induced pluripotent stem cell

诱导多能干细胞
  • 文章类型: Editorial
    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
    肌萎缩侧索硬化症(ALS)是成人最常见的运动神经元疾病,导致运动神经元进行性变性,导致肌肉萎缩,呼吸衰竭并最终导致患者死亡。ALS的发病机制复杂,广泛的努力集中在揭示潜在的分子机制上,重点放在垂死的运动神经元上。然而,最近的重点转移到支持神经胶质细胞的人群已经揭示了ALS的巨大贡献和影响,强调需要更详细地探索这一领域。特别是对星形胶质细胞的研究,居住的神经元稳态支持细胞,在ALS中发现了明显的星形细胞功能障碍,因此可以为新的和有希望的治疗切入点提供靶标。在这次审查中,我们提供了一般星形胶质细胞功能的概述,并通过对潜在的潜在分子机制进行分类,总结了有关星形胶质细胞在ALS中的作用的现有文献。我们讨论了星形胶质细胞靶向治疗的当前努力,并强调可用模型的潜力和缺点。
    Amyotrophic lateral sclerosis (ALS) is the most common motor neuron disease in adults, causing progressive degeneration of motor neurons, which results in muscle atrophy, respiratory failure and ultimately death of the patients. The pathogenesis of ALS is complex, and extensive efforts have focused on unravelling the underlying molecular mechanisms with a large emphasis on the dying motor neurons. However, a recent shift in focus towards the supporting glial population has revealed a large contribution and influence in ALS, which stresses the need to explore this area in more detail. Especially studies into astrocytes, the residential homeostatic supporter cells of neurons, have revealed a remarkable astrocytic dysfunction in ALS, and therefore could present a target for new and promising therapeutic entry points. In this review, we provide an overview of general astrocyte function and summarize the current literature on the role of astrocytes in ALS by categorizing the potentially underlying molecular mechanisms. We discuss the current efforts in astrocyte-targeted therapy, and highlight the potential and shortcomings of available models.
    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:尽管多能干细胞(PSC)疗法以快速的速度向临床和商业应用发展,制造再现性和稳健性是监管批准的显著瓶颈。PSC的治疗应用需要在高度稳健的情况下产生大量细胞,定义明确,和经济上可行的条件。小规模和短期工艺优化,然而,通常以线性方式进行,其不考虑验证所使用的生物过程方案和分析方法所需的时间。可重复和强大的生物过程的设计应该是动态的,并且包括持续的努力,以了解该过程在过渡到应力将被放大的大规模生产之前,随着时间的推移和对不同应力的反应。
    方法:本研究采用基线方案,为在Vertical-Wheel®生物反应器中短期培养PSC聚集体而开发,通过长期(连续传代)悬浮培养评估关键过程属性。这样做是为了在用各种市售的培养基和细胞系进行时获得整体过程的稳健性。过程输出变量,包括生长动力学,聚集体形态,收获效率,基因组稳定性,功能多能性通过短期和长期培养进行评估。
    结果:在六天培养期内证明了扩增方案的稳健性质,其中对于测试的所有五种商业培养基,观察到球形聚集体形成和扩增具有高倍扩增。只有通过长期连续扩增和血管内解离操作才能揭示细胞生长和质量的深刻差异。测试的一些商业培养基制剂证明了细胞生长速率的维持,聚集体形态,通过使用多个PSC系的三个生物反应器串联通道和高收获回收效率。在10个连续的生物反应器通道中,持续的生长和质量维护甚至证明了出色的生物过程稳健性。然而,一些测试的商业培养基被证明不太适合在生物反应器中连续传代培养,因为培养物在解离过程中导致细胞裂解,增长率降低,和聚集体形态的损失。
    结论:本研究证明了系统选择和测试生物过程输入变量的重要性,与多个生物过程输出变量通过连续通道创建一个真正的可重复和强大的方案临床和商业PSC生产使用可扩展的生物反应器系统。
    BACKGROUND: While pluripotent stem cell (PSC) therapies move toward clinical and commercial applications at a rapid rate, manufacturing reproducibility and robustness are notable bottlenecks in regulatory approval. Therapeutic applications of PSCs require large cell quantities to be generated under highly robust, well-defined, and economically viable conditions. Small-scale and short-term process optimization, however, is often performed in a linear fashion that does not account for time needed to verify the bioprocess protocols and analysis methods used. Design of a reproducible and robust bioprocess should be dynamic and include a continuous effort to understand how the process will respond over time and to different stresses before transitioning into large-scale production where stresses will be amplified.
    METHODS: This study utilizes a baseline protocol, developed for the short-term culture of PSC aggregates in Vertical-Wheel® bioreactors, to evaluate key process attributes through long-term (serial passage) suspension culture. This was done to access overall process robustness when performed with various commercially available media and cell lines. Process output variables including growth kinetics, aggregate morphology, harvest efficiency, genomic stability, and functional pluripotency were assessed through short and long-term culture.
    RESULTS: The robust nature of the expansion protocol was demonstrated over a six-day culture period where spherical aggregate formation and expansion were observed with high-fold expansions for all five commercial media tested. Profound differences in cell growth and quality were revealed only through long-term serial expansion and in-vessel dissociation operations. Some commercial media formulations tested demonstrated maintenance of cell growth rates, aggregate morphology, and high harvest recovery efficiencies through three bioreactor serial passages using multiple PSC lines. Exceptional bioprocess robustness was even demonstrated with sustained growth and quality maintenance over 10 serial bioreactor passages. However, some commercial media tested proved less equipped for serial passage cultures in bioreactors as cultures led to cell lysis during dissociation, reduction in growth rates, and a loss of aggregate morphology.
    CONCLUSIONS: This study demonstrates the importance of systematic selection and testing of bioprocess input variables, with multiple bioprocess output variables through serial passages to create a truly reproducible and robust protocol for clinical and commercial PSC production using scalable bioreactor systems.
    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
    脊髓损伤(SCI)后,神经再生和回路重建仍然是一个挑战。皮质脊髓锥体神经元具有很强的轴突投射能力。在这项研究中,通过向培养物中添加小分子dorsomorphin,将人诱导多能干细胞(iPSCs)分化为锥体神经元前体(PNP)。在损伤的同一天,将iPSC衍生的PNP急性移植到大鼠挫伤SCI模型中。雕刻后,行为测试显示,与载体对照组相比,SCI大鼠的运动功能显着改善。植入八周后,PNP成熟为皮质脊髓锥体神经元,并延伸轴突进入远处宿主脊髓组织,主要是在尾部方向。病变部位的宿主神经元也将轴突生长到移植物中。作为桥接中继的可能的突触连接可能已经在宿主和移植物衍生的神经元之间形成,如突触前和突触后标记染色和化学遗传调节系统的调节所示。PNP移植物在损伤部位显示出抗炎作用,并且可能使小胶质细胞/巨噬细胞偏向M2表型。此外,PNP移植物是安全的,移植到免疫缺陷小鼠和SCI大鼠中后未检测到肿瘤形成。重建整个病变部位的神经元中继电路并调节SCI微环境的潜力使PNP成为治疗SCI的有希望的细胞候选者。
    Nerve regeneration and circuit reconstruction remain a challenge following spinal cord injury (SCI). Corticospinal pyramidal neurons possess strong axon projection ability. In this study, human induced pluripotent stem cells (iPSCs) were differentiated into pyramidal neuronal precursors (PNPs) by addition of small molecule dorsomorphin into the culture. iPSC-derived PNPs were transplanted acutely into a rat contusion SCI model on the same day of injury. Following engraftment, the SCI rats showed significantly improved motor functions compared with vehicle control group as revealed by behavioral tests. Eight weeks following engraftment, the PNPs matured into corticospinal pyramidal neurons and extended axons into distant host spinal cord tissues, mostly in a caudal direction. Host neurons rostral to the lesion site also grew axons into the graft. Possible synaptic connections as a bridging relay may have been formed between host and graft-derived neurons, as indicated by pre- and post-synaptic marker staining and the regulation of chemogenetic regulatory systems. PNP graft showed an anti-inflammatory effect at the injury site and could bias microglia/macrophages towards a M2 phenotype. In addition, PNP graft was safe and no tumor formation was detected after transplantation into immunodeficient mice and SCI rats. The potential to reconstruct a neuronal relay circuitry across the lesion site and to modulate the microenvironment in SCI makes PNPs a promising cellular candidate for treatment of SCI.
    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
    肥大细胞(MC)来源于CD34+造血祖细胞,由不同的亚型组成,涉及几种炎症。然而,由于缺乏合适的细胞模型系统,我们对人类MC发育轨迹和亚型的理解受到限制。在这里,我们建立了诱导多能干细胞(iPSC)分化的体外模型,以研究人MC分化轨迹.来自骨髓细胞形成复合物(MCFC)的造血细胞的流式细胞术表征显示,在第1-3周内,Lin-CD34造血祖细胞的初始增加,随后是CD34-CD45RA-SSCLow和SSChigh造血细胞的增加。Lin-CD34造血祖细胞由SSClowCD45RA-CD123±c-KitFcERI群体组成,该群体是与CMPFceRI细胞一致的β7-整合素高CD203c和β7-整合素高CD203c-细胞。CD34-Lin-(SSClow)群体的流式细胞术和细胞学分析揭示了下颗粒细胞群体,主要特征为CD45RA-CD123±c-Kit+FcERI-β7-整合素和CD45RA-CD123±c-Kit-FcERI+β7-整合素Mid细胞。超颗粒SSChigh细胞的分析鉴定了Lin-CD34-CD45RA-c-Kit+FceRI-和Lin-CD34-CD45RA-c-Kit+FceRI+细胞。在MCFC培养的第4周收获的细胞的scRNAseq分析显示存在单核细胞和粒细胞祖细胞(n=547个细胞,26.7%),红细胞/未知(n=85,4.1%),中性粒细胞/粒细胞(n=211细胞,10.2%),肥大细胞祖细胞1(n=599,29.1%),肥大细胞祖细胞2(n=152,7.4%),承诺的肥大细胞前体(n=113,5.5%),肥大细胞(n=353,17.1%)。对MC前体和成熟MC种群的计算机分析显示,转录上不同的MC前体亚型和成熟MC状态(CMA1和CMA1-亚型)。在MC成熟培养基(肥大细胞培养基II)中培养MC前体群体导致同源成熟MC群体,高亲和力IgE受体的高表达证明了这一点。变色颗粒,在P物质刺激和FceRI交联后,MC颗粒蛋白(类胰蛋白酶和糜酶)的存在和活化。这种基于人iPSC的方法产生MC前体和表型成熟和功能性MC群体。该系统将是生成人类MC种群并拓宽我们对MC生物学和MC分化轨迹的转录调节的理解的有用模型。
    Mast cells (MCs) are derived from CD34+ hematopoietic progenitors, consist of different subtypes and are involved in several inflammatory conditions. However, our understanding of human MC developmental trajectories and subtypes have been limited by a scarcity of suitable cellular model systems. Herein, we developed an in vitro model of human MC differentiation from induced pluripotent stem cells (iPSC) to study human MC differentiation trajectories. Flow cytometry characterization of hemopoietic cells derived from the myeloid cells-forming complex (MCFC) revealed an initial increase in Lin- CD34+ hematopoietic progenitors within Weeks 1-3, followed by an increase in CD34- CD45RA- SSCLow and SSChigh hematopoietic cells. The Lin- CD34+ hematopoietic progenitors consisted of SSClow CD45RA- CD123± c-Kit+ FcERI+ population that was β7-integrinhigh CD203c+ and β7-integrinhigh CD203c- cells consistent with CMPFceRI+ cells. Flow cytometry and cytologic analyses of the CD34- Lin- (SSClow) population revealed hypogranular cell populations, predominantly characterized by CD45RA- CD123± c-Kit+ FcERI- β7-integrinlow and CD45RA- CD123± c-Kit- FcERI+ β7-integrinMid cells. Analyses of hypergranular SSChigh cells identified Lin- CD34- CD45RA- c-Kit+ FceRI- and Lin- CD34- CD45RA- c-Kit+ FceRI+ cells. scRNA seq analysis of the cells harvested at week 4 of the MCFC culture revealed the presence of monocyte and granulocyte progenitors (n = 547 cells, 26.7 %), Erythrocyte / unknown (n = 85, 4.1 %), neutrophils / myelocytes (n = 211 cells, 10.2 %), Mast cell progenitor 1 (n = 599, 29.1 %), Mast cell progenitor 2 (n = 152, 7.4 %), Committed Mast cell precursor (n = 113, 5.5 %), and mast cells (n = 353, 17.1 %). In silico analyses of the MC precursor and mature MC populations revealed transcriptionally distinct MC precursor subtype and mature MC states (CMA1+ and CMA1- subtypes). Culturing MC precursor populations in MC maturation media (mast cell media II) led to homogenous mature MC populations as evidenced by high expression of high affinity IgE receptor, metachromatic granules, presence of MC granule proteins (Tryptase and Chymase) and activation following substance P stimulation and FceRI crosslinking. This human iPSC-based approach generates MC precursors and phenotypically mature and functional MC populations. This system will be a useful model to generate human MC populations and broaden our understanding of MC biology and transcriptional regulation of MC differentiation trajectories.
    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
    心脏病是美国的主要死因,然而,由于无法培养原代心脏细胞,研究受到限制。源自人诱导多能干细胞(iPSC)的心肌细胞(CM)是药物筛选和疾病建模的有希望的解决方案。
    诱导多能干细胞衍生的CM(iPSC-CM)分化和成熟研究通常使用异质底物进行生长和破坏性验证方法。可重复,需要可调谐基板和无接触监测来识别理想条件以产生均匀,功能性CM。
    我们生成了基于聚乙二醇的合成水凝胶,用于iPSC-CM的分化和成熟。肽浓度,组合,和凝胶刚度独立调整。无标记光学氧化还原成像(ORI)在宽视场显微镜上在凝胶制剂的96孔筛网中进行。我们在整个分化和早期到晚期成熟过程中进行了活细胞成像,以确定关键的代谢变化。
    无标记ORI证实了在整个iPSC-CM在合成水凝胶上的分化和成熟过程中预期的向氧化磷酸化的代谢转移。此外,ORI在心脏祖细胞阶段区分了高分化效率和低分化效率的细胞批次。
    我们建立了合成水凝胶条件的培养基通量筛选工作流程,能够进行重复的活细胞测量并确认预期的代谢变化。这些方法对生物制造中可重复的iPSC-CM产生具有意义。
    UNASSIGNED: Heart disease is the leading cause of death in the United States, yet research is limited by the inability to culture primary cardiac cells. Cardiomyocytes (CMs) derived from human induced pluripotent stem cells (iPSCs) are a promising solution for drug screening and disease modeling.
    UNASSIGNED: Induced pluripotent stem cell-derived CM (iPSC-CM) differentiation and maturation studies typically use heterogeneous substrates for growth and destructive verification methods. Reproducible, tunable substrates and touch-free monitoring are needed to identify ideal conditions to produce homogenous, functional CMs.
    UNASSIGNED: We generated synthetic polyethylene glycol-based hydrogels for iPSC-CM differentiation and maturation. Peptide concentrations, combinations, and gel stiffness were tuned independently. Label-free optical redox imaging (ORI) was performed on a widefield microscope in a 96-well screen of gel formulations. We performed live-cell imaging throughout differentiation and early to late maturation to identify key metabolic shifts.
    UNASSIGNED: Label-free ORI confirmed the expected metabolic shifts toward oxidative phosphorylation throughout the differentiation and maturation processes of iPSC-CMs on synthetic hydrogels. Furthermore, ORI distinguished high and low differentiation efficiency cell batches in the cardiac progenitor stage.
    UNASSIGNED: We established a workflow for medium throughput screening of synthetic hydrogel conditions with the ability to perform repeated live-cell measurements and confirm expected metabolic shifts. These methods have implications for reproducible iPSC-CM generation in biomanufacturing.
    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
    自从发现诱导多能干细胞(iPSC)技术以来,已经有许多尝试创建遗传性视网膜疾病(IRD)的细胞模型,用于研究致病过程,以促进靶标发现和验证活动.一致性仍然是决定这些发现效用的关键。尽管一致性很重要,质量控制指标仍未广泛使用。在这次审查中,一个利用iPSC技术产生感光器的工具包,视网膜色素上皮细胞,提供了类器官疾病模型。在开发iPSC衍生的IRD模型时的注意事项,例如iPSC起源,重新编程方法,质量控制指标,控制策略,并讨论了区分方案。解剖了各种iPSCIRD模型,并讨论了基于iPSC的疾病建模的科学障碍,以概述该领域的当前方法和未来方向。
    Since the discovery of induced pluripotent stem cell (iPSC) technology, there have been many attempts to create cellular models of inherited retinal diseases (IRDs) for investigation of pathogenic processes to facilitate target discovery and validation activities. Consistency remains key in determining the utility of these findings. Despite the importance of consistency, quality control metrics are still not widely used. In this review, a toolkit for harnessing iPSC technology to generate photoreceptor, retinal pigment epithelial cell, and organoid disease models is provided. Considerations while developing iPSC-derived IRD models such as iPSC origin, reprogramming methods, quality control metrics, control strategies, and differentiation protocols are discussed. Various iPSC IRD models are dissected and the scientific hurdles of iPSC-based disease modeling are discussed to provide an overview of current methods and future directions in this field.
    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
    COVID-19患者的不良心脏结局,特别是那些已经有心脏病的人,激发基于人类细胞的芯片上器官模型的发展,以概括心脏损伤和功能障碍,并筛选心脏保护性疗法。这里,我们开发了一种芯片心脏模型来研究SARS-CoV-2在健康心肌中的发病机理,该模型由人诱导多能干细胞(iPSC)衍生的心肌细胞和心功能不全模型建立,模仿血管紧张素II(AngII)引起的先前存在的高血压疾病的方面。我们概述了SARS-CoV-2引起的心脏损伤的细胞病变特征,包括逐渐受损的收缩功能和钙处理,凋亡,和肌节混乱。与单独的SARS-CoV-2相比,在AngII治疗的芯片心脏中存在SARS-CoV-2会降低收缩力,并较早地发生收缩功能障碍,并大大增强了炎性细胞因子。为了开发潜在的疗法,我们评估了来自人类iPSC的细胞外囊泡(EV)的心脏保护作用,该作用减轻了收缩力的损害,细胞凋亡减少,减少肌节蛋白的破坏,增强β-氧化基因表达。AngII或EV治疗均不影响病毒载量。我们鉴定了微小RNAmiR-20a-5p和miR-19a-3p作为这些EV的心脏保护作用的潜在介质。
    Adverse cardiac outcomes in COVID-19 patients, particularly those with preexisting cardiac disease, motivate the development of human cell-based organ-on-a-chip models to recapitulate cardiac injury and dysfunction and for screening of cardioprotective therapeutics. Here, we developed a heart-on-a-chip model to study the pathogenesis of SARS-CoV-2 in healthy myocardium established from human induced pluripotent stem cell (iPSC)-derived cardiomyocytes and a cardiac dysfunction model, mimicking aspects of preexisting hypertensive disease induced by angiotensin II (Ang II). We recapitulated cytopathic features of SARS-CoV-2-induced cardiac damage, including progressively impaired contractile function and calcium handling, apoptosis, and sarcomere disarray. SARS-CoV-2 presence in Ang II-treated hearts-on-a-chip decreased contractile force with earlier onset of contractile dysfunction and profoundly enhanced inflammatory cytokines compared to SARS-CoV-2 alone. Toward the development of potential therapeutics, we evaluated the cardioprotective effects of extracellular vesicles (EVs) from human iPSC which alleviated the impairment of contractile force, decreased apoptosis, reduced the disruption of sarcomeric proteins, and enhanced beta-oxidation gene expression. Viral load was not affected by either Ang II or EV treatment. We identified MicroRNAs miR-20a-5p and miR-19a-3p as potential mediators of cardioprotective effects of these EVs.
    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
    视网膜疾病的患病率不断上升-尤其是,年龄相关性黄斑变性和遗传性视网膜疾病对眼科医学提出了严峻的挑战,通常导致不可逆转的视力丧失。目前的治疗是有限的,并且通常不能解决视网膜细胞的潜在损失。本文探讨了基于干细胞的疗法作为视网膜再生的有希望的途径的潜力。我们回顾了干细胞技术的最新进展,专注于胚胎干细胞(ESC),多能干细胞(PSC),和间充质干细胞(MSCs),以及它们分化成视网膜细胞类型的能力。我们讨论了干细胞移植的挑战,比如免疫排斥,整合到宿主视网膜中,功能恢复。检查了先前和正在进行的临床试验,以突出这些新疗法的疗效和安全性。此外,我们讨论干细胞研究的伦理考虑和监管框架。我们的分析表明,尽管基于干细胞的疗法为治疗视网膜疾病提供了开创性的方法,需要进一步研究以确保长期安全性并优化治疗结果.这篇综述总结了干细胞治疗的临床证据和目前利用干细胞治疗视网膜变性的局限性。如年龄相关性黄斑变性,视网膜色素变性,和Stargardt的病.
    The escalating prevalence of retinal diseases-notably, age-related macular degeneration and hereditary retinal disorders-poses an intimidating challenge to ophthalmic medicine, often culminating in irreversible vision loss. Current treatments are limited and often fail to address the underlying loss of retinal cells. This paper explores the potential of stem-cell-based therapies as a promising avenue for retinal regeneration. We review the latest advancements in stem cell technology, focusing on embryonic stem cells (ESCs), pluripotent stem cells (PSCs), and mesenchymal stem cells (MSCs), and their ability to differentiate into retinal cell types. We discuss the challenges in stem cell transplantation, such as immune rejection, integration into the host retina, and functional recovery. Previous and ongoing clinical trials are examined to highlight the therapeutic efficacy and safety of these novel treatments. Additionally, we address the ethical considerations and regulatory frameworks governing stem cell research. Our analysis suggests that while stem-cell-based therapies offer a groundbreaking approach to treating retinal diseases, further research is needed to ensure long-term safety and to optimize therapeutic outcomes. This review summarizes the clinical evidence of stem cell therapy and current limitations in utilizing stem cells for retinal degeneration, such as age-related macular degeneration, retinitis pigmentosa, and Stargardt\'s 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
    人类诱导多能干细胞(hiPSC)来源的肠道类器官是研究发育生物学和个性化治疗的有价值的工具,但是它们的封闭拓扑和相对不成熟的状态限制了应用。这里,我们使用芯片上器官技术在更生理的体外微环境中开发出具有顶端和基底外侧通路的hiPSC衍生肠屏障。为了沿着隐窝-绒毛轴复制生长因子梯度,我们将细胞局部暴露于扩增和分化培养基中。在这些条件下,肠上皮细胞自组织成具有生理屏障完整性的绒毛样褶皱,肌成纤维细胞和神经元在底部通道中出现并形成上皮下组织。生长因子梯度有效地平衡分裂和成熟细胞类型,并诱导肠上皮成分,包括吸收和分泌谱系,类似于人类小肠的成分。这种特征良好的hiPSC衍生的芯片肠系统可以促进对人类小肠中的生理过程和治疗开发的个性化研究。
    Human induced pluripotent stem cell (hiPSC)-derived intestinal organoids are valuable tools for researching developmental biology and personalized therapies, but their closed topology and relative immature state limit applications. Here, we use organ-on-chip technology to develop a hiPSC-derived intestinal barrier with apical and basolateral access in a more physiological in vitro microenvironment. To replicate growth factor gradients along the crypt-villus axis, we locally expose the cells to expansion and differentiation media. In these conditions, intestinal epithelial cells self-organize into villus-like folds with physiological barrier integrity, and myofibroblasts and neurons emerge and form a subepithelial tissue in the bottom channel. The growth factor gradients efficiently balance dividing and mature cell types and induce an intestinal epithelial composition, including absorptive and secretory lineages, resembling the composition of the human small intestine. This well-characterized hiPSC-derived intestine-on-chip system can facilitate personalized studies on physiological processes and therapy development in the human small intestine.
    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