Kidney organoid

肾类器官
  • 文章类型: Journal Article
    在过去的十年里,基于在发育性肾病学中获得的知识,已经建立了来自多能干细胞(PSC)的两种类型的肾脏类器官(肾单位类器官和输尿管芽类器官)的诱导方案.肾脏类器官现在用于疾病建模和药物筛选,但它们也有可能作为临床移植治疗的工具。实现这一目标的选择之一是组装多个肾祖细胞(肾单位祖细胞,输尿管芽,基质祖细胞)从PSC复制器官型肾脏结构。至少从小鼠PSC,所有这三个祖细胞都被诱导并组装成这样的“高阶”肾类器官。我们将概述诱导肾祖细胞所需的发育肾病学,并讨论临床移植治疗中肾脏类器官的最新进展和剩余挑战。
    Over the past decade, the induction protocols for the two types of kidney organoids (nephron organoids and ureteric bud organoids) from pluripotent stem cells (PSCs) have been established based on the knowledge gained in developmental nephrology. Kidney organoids are now used for disease modeling and drug screening, but they also have potential as tools for clinical transplantation therapy. One of the options to achieve this goal would be to assemble multiple renal progenitor cells (nephron progenitor, ureteric bud, stromal progenitor) to reproduce the organotypic kidney structure from PSCs. At least from mouse PSCs, all the three progenitors have been induced and assembled into such \"higher order\" kidney organoids. We will provide an overview of the developmental nephrology required for the induction of renal progenitors and discuss recent advances and remaining challenges of kidney organoids for clinical transplantation 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
    遗传性或遗传性肾脏疾病是慢性肾脏疾病(CKD)的关键原因。DNA检测在临床环境中的增殖和广泛使用显着缓解了遗传性肾脏疾病的诊断,这些疾病曾经难以捉摸,但现在在以前被认为病因不明的CKD病例中越来越多地被发现。然而,尽管有这些诊断上的进步,对疾病发病机制和新药开发的研究面临重大障碍,主要是由于缺乏适当的动物模型以及临床研究中有限的患者队列带来的挑战。相反,人类诱导多能干细胞(hiPSCs)的出现和利用为遗传性肾病研究提供了一条有希望的途径.特别是,hiPSC来源的肾脏类器官系统的发展为研究各种形式的遗传性肾脏疾病提供了新的平台。此外,将CRISPR/Cas9技术整合到该系统中对于有效研究遗传性肾脏疾病具有巨大潜力.本文旨在探讨由hiPSCs产生的体外肾脏类器官在多种遗传性肾脏疾病研究中的应用。此外,它将深入研究这个研究平台的局限性,并概述推进这一关键领域研究的未来前景。
    Genetic or hereditary kidney disease stands as a pivotal cause of chronic kidney disease (CKD). The proliferation and widespread utilization of DNA testing in clinical settings have notably eased the diagnosis of genetic kidney diseases, which were once elusive but are now increasingly identified in cases previously deemed CKD of unknown etiology. However, despite these diagnostic strides, research into disease pathogenesis and novel drug development faces significant hurdles, chiefly due to the dearth of appropriate animal models and the challenges posed by limited patient cohorts in clinical studies. Conversely, the advent and utilization of human-induced pluripotent stem cells (hiPSCs) offer a promising avenue for genetic kidney disease research. Particularly, the development of hiPSC-derived kidney organoid systems presents a novel platform for investigating various forms of genetic kidney diseases. Moreover, the integration of the CRISPR/Cas9 technique into this system holds immense potential for efficient research on genetic kidney diseases. This review aims to explore the applications of in vitro kidney organoids generated from hiPSCs in the study of diverse genetic kidney diseases. Additionally, it will delve into the limitations of this research platform and outline future perspectives for advancing research in this crucial area.
    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
    幼年性肾单位网是一种遗传性肾性纤毛病伴囊性肾病,肾纤维化,以及儿童和年轻人的终末期肾衰竭。编码肾细胞素-1蛋白的NPHP1基因中的突变已被鉴定为最常见的负责基因,并导致肾髓质中囊肿的形成。青少年肾单位的分子发病机制仍然难以捉摸,即使在今天,也没有有效的药物来预防终末期肾衰竭。目前还没有人类细胞模型。这里,我们报道了第一个使用患者来源和基因编辑的人诱导性多能干细胞(hiPSCs)和这些hiPSCs来源的肾脏类器官的幼年肾单位的疾病模型.我们建立了来自患者来源的hiPSC的NPHP1过表达hiPSC和来自健康供体hiPSC的NPHP1缺陷型hiPSC。比较这些系列的hiPSC,我们发现与hiPSCs中NPHP1缺乏相关的原发性纤毛异常。从缺乏NPHP1的hiPSC产生的肾脏类器官在具有恒定旋转的悬浮培养物中经常形成肾囊肿。NPHP1的过表达挽救了患者来源的肾类器官中的这种囊肿形成。对这些肾脏类器官的转录组分析显示,NPHP1的丢失导致与上皮细胞中初级纤毛相关的基因表达降低,与细胞周期相关的基因表达升高。这些发现表明NPHP1丢失引起的原发性纤毛异常与肾囊肿形成中的异常增殖特征之间的关系。这些发现表明,基于hiPSC的青少年肾单位的系统疾病建模有助于阐明分子发病机理和开发新的疗法。
    Juvenile nephronophthisis is an inherited renal ciliopathy with cystic kidney disease, renal fibrosis, and end-stage renal failure in children and young adults. Mutations in the NPHP1 gene encoding nephrocystin-1 protein have been identified as the most frequently responsible gene and cause the formation of cysts in the renal medulla. The molecular pathogenesis of juvenile nephronophthisis remains elusive, and no effective medicines to prevent end-stage renal failure exist even today. No human cellular models have been available yet. Here, we report a first disease model of juvenile nephronophthisis using patient-derived and gene-edited human induced pluripotent stem cells (hiPSCs) and kidney organoids derived from these hiPSCs. We established NPHP1-overexpressing hiPSCs from patient-derived hiPSCs and NPHP1-deficient hiPSCs from healthy donor hiPSCs. Comparing these series of hiPSCs, we found abnormalities in primary cilia associated with NPHP1 deficiency in hiPSCs. Kidney organoids generated from the hiPSCs lacking NPHP1 formed renal cysts frequently in suspension culture with constant rotation. This cyst formation in patient-derived kidney organoids was rescued by overexpression of NPHP1. Transcriptome analysis on these kidney organoids revealed that loss of NPHP1 caused lower expression of genes related to primary cilia in epithelial cells and higher expression of genes related to the cell cycle. These findings suggested the relationship between abnormality in primary cilia induced by NPHP1 loss and abnormal proliferative characteristics in the formation of renal cysts. These findings demonstrated that hiPSC-based systematic disease modeling of juvenile nephronophthisis contributed to elucidating the molecular pathogenesis and developing new therapies.
    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
    Wilms肿瘤-1(WT1)是调节足细胞发育的关键转录因子。然而,足细胞发育过程中WT1功能的表观基因组机制尚未完全阐明。这里,生成胎儿肾脏和肾脏类器官的单细胞染色质可及性和基因表达图。WT1靶向基因的功能意义,对足细胞的发育和结构的维持至关重要,包括调节WNT信号通路的BMPER/PAX2/MAGI2,鉴定了维持肌动蛋白丝组织的MYH9和调节细胞连接组装的NPHS1。为了进一步说明WT1介导的转录调控在足细胞发育过程中的功能重要性,产生了培养和植入的患者来源的肾脏类器官,这些器官来源于WT1中具有杂合错义突变的患者的诱导多能干细胞(iPSC)。单细胞RNA测序(scRNA-seq)和功能测定的结果证实,WT1突变导致足细胞发育延迟,并导致细胞结构受损。由于其未能激活靶向基因MAGI2,MYH9和NPHS1。值得注意的是,使用CRISPR-Cas9基因编辑纠正患者iPSCs中的突变可以挽救足细胞表型.总的来说,这项工作阐明了与人类足细胞发育有关的WT1相关的表观基因组景观,并确定了WT1突变的致病作用。
    Wilms tumor-1(WT1) is a crucial transcription factor that regulates podocyte development. However, the epigenomic mechanism underlying the function of WT1 during podocyte development has yet to be fully elucidated. Here, single-cell chromatin accessibility and gene expression maps of foetal kidneys and kidney organoids are generated. Functional implications of WT1-targeted genes, which are crucial for the development of podocytes and the maintenance of their structure, including BMPER/PAX2/MAGI2 that regulates WNT signaling pathway, MYH9 that maintains actin filament organization and NPHS1 that modulates cell junction assembly are identified. To further illustrate the functional importance of WT1-mediated transcriptional regulation during podocyte development, cultured and implanted patient-derived kidney organoids derived from the Induced Pluripotent Stem Cell (iPSCs) of a patient with a heterozygous missense mutation in WT1 are generated. Results from single-cell RNA sequencing (scRNA-seq) and functional assays confirm that the WT1 mutation leads to delays in podocyte development and causes damage to cell structures, due to its failure to activate the targeting genes MAGI2, MYH9, and NPHS1. Notably, correcting the mutation in the patient iPSCs using CRISPR-Cas9 gene editing rescues the podocyte phenotype. Collectively, this work elucidates the WT1-related epigenomic landscape with respect to human podocyte development and identifies the disease-causing role of a WT1 mutation.
    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
    肾祖细胞(NPCs)自我更新并分化为肾单位,肾脏的功能单位。这里,p38和YAP活性的操纵允许原代小鼠和人NPC的长期克隆扩增以及来自人多能干细胞(hPSC)的诱导NPC(iNPC)。分子分析表明培养的iNPC非常类似于原代人NPC。相对于已发表的人类肾类器官方案,iNPC产生的肾单位类器官具有最少的脱靶细胞类型和增强的足细胞成熟。令人惊讶的是,NPC培养基揭示了人类足细胞程序中的可塑性,使足细胞重新编程为类似NPC的状态。基因组编辑的可扩展性和易用性促进了NPC培养中的全基因组CRISPR筛选,发现与肾脏发育和疾病相关的基因。Further,NPC指导的常染色体显性遗传多囊肾病(ADPKD)模型确定了一种小分子的膀胱形成抑制剂。这些发现突出了报告的iNPC平台在肾脏发育研究中的广泛应用,疾病,可塑性,和再生。
    Nephron progenitor cells (NPCs) self-renew and differentiate into nephrons, the functional units of the kidney. Here, manipulation of p38 and YAP activity allowed for long-term clonal expansion of primary mouse and human NPCs and induced NPCs (iNPCs) from human pluripotent stem cells (hPSCs). Molecular analyses demonstrated that cultured iNPCs closely resemble primary human NPCs. iNPCs generated nephron organoids with minimal off-target cell types and enhanced maturation of podocytes relative to published human kidney organoid protocols. Surprisingly, the NPC culture medium uncovered plasticity in human podocyte programs, enabling podocyte reprogramming to an NPC-like state. Scalability and ease of genome editing facilitated genome-wide CRISPR screening in NPC culture, uncovering genes associated with kidney development and disease. Further, NPC-directed modeling of autosomal-dominant polycystic kidney disease (ADPKD) identified a small-molecule inhibitor of cystogenesis. These findings highlight a broad application for the reported iNPC platform in the study of kidney development, disease, plasticity, and regeneration.
    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
    类器官技术正在迅速发展为器官(病理)生理学研究。肾小管是从成人肾脏组织或尿液中生长出来的长期扩张的类器官。扩张肾小管的祖先状态是以分化为代价的。这里,我们区分肾小管以模拟远端肾单位和收集管,肾脏的重要功能部位。分化抑制祖先性状并上调功能所需的基因。单细胞图谱显示,分化主要产生厚的上行肢体和主细胞。分化的人肾小管表达能够抑制利尿剂抑制电解质摄取的管腔NKCC2和ENaC,并能够进行疾病建模,如锂诱导的肾小管病模型所证明的。锂导致AQP2的标志性损失,诱导增殖,并上调炎症介质,如在体内所见。锂还抑制多段中的电解质传输。总之,这种肾小管样蛋白模型可以对健康和疾病中的人远端肾单位和集合管进行建模,并为开发改进的疗法提供了机会。
    Organoid technology is rapidly gaining ground for studies on organ (patho)physiology. Tubuloids are long-term expanding organoids grown from adult kidney tissue or urine. The progenitor state of expanding tubuloids comes at the expense of differentiation. Here, we differentiate tubuloids to model the distal nephron and collecting ducts, essential functional parts of the kidney. Differentiation suppresses progenitor traits and upregulates genes required for function. A single-cell atlas reveals that differentiation predominantly generates thick ascending limb and principal cells. Differentiated human tubuloids express luminal NKCC2 and ENaC capable of diuretic-inhibitable electrolyte uptake and enable disease modeling as demonstrated by a lithium-induced tubulopathy model. Lithium causes hallmark AQP2 loss, induces proliferation, and upregulates inflammatory mediators, as seen in vivo. Lithium also suppresses electrolyte transport in multiple segments. In conclusion, this tubuloid model enables modeling of the human distal nephron and collecting duct in health and disease and provides opportunities to develop improved 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
    干细胞的定向分化是产生用于再生疗法的肾组织的有吸引力的方法。目前,测试该组织再生潜力的最有用的平台是将其植入免疫功能低下的啮齿动物的肾脏。干细胞来源的肾脏组织在植入后血管化,但是,对于尿液从移植物进入宿主收集系统至关重要的上皮小管之间的联系尚未得到证实。我们表明,小管融合的一个重要障碍是纤维状胶原蛋白在移植物和宿主之间的界面处的积累。作为一种筛选策略,以确定可以防止这种胶原蛋白积累的因素,我们建议将实验室生长的肾组织封装在补充有候选化合物如重组蛋白的纤维蛋白水凝胶中,小分子,饲养细胞,和基因治疗载体来调节局部移植物环境。我们证明了AAV-DJ血清型是囊膜下区域的有效基因治疗载体,并且对该隔室中的间质细胞具有特异性。除了上皮小管融合的组织学评估,我们证明了两种尿液生物标志物测定的特异性,可用于检测近端肾单位(CD59)和远端肾单位(尿调节素)的人类特异性标志物,我们证明了人类移植物来源的尿液沉积到小鼠收集系统中。使用本报告中描述的测试平台,有可能通过功能性活体读出系统筛选促进移植物和宿主组织上皮融合的潜在因子.
    Directed differentiation of stem cells is an attractive approach to generate kidney tissue for regenerative therapies. Currently, the most informative platform to test the regenerative potential of this tissue is engraftment into kidneys of immunocompromised rodents. Stem cell-derived kidney tissue is vascularized following engraftment, but the connection between epithelial tubules that is critical for urine to pass from the graft to the host collecting system has not yet been demonstrated. We show that one significant obstacle to tubule fusion is the accumulation of fibrillar collagens at the interface between the graft and the host. As a screening strategy to identify factors that can prevent this collagen accumulation, we propose encapsulating laboratory-grown kidney tissue in fibrin hydrogels supplemented with candidate compounds such as recombinant proteins, small molecules, feeder cells, and gene therapy vectors to condition the local graft environment. We demonstrate that the AAV-DJ serotype is an efficient gene therapy vector for the subcapsular region and that it is specific for interstitial cells in this compartment. In addition to the histological evaluation of epithelial tubule fusion, we demonstrate the specificity of two urine biomarker assays that can be used to detect human-specific markers of the proximal nephron (CD59) and the distal nephron (uromodulin), and we demonstrate the deposition of human graft-derived urine into the mouse collecting system. Using the testing platform described in this report, it will be possible to systematically screen factors for their potential to promote epithelial fusion of graft and host tissue with a functional intravital read-out.
    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:源自人多能干细胞(HiPSCs)的肾脏类器官在药物筛选中具有巨大的应用,疾病建模,和细胞移植治疗。然而,这些应用是有限的,因为肾脏类器官不能像人类肾脏一样保持完整的形态和功能。肾脏的器官没有很好的分化,因为器官的核心缺乏氧气,营养,和脉管系统,这创造了必要的利基。低氧诱导因子-1α(HIF-1α)是低氧环境下血管形成和细胞存活的关键调节因子。在这方面,关于HIF-1α在肾脏类器官中的作用知之甚少。本研究试图探讨HIF-1α在肾脏器官血管化和相关疾病建模中的作用。
    方法:对于血管化研究,肾脏的类器官是从人类诱导的多能干细胞中产生的。我们通过质粒转染或处理的DMOG过表达HIF-1α(二甲基氧基烯丙基甘氨酸,HIF-1α稳定和积累的试剂)在肾祖细胞中检测内皮。对于疾病建模研究,我们在缺氧环境下用顺铂治疗肾类器官,用额外的HIF-1α转染。
    结果:HIF-1α过表达引起肾脏类器官血管化。在HIF-1α质粒转染和DMOG处理的类器官中,内皮细胞和血管紧张素分析参数增加。这些血管生成过程被VEGFR抑制剂部分阻断,semaxanib或axitinib。HIF-1α通过上调CD31和SOD2来保护顺铂诱导的肾损伤(裂解的半胱天冬酶3)。
    结论:我们证明HIF-1α引起了肾脏类器官血管化的过程,并在缺氧环境中保护了顺铂引起的肾脏类器官损伤。
    Kidney organoids derived from human pluripotent stem cells (HiPSCs) hold huge applications for drug screening, disease modeling, and cell transplanting therapy. However, these applications are limited since kidney organoid cannot maintain complete morphology and function like human kidney. Kidney organoids are not well differentiated since the core of the organoid lacked oxygen, nutrition, and vasculature, which creates essential niches. Hypoxia-inducible factor-1 α (HIF-1α) serves as a critical regulator in vascularization and cell survival under hypoxia environment. Less is known about the role of HIF-1α in kidney organoids in this regard. This study tried to investigate the effect of HIF-1α in kidney organoid vascularization and related disease modeling.
    For the vascularization study, kidney organoids were generated from human induced pluripotent stem cells. We overexpressed HIF-1α via plasmid transfection or treated DMOG (Dimethyloxallyl Glycine, an agent for HIF-1α stabilization and accumulation) in kidney progenitor cells to detect the endothelium. For the disease modeling study, we treated kidney organoid with cisplatin under hypoxia environment, with additional HIF-1α transfection.
    HIF-1α overexpression elicited kidney organoid vascularization. The endothelial cells and angiotool analysis parameters were increased in HIF-1α plasmid-transfected and DMOG-treated organoids. These angiogenesis processes were partially blocked by VEGFR inhibitors, semaxanib or axitinib. Cisplatin-induced kidney injury (Cleaved caspase 3) was protected by HIF-1α through the upregulation of CD31 and SOD2.
    We demonstrated that HIF-1α elicited the process of kidney organoid vascularization and protected against cisplatin-induced kidney organoid injury in hypoxia environment.
    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
    核型间质性肾炎(KIN)是由15q13.3上的FANCD2/FANCI相关核酸酶1(FAN1)基因突变引起的遗传性肾脏疾病,通过DNA损伤的不完全修复导致肾细胞核型肿大和纤维化。这项研究的目的是探索使用人类诱导性多能干细胞(hiPSC)衍生的肾脏类器官系统建模FAN1缺乏的肾脏疾病的可能性,也被称为KIN。我们使用WTC-11(野生型)hiPSC和FAN1-突变型hiPSC(包括KIN患者来源的hiPSC和FAN1编辑的hiPSC(WTC-11FAN1+/-))产生了肾脏类器官,在WTC-11-hiPSC中使用CRISPR/Cas9系统创建。每组的肾脏类器官用20nM丝裂霉素C(MMC)处理24或48小时,和Ki67和H2A组蛋白家族成员X(H2A。分析X)以检测DNA损伤并评估肾类器官内细胞的活力。WTC-11-hiPSCs和FAN1-突变型hiPSCs均成功分化成肾类器官而没有结构畸形。MMC处理48h显著增加DNA损伤标记的表达,而两种FAN1突变型肾类器官的细胞活力均降低。然而,这些发现在WTC-11肾类器官中观察到。这些结果表明,FAN1突变的肾脏类器官可以概括FAN1缺陷性肾脏疾病的表型。
    Karyomegalic interstitial nephritis (KIN) is a genetic kidney disease caused by mutations in the FANCD2/FANCI-Associated Nuclease 1 (FAN1) gene on 15q13.3, which results in karyomegaly and fibrosis of kidney cells through the incomplete repair of DNA damage. The aim of this study was to explore the possibility of using a human induced pluripotent stem cell (hiPSC)-derived kidney organoid system for modeling FAN1-deficient kidney disease, also known as KIN. We generated kidney organoids using WTC-11 (wild-type) hiPSCs and FAN1-mutant hiPSCs which include KIN patient-derived hiPSCs and FAN1-edited hiPSCs (WTC-11 FAN1+/-), created using the CRISPR/Cas9 system in WTC-11-hiPSCs. Kidney organoids from each group were treated with 20 nM of mitomycin C (MMC) for 24 or 48 h, and the expression levels of Ki67 and H2A histone family member X (H2A.X) were analyzed to detect DNA damage and assess the viability of cells within the kidney organoids. Both WTC-11-hiPSCs and FAN1-mutant hiPSCs were successfully differentiated into kidney organoids without structural deformities. MMC treatment for 48 h significantly increased the expression of DNA damage markers, while cell viability in both FAN1-mutant kidney organoids was decreased. However, these findings were observed in WTC-11-kidney organoids. These results suggest that FAN1-mutant kidney organoids can recapitulate the phenotype of FAN1-deficient kidney 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
    肾脏类器官已经实现了人类发育和疾病的建模。虽然产生肾单位谱系的方法已经建立,新的协议来诱导另一个谱系,输尿管芽/收集管,在过去的5年里有报道。许多报道描述了各种遗传性肾脏疾病的建模,多囊肾病是典型的疾病,通过使用患者来源或基因组编辑的肾脏类器官。更多器官型肾脏的产生也变得可行。在这次审查中,我还讨论了更复杂的疾病建模和实现产生可移植合成肾脏的宏伟目标的重大挑战。
    Kidney organoids have enabled modeling of human development and disease. While methods of generating the nephron lineage are well established, new protocols to induce another lineage, the ureteric bud/collecting duct, have been reported in the past 5 years. Many reports have described modeling of various hereditary kidney diseases, with polycystic kidney disease serving as the archetypal disease, by using patient-derived or genome-edited kidney organoids. The generation of more organotypic kidneys is also becoming feasible. In this review, I also discuss the significant challenges for more sophisticated disease modeling and for realizing the ambitious goal of generating transplantable synthetic kidneys.
    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