lung progenitors

  • 文章类型: Journal Article
    类器官本质上是一种体外(实验室培养的)三维组织培养系统模型,可精确复制人体器官的结构和生理。目前类器官的一些应用是在基础生物学研究领域,分子医学和药物测试。在药物发现过程中,类器官在连接动物模型和人体临床试验之间的差距方面至关重要,这显著降低了与每个测试阶段相关的持续时间和成本。同样,它们可以用来理解细胞间的相互作用,组织生物学和再生的关键方面,并在疾病的各个阶段对疾病的发病机理进行建模。肺类器官可用于探索肺的许多病理生理活动,因为它们与其功能具有相似性。研究人员一直试图通过开发各种“肺类器官”模型来重建肺的复杂性质。本文是对肺类器官及其潜在祖细胞的各种发展的系统综述。它还涵盖了肺部类器官的深入应用,以促进转化研究。这篇综述讨论了建立不同类型的肺类器官的方法,以研究呼吸系统的再生能力和理解各种呼吸系统疾病。呼吸系统疾病是全世界最常见的疾病之一,必须立即解决日益增长的负担。肺类器官以及各种生物工程工具和技术将作为研究各种呼吸系统疾病的病理生理学和药物筛选目的的新模型。
    Organoids are essentially an in vitro (lab-grown) three-dimensional tissue culture system model that meticulously replicates the structure and physiology of human organs. A few of the present applications of organoids are in the basic biological research area, molecular medicine and pharmaceutical drug testing. Organoids are crucial in connecting the gap between animal models and human clinical trials during the drug discovery process, which significantly lowers the time duration and cost associated with each stage of testing. Likewise, they can be used to understand cell-to-cell interactions, a crucial aspect of tissue biology and regeneration, and to model disease pathogenesis at various stages of the disease. Lung organoids can be utilized to explore numerous pathophysiological activities of a lung since they share similarities with its function. Researchers have been trying to recreate the complex nature of the lung by developing various \"Lung organoids\" models.This article is a systematic review of various developments of lung organoids and their potential progenitors. It also covers the in-depth applications of lung organoids for the advancement of translational research. The review discusses the methodologies to establish different types of lung organoids for studying the regenerative capability of the respiratory system and comprehending various respiratory diseases.Respiratory diseases are among the most common worldwide, and the growing burden must be addressed instantaneously. Lung organoids along with diverse bio-engineering tools and technologies will serve as a novel model for studying the pathophysiology of various respiratory diseases and for drug screening purposes.
    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
    多能干细胞(PSC)衍生物的远端肺上皮的持久重建,如果意识到,将代表一种有希望的治疗由肺泡损伤引起的疾病的方法。这里,我们将鼠PSC分化为自我更新的肺上皮祖细胞,能够移植到免疫活性受损的远端肺上皮,同系小鼠接受者。移植后,这些祖细胞在远端肺成熟,假设肺泡2型(AT2)和1型(AT1)细胞的分子表型。在体内几个月后,供体来源的细胞保留其成熟表型,以单细胞RNA测序(scRNA-seq)为特征,组织学分析,和功能评估,证明移植细胞的持续增殖和分化能力。这些结果表明远端肺兼性祖细胞和分化上皮细胞区室与PSC衍生细胞的持久重建,因此建立了一种新的肺细胞治疗模型,可用于更好地理解移植的机制和效用。
    Durable reconstitution of the distal lung epithelium with pluripotent stem cell (PSC) derivatives, if realized, would represent a promising therapy for diseases that result from alveolar damage. Here, we differentiate murine PSCs into self-renewing lung epithelial progenitors able to engraft into the injured distal lung epithelium of immunocompetent, syngeneic mouse recipients. After transplantation, these progenitors mature in the distal lung, assuming the molecular phenotypes of alveolar type 2 (AT2) and type 1 (AT1) cells. After months in vivo, donor-derived cells retain their mature phenotypes, as characterized by single-cell RNA sequencing (scRNA-seq), histologic profiling, and functional assessment that demonstrates continued capacity of the engrafted cells to proliferate and differentiate. These results indicate durable reconstitution of the distal lung\'s facultative progenitor and differentiated epithelial cell compartments with PSC-derived cells, thus establishing a novel model for pulmonary cell therapy that can be utilized to better understand the mechanisms and utility of engraftment.
    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
    许多肺部病变存在性别差异,包括COVID-19和肺纤维化,但其机制基础尚不清楚。肺泡2型细胞(AT2s),在肺泡肺再生中起关键作用,表达在肺修复和SARS-CoV-2发病机制中起作用的X连锁Ace2基因,表明AT2中X染色体失活(XCI)可能会影响性别偏倚的肺病理。在这里,我们研究了使用男性和女性AT2的XCI维持和性别特异性基因表达谱。值得注意的是,非活性X染色体(Xi)缺乏强大的经典XistRNA“云”,并且在人类和小鼠AT2中缺乏异色修饰。我们证明,在小鼠AT2s中大约68%的表达X连锁基因,包括Ace2,逃离XCI。男性和女性AT2之间存在全基因组表达差异,可能影响肺生理和病理生理反应。这些研究支持对AT2的重新关注,AT2是导致肺部疾病性别偏见差异的潜在因素。
    Sex differences exist for many lung pathologies, including COVID-19 and pulmonary fibrosis, but the mechanistic basis for this remains unclear. Alveolar type 2 cells (AT2s), which play a key role in alveolar lung regeneration, express the X-linked Ace2 gene that has roles in lung repair and SARS-CoV-2 pathogenesis, suggesting that X chromosome inactivation (XCI) in AT2s might impact sex-biased lung pathology. Here we investigate XCI maintenance and sex-specific gene expression profiles using male and female AT2s. Remarkably, the inactive X chromosome (Xi) lacks robust canonical Xist RNA \"clouds\" and less enrichment of heterochromatic modifications in human and mouse AT2s. We demonstrate that about 68% of expressed X-linked genes in mouse AT2s, including Ace2, escape XCI. There are genome-wide expression differences between male and female AT2s, likely influencing both lung physiology and pathophysiologic responses. These studies support a renewed focus on AT2s as a potential contributor to sex-biased differences in lung 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
    呼吸道上皮由多个,功能上不同的细胞类型,并由损伤后修复上皮的区域特异性祖细胞群维持。存在几种使用原代鼠肺细胞研究肺上皮修复的体外方法,但是分离方法由于缺乏区分呼吸道上皮上皮祖细胞的表面标记而受到阻碍。这里,我们开发了一种3D打印的叶分隔器(3DLD),以帮助从多个体外测定中产生分化上皮的单个小鼠中同时分离近端和远端肺上皮祖细胞。与3DLD分离的远端祖细胞相反,常用的手动气管结扎方法,然后去除肺叶,导致罕见的近端细胞与远端细胞的共同分离,改变了远端类器官的转录景观和大小分布。3DLD有助于远端与近端祖细胞的可重复分离,并最大程度地减少了污染人群混淆体外测定的可能性。
    The respiratory epithelium consists of multiple, functionally distinct cell types and is maintained by regionally specific progenitor populations that repair the epithelium following injury. Several in vitro methods exist for studying lung epithelial repair using primary murine lung cells, but isolation methods are hampered by a lack of surface markers distinguishing epithelial progenitors along the respiratory epithelium. Here, we developed a 3D printed lobe divider (3DLD) to aid in simultaneous isolation of proximal versus distal lung epithelial progenitors from individual mice that give rise to differentiated epithelia in multiple in vitro assays. In contrast to 3DLD-isolated distal progenitor cells, commonly used manual tracheal ligation methods followed by lobe removal resulted in co-isolation of rare proximal cells with distal cells, which altered the transcriptional landscape and size distribution of distal organoids. The 3DLD aids in reproducible isolation of distal versus proximal progenitor populations and minimizes the potential for contaminating populations to confound in vitro assays.
    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
    吸烟已被确定有助于各种呼吸系统疾病的发病机理,包括慢性阻塞性肺疾病(COPD),肺癌,和哮喘。然而,关于吸烟在人肺中的作用的机械研究仍然存在主要障碍,部分原因是缺乏离体实验模型和动物模型的模糊数据,这些数据可以最好地概括人肺的结构和病理生理学。肺类器官培养系统的最新发展为呼吸系统疾病研究开辟了新的途径,因为类器官被证明是一种复杂的离体模型,其在功能和结构上比其他传统使用的模型更好地模拟人肺。这篇综述将讨论肺类器官系统的最新进展如何帮助我们更好地确定吸烟对人类肺的伤害和免疫影响,并将为未来的研究方向提供一些建议。
    Tobacco smoking has been established to contribute to the pathogenesis of various respiratory diseases including chronic obstructive pulmonary disease (COPD), lung cancer, and asthma. However, major hurdles in mechanistic studies on the role of smoking in human lungs remain in part due to the lack of ex vivo experimental models and ambiguous data from animal models that can best recapitulate the architecture and pathophysiology of the human lung. Recent development of the lung organoid culture system has opened new avenues for respiratory disease research as organoids are proving to be a sophisticated ex vivo model that functionally and structurally mimics the human lungs better than other traditionally used models. This review will discuss how recent advances in lung organoid systems may help us better determine the injurious and immunological effect of smoking on human lungs and will provide some suggestions for future research directions.
    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:健康的肺泡上皮对肺的气体交换功能至关重要。作为肺泡上皮细胞的主要细胞类型,肺泡2型(AT2)细胞在肺损伤期间作为肺泡祖细胞,在维持肺稳态中起关键作用,炎症,和修复。AT2细胞的失调可能导致急慢性肺部疾病和癌症的发展。缺乏临床相关的AT2细胞模型阻碍了我们了解肺部疾病的能力。这里,我们试图建立可逆永生化的小鼠肺泡2型细胞(imPAC2),并研究其在形成肺泡类器官以模拟肺部疾病方面的潜力.
    方法:分离原代小鼠肺泡细胞(mPAC),并通过逆转录病毒表达SV40大T抗原(LTA)使其永生化。通过结晶紫染色和WST-1测定评估细胞增殖和存活。通过qPCR评估标志物基因表达,西方印迹,和/或免疫染色。通过使用基质胶产生肺泡类器官。Ad-TGF-β1用于瞬时表达TGF-β1。通过使用逆转录病毒载体实现稳定沉默β-连环蛋白或突变体KRAS和TP53的过表达。在无胸腺裸鼠中进行皮下细胞植入。对取回的组织块进行H&E组织学评价。
    结果:我们用SV40LTA使原发性mPAC永生化,以产生非致瘤性并保持长期增殖活性的imPAC,该活性通过FLP介导的SV40LTA去除是可逆的。富含EpCAM+AT2的亚群(即,IMPAC2)是从IMPAC中挑选出来的,并显示表达AT2标记并形成肺泡类器官。功能上,沉默β-catenin降低了IMPAC2细胞中AT2标记的表达,而TGF-β1通过调节imPAC2细胞中上皮-间质转化标志物的表达诱导纤维化样反应。最后,致癌KRAS和突变型TP53的同时表达使imPAC2细胞成为肿瘤样表型,并激活了肺癌相关通路.总的来说,我们的结果表明,imPAC2细胞可能忠实地代表AT2群体,可进一步探索以模拟肺部疾病.
    BACKGROUND: A healthy alveolar epithelium is critical to the gas exchange function of the lungs. As the major cell type of alveolar epithelium, alveolar type 2 (AT2) cells play a critical role in maintaining pulmonary homeostasis by serving as alveolar progenitors during lung injury, inflammation, and repair. Dysregulation of AT2 cells may lead to the development of acute and chronic lung diseases and cancer. The lack of clinically relevant AT2 cell models hampers our ability to understand pulmonary diseases. Here, we sought to establish reversibly immortalized mouse pulmonary alveolar type 2 cells (imPAC2) and investigate their potential in forming alveolar organoids to model pulmonary diseases.
    METHODS: Primary mouse pulmonary alveolar cells (mPACs) were isolated and immortalized with a retroviral expression of SV40 Large T antigen (LTA). Cell proliferation and survival was assessed by crystal violet staining and WST-1 assays. Marker gene expression was assessed by qPCR, Western blotting, and/or immunostaining. Alveolar organoids were generated by using matrigel. Ad-TGF-β1 was used to transiently express TGF-β1. Stable silencing β-catenin or overexpression of mutant KRAS and TP53 was accomplished by using retroviral vectors. Subcutaneous cell implantations were carried out in athymic nude mice. The retrieved tissue masses were subjected to H & E histologic evaluation.
    RESULTS: We immortalized primary mPACs with SV40 LTA to yield the imPACs that were non-tumorigenic and maintained long-term proliferative activity that was reversible by FLP-mediated removal of SV40 LTA. The EpCAM+ AT2-enriched subpopulation (i.e., imPAC2) was sorted out from the imPACs, and was shown to express AT2 markers and form alveolar organoids. Functionally, silencing β-catenin decreased the expression of AT2 markers in imPAC2 cells, while TGF-β1 induced fibrosis-like response by regulating the expression of epithelial-mesenchymal transition markers in the imPAC2 cells. Lastly, concurrent expression of oncogenic KRAS and mutant TP53 rendered the imPAC2 cells a tumor-like phenotype and activated lung cancer-associated pathways. Collectively, our results suggest that the imPAC2 cells may faithfully represent AT2 populations that can be further explored to model pulmonary diseases.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Editorial
    《病理学杂志》2022年年度评论,病理学的最新进展,包含15篇关于病理学中日益重要的研究领域的特邀评论。今年,这些文章包括那些专注于数字病理学的文章,采用现代成像技术和软件来改进诊断和研究应用,以研究人类疾病。该主题领域包括通过其诱导的形态变化来识别特定遗传改变的能力,以及将数字和计算病理学与组学技术集成。本期的其他评论包括对癌症突变模式(突变特征)的最新评估,谱系追踪在人体组织中的应用,和单细胞测序技术来揭示肿瘤进化和肿瘤异质性。组织微环境包含在专门处理表皮分化的蛋白水解控制的综述中,癌症相关成纤维细胞,场抵消,和决定肿瘤免疫的宿主因子。本期中包含的所有评论都是受邀专家的工作,这些专家被选中讨论各自领域的最新进展,并且可以在线免费获得(https://onlinelibrary。wiley.com/journal/10969896)。©2022英国和爱尔兰病理学会。由JohnWiley&Sons出版,Ltd.
    The 2022 Annual Review Issue of The Journal of Pathology, Recent Advances in Pathology, contains 15 invited reviews on research areas of growing importance in pathology. This year, the articles include those that focus on digital pathology, employing modern imaging techniques and software to enable improved diagnostic and research applications to study human diseases. This subject area includes the ability to identify specific genetic alterations through the morphological changes they induce, as well as integrating digital and computational pathology with \'omics technologies. Other reviews in this issue include an updated evaluation of mutational patterns (mutation signatures) in cancer, the applications of lineage tracing in human tissues, and single cell sequencing technologies to uncover tumour evolution and tumour heterogeneity. The tissue microenvironment is covered in reviews specifically dealing with proteolytic control of epidermal differentiation, cancer-associated fibroblasts, field cancerisation, and host factors that determine tumour immunity. All of the reviews contained in this issue are the work of invited experts selected to discuss the considerable recent progress in their respective fields and are freely available online (https://onlinelibrary.wiley.com/journal/10969896). © 2022 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
    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
    在正常健康状态下,人肺是相对静止的器官,但含有有助于正常组织维持以及响应于损伤和疾病的修复或重塑的干/祖细胞。维持或修复导致功能性肺组织的适当恢复和生理功能的维持,重塑导致通常与疾病相关的结构和功能改变。有助于肺组织维持和修复/重塑的细胞类型的知识在很大程度上依赖于损伤修复的小鼠模型和局部祖细胞的谱系追踪。因此,人类肺部疾病重塑的许多功能改变仍未明确。然而,以单细胞分辨率定义肺细胞分子表型的先进基因组学方法的出现,为我们对正常人肺中存在的细胞类型和伴随疾病的变化的理解的快速进展铺平了道路.在这里,我们总结了从单细胞转录组学研究中出现的人类肺上皮分子表型疾病相关变化的最新进展。我们将注意力集中在上皮过渡状态的新兴概念上,这些概念表征了伴随慢性肺部疾病的病理性重塑,包括特发性肺纤维化,慢性阻塞性肺疾病,囊性纤维化,和哮喘。这些研究产生的概念正在积极发展,需要确凿的研究来提高我们对疾病机制的理解。只要有可能,我们强调在这个快速发展的研究领域提供统一命名法的机会。©2022英国和爱尔兰病理学会。
    The human lung is a relatively quiescent organ in the normal healthy state but contains stem/progenitor cells that contribute to normal tissue maintenance and either repair or remodeling in response to injury and disease. Maintenance or repair lead to proper restoration of functional lung tissue and maintenance of physiological functions, with remodeling resulting in altered structure and function that is typically associated with disease. Knowledge of cell types contributing to lung tissue maintenance and repair/remodeling have largely relied on mouse models of injury-repair and lineage tracing of local progenitors. Therefore, many of the functional alterations underlying remodeling in human lung disease have remained poorly defined. However, the advent of advanced genomics approaches to define the molecular phenotype of lung cells at single-cell resolution has paved the way for rapid advances in our understanding of cell types present within the normal human lung and changes that accompany disease. Here we summarize recent advances in our understanding of disease-related changes in the molecular phenotype of human lung epithelium that have emerged from single-cell transcriptomic studies. We focus attention on emerging concepts of epithelial transitional states that characterize the pathological remodeling that accompanies chronic lung diseases, including idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, cystic fibrosis, and asthma. Concepts arising from these studies are actively evolving and require corroborative studies to improve our understanding of disease mechanisms. Whenever possible, we highlight opportunities for providing a unified nomenclature in this rapidly advancing field of research. © 2022 The Pathological Society of Great Britain and Ireland.
    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
    Over the last decades, several studies demonstrated the possibility of lung regeneration through transplantation of various lung progenitor populations. Recently, we showed in mice that fetal or adult lung progenitors could potentially provide donor cells for transplantation, provided that the lung stem cell niche in the recipient is vacated of endogenous lung progenitors by adequate conditioning. Accordingly, marked lung regeneration could be attained following i.v. infusion of a single cell suspension of lung cells into recipient mice conditioned with naphthalene (NA) and 6Gy total body irradiation (TBI). As clinical translation of this approach requires the use of allogenic donors, we more recently developed a novel transplantation modality based on co-infusion of hematopoietic and lung progenitors from the same donor. Thus, by virtue of hematopoietic chimerism, which leads to immune tolerance toward donor antigens, the lung progenitors can be successfully engrafted without any need for post-transplant immune suppression. In the present study, we demonstrate that it is possible to replace NA in the conditioning regimen with Cyclophosphamide (CY), approved for the treatment of many diseases and that a lower dose of 2 GY TBI can successfully enable engraftment of donor-derived hematopoietic and lung progenitors when CY is administered in 2 doses after the stem cell infusion. Taken together, our results suggest a feasible and relatively safe protocol that could potentially be translated to clinical transplantation of lung progenitors across major MHC barriers in patients with terminal lung diseases.
    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
    肺移植是治疗终末期肺部疾病的唯一方法;捐助者短缺是一个全球性问题。使用人多能干细胞(hPSC)进行器官再生是一种有前途的方法。然而,用于移植目的的分离的hPSC衍生的肺祖细胞(hLPs)的扩增方法尚未被报道。在这里,基于hLPs在核壳水凝胶微纤维中的三维培养,确保维持其分化为肺泡和气道上皮细胞的双能性,包括肺泡II型(AT2)细胞。Further,我们使用内窥镜辅助的经气管给药系统开发了一种有效的体内移植方法;观察到hLPs的成功植入和体内分化为肺泡上皮细胞(掺入肺泡)。重要的是,在微纤维的情况下,扩大的hLP被成功地移植到鼠肺中,为肺部疾病的细胞疗法开辟了道路。因此,我们的新方法具有潜在的再生医学应用;此外,通过基于微纤维的技术产生的高质量hLPs和AT2细胞对于药物发现目的很有价值.
    Lung transplantation is the only treatment available for end-stage lung diseases; however, donor shortage is a global issue. The use of human pluripotent stem cells (hPSCs) for organ regeneration is a promising approach. Nevertheless, methods for the expansion of isolated hPSC-derived lung progenitors (hLPs) for transplantation purposes have not yet been reported. Herein, we established an expansion system of hLPs based on their three-dimensional culture in core-shell hydrogel microfibers, that ensures the maintenance of their bipotency for differentiation into alveolar and airway epithelial cells including alveolar type II (AT2) cells. Further, we developed an efficient in vivo transplantation method using an endoscope-assisted transtracheal administration system; the successful engraftment and in vivo differentiation of hLPs into alveolar epithelial cells (incorporated into the alveoli) was observed. Importantly, expanded hLPs in the context of microfibers were successfully transplanted into the murine lungs, opening avenues for cell-based therapies of lung diseases. Therefore, our novel method has potential regenerative medicine applications; additionally, the high-quality hLPs and AT2 cells generated via the microfiber-based technology are valuable for drug discovery purposes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号