Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Nat Commun ; 13(1): 5943, 2022 10 08.
Article in English | MEDLINE | ID: mdl-36209212

ABSTRACT

While pluripotent stem cell-derived kidney organoids are now being used to model renal disease, the proximal nephron remains immature with limited evidence for key functional solute channels. This may reflect early mispatterning of the nephrogenic mesenchyme and/or insufficient maturation. Here we show that enhanced specification to metanephric nephron progenitors results in elongated and radially aligned proximalised nephrons with distinct S1 - S3 proximal tubule cell types. Such PT-enhanced organoids possess improved albumin and organic cation uptake, appropriate KIM-1 upregulation in response to cisplatin, and improved expression of SARS-CoV-2 entry factors resulting in increased viral replication. The striking proximo-distal orientation of nephrons resulted from localized WNT antagonism originating from the organoid stromal core. PT-enhanced organoids represent an improved model to study inherited and acquired proximal tubular disease as well as drug and viral responses.


Subject(s)
COVID-19 , Communicable Diseases , Albumins/metabolism , Cell Differentiation/physiology , Cisplatin/metabolism , Cisplatin/pharmacology , Communicable Diseases/metabolism , Humans , Kidney , Nephrons/metabolism , Organoids/metabolism , SARS-CoV-2
2.
bioRxiv ; 2022 May 27.
Article in English | MEDLINE | ID: mdl-35665006

ABSTRACT

While pluripotent stem cell-derived kidney organoids are now being used to model renal disease, the proximal nephron remains immature with limited evidence for key functional solute channels. This may reflect early mispatterning of the nephrogenic mesenchyme and/or insufficient maturation. Here we show that enhanced specification to metanephric nephron progenitors results in elongated and radially aligned proximalised nephrons with distinct S1 - S3 proximal tubule cell types. Such PT-enhanced organoids possess improved albumin and organic cation uptake, appropriate KIM-1 upregulation in response to cisplatin, and improved expression of SARS-CoV-2 entry factors resulting in increased viral replication. The striking proximo-distal orientation of nephrons resulted from localized WNT antagonism originating from the organoid stromal core. PT-enhanced organoids represent an improved model to study inherited and acquired proximal tubular disease as well as drug and viral responses.

3.
Genome Med ; 14(1): 19, 2022 02 22.
Article in English | MEDLINE | ID: mdl-35189942

ABSTRACT

BACKGROUND: While single-cell transcriptional profiling has greatly increased our capacity to interrogate biology, accurate cell classification within and between datasets is a key challenge. This is particularly so in pluripotent stem cell-derived organoids which represent a model of a developmental system. Here, clustering algorithms and selected marker genes can fail to accurately classify cellular identity while variation in analyses makes it difficult to meaningfully compare datasets. Kidney organoids provide a valuable resource to understand kidney development and disease. However, direct comparison of relative cellular composition between protocols has proved challenging. Hence, an unbiased approach for classifying cell identity is required. METHODS: The R package, scPred, was trained on multiple single cell RNA-seq datasets of human fetal kidney. A hierarchical model classified cellular subtypes into nephron, stroma and ureteric epithelial elements. This model, provided in the R package DevKidCC ( github.com/KidneyRegeneration/DevKidCC ), was then used to predict relative cell identity within published kidney organoid datasets generated using distinct cell lines and differentiation protocols, interrogating the impact of such variations. The package contains custom functions for the display of differential gene expression within cellular subtypes. RESULTS: DevKidCC was used to directly compare between distinct kidney organoid protocols, identifying differences in relative proportions of cell types at all hierarchical levels of the model and highlighting variations in stromal and unassigned cell types, nephron progenitor prevalence and relative maturation of individual epithelial segments. Of note, DevKidCC was able to distinguish distal nephron from ureteric epithelium, cell types with overlapping profiles that have previously confounded analyses. When applied to a variation in protocol via the addition of retinoic acid, DevKidCC identified a consequential depletion of nephron progenitors. CONCLUSIONS: The application of DevKidCC to kidney organoids reproducibly classifies component cellular identity within distinct single-cell datasets. The application of the tool is summarised in an interactive Shiny application, as are examples of the utility of in-built functions for data presentation. This tool will enable the consistent and rapid comparison of kidney organoid protocols, driving improvements in patterning to kidney endpoints and validating new approaches.


Subject(s)
Organoids , Pluripotent Stem Cells , Cell Differentiation/genetics , Humans , Kidney , Organogenesis/genetics , Pluripotent Stem Cells/metabolism
4.
Trends Mol Med ; 27(12): 1135-1158, 2021 12.
Article in English | MEDLINE | ID: mdl-34657800

ABSTRACT

Pluripotent stem cells underpin a growing sector that leverages their differentiation potential for research, industry, and clinical applications. This review evaluates the landscape of methods in single-cell transcriptomics that are enabling accelerated discovery in stem cell science. We focus on strategies for scaling stem cell differentiation through multiplexed single-cell analyses, for evaluating molecular regulation of cell differentiation using new analysis algorithms, and methods for integration and projection analysis to classify and benchmark stem cell derivatives against in vivo cell types. By discussing the available methods, comparing their strengths, and illustrating strategies for developing integrated analysis pipelines, we provide user considerations to inform their implementation and interpretation.


Subject(s)
Genomics , Pluripotent Stem Cells , Cell Differentiation/genetics , Humans , Pluripotent Stem Cells/physiology , Single-Cell Analysis/methods , Transcriptome
5.
Development ; 148(19)2021 10 01.
Article in English | MEDLINE | ID: mdl-34553766

ABSTRACT

The postnatal kidney is predominantly composed of nephron epithelia with the interstitial components representing a small proportion of the final organ, except in the diseased state. This is in stark contrast to the developing organ, which arises from the mesoderm and comprises an expansive stromal population with distinct regional gene expression. In many organs, the identity and ultimate function of an epithelium is tightly regulated by the surrounding stroma during development. However, although the presence of a renal stromal stem cell population has been demonstrated, the focus has been on understanding the process of nephrogenesis whereas the role of distinct stromal components during kidney morphogenesis is less clear. In this Review, we consider what is known about the role of the stroma of the developing kidney in nephrogenesis, where these cells come from as well as their heterogeneity, and reflect on how this information may improve human kidney organoid models.


Subject(s)
Embryonic Stem Cells/metabolism , Kidney/embryology , Animals , Cell Differentiation , Embryonic Stem Cells/cytology , Gene Expression Regulation, Developmental , Humans , Kidney/cytology , Kidney/metabolism , Organogenesis
6.
Cell Stem Cell ; 28(4): 671-684.e6, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33378647

ABSTRACT

During development, distinct progenitors contribute to the nephrons versus the ureteric epithelium of the kidney. Indeed, previous human pluripotent stem-cell-derived models of kidney tissue either contain nephrons or pattern specifically to the ureteric epithelium. By re-analyzing the transcriptional distinction between distal nephron and ureteric epithelium in human fetal kidney, we show here that, while existing nephron-containing kidney organoids contain distal nephron epithelium and no ureteric epithelium, this distal nephron segment alone displays significant in vitro plasticity and can adopt a ureteric epithelial tip identity when isolated and cultured in defined conditions. "Induced" ureteric epithelium cultures can be cryopreserved, serially passaged without loss of identity, and transitioned toward a collecting duct fate. Cultures harboring loss-of-function mutations in PKHD1 also recapitulate the cystic phenotype associated with autosomal recessive polycystic kidney disease.


Subject(s)
Organogenesis , Organoids , Cell Differentiation , Epithelium , Humans , Kidney , Nephrons
7.
Nat Mater ; 20(2): 260-271, 2021 02.
Article in English | MEDLINE | ID: mdl-33230326

ABSTRACT

Directed differentiation of human pluripotent stem cells to kidney organoids brings the prospect of drug screening, disease modelling and the generation of tissue for renal replacement. Currently, these applications are hampered by organoid variability, nephron immaturity, low throughput and limited scale. Here, we apply extrusion-based three-dimensional cellular bioprinting to deliver rapid and high-throughput generation of kidney organoids with highly reproducible cell number and viability. We demonstrate that manual organoid generation can be replaced by 6- or 96-well organoid bioprinting and evaluate the relative toxicity of aminoglycosides as a proof of concept for drug testing. In addition, three-dimensional bioprinting enables precise manipulation of biophysical properties, including organoid size, cell number and conformation, with modification of organoid conformation substantially increasing nephron yield per starting cell number. This facilitates the manufacture of uniformly patterned kidney tissue sheets with functional proximal tubular segments. Hence, automated extrusion-based bioprinting for kidney organoid production delivers improvements in throughput, quality control, scale and structure, facilitating in vitro and in vivo applications of stem cell-derived human kidney tissue.


Subject(s)
Bioprinting , Kidney Tubules, Proximal/metabolism , Organoids/metabolism , Pluripotent Stem Cells/metabolism , Humans , Kidney Tubules, Proximal/cytology , Organoids/cytology , Pluripotent Stem Cells/cytology
8.
Nat Commun ; 11(1): 1327, 2020 Mar 09.
Article in English | MEDLINE | ID: mdl-32152305

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

9.
J Am Soc Nephrol ; 30(10): 1811-1823, 2019 10.
Article in English | MEDLINE | ID: mdl-31492807

ABSTRACT

BACKGROUND: The generation of reporter lines for cell identity, lineage, and physiologic state has provided a powerful tool in advancing the dissection of mouse kidney morphogenesis at a molecular level. Although use of this approach is not an option for studying human development in vivo, its application in human induced pluripotent stem cells (iPSCs) is now feasible. METHODS: We used CRISPR/Cas9 gene editing to generate ten fluorescence reporter iPSC lines designed to identify nephron progenitors, podocytes, proximal and distal nephron, and ureteric epithelium. Directed differentiation to kidney organoids was performed according to published protocols. Using immunofluorescence and live confocal microscopy, flow cytometry, and cell sorting techniques, we investigated organoid patterning and reporter expression characteristics. RESULTS: Each iPSC reporter line formed well patterned kidney organoids. All reporter lines showed congruence of endogenous gene and protein expression, enabling isolation and characterization of kidney cell types of interest. We also demonstrated successful application of reporter lines for time-lapse imaging and mouse transplantation experiments. CONCLUSIONS: We generated, validated, and applied a suite of fluorescence iPSC reporter lines for the study of morphogenesis within human kidney organoids. This fluorescent iPSC reporter toolbox enables the visualization and isolation of key populations in forming kidney organoids, facilitating a range of applications, including cellular isolation, time-lapse imaging, protocol optimization, and lineage-tracing approaches. These tools offer promise for enhancing our understanding of this model system and its correspondence with human kidney morphogenesis.


Subject(s)
Induced Pluripotent Stem Cells , Kidney/cytology , Organoids , Animals , Female , Mice , Organogenesis
10.
EMBO Rep ; 20(4)2019 04.
Article in English | MEDLINE | ID: mdl-30858339

ABSTRACT

Nephron formation continues throughout kidney morphogenesis in both mice and humans. Lineage tracing studies in mice identified a self-renewing Six2-expressing nephron progenitor population able to give rise to the full complement of nephrons throughout kidney morphogenesis. To investigate the origin of nephrons within human pluripotent stem cell-derived kidney organoids, we performed a similar fate-mapping analysis of the SIX2-expressing lineage in induced pluripotent stem cell (iPSC)-derived kidney organoids to explore the feasibility of investigating lineage relationships in differentiating iPSCs in vitro Using CRISPR/Cas9 gene-edited lineage reporter lines, we show that SIX2-expressing cells give rise to nephron epithelial cell types but not to presumptive ureteric epithelium. The use of an inducible (CreERT2) line revealed a declining capacity for SIX2+ cells to contribute to nephron formation over time, but retention of nephron-forming capacity if provided an exogenous WNT signal. Hence, while human iPSC-derived kidney tissue appears to maintain lineage relationships previously identified in developing mouse kidney, unlike the developing kidney in vivo, kidney organoids lack a nephron progenitor niche capable of both self-renewal and ongoing nephrogenesis.


Subject(s)
Chromosome Mapping , Gene Expression Profiling , Genes, Reporter , Nephrons/embryology , Nephrons/metabolism , Organogenesis/genetics , Biomarkers , CRISPR-Cas Systems , Cell Culture Techniques , Cell Differentiation , Homeodomain Proteins/genetics , Humans , Nerve Tissue Proteins/genetics , Organoids , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Single-Cell Analysis
11.
Kidney Int ; 95(5): 1153-1166, 2019 05.
Article in English | MEDLINE | ID: mdl-30827514

ABSTRACT

All nephrons in the mammalian kidney arise from a transient nephron progenitor population that is lost close to the time of birth. The generation of new nephron progenitors and their maintenance in culture are central to the success of kidney regenerative strategies. Using a lentiviral screening approach, we previously generated a human induced nephron progenitor-like state in vitro using a pool of six transcription factors. Here, we sought to develop a more efficient approach for direct reprogramming of human cells that could be applied in vivo. PiggyBac transposons are a non-viral integrating gene delivery system that is suitable for in vivo use and allows for simultaneous delivery of multiple genes. Using an inducible piggyBac transposon system, we optimized a protocol for the direct reprogramming of HK2 cells to induced nephron progenitor-like cells with expression of only 3 transcription factors (SNAI2, EYA1, and SIX1). Culture in conditions supportive of the nephron progenitor state further increased the expression of nephron progenitor genes. The refined protocol was then applied to primary human renal epithelial cells, which integrated into developing nephron structures in vitro and in vivo. Such inducible reprogramming to nephron progenitor-like cells could facilitate direct cellular reprogramming for kidney regeneration.


Subject(s)
Cellular Reprogramming/genetics , DNA Transposable Elements/genetics , Genetic Engineering/methods , Nephrons/physiology , Regeneration/genetics , Cells, Cultured , Gene Transfer Techniques , Homeodomain Proteins/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Nuclear Proteins/genetics , Primary Cell Culture , Protein Tyrosine Phosphatases/genetics , Snail Family Transcription Factors/genetics
12.
Nat Commun ; 10(1): 168, 2019 01 11.
Article in English | MEDLINE | ID: mdl-30635573

ABSTRACT

In humans and in mice the formation of nephrons during embryonic development reaches completion near the end of gestation, after which no new nephrons are formed. The final nephron complement can vary 10-fold, with reduced nephron number predisposing individuals to hypertension, renal, and cardiovascular diseases in later life. While the heterochronic genes lin28 and let-7 are well-established regulators of developmental timing in invertebrates, their role in mammalian organogenesis is not fully understood. Here we report that the Lin28b/let-7 axis controls the duration of kidney development in mice. Suppression of let-7 miRNAs, directly or via the transient overexpression of LIN28B, can prolong nephrogenesis and enhance kidney function potentially via upregulation of the Igf2/H19 locus. In contrast, kidney-specific loss of Lin28b impairs renal development. Our study reveals mechanisms regulating persistence of nephrogenic mesenchyme and provides a rationale for therapies aimed at increasing nephron mass.


Subject(s)
DNA-Binding Proteins/metabolism , Kidney/embryology , MicroRNAs/metabolism , RNA-Binding Proteins/metabolism , Animals , Female , Insulin-Like Growth Factor II/metabolism , Kidney/metabolism , Kidney Function Tests , Male , Mice, Transgenic , RNA, Long Noncoding/metabolism
13.
Proc Natl Acad Sci U S A ; 115(23): 5998-6003, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29784808

ABSTRACT

Nephrogenesis concludes by the 36th week of gestation in humans and by the third day of postnatal life in mice. Extending the nephrogenic period may reduce the onset of adult renal and cardiovascular disease associated with low nephron numbers. We conditionally deleted either Mtor or Tsc1 (coding for hamartin, an inhibitor of Mtor) in renal progenitor cells. Loss of one Mtor allele caused a reduction in nephron numbers; complete deletion led to severe paucity of glomeruli in the kidney resulting in early death after birth. By contrast, loss of one Tsc1 allele from renal progenitors resulted in a 25% increase in nephron endowment with no adverse effects. Increased progenitor engraftment rates ex vivo relative to controls correlated with prolonged nephrogenesis through the fourth postnatal day. Complete loss of both Tsc1 alleles in renal progenitors led to a lethal tubular lesion. The hamartin phenotypes are not dependent on the inhibitory effect of TSC on the Mtor complex but are dependent on Raptor.


Subject(s)
Nephrons , Organogenesis/physiology , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Female , Male , Mice , Mice, Transgenic , Nephrons/chemistry , Nephrons/cytology , Nephrons/growth & development , Nephrons/physiology , TOR Serine-Threonine Kinases/genetics , Tuberous Sclerosis Complex 1 Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...