应。这篇综述概述了iPSCs和FILD,其次是FILD的基于细胞的治疗现状,然后讨论了使用iPSC衍生细胞进行FILD治疗的可能性和观点。
    Fibrosing interstitial lung diseases (FILDs), e.g., due to idiopathic pulmonary fibrosis (IPF), are chronic progressive diseases with a poor prognosis. The management of these diseases is challenging and focuses mainly on the suppression of progression with anti-fibrotic drugs. Therefore, novel FILD treatments are needed. In recent years, cell-based therapy with various stem cells has been investigated for FILD, and the use of mesenchymal stem cells (MSCs) has been widely reported and clinical studies are also ongoing. Induced pluripotent stem cells (iPSCs) have also been reported to have an anti-fibrotic effect in FILD; however, these have not been as well studied as MSCs in terms of the mechanisms and side effects. While MSCs show a potent anti-fibrotic effect, the possibility of quality differences between donors and a stable supply in the case of donor shortage or reduced proliferative capacity after cell passaging needs to be considered. The application of iPSC-derived cells has the potential to overcome these problems and may lead to consistent quality of the cell product and stable product supply. This review provides an overview of iPSCs and FILD, followed by the current status of cell-based therapy for FILD, and then discusses the possibilities and perspectives of FILD therapy with iPSC-derived cells.
    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
    先天性心脏病(CHD)是出生缺陷相关死亡的主要原因。尽管取得了重大进展,冠心病发展的潜在机制是复杂的,由于缺乏效率,仍然难以捉摸,可重复,和平移模型系统。由于种间差异,依赖于动物模型的研究具有固有的局限性。人类诱导多能干细胞(iPSC)已成为疾病建模的有效平台。iPSC允许产生患者特异性体细胞的无限供应,从而促进心血管精准医学的发展。在过去的十年里,研究人员已经开发了将iPSCs分化为多个心血管谱系的协议,以及增强这些细胞的成熟度和功能。随着生理三维心脏器官的发展,iPSC代表了机械解剖CHD的强大平台,并作为未来转化研究的基础。
    Congenital heart disease (CHD) is a leading cause of birth defect-related death. Despite significant advances, the mechanisms underlying the development of CHD are complex and remain elusive due to a lack of efficient, reproducible, and translational model systems. Investigations relied on animal models have inherent limitations due to interspecies differences. Human induced pluripotent stem cells (iPSCs) have emerged as an effective platform for disease modeling. iPSCs allow for the production of a limitless supply of patient-specific somatic cells that enable advancement in cardiovascular precision medicine. Over the past decade, researchers have developed protocols to differentiate iPSCs to multiple cardiovascular lineages, as well as to enhance the maturity and functionality of these cells. With the development of physiologic three-dimensional cardiac organoids, iPSCs represent a powerful platform to mechanistically dissect CHD and serve as a foundation for future translational research.
    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
    动物和植物已经开发出了恢复力机制,可以在其整个生命周期中有效地承受和克服物理损伤和环境挑战。为了维持他们的活力,动物和植物都使用机制来补充受损的细胞,要么直接,涉及成人干细胞的活动,或者间接地,通过诱导恢复到干细胞状态并随后再分化的体细胞的去分化。多年来,干细胞研究一直是动物研究中快速发展的领域,受到其在人类治疗方面有前途的潜力的驱动,包括组织再生和药物开发。一个重大突破是发现了诱导多能干细胞(iPS),通过表达一组有限的转录因子从体细胞重编程。这一发现使得能够产生可分化成特定细胞类型和组织的无限供应的细胞。同样,在过去的十年中,人们对植物干细胞与再生之间的联系产生了浓厚的兴趣,在提高植物性状如产量的需求的推动下,对病原体的抗性和CRISPR/Cas介导的基因编辑提供的机会。在这里,我们讨论干细胞生物学知识如何有益于再生技术,我们推测为植物创建了一个通用的不依赖基因型的iPS细胞系统,以克服再生的顽抗。
    Animals and plants have developed resilience mechanisms to effectively endure and overcome physical damage and environmental challenges throughout their life span. To sustain their vitality, both animals and plants employ mechanisms to replenish damaged cells, either directly, involving the activity of adult stem cells, or indirectly, via dedifferentiation of somatic cells that are induced to revert to a stem cell state and subsequently redifferentiate. Stem cell research has been a rapidly advancing field in animal studies for many years, driven by its promising potential in human therapeutics, including tissue regeneration and drug development. A major breakthrough was the discovery of induced pluripotent stem cells (iPSCs), which are reprogrammed from somatic cells by expressing a limited set of transcription factors. This discovery enabled the generation of an unlimited supply of cells that can be differentiated into specific cell types and tissues. Equally, a keen interest in the connection between plant stem cells and regeneration has been developed in the last decade, driven by the demand to enhance plant traits such as yield, resistance to pathogens, and the opportunities provided by CRISPR/Cas-mediated gene editing. Here we discuss how knowledge of stem cell biology benefits regeneration technology, and we speculate on the creation of a universal genotype-independent iPSC system for plants to overcome regenerative recalcitrance.
    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
    随着特定遗传因素的使用和细胞重编程的最新进展,现在可以从容易获得和常见的体细胞类型产生谱系定型细胞或诱导多能干细胞(iPSC)。然而,对于当前用于重编程细胞的遗传方法的安全性和有效性仍然存在很大的疑问,以及维持干细胞的常规培养方法。靶向特定表观遗传过程的小分子,信号通路,和其他细胞过程可以用作操纵细胞命运以实现所需目标的补充方法。已经发现,越来越多的小分子可以支持谱系分化,维持干细胞自我更新潜能,并通过提高重编程效率或充当基因重编程因子替代品来促进重编程。然而,持续的挑战包括提高重新编程效率,确保小分子的安全,并解决不完全表观遗传重置的问题。小分子iPSC在再生医学和个性化治疗中具有重要的临床应用价值。这篇综述强调了小分子在克服与iPSCs重编程过程相关的挑战方面的多功能性和潜在的安全益处。
    With the use of specific genetic factors and recent developments in cellular reprogramming, it is now possible to generate lineage-committed cells or induced pluripotent stem cells (iPSCs) from readily available and common somatic cell types. However, there are still significant doubts regarding the safety and effectiveness of the current genetic methods for reprogramming cells, as well as the conventional culture methods for maintaining stem cells. Small molecules that target specific epigenetic processes, signaling pathways, and other cellular processes can be used as a complementary approach to manipulate cell fate to achieve a desired objective. It has been discovered that a growing number of small molecules can support lineage differentiation, maintain stem cell self-renewal potential, and facilitate reprogramming by either increasing the efficiency of reprogramming or acting as a genetic reprogramming factor substitute. However, ongoing challenges include improving reprogramming efficiency, ensuring the safety of small molecules, and addressing issues with incomplete epigenetic resetting. Small molecule iPSCs have significant clinical applications in regenerative medicine and personalized therapies. This review emphasizes the versatility and potential safety benefits of small molecules in overcoming challenges associated with the iPSCs reprogramming process.
    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