human stem cells

  • 文章类型: Journal Article
    肺泡成纤维细胞和上皮细胞之间的相互作用对肺稳态至关重要。损伤修复,和纤维发生,但潜在的机制仍不清楚。为了调查,我们给予成纤维细胞选择性TGFβ1信号抑制剂,表没食子儿茶素没食子酸酯(EGCG),间质性肺病(ILD)患者接受诊断性肺活检,并对备用组织进行单细胞RNA测序。与非疾病供体或终末期ILD组织相比,未治疗患者的活检显示出更高的成纤维细胞TGFβ1信号传导。在体内,EGCG下调活检样品中的TGFβ1信号传导和几种促炎和应激途径。值得注意的是,EGCG减少成纤维细胞分泌的卷曲样受体蛋白2(sFRP2),未识别的TGFβ1成纤维细胞靶基因原位诱导II型肺泡上皮细胞(AEC2s)附近。使用AEC2-成纤维细胞共培养的类器官和来自非患病供体的精确切割的肺切片(PCLS),我们发现TGFβ1信号传导促进了AEC2KRT17+基底细胞状态的扩散,然后sFRP2激活成熟的Krt5+基础细胞程序。sFRP2诱导的核NFATc3积累和KRT5表达需要Wnt受体卷曲蛋白5(Fzd5)表达和下游钙调磷酸酶信号传导。这些发现强调了ILD中的阶段特异性TGFβ1信号传导,EGCG在减少IPF相关转录变化方面的治疗潜力,并通过sFRP2鉴定TGFβ1-非经典Wnt通路串扰是特发性肺纤维化/ILD中功能失调的上皮信号传导的新机制。
    Reciprocal interactions between alveolar fibroblasts and epithelial cells are crucial for lung homeostasis, injury repair, and fibrogenesis, but underlying mechanisms remain unclear. To investigate, we administered the fibroblast-selective TGFβ1 signaling inhibitor, epigallocatechin gallate (EGCG), to Interstitial Lung Disease (ILD) patients undergoing diagnostic lung biopsy and conducted single-cell RNA sequencing on spare tissue. Biopsies from untreated patients showed higher fibroblast TGFβ1 signaling compared to non-disease donor or end-stage ILD tissues. In vivo, EGCG downregulated TGFβ1 signaling and several pro-inflammatory and stress pathways in biopsy samples. Notably, EGCG reduced fibroblast secreted frizzle-like receptor protein 2 (sFRP2), an unrecognized TGFβ1 fibroblast target gene induced near type II alveolar epithelial cells (AEC2s) in situ. Using AEC2-fibroblast coculture organoids and precision cut lung slices (PCLS) from non-diseased donors, we found TGFβ1 signaling promotes a spread AEC2 KRT17+ basaloid state, whereupon sFRP2 then activates a mature Krt5+ basal cell program. Wnt-receptor Frizzled 5 (Fzd5) expression and downstream calcineurin signaling were required for sFRP2-induced nuclear NFATc3 accumulation and KRT5 expression. These findings highlight stage-specific TGFβ1 signaling in ILD, the therapeutic potential of EGCG in reducing IPF-related transcriptional changes, and identify TGFβ1-non-canonical Wnt pathway crosstalk via sFRP2 as a novel mechanism for dysfunctional epithelial signaling in Idiopathic Pulmonary Fibrosis/ILD.
    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
    在这项跨学科研究中,我们证明了6D打印机在软组织工程模型中的适用性。为此,建造了一个特殊的工厂,将6D打印的技术要求与生物必需品相结合,尤其是软组织.因此,商用6D机器人手臂与可灭菌外壳(包括高效微粒空气(HEPA)过滤器和紫外线辐射(UVC)灯)以及定制的打印头和打印床相结合。这两个组件都允许冷却和加热,这对于使用活细胞是理想的。此外,安装了一个喷雾装置,允许液体的细小液滴的分布。可以使用所有六个轴创建不平坦或成角度的表面上的高级几何形状。基于软组织工程领域经常使用的生物墨水(结冷胶,胶原蛋白,和明胶甲基丙烯酰基)具有非常不同的材料特性,我们可以展示打印系统的灵活性。此外,使用原代人脂肪来源的干细胞(ASC)的含细胞的构建体可以以自动化方式产生。除了细胞存活,沿着脂肪谱系分化的能力也可以被证明是软组织工程的代表。
    Within this interdisciplinary study, we demonstrate the applicability of a 6D printer for soft tissue engineering models. For this purpose, a special plant was constructed, combining the technical requirements for 6D printing with the biological necessities, especially for soft tissue. Therefore, a commercial 6D robot arm was combined with a sterilizable housing (including a high-efficiency particulate air (HEPA) filter and ultraviolet radiation (UVC) lamps) and a custom-made printhead and printbed. Both components allow cooling and heating, which is desirable for working with viable cells. In addition, a spraying unit was installed that allows the distribution of fine droplets of a liquid. Advanced geometries on uneven or angled surfaces can be created with the use of all six axes. Based on often used bioinks in the field of soft tissue engineering (gellan gum, collagen, and gelatin methacryloyl) with very different material properties, we could demonstrate the flexibility of the printing system. Furthermore, cell-containing constructs using primary human adipose-derived stem cells (ASCs) could be produced in an automated manner. In addition to cell survival, the ability to differentiate along the adipogenic lineage could also be demonstrated as a representative of soft tissue engineering.
    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
    银河宇宙辐射(GCR)是宇航员在执行月球和火星任务期间面临的最严重风险之一。能够概括人体生理的实验模型对于理解辐射对人体器官的影响以及制定针对太空旅行暴露的辐射防护措施至关重要。使用包含人类骨髓(造血和急性辐射损伤部位)的工程组织模型的多器官芯片(multi-OoC)平台研究了全身辐射的影响,心肌(慢性辐射损伤部位)和肝脏(代谢部位),通过血管循环与内皮屏障相连,将各个组织腔与血管灌注液分开。在长期的中子辐射之后,深空中最具破坏性的辐射成分,与急性递送的相同累积剂量相比,观察到更大的组织功能偏差。Further,通过表征循环中的工程骨髓(eBM)衍生的免疫细胞,鉴定了58个特定于延长中子剂量效应的独特基因,与急性照射和健康组织相比。它提出,这个生物工程平台允许在“宇航员芯片”模型中研究人类对延长辐射暴露的反应,该模型可以为减轻宇宙辐射伤害的措施提供信息。
    Galactic cosmic radiation (GCR) is one of the most serious risks posed to astronauts during missions to the Moon and Mars. Experimental models capable of recapitulating human physiology are critical to understanding the effects of radiation on human organs and developing radioprotective measures against space travel exposures. The effects of systemic radiation are studied using a multi-organ-on-a-chip (multi-OoC) platform containing engineered tissue models of human bone marrow (site of hematopoiesis and acute radiation damage), cardiac muscle (site of chronic radiation damage) and liver (site of metabolism), linked by vascular circulation with an endothelial barrier separating individual tissue chambers from the vascular perfusate. Following protracted neutron radiation, the most damaging radiation component in deep space, a greater deviation of tissue function is observed as compared to the same cumulative dose delivered acutely. Further, by characterizing engineered bone marrow (eBM)-derived immune cells in circulation, 58 unique genes specific to the effects of protracted neutron dosing are identified, as compared to acutely irradiated and healthy tissues. It propose that this bioengineered platform allows studies of human responses to extended radiation exposure in an \"astronaut-on-a-chip\" model that can inform measures for mitigating cosmic radiation injury.
    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:结节性硬化症(TSC)是一种多系统遗传疾病,可在大脑和其他重要器官中引起良性肿瘤。最令人衰弱的症状是由中枢神经系统受累引起的,并导致许多严重的症状,包括癫痫发作。智力残疾,自闭症,和行为问题。TSC是由TSC1或TSC2基因的杂合突变引起的,mTOR激酶的失调及其多方面的下游信号改变是疾病发病机理的核心。尽管这种疾病的神经系统后遗症已经得到了很好的证实,关于这些突变如何影响细胞成分和血脑屏障(BBB)的功能知之甚少。
    方法:我们通过利用人类诱导多能干细胞和微流控细胞培养技术,产生了BBB的TSC疾病特异性细胞模型。
    结果:使用微生理系统,我们证明从TSC2杂合突变细胞产生的BBB显示增加的通透性。这可以通过野生型星形胶质细胞或用雷帕霉素治疗来拯救,mTOR激酶抑制剂。
    结论:我们的研究结果表明,微生理系统可用于研究人类神经系统疾病,并提高我们对有助于TSC发病机理的细胞谱系的认识,并为未来的治疗提供信息。
    BACKGROUND: Tuberous sclerosis complex (TSC) is a multi-system genetic disease that causes benign tumors in the brain and other vital organs. The most debilitating symptoms result from involvement of the central nervous system and lead to a multitude of severe symptoms including seizures, intellectual disability, autism, and behavioral problems. TSC is caused by heterozygous mutations of either the TSC1 or TSC2 gene and dysregulation of mTOR kinase with its multifaceted downstream signaling alterations is central to disease pathogenesis. Although the neurological sequelae of the disease are well established, little is known about how these mutations might affect cellular components and the function of the blood-brain barrier (BBB).
    METHODS: We generated TSC disease-specific cell models of the BBB by leveraging human induced pluripotent stem cell and microfluidic cell culture technologies.
    RESULTS: Using microphysiological systems, we demonstrate that a BBB generated from TSC2 heterozygous mutant cells shows increased permeability. This can be rescued by wild type astrocytes or by treatment with rapamycin, an mTOR kinase inhibitor.
    CONCLUSIONS: Our results demonstrate the utility of microphysiological systems to study human neurological disorders and advance our knowledge of cell lineages contributing to TSC pathogenesis and informs future therapeutics.
    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
    我们先前报道,在免疫缺陷啮齿动物模型中,人肌肉衍生的干细胞(hMuStemcells)在局部施用到受伤的骨骼肌或梗塞的心脏中后有助于组织修复。然而,将这些发现外推到临床背景是有问题的,因为在人类和啮齿动物的体内发现之间经常出现相当大的差异。因此,我们调查了hMuStem细胞的肌肉再生行为在临床相关的移植环境中是否得以维持.通过高密度显微注射基质将人MuStem细胞肌肉内施用到接受基于他克莫司的免疫抑制的非人灵长类动物中,从而再现了迄今为止在肌病细胞疗法的临床试验中产生最佳结果的方案。给药后4周和9周,细胞注射部位的组织学分析显示,在所有情况下都有大量的hMuStem细胞衍生的细胞核。大多数移植物衍生的细胞核分布在小的肌纤维组中,其中没有观察到特定免疫反应的迹象。重要的是,hMuStem细胞主要通过与宿主肌纤维融合促进猿猴组织修复,在这个模型中证明了它们的肌纤维再生能力。一起,在有效的临床前模型中获得的这些发现为hMuStem细胞在肌肉疾病的未来细胞治疗中的潜力提供了新的见解.
    We previously reported that human muscle-derived stem cells (hMuStem cells) contribute to tissue repair after local administration into injured skeletal muscle or infarcted heart in immunodeficient rodent models. However, extrapolation of these findings to a clinical context is problematic owing to the considerable differences often seen between in vivo findings in humans versus rodents. Therefore, we investigated whether the muscle regenerative behavior of hMuStem cells is maintained in a clinically relevant transplantation context. Human MuStem cells were intramuscularly administered by high-density microinjection matrices into nonhuman primates receiving tacrolimus-based immunosuppression thereby reproducing the protocol that has so far produced the best results in clinical trials of cell therapy in myopathies. Four and 9 weeks after administration, histological analysis of cell injection sites revealed large numbers of hMuStem cell-derived nuclei in all cases. Most graft-derived nuclei were distributed in small myofiber groups in which no signs of a specific immune response were observed. Importantly, hMuStem cells contributed to simian tissue repair by fusing mainly with host myofibers, demonstrating their capacity for myofiber regeneration in this model. Together, these findings obtained in a valid preclinical model provide new insights supporting the potential of hMuStem cells in future cell therapies for muscle 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
    利用组织特异性细胞外基质(ECM)对于复制天然组织的组成和开发生物学相关的生物材料至关重要。人类或动物来源的供体组织和器官是目前这些ECM来源的黄金标准。包括供体组织的高度有限的可用性,细胞衍生的ECM为工程组织特异性生物材料提供了一种替代方法,例如用于三维(3D)生物打印的生物墨水。3D生物打印是一种最先进的生物制造技术,可满足全球对供体组织和器官的需求。事实上,全球对用于治疗角膜失明的人类供体角膜有巨大的需求,通常是由于角膜基质微结构的损伤。脂肪组织是人体最丰富且易于获取的组织之一,和脂肪组织来源的干细胞(hASC)是用于组织工程的高度有利的细胞类型。此外,hASC已经在治疗角膜基质病理学的临床试验中进行了研究。在这项研究中,通过将hASC分化为角膜基质角膜细胞(hASC-CSKs),在不需要供体角膜的情况下对角膜基质特异性ECM进行工程改造。此外,该ECM被用作角膜基质特异性生物墨水的组成部分,其中hASC-CSK被打印以产生角膜基质结构。这种具有成本效益的方法与临床相关的细胞类型相结合,为开发更可持续的组织特异性解决方案提供了有价值的信息,并推进了角膜组织工程领域。
    Utilizing tissue-specific extracellular matrices (ECMs) is vital for replicating the composition of native tissues and developing biologically relevant biomaterials. Human- or animal-derived donor tissues and organs are the current gold standard for the source of these ECMs. To overcome the several limitations related to these ECM sources, including the highly limited availability of donor tissues, cell-derived ECM offers an alternative approach for engineering tissue-specific biomaterials, such as bioinks for three-dimensional (3D) bioprinting. 3D bioprinting is a state-of-the-art biofabrication technology that addresses the global need for donor tissues and organs. In fact, there is a vast global demand for human donor corneas that are used for treating corneal blindness, often resulting from damage in the corneal stromal microstructure. Human adipose tissue is one of the most abundant tissues and easy to access, and adipose tissue-derived stem cells (hASCs) are a highly advantageous cell type for tissue engineering. Furthermore, hASCs have already been studied in clinical trials for treating corneal stromal pathologies. In this study, a corneal stroma-specific ECM was engineered without the need for donor corneas by differentiating hASCs toward corneal stromal keratocytes (hASC-CSKs). Furthermore, this ECM was utilized as a component for corneal stroma-specific bioink where hASC-CSKs were printed to produce corneal stroma structures. This cost-effective approach combined with a clinically relevant cell type provides valuable information on developing more sustainable tissue-specific solutions and advances the field of corneal tissue engineering.
    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
    ATP结合盒A3(ABCA3)中的突变,对肺泡II型上皮细胞(AEC2s)中的表面活性剂稳态至关重要的磷脂转运蛋白,是儿童间质性肺病(chILD)最常见的遗传原因。对ABCA3突变病理变异患者的治疗是有限的,部分原因是由于无法从受影响儿童获得原发性AEC2导致对疾病发病机制缺乏了解。这里,我们报道了携带多个ABCA3突变纯合版本的患者诱导多能干细胞(iPSCs)产生AEC2的过程.我们产生了用于体外疾病建模的同基因CRISPR/Cas9基因校正和未校正的iPSC和ABCA3-突变体敲入ABCA3:GFP融合报告系。我们观察到ABCA3突变iPSC衍生的AEC2s(iAEC2s)中表面活性剂分泌的预期能力降低,但我们也在突变iAEC2s中发现了一个意想不到的上皮内在异常表型,表现为祖先潜力减弱,NFκB信号增加,和促炎细胞因子的产生。ABCA3:GFP融合报告基因允许突变体特异性,层状体大小和ABCA3蛋白运输的可量化表征,根据ABCA3突变类型被扰乱的功能特征。我们的疾病模型为理解ABCA3突变介导的肺泡上皮细胞功能障碍机制提供了一个平台,该机制可能引发chILD发病机制。
    Mutations in ATP-binding cassette A3 (ABCA3), a phospholipid transporter critical for surfactant homeostasis in pulmonary alveolar type II epithelial cells (AEC2s), are the most common genetic causes of childhood interstitial lung disease (chILD). Treatments for patients with pathological variants of ABCA3 mutations are limited, in part due to a lack of understanding of disease pathogenesis resulting from an inability to access primary AEC2s from affected children. Here, we report the generation of AEC2s from affected patient induced pluripotent stem cells (iPSCs) carrying homozygous versions of multiple ABCA3 mutations. We generated syngeneic CRISPR/Cas9 gene-corrected and uncorrected iPSCs and ABCA3-mutant knockin ABCA3:GFP fusion reporter lines for in vitro disease modeling. We observed an expected decreased capacity for surfactant secretion in ABCA3-mutant iPSC-derived AEC2s (iAEC2s), but we also found an unexpected epithelial-intrinsic aberrant phenotype in mutant iAEC2s, presenting as diminished progenitor potential, increased NFκB signaling, and the production of pro-inflammatory cytokines. The ABCA3:GFP fusion reporter permitted mutant-specific, quantifiable characterization of lamellar body size and ABCA3 protein trafficking, functional features that are perturbed depending on ABCA3 mutation type. Our disease model provides a platform for understanding ABCA3 mutation-mediated mechanisms of alveolar epithelial cell dysfunction that may trigger chILD pathogenesis.
    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
    三维(3D)生物打印提供了一种自动化、可定制的解决方案,以制造高度详细的3D组织构造,并为再生医学提供巨大的希望,以解决供体组织和器官的严重全球短缺。然而,单材料3D生物打印不足以制造具有类似天然微结构的异质3D构建体,因此,需要创新的多材料解决方案。这里,我们开发了一种用于生物打印人角膜基质的新型多材料3D生物打印策略。人类角膜是眼睛的透明外层,角膜盲导致的视力丧失严重影响个体的生活质量。角膜失明的主要原因之一是角膜基质的详细组织中的损伤,其中胶原纤维在彼此垂直的层中排列并且角膜基质细胞沿着原纤维生长。用于治疗角膜失明的供体角膜很少,目前的组织工程(TE)技术无法生产具有天然角膜基质复杂微结构的人工角膜。为了解决这个问题,我们开发了一种新的多材料3D生物打印策略来模拟角膜基质的详细组织。这些具有异质设计的多材料3D结构通过使用具有不同硬度的人脂肪组织来源的干细胞(hASCs)和基于透明质酸(HA)的生物墨水进行生物打印。在我们新颖的3D模型设计中,无细胞更硬的HA-生物墨水和载有细胞的更软的HA-生物墨水以交替的细丝打印,并且在交替层中垂直印刷长丝。多材料生物打印策略首次应用于角膜基质3D生物打印以模拟天然微结构。因此,软生物墨水促进了多材料3D生物打印复合材料中hASCs的细胞生长和组织形成,而坚硬的生物墨水在培养时提供了机械支持以及细胞组织的指导。有趣的是,细胞生长和组织形成显着改变了生物打印复合构建体的机械性能。重要的是,在离体角膜器官培养模型中,生物打印的复合结构显示出与宿主组织的良好整合。作为结论,开发的多材料生物打印策略作为有组织的制造的生物制造解决方案提供了巨大的潜力,天然组织的异质微结构。据我们所知,这种多材料生物打印策略从未应用于角膜生物打印.因此,我们的工作推进了增材制造的技术成果,并将角膜TE领域提升到一个新的水平。
    Three-dimensional (3D) bioprinting offers an automated, customizable solution to manufacture highly detailed 3D tissue constructs and holds great promise for regenerative medicine to solve the severe global shortage of donor tissues and organs. However, uni-material 3D bioprinting is not sufficient for manufacturing heterogenous 3D constructs with native-like microstructures and thus, innovative multi-material solutions are required. Here, we developed a novel multi-material 3D bioprinting strategy for bioprinting human corneal stroma. The human cornea is the transparent outer layer of your eye, and vision loss due to corneal blindness has serious effects on the quality of life of individuals. One of the main reasons for corneal blindness is the damage in the detailed organization of the corneal stroma where collagen fibrils are arranged in layers perpendicular to each other and the corneal stromal cells grow along the fibrils. Donor corneas for treating corneal blindness are scarce, and the current tissue engineering (TE) technologies cannot produce artificial corneas with the complex microstructure of native corneal stroma. To address this, we developed a novel multi-material 3D bioprinting strategy to mimic detailed organization of corneal stroma. These multi-material 3D structures with heterogenous design were bioprinted by using human adipose tissue -derived stem cells (hASCs) and hyaluronic acid (HA) -based bioinks with varying stiffnesses. In our novel design of 3D models, acellular stiffer HA-bioink and cell-laden softer HA-bioink were printed in alternating filaments, and the filaments were printed perpendicularly in alternating layers. The multi-material bioprinting strategy was applied for the first time in corneal stroma 3D bioprinting to mimic the native microstructure. As a result, the soft bioink promoted cellular growth and tissue formation of hASCs in the multi-material 3D bioprinted composites, whereas the stiff bioink provided mechanical support as well as guidance of cellular organization upon culture. Interestingly, cellular growth and tissue formation altered the mechanical properties of the bioprinted composite constructs significantly. Importantly, the bioprinted composite structures showed good integration to the host tissue in ex vivo cornea organ culture model. As a conclusion, the developed multi-material bioprinting strategy provides great potential as a biofabrication solution for manufacturing organized, heterogenous microstructures of native tissues. To the best of our knowledge, this multi-material bioprinting strategy has never been applied in corneal bioprinting. Therefore, our work advances the technological achievements in additive manufacturing and brings the field of corneal TE to a new level.
    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
    结节性硬化症(TSC)是一种多系统遗传疾病,可引起大脑和其他重要器官的良性肿瘤。最令人衰弱的症状是由中枢神经系统受累引起的,并导致许多严重的症状,包括癫痫发作。智力残疾,自闭症,和行为问题。TSC是由TSC1或TSC2基因的杂合突变引起的。mTOR激酶的失调及其多方面的下游信号改变是疾病发病机制的核心。尽管这种疾病的神经系统后遗症已经得到了很好的证实,关于这些突变如何影响细胞成分和血脑屏障(BBB)的功能知之甚少。我们通过利用人类诱导多能干细胞和微流控细胞培养技术产生了BBB的疾病特异性细胞模型。利用这些微生理系统,我们证明,从TSC2杂合突变细胞产生的BBB显示出增加的通透性,这可以通过野生型星形胶质细胞和雷帕霉素治疗来拯救,mTOR激酶抑制剂。我们的结果进一步证明了微生理系统在研究人类神经系统疾病和增进我们对有助于TSC发病机理的细胞谱系的认识方面的实用性。
    Tuberous sclerosis complex (TSC) is a multi-system genetic disease that causes benign tumors in the brain and other vital organs. The most debilitating symptoms result from involvement of the central nervous system and lead to a multitude of severe symptoms including seizures, intellectual disability, autism, and behavioral problems. TSC is caused by heterozygous mutations of either the TSC1 or TSC2 gene. Dysregulation of mTOR kinase with its multifaceted downstream signaling alterations is central to disease pathogenesis. Although the neurological sequelae of the disease are well established, little is known about how these mutations might affect cellular components and the function of the blood-brain barrier (BBB). We generated disease-specific cell models of the BBB by leveraging human induced pluripotent stem cell and microfluidic cell culture technologies. Using these microphysiological systems, we demonstrate that the BBB generated from TSC2 heterozygous mutant cells shows increased permeability which can be rescued by wild type astrocytes and with treatment with rapamycin, an mTOR kinase inhibitor. Our results further demonstrate the utility of microphysiological systems to study human neurological disorders and advance our knowledge of the cell lineages contributing to TSC pathogenesis.
    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
    探讨人类干细胞(hSC)在眼科应用中的图像分析和计算建模方面的进展和未来研究方向。
    hSCs在眼部再生医学中具有巨大潜力,因为它们在基于细胞的治疗以及使用最先进的2D和3D类器官模型进行疾病建模和药物发现中的应用。然而,对他们的情结有更深刻的描述,需要多尺度属性来优化它们到临床实践的翻译。图像分析与计算建模相结合是探索hSC行为机制并帮助临床诊断和治疗的有力工具。
    许多计算模型借鉴了各种技术,通常混合连续和离散方法,并被用来描述细胞分化和自组织。机器学习工具在模型开发和改善临床诊断和治疗的图像分类过程中产生了重大影响,并将成为未来许多研究的重点。
    UNASSIGNED: To explore the advances and future research directions in image analysis and computational modelling of human stem cells (hSCs) for ophthalmological applications.
    UNASSIGNED: hSCs hold great potential in ocular regenerative medicine due to their application in cell-based therapies and in disease modelling and drug discovery using state-of-the-art 2D and 3D organoid models. However, a deeper characterisation of their complex, multi-scale properties is required to optimise their translation to clinical practice. Image analysis combined with computational modelling is a powerful tool to explore mechanisms of hSC behaviour and aid clinical diagnosis and therapy.
    UNASSIGNED: Many computational models draw on a variety of techniques, often blending continuum and discrete approaches, and have been used to describe cell differentiation and self-organisation. Machine learning tools are having a significant impact in model development and improving image classification processes for clinical diagnosis and treatment and will be the focus of much future research.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号