101
|
Howden SE, Vanslambrouck JM, Wilson SB, Tan KS, Little MH. Reporter-based fate mapping in human kidney organoids confirms nephron lineage relationships and reveals synchronous nephron formation. EMBO Rep 2019; 20:embr.201847483. [PMID: 30858339 DOI: 10.15252/embr.201847483] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 02/05/2019] [Accepted: 02/08/2019] [Indexed: 01/15/2023] Open
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.
Collapse
Affiliation(s)
- Sara E Howden
- Murdoch Children's Research Institute, Parkville, Vic., Australia .,Department of Paediatrics, The University of Melbourne, Melbourne, Vic., Australia
| | - Jessica M Vanslambrouck
- Murdoch Children's Research Institute, Parkville, Vic., Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, Vic., Australia
| | - Sean B Wilson
- Murdoch Children's Research Institute, Parkville, Vic., Australia
| | - Ker Sin Tan
- Murdoch Children's Research Institute, Parkville, Vic., Australia
| | - Melissa H Little
- Murdoch Children's Research Institute, Parkville, Vic., Australia .,Department of Paediatrics, The University of Melbourne, Melbourne, Vic., Australia.,Department of Anatomy and Neuroscience, The University of Melbourne, Melbourne, Vic., Australia
| |
Collapse
|
102
|
Munro DAD, Wineberg Y, Tarnick J, Vink CS, Li Z, Pridans C, Dzierzak E, Kalisky T, Hohenstein P, Davies JA. Macrophages restrict the nephrogenic field and promote endothelial connections during kidney development. eLife 2019; 8:43271. [PMID: 30758286 PMCID: PMC6374076 DOI: 10.7554/elife.43271] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/29/2019] [Indexed: 12/17/2022] Open
Abstract
The origins and functions of kidney macrophages in the adult have been explored, but their roles during development remain largely unknown. Here we characterise macrophage arrival, localisation, heterogeneity, and functions during kidney organogenesis. Using genetic approaches to ablate macrophages, we identify a role for macrophages in nephron progenitor cell clearance as mouse kidney development begins. Throughout renal organogenesis, most kidney macrophages are perivascular and express F4/80 and CD206. These macrophages are enriched for mRNAs linked to developmental processes, such as blood vessel morphogenesis. Using antibody-mediated macrophage-depletion, we show macrophages support vascular anastomoses in cultured kidney explants. We also characterise a subpopulation of galectin-3+ (Gal3+) myeloid cells within the developing kidney. Our findings may stimulate research into macrophage-based therapies for renal developmental abnormalities and have implications for the generation of bioengineered kidney tissues. The kidneys clean our blood by filtering out waste products while ensuring that useful components, like nutrients, remain in the bloodstream. Blood enters the kidneys through a network of intricately arranged blood vessels, which associate closely with the ‘cleaning tubes’ that carry out filtration. Human kidneys start developing during the early phases of embryonic development. During this process, the newly forming blood vessels and cleaning tubes must grow in the right places for the adult kidney to work properly. Macrophages are cells of the immune system that clear away foreign, diseased, or damaged cells. They are also thought to encourage growth of the developing kidney, but how exactly they do this has remained unknown. Munro et al. therefore wanted to find out when macrophages first appeared in the embryonic kidney and how they might help control their development. Experiments using mice revealed that the first macrophages arrived in the kidney early during its development, alongside newly forming blood vessels. Further investigation using genetically modified mice that did not have macrophages revealed that these immune cells were needed at this stage to clear away misplaced kidney cells and help ‘set the scene’ for future development. At later stages, macrophages in the kidney interacted closely with growing blood vessels. As well as producing molecules linked with blood vessel formation, the macrophages wrapped around the vessels themselves, sometimes even eating cells lining the vessels and the blood cells carried within them. These observations suggested that macrophages actively shaped the network of blood vessels developing within the kidneys. Experiments removing macrophages from kidney tissue confirmed this: in normal kidneys, the blood vessels grew into a continuous network, but in kidneys lacking macrophages, far fewer connections formed between the vessels. This work sheds new light on how the complex structures in the adult kidney first arise and could be useful in future research. For example, adding macrophages to simplified, laboratory-grown ‘mini-kidneys’ could make them better models to study kidney growth, while patients suffering from kidney diseases might benefit from new drugs targeting macrophages.
Collapse
Affiliation(s)
- David AD Munro
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| | - Yishay Wineberg
- Department of Bioengineering, Bar-Ilan University, Ramat Gan, Israel.,Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan, Israel
| | - Julia Tarnick
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| | - Chris S Vink
- Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Zhuan Li
- Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Clare Pridans
- Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Elaine Dzierzak
- Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Tomer Kalisky
- Department of Bioengineering, Bar-Ilan University, Ramat Gan, Israel.,Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan, Israel
| | - Peter Hohenstein
- Leiden University Medical Center, Leiden University, Leiden, The Netherlands.,The Roslin Institute, The University of Edinburgh, Midlothian, United Kingdom
| | - Jamie A Davies
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
103
|
Lawlor KT, Zappia L, Lefevre J, Park JS, Hamilton NA, Oshlack A, Little MH, Combes AN. Nephron progenitor commitment is a stochastic process influenced by cell migration. eLife 2019; 8:41156. [PMID: 30676318 PMCID: PMC6363379 DOI: 10.7554/elife.41156] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 01/23/2019] [Indexed: 12/31/2022] Open
Abstract
Progenitor self-renewal and differentiation is often regulated by spatially restricted cues within a tissue microenvironment. Here, we examine how progenitor cell migration impacts regionally induced commitment within the nephrogenic niche in mice. We identify a subset of cells that express Wnt4, an early marker of nephron commitment, but migrate back into the progenitor population where they accumulate over time. Single cell RNA-seq and computational modelling of returning cells reveals that nephron progenitors can traverse the transcriptional hierarchy between self-renewal and commitment in either direction. This plasticity may enable robust regulation of nephrogenesis as niches remodel and grow during organogenesis.
Collapse
Affiliation(s)
- Kynan T Lawlor
- Murdoch Children's Research Institute, Parkville, Australia
| | - Luke Zappia
- Murdoch Children's Research Institute, Parkville, Australia.,School of Biosciences, University of Melbourne, Melbourne, Australia
| | - James Lefevre
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Joo-Seop Park
- Division of Pediatric Urology and Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, United States
| | - Nicholas A Hamilton
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Alicia Oshlack
- Murdoch Children's Research Institute, Parkville, Australia.,School of Biosciences, University of Melbourne, Melbourne, Australia
| | - Melissa H Little
- Murdoch Children's Research Institute, Parkville, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, Australia.,Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Australia
| | - Alexander N Combes
- Murdoch Children's Research Institute, Parkville, Australia.,Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Australia
| |
Collapse
|
104
|
Combes AN, Zappia L, Er PX, Oshlack A, Little MH. Single-cell analysis reveals congruence between kidney organoids and human fetal kidney. Genome Med 2019; 11:3. [PMID: 30674341 PMCID: PMC6345028 DOI: 10.1186/s13073-019-0615-0] [Citation(s) in RCA: 149] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 01/14/2019] [Indexed: 01/12/2023] Open
Abstract
Background Human kidney organoids hold promise for studying development, disease modelling and drug screening. However, the utility of stem cell-derived kidney tissues will depend on how faithfully these replicate normal fetal development at the level of cellular identity and complexity. Methods Here, we present an integrated analysis of single cell datasets from human kidney organoids and human fetal kidney to assess similarities and differences between the component cell types. Results Clusters in the combined dataset contained cells from both organoid and fetal kidney with transcriptional congruence for key stromal, endothelial and nephron cell type-specific markers. Organoid enriched neural, glial and muscle progenitor populations were also evident. Major transcriptional differences between organoid and human tissue were likely related to technical artefacts. Cell type-specific comparisons revealed differences in stromal, endothelial and nephron progenitor cell types including expression of WNT2B in the human fetal kidney stroma. Conclusions This study supports the fidelity of kidney organoids as models of the developing kidney and affirms their potential in disease modelling and drug screening. Electronic supplementary material The online version of this article (10.1186/s13073-019-0615-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alexander N Combes
- Department of Anatomy & Neuroscience, University of Melbourne, Melbourne, VIC, Australia. .,Murdoch Children's Research Institute, Melbourne, VIC, Australia.
| | - Luke Zappia
- Murdoch Children's Research Institute, Melbourne, VIC, Australia.,School of Biosciences, The University of Melbourne, Melbourne, VIC, Australia
| | - Pei Xuan Er
- Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | - Alicia Oshlack
- Murdoch Children's Research Institute, Melbourne, VIC, Australia.,School of Biosciences, The University of Melbourne, Melbourne, VIC, Australia
| | - Melissa H Little
- Department of Anatomy & Neuroscience, University of Melbourne, Melbourne, VIC, Australia. .,Murdoch Children's Research Institute, Melbourne, VIC, Australia. .,School of Biosciences, The University of Melbourne, Melbourne, VIC, Australia. .,Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
105
|
Image-based modeling of kidney branching morphogenesis reveals GDNF-RET based Turing-type mechanism and pattern-modulating WNT11 feedback. Nat Commun 2019; 10:239. [PMID: 30651543 PMCID: PMC6484223 DOI: 10.1038/s41467-018-08212-8] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 12/22/2018] [Indexed: 11/08/2022] Open
Abstract
Branching patterns and regulatory networks differ between branched organs. It has remained unclear whether a common regulatory mechanism exists and how organ-specific patterns can emerge. Of all previously proposed signalling-based mechanisms, only a ligand-receptor-based Turing mechanism based on FGF10 and SHH quantitatively recapitulates the lung branching patterns. We now show that a GDNF-dependent ligand-receptor-based Turing mechanism quantitatively recapitulates branching of cultured wildtype and mutant ureteric buds, and achieves similar branching patterns when directing domain outgrowth in silico. We further predict and confirm experimentally that the kidney-specific positive feedback between WNT11 and GDNF permits the dense packing of ureteric tips. We conclude that the ligand-receptor based Turing mechanism presents a common regulatory mechanism for lungs and kidneys, despite the differences in the molecular implementation. Given its flexibility and robustness, we expect that the ligand-receptor-based Turing mechanism constitutes a likely general mechanism to guide branching morphogenesis and other symmetry breaks during organogenesis. Many organs develop through branching morphogenesis, but whether the underlying mechanisms are shared is unknown. Here, the authors show that a ligand-receptor based Turing mechanisms, similar to that observed in lung development, likely underlies branching morphogenesis of the kidney.
Collapse
|
106
|
Lin28 and let-7 regulate the timing of cessation of murine nephrogenesis. Nat Commun 2019; 10:168. [PMID: 30635573 PMCID: PMC6329821 DOI: 10.1038/s41467-018-08127-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 12/12/2018] [Indexed: 01/10/2023] Open
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. Nephrogenesis ceases after postnatal day 2 in the mouse or after the 36th week of gestation in humans, but how this is regulated is unclear. Here, the authors identify a role for the RNA-binding protein Lin28 and suppression of let-7 microRNA in regulating the duration of nephrogenesis.
Collapse
|
107
|
Wanner N, Vornweg J, Combes A, Wilson S, Plappert J, Rafflenbeul G, Puelles VG, Rahman RU, Liwinski T, Lindner S, Grahammer F, Kretz O, Wlodek ME, Romano T, Moritz KM, Boerries M, Busch H, Bonn S, Little MH, Bechtel-Walz W, Huber TB. DNA Methyltransferase 1 Controls Nephron Progenitor Cell Renewal and Differentiation. J Am Soc Nephrol 2019; 30:63-78. [PMID: 30518531 PMCID: PMC6317605 DOI: 10.1681/asn.2018070736] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 10/22/2018] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Nephron number is a major determinant of long-term renal function and cardiovascular risk. Observational studies suggest that maternal nutritional and metabolic factors during gestation contribute to the high variability of nephron endowment. However, the underlying molecular mechanisms have been unclear. METHODS We used mouse models, including DNA methyltransferase (Dnmt1, Dnmt3a, and Dnmt3b) knockout mice, optical projection tomography, three-dimensional reconstructions of the nephrogenic niche, and transcriptome and DNA methylation analysis to characterize the role of DNA methylation for kidney development. RESULTS We demonstrate that DNA hypomethylation is a key feature of nutritional kidney growth restriction in vitro and in vivo, and that DNA methyltransferases Dnmt1 and Dnmt3a are highly enriched in the nephrogenic zone of the developing kidneys. Deletion of Dnmt1 in nephron progenitor cells (in contrast to deletion of Dnmt3a or Dnm3b) mimics nutritional models of kidney growth restriction and results in a substantial reduction of nephron number as well as renal hypoplasia at birth. In Dnmt1-deficient mice, optical projection tomography and three-dimensional reconstructions uncovered a significant reduction of stem cell niches and progenitor cells. RNA sequencing analysis revealed that global DNA hypomethylation interferes in the progenitor cell regulatory network, leading to downregulation of genes crucial for initiation of nephrogenesis, Wt1 and its target Wnt4. Derepression of germline genes, protocadherins, Rhox genes, and endogenous retroviral elements resulted in the upregulation of IFN targets and inhibitors of cell cycle progression. CONCLUSIONS These findings establish DNA methylation as a key regulatory event of prenatal renal programming, which possibly represents a fundamental link between maternal nutritional factors during gestation and reduced nephron number.
Collapse
Affiliation(s)
| | - Julia Vornweg
- Faculty of Medicine, Department of Medicine IV, Medical Center-University of Freiburg, and
- Faculty of Biology
| | - Alexander Combes
- Anatomy and Neuroscience
- Cell Biology Theme, Murdoch Children's Research Institute, Melbourne, Australia
| | | | - Julia Plappert
- Faculty of Medicine, Department of Medicine IV, Medical Center-University of Freiburg, and
| | - Gesa Rafflenbeul
- Faculty of Medicine, Department of Medicine IV, Medical Center-University of Freiburg, and
| | | | - Raza-Ur Rahman
- Institute of Medical Systems Biology, Center for Molecular Neurobiology, and
| | - Timur Liwinski
- Institute of Medical Systems Biology, Center for Molecular Neurobiology, and
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Saskia Lindner
- Faculty of Medicine, Department of Medicine IV, Medical Center-University of Freiburg, and
| | | | - Oliver Kretz
- III. Department of Medicine
- Department of Neuroanatomy, University of Freiburg, Freiburg, Germany
| | | | - Tania Romano
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Karen M Moritz
- Child Health Research Centre and School of Biomedical Sciences, University of Queensland, St. Lucia, Queensland, Australia
| | - Melanie Boerries
- German Cancer Consortium, Heidelberg, Germany
- German Cancer Research Center, Heidelberg, Germany
- Institute of Molecular Medicine and Cell Research
| | - Hauke Busch
- Institute of Molecular Medicine and Cell Research
- Lübeck Institute of Experimental Dermatology, Lübeck, Germany; and
| | - Stefan Bonn
- Institute of Molecular Medicine and Cell Research
- Laboratory of Computational Systems Biology, German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Melissa H Little
- Cell Biology Theme, Murdoch Children's Research Institute, Melbourne, Australia
- Pediatrics, University of Melbourne, Melbourne, Australia
| | - Wibke Bechtel-Walz
- Faculty of Medicine, Department of Medicine IV, Medical Center-University of Freiburg, and
| | - Tobias B Huber
- III. Department of Medicine,
- Faculty of Medicine, Department of Medicine IV, Medical Center-University of Freiburg, and
- Centre for Biological Signalling Studies (BIOSS) and Center for Biological Systems Analysis (ZBSA), and
- Freiburg Institute for Advanced Studies, Albert Ludwig University of Freiburg, Freiburg, Germany; Departments of
| |
Collapse
|
108
|
Abstract
Kidney organogenesis has been a widely used classical model system to study inductive tissue interactions that guide differentiation of many organs. The basis for this is in the pioneering work done during the early 1950s when the conditions of how to support ex vivo growth and differentiation of developing kidneys were revealed. Importantly, culturing developing kidneys remains as an essential instrument to advance our understanding of molecular and cellular regulation of morphogenesis even today. Despite the fact that embryonic kidneys have been cultured for decades, it is not a trivial method and requires specific anatomical and developmental biology knowledge. This chapter outlines the general steps in organ culture and details the requirements for successful kidney explant differentiation.
Collapse
|
109
|
Lang C, Conrad L, Michos O. Mathematical Approaches of Branching Morphogenesis. Front Genet 2018; 9:673. [PMID: 30631344 PMCID: PMC6315180 DOI: 10.3389/fgene.2018.00673] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 12/04/2018] [Indexed: 12/16/2022] Open
Abstract
Many organs require a high surface to volume ratio to properly function. Lungs and kidneys, for example, achieve this by creating highly branched tubular structures during a developmental process called branching morphogenesis. The genes that control lung and kidney branching share a similar network structure that is based on ligand-receptor reciprocal signalling interactions between the epithelium and the surrounding mesenchyme. Nevertheless, the temporal and spatial development of the branched epithelial trees differs, resulting in organs of distinct shape and size. In the embryonic lung, branching morphogenesis highly depends on FGF10 signalling, whereas GDNF is the driving morphogen in the kidney. Knockout of Fgf10 and Gdnf leads to lung and kidney agenesis, respectively. However, FGF10 plays a significant role during kidney branching and both the FGF10 and GDNF pathway converge on the transcription factors ETV4/5. Although the involved signalling proteins have been defined, the underlying mechanism that controls lung and kidney branching morphogenesis is still elusive. A wide range of modelling approaches exists that differ not only in the mathematical framework (e.g., stochastic or deterministic) but also in the spatial scale (e.g., cell or tissue level). Due to advancing imaging techniques, image-based modelling approaches have proven to be a valuable method for investigating the control of branching events with respect to organ-specific properties. Here, we review several mathematical models on lung and kidney branching morphogenesis and suggest that a ligand-receptor-based Turing model represents a potential candidate for a general but also adaptive mechanism to control branching morphogenesis during development.
Collapse
Affiliation(s)
| | | | - Odyssé Michos
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| |
Collapse
|
110
|
Tham MS, Smyth IM. Cellular and molecular determinants of normal and abnormal kidney development. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2018; 8:e338. [DOI: 10.1002/wdev.338] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 11/07/2018] [Accepted: 11/14/2018] [Indexed: 01/21/2023]
Affiliation(s)
- Ming S. Tham
- Department of Anatomy and Developmental Biology Monash Biomedicine Discovery Institute, Monash University Melbourne Victoria Australia
| | - Ian M. Smyth
- Department of Anatomy and Developmental Biology Monash Biomedicine Discovery Institute, Monash University Melbourne Victoria Australia
- Department of Biochemistry and Molecular Biology Monash Biomedicine Discovery Institute, Monash University Melbourne Victoria Australia
| |
Collapse
|
111
|
O'Brien LL, Combes AN, Short KM, Lindström NO, Whitney PH, Cullen-McEwen LA, Ju A, Abdelhalim A, Michos O, Bertram JF, Smyth IM, Little MH, McMahon AP. Wnt11 directs nephron progenitor polarity and motile behavior ultimately determining nephron endowment. eLife 2018; 7:e40392. [PMID: 30516471 PMCID: PMC6281319 DOI: 10.7554/elife.40392] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 11/16/2018] [Indexed: 01/09/2023] Open
Abstract
A normal endowment of nephrons in the mammalian kidney requires a balance of nephron progenitor self-renewal and differentiation throughout development. Here, we provide evidence for a novel action of ureteric branch tip-derived Wnt11 in progenitor cell organization and interactions within the nephrogenic niche, ultimately determining nephron endowment. In Wnt11 mutants, nephron progenitors dispersed from their restricted niche, intermixing with interstitial progenitors. Nephron progenitor differentiation was accelerated, kidneys were significantly smaller, and the nephron progenitor pool was prematurely exhausted, halving the final nephron count. Interestingly, RNA-seq revealed no significant differences in gene expression. Live imaging of nephron progenitors showed that in the absence of Wnt11 they lose stable attachments to the ureteric branch tips, continuously detaching and reattaching. Further, the polarized distribution of several markers within nephron progenitors is disrupted. Together these data highlight the importance of Wnt11 signaling in directing nephron progenitor behavior which determines a normal nephrogenic program.
Collapse
Affiliation(s)
- Lori L O'Brien
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell ResearchKeck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Alexander N Combes
- Institute for Molecular BioscienceThe University of QueenslandBrisbaneAustralia
- Department of Anatomy and NeuroscienceThe University of MelbourneMelbourneAustralia
- Murdoch Children’s Research InstituteRoyal Children's HospitalMelbourneAustralia
| | - Kieran M Short
- Department of Anatomy and Neuroscience, Monash Biomedicine Discovery InstituteMonash UniversityMelbourneAustralia
- Development and Stem Cells Program, Monash Biomedicine Discovery InstituteMonash UniversityMelbourneAustralia
| | - Nils O Lindström
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell ResearchKeck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Peter H Whitney
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell ResearchKeck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Luise A Cullen-McEwen
- Department of Anatomy and Neuroscience, Monash Biomedicine Discovery InstituteMonash UniversityMelbourneAustralia
| | - Adler Ju
- Institute for Molecular BioscienceThe University of QueenslandBrisbaneAustralia
| | - Ahmed Abdelhalim
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell ResearchKeck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Odyssé Michos
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell ResearchKeck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - John F Bertram
- Department of Anatomy and Neuroscience, Monash Biomedicine Discovery InstituteMonash UniversityMelbourneAustralia
| | - Ian M Smyth
- Department of Anatomy and Neuroscience, Monash Biomedicine Discovery InstituteMonash UniversityMelbourneAustralia
- Development and Stem Cells Program, Monash Biomedicine Discovery InstituteMonash UniversityMelbourneAustralia
| | - Melissa H Little
- Institute for Molecular BioscienceThe University of QueenslandBrisbaneAustralia
- Department of Anatomy and NeuroscienceThe University of MelbourneMelbourneAustralia
- Murdoch Children’s Research InstituteRoyal Children's HospitalMelbourneAustralia
- Department of PediatricsUniversity of MelbourneParkvilleAustralia
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell ResearchKeck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| |
Collapse
|
112
|
Changes in cell fate determine the regenerative and functional capacity of the developing kidney before and after release of obstruction. Clin Sci (Lond) 2018; 132:2519-2545. [PMID: 30442812 DOI: 10.1042/cs20180623] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 10/23/2018] [Accepted: 11/14/2018] [Indexed: 12/14/2022]
Abstract
Congenital obstructive nephropathy is a major cause of chronic kidney disease (CKD) in children. The contribution of changes in the identity of renal cells to the pathology of obstructive nephropathy is poorly understood. Using a partial unilateral ureteral obstruction (pUUO) model in genetically modified neonatal mice, we traced the fate of cells derived from the renal stroma, cap mesenchyme, ureteric bud (UB) epithelium, and podocytes using Foxd1Cre, Six2Cre, HoxB7Cre, and Podocyte.Cre mice respectively, crossed with double fluorescent reporter (membrane-targetted tandem dimer Tomato (mT)/membrane-targetted GFP (mG)) mice. Persistent obstruction leads to a significant loss of tubular epithelium, rarefaction of the renal vasculature, and decreased renal blood flow (RBF). In addition, Forkhead Box D1 (Foxd1)-derived pericytes significantly expanded in the interstitial space, acquiring a myofibroblast phenotype. Degeneration of Sine Oculis Homeobox Homolog 2 (Six2) and HoxB7-derived cells resulted in significant loss of glomeruli, nephron tubules, and collecting ducts. Surgical release of obstruction resulted in striking regeneration of tubules, arterioles, interstitium accompanied by an increase in blood flow to the level of sham animals. Contralateral kidneys with remarkable compensatory response to kidney injury showed an increase in density of arteriolar branches. Deciphering the mechanisms involved in kidney repair and regeneration post relief of obstruction has potential therapeutic implications for infants and children and the growing number of adults suffering from CKD.
Collapse
|
113
|
Sánchez N, Inostroza V, Pérez MC, Moya P, Ubilla A, Besa J, Llaguno E, Vera P-G C, Inzunza O, Gaete M. Tracking morphological complexities of organ development in culture. Mech Dev 2018; 154:179-192. [DOI: 10.1016/j.mod.2018.07.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 07/03/2018] [Accepted: 07/13/2018] [Indexed: 12/14/2022]
|
114
|
Wu H, Uchimura K, Donnelly EL, Kirita Y, Morris SA, Humphreys BD. Comparative Analysis and Refinement of Human PSC-Derived Kidney Organoid Differentiation with Single-Cell Transcriptomics. Cell Stem Cell 2018; 23:869-881.e8. [PMID: 30449713 DOI: 10.1016/j.stem.2018.10.010] [Citation(s) in RCA: 396] [Impact Index Per Article: 56.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 08/09/2018] [Accepted: 10/06/2018] [Indexed: 12/18/2022]
Abstract
Kidney organoids derived from human pluripotent stem cells have great utility for investigating organogenesis and disease mechanisms and, potentially, as a replacement tissue source, but how closely organoids derived from current protocols replicate adult human kidney is undefined. We compared two directed differentiation protocols by single-cell transcriptomics of 83,130 cells from 65 organoids with single-cell transcriptomes of fetal and adult kidney cells. Both protocols generate a diverse range of kidney cells with differing ratios, but organoid-derived cell types are immature, and 10%-20% of cells are non-renal. Reconstructing lineage relationships by pseudotemporal ordering identified ligands, receptors, and transcription factor networks associated with fate decisions. Brain-derived neurotrophic factor (BDNF) and its cognate receptor NTRK2 were expressed in the neuronal lineage during organoid differentiation. Inhibiting this pathway improved organoid formation by reducing neurons by 90% without affecting kidney differentiation, highlighting the power of single-cell technologies to characterize and improve organoid differentiation.
Collapse
Affiliation(s)
- Haojia Wu
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Kohei Uchimura
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Erinn L Donnelly
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Yuhei Kirita
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Samantha A Morris
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA; Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Benjamin D Humphreys
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA; Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
115
|
Kurtzeborn K, Cebrian C, Kuure S. Regulation of Renal Differentiation by Trophic Factors. Front Physiol 2018; 9:1588. [PMID: 30483151 PMCID: PMC6240607 DOI: 10.3389/fphys.2018.01588] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 10/23/2018] [Indexed: 12/11/2022] Open
Abstract
Classically, trophic factors are considered as proteins which support neurons in their growth, survival, and differentiation. However, most neurotrophic factors also have important functions outside of the nervous system. Especially essential renal growth and differentiation regulators are glial cell line-derived neurotrophic factor (GDNF), bone morphogenetic proteins (BMPs), and fibroblast growth factors (FGFs). Here we discuss how trophic factor-induced signaling contributes to the control of ureteric bud (UB) branching morphogenesis and to maintenance and differentiation of nephrogenic mesenchyme in embryonic kidney. The review includes recent advances in trophic factor functions during the guidance of branching morphogenesis and self-renewal versus differentiation decisions, both of which dictate the control of kidney size and nephron number. Creative utilization of current information may help better recapitulate renal differentiation in vitro, but it is obvious that significantly more basic knowledge is needed for development of regeneration-based renal therapies.
Collapse
Affiliation(s)
- Kristen Kurtzeborn
- Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Medicum, University of Helsinki, Helsinki, Finland
| | - Cristina Cebrian
- Developmental Biology Division, Cincinnati Children’s Hospital, Cincinnati, OH, United States
| | - Satu Kuure
- Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Medicum, University of Helsinki, Helsinki, Finland
- GM-Unit, Laboratory Animal Centre, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| |
Collapse
|
116
|
Rabadi MM, Abdulmahdi W, Nesi L, Jules E, Marghani Y, Sheinin E, Tilzer J, Gupta S, Chen S, Cassimatis ND, Lipphardt M, Kozlowski PB, Ratliff BB. Maternal malnourishment induced upregulation of fetuin-B blunts nephrogenesis in the low birth weight neonate. Dev Biol 2018; 443:78-91. [DOI: 10.1016/j.ydbio.2018.09.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 08/21/2018] [Accepted: 09/01/2018] [Indexed: 11/16/2022]
|
117
|
Ihermann-Hella A, Hirashima T, Kupari J, Kurtzeborn K, Li H, Kwon HN, Cebrian C, Soofi A, Dapkunas A, Miinalainen I, Dressler GR, Matsuda M, Kuure S. Dynamic MAPK/ERK Activity Sustains Nephron Progenitors through Niche Regulation and Primes Precursors for Differentiation. Stem Cell Reports 2018; 11:912-928. [PMID: 30220628 PMCID: PMC6178244 DOI: 10.1016/j.stemcr.2018.08.012] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 08/20/2018] [Accepted: 08/20/2018] [Indexed: 12/24/2022] Open
Abstract
The in vivo niche and basic cellular properties of nephron progenitors are poorly described. Here we studied the cellular organization and function of the MAPK/ERK pathway in nephron progenitors. Live-imaging of ERK activity by a Förster resonance energy transfer biosensor revealed a dynamic activation pattern in progenitors, whereas differentiating precursors exhibited sustained activity. Genetic experiments demonstrate that MAPK/ERK activity controls the thickness, coherence, and integrity of the nephron progenitor niche. Molecularly, MAPK/ERK activity regulates niche organization and communication with extracellular matrix through PAX2 and ITGA8, and is needed for CITED1 expression denoting undifferentiated status. MAPK/ERK activation in nephron precursors propels differentiation by priming cells for distal and proximal fates induced by the Wnt and Notch pathways. Thus, our results demonstrate a mechanism through which MAPK/ERK activity controls both progenitor maintenance and differentiation by regulating a distinct set of targets, which maintain the biomechanical milieu of tissue-residing progenitors and prime precursors for nephrogenesis.
Collapse
Affiliation(s)
| | - Tsuyoshi Hirashima
- Department of Pathology and Biology of Diseases, Graduate School of Medicine & Laboratory of Bioimaging and Cell Signaling, Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| | - Jussi Kupari
- HiLIFE and Medicum, University of Helsinki, Helsinki FIN-00014, Finland
| | | | - Hao Li
- HiLIFE and Medicum, University of Helsinki, Helsinki FIN-00014, Finland
| | - Hyuk Nam Kwon
- HiLIFE and Medicum, University of Helsinki, Helsinki FIN-00014, Finland
| | - Cristina Cebrian
- Developmental Biology Division, Cincinnati Children's Hospital, Cincinnati, OH 45229, USA
| | - Abdul Soofi
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Arvydas Dapkunas
- Medicum and Meilahti Clinical Proteomics Core Facility/HiLIFE, University of Helsinki, Helsinki FIN-00014, Finland
| | - Ilkka Miinalainen
- Department of Pathology (Biocenter Oulu), University of Oulu, Oulu 90220, Finland
| | - Gregory R Dressler
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Michiyuki Matsuda
- Department of Pathology and Biology of Diseases, Graduate School of Medicine & Laboratory of Bioimaging and Cell Signaling, Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| | - Satu Kuure
- HiLIFE and Medicum, University of Helsinki, Helsinki FIN-00014, Finland; GM-Unit, LAC/ HiLIFE, and Medicum, University of Helsinki, Helsinki FIN-00014, Finland.
| |
Collapse
|
118
|
Ishiyama H, Ishikawa A, Kitazawa H, Fujii S, Matsubayashi J, Yamada S, Takakuwa T. Branching morphogenesis of the urinary collecting system in the human embryonic metanephros. PLoS One 2018; 13:e0203623. [PMID: 30192900 PMCID: PMC6128595 DOI: 10.1371/journal.pone.0203623] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 08/23/2018] [Indexed: 11/23/2022] Open
Abstract
An elaborate system of ducts collects urine from all nephrons, and this structure is known as the urinary collecting system (UCS). This study focused on how the UCS is formed during human embryogenesis. Fifty human embryos between the Carnegie stage (CS) 14 and CS23 were selected from the Kyoto Collection at the Congenital Anomaly Research Center of Kyoto University, Japan. Metanephroses, including the UCS, were segmented on serial digital virtual histological sections. Three-dimensional images were computationally reconstructed for morphological and quantitative analyses. A CS timeline was plotted. It consisted of the 3-D structural morphogenesis of UCS and quantification of the total amount of end-branching, average and maximum numbers of generations, deviation in the metanephros, differentiation of the urothelial epithelium in the renal pelvis, and timing of the rapid expansion of the renal pelvis. The first UCS branching generation occurred by CS16. The average branching generation reached a maximum of 8.74 ± 1.60 and was already the twelfth in CS23. The total end-branching number squared between the start and the end of the embryonic period. UCS would reach the fifteenth branching generation soon after CS23. The number of nephrons per UCS end-branch was low (0.21 ± 0.14 at CS19, 1.34 ± 0.49 at CS23), indicating that the bifid branching occurred rapidly and that the formation of nephrons followed after. The renal pelvis expanded mainly in CS23, which was earlier than that reported in a previous study. The number of nephrons connected to the UCS in the expanded group (246.0 ± 13.2) was significantly larger than that of the pre-expanded group (130.8 ± 80.1) (P < 0.05). The urothelial epithelium differentiated from the zeroth to the third generations at CS23. Differentiation may have continued up until the tenth generation to allow for renal pelvis expansion. The branching speed was not uniform. There were significantly more branching generations in the polar- than in the interpolar regions (P < 0.05). Branching speed reflects the growth orientation required to form the metanephros. Further study will be necessary to understand the renal pelvis expansion mechanism in CS23. Our CS-based timeline enabled us to map UCS formation and predict functional renal capacity after differentiation and growth.
Collapse
Affiliation(s)
- Hana Ishiyama
- Human Health Science, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Aoi Ishikawa
- Human Health Science, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Haruka Kitazawa
- Human Health Science, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Sena Fujii
- Human Health Science, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Jun Matsubayashi
- Human Health Science, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shigehito Yamada
- Human Health Science, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Congenital Anomaly Research Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tetsuya Takakuwa
- Human Health Science, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- * E-mail:
| |
Collapse
|
119
|
Abstract
The nephron is a multifunctional filtration device equipped with an array of sophisticated sensors. For appropriate physiological function in the human and mouse, nephrons must be stereotypically arrayed in large numbers, and this essential structural property that defines the kidney is determined during its fetal development. This review explores the process of nephron determination in the fetal kidney, providing an overview of the foundational literature in the field as well as exploring new developments in this dynamic research area. Mechanisms that ensure that a large number of nephrons can be formed from a small initial number of progenitor cells are central to this process, and the question of how the nephron progenitor cell population balances epithelial differentiation with renewal in the progenitor state is a subject of particular interest. Key growth factor signaling pathways and transcription factor networks are discussed.
Collapse
Affiliation(s)
- Leif Oxburgh
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine 04074, USA;
| |
Collapse
|
120
|
Short KM, Combes AN, Lisnyak V, Lefevre JG, Jones LK, Little MH, Hamilton NA, Smyth IM. Branching morphogenesis in the developing kidney is not impacted by nephron formation or integration. eLife 2018; 7:38992. [PMID: 30063208 PMCID: PMC6115188 DOI: 10.7554/elife.38992] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 07/31/2018] [Indexed: 02/07/2023] Open
Abstract
Branching morphogenesis of the ureteric bud is integral to kidney development; establishing the collecting ducts of the adult organ and driving organ expansion via peripheral interactions with nephron progenitor cells. A recent study suggested that termination of tip branching within the developing kidney involved stochastic exhaustion in response to nephron formation, with such a termination event representing a unifying developmental process evident in many organs. To examine this possibility, we have profiled the impact of nephron formation and maturation on elaboration of the ureteric bud during mouse kidney development. We find a distinct absence of random branch termination events within the kidney or evidence that nephrogenesis impacts the branching program or cell proliferation in either tip or progenitor cell niches. Instead, organogenesis proceeds in a manner indifferent to the development of these structures. Hence, stochastic cessation of branching is not a unifying developmental feature in all branching organs. During development and before birth, many organs develop from branched tubes. Whether forming the airways of the lungs, the collecting ducts of the kidneys or the milk ducts of the breast, there are many similarities between these structures. Given their shared tree-like structures, one possibility is that these tissues all form through the same general process. A key challenge is understanding why branched networks develop and pattern in such a way as to assume their functional roles in the adult organ. A unifying theory, which proposes that certain tips stop growing in a random manner, has been proposed to solve this problem. In this theory, the branched mammary gland structures stop growing when the tips of the structure impinge on neighbouring branches. In the kidney, this cessation has been proposed to occur when nephrons – the structures that filter urine from blood – form near the end of the collecting ducts. By growing kidneys in the laboratory and studying developing kidneys in mice, Short et al. investigated whether nephrons do affect collecting duct growth and branch development. The results of these experiments instead suggest that nephron formation has no effect on duct growth or branching. The nephrons also do not appear to affect how quickly the duct cells grow and divide. Moreover, there is no evidence that the cell proliferation in individual branch tips ceases randomly by any other mechanism. Overall, the experiments Short et al. performed suggest that a unifying theory of branching in developing organs may not hold true, at least not in the way that has been envisioned previously.
Collapse
Affiliation(s)
- Kieran M Short
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Alexander N Combes
- Murdoch Children's Research Institute, Parkville, Australia.,Department of Anatomy and Neuroscience, School of Biomedical Sciences, University of Melbourne, Parkville, Australia
| | - Valerie Lisnyak
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - James G Lefevre
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Lynelle K Jones
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Melissa H Little
- Murdoch Children's Research Institute, Parkville, Australia.,Department of Anatomy and Neuroscience, School of Biomedical Sciences, University of Melbourne, Parkville, Australia.,Department of Pediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Australia
| | - Nicholas A Hamilton
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Ian M Smyth
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia.,Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| |
Collapse
|
121
|
O'Brien LL. Nephron progenitor cell commitment: Striking the right balance. Semin Cell Dev Biol 2018; 91:94-103. [PMID: 30030141 DOI: 10.1016/j.semcdb.2018.07.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 06/29/2018] [Accepted: 07/16/2018] [Indexed: 10/28/2022]
Abstract
The filtering component of the kidney, the nephron, arises from a single progenitor population. These nephron progenitor cells (NPCs) both self-renew and differentiate throughout the course of kidney development ensuring sufficient nephron endowment. An appropriate balance of these processes must be struck as deficiencies in nephron numbers are associated with hypertension and kidney disease. This review will discuss the mechanisms and molecules supporting NPC maintenance and differentiation. A focus on recent work will highlight new molecular insights into NPC regulation and their dynamic behavior in both space and time.
Collapse
Affiliation(s)
- Lori L O'Brien
- Department of Cell Biology and Physiology, UNC Kidney Center, University of North Carolina at Chapel Hill, 111 Mason Farm Road, Chapel Hill, NC, 27599, United States.
| |
Collapse
|
122
|
Deconstructing the principles of ductal network formation in the pancreas. PLoS Biol 2018; 16:e2002842. [PMID: 30048442 PMCID: PMC6080801 DOI: 10.1371/journal.pbio.2002842] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 08/07/2018] [Accepted: 07/16/2018] [Indexed: 12/18/2022] Open
Abstract
The mammalian pancreas is a branched organ that does not exhibit stereotypic branching patterns, similarly to most other glands. Inside branches, it contains a network of ducts that undergo a transition from unconnected microlumen to a mesh of interconnected ducts and finally to a treelike structure. This ductal remodeling is poorly understood, both on a microscopic and macroscopic level. In this article, we quantify the network properties at different developmental stages. We find that the pancreatic network exhibits stereotypic traits at each stage and that the network properties change with time toward the most economical and optimized delivery of exocrine products into the duodenum. Using in silico modeling, we show how steps of pancreatic network development can be deconstructed into two simple rules likely to be conserved for many other glands. The early stage of the network is explained by noisy, redundant duct connection as new microlumens form. The later transition is attributed to pruning of the network based on the flux of fluid running through the pancreatic network into the duodenum. In the pancreas of mammals, digestive enzymes are transported from their production site in acini (clusters of cells that secrete the enzymes) to the intestine via a network of ducts. During organ development in fetuses, the ducts initially form by the coordinated polarization of cells to form small holes, which will connect and fuse, to constitute a meshwork. This hyperconnected network further develops into a treelike structure by the time of birth. In this article, we use methods originally developed to analyze road, rail, web, or river networks to quantify the network properties at different developmental stages. We find that the pancreatic network properties are similar between individuals at specific time points but eventually change to achieve the most economical and optimized structure to deliver pancreatic juice into the duodenum. Using in silico modeling, we show how the stages of pancreatic network development follow two simple rules, which are likely to be conserved for the development of other glands. The early stage of the network is explained by noisy, redundant duct connection as new small ductal holes form. Later on, the secretion of fluid that runs through the pancreatic network into the duodenum leads to the widening of ducts with the greatest flow, while nonnecessary ducts are eliminated, akin to how river beds are formed.
Collapse
|
123
|
Zhang L, Ettou S, Khalid M, Taglienti M, Jain D, Jung YL, Seager C, Liu Y, Ng KH, Park PJ, Kreidberg JA. EED, a member of the polycomb group, is required for nephron differentiation and the maintenance of nephron progenitor cells. Development 2018; 145:dev.157149. [PMID: 29945864 DOI: 10.1242/dev.157149] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 06/14/2018] [Indexed: 12/26/2022]
Abstract
Epigenetic regulation of gene expression has a crucial role allowing for the self-renewal and differentiation of stem and progenitor populations during organogenesis. The mammalian kidney maintains a population of self-renewing stem cells that differentiate to give rise to thousands of nephrons, which are the functional units that carry out filtration to maintain physiological homeostasis. The polycomb repressive complex 2 (PRC2) epigenetically represses gene expression during development by placing the H3K27me3 mark on histone H3 at promoter and enhancer sites, resulting in gene silencing. To understand the role of PRC2 in nephron differentiation, we conditionally inactivated the Eed gene, which encodes a nonredundant component of the PRC2 complex, in nephron progenitor cells. Resultant kidneys were smaller and showed premature loss of progenitor cells. The progenitors in Eed mutant mice that were induced to differentiate did not develop into properly formed nephrons. Lhx1, normally expressed in the renal vesicle, was overexpressed in kidneys of Eed mutant mice. Thus, PRC2 has a crucial role in suppressing the expression of genes that maintain the progenitor state, allowing nephron differentiation to proceed.
Collapse
Affiliation(s)
- Le Zhang
- Department of Urology, Boston Children's Hospital, Boston, MA 02115, USA.,Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Sandrine Ettou
- Department of Urology, Boston Children's Hospital, Boston, MA 02115, USA.,Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Myda Khalid
- Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Mary Taglienti
- Department of Urology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Dhawal Jain
- Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Youngsook L Jung
- Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Catherine Seager
- Department of Urology, Boston Children's Hospital, Boston, MA 02115, USA.,Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Yongqing Liu
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Kar-Hui Ng
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Peter J Park
- Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Jordan A Kreidberg
- Department of Urology, Boston Children's Hospital, Boston, MA 02115, USA .,Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Department of Surgery, Harvard Medical School, Boston, MA 02115, USA.,Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.,Harvard Stem Cell Institute, Cambridge, MA 02139, USA
| |
Collapse
|
124
|
Shaw I, Rider S, Mullins J, Hughes J, Péault B. Pericytes in the renal vasculature: roles in health and disease. Nat Rev Nephrol 2018; 14:521-534. [DOI: 10.1038/s41581-018-0032-4] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
125
|
Lambert B, MacLean AL, Fletcher AG, Combes AN, Little MH, Byrne HM. Bayesian inference of agent-based models: a tool for studying kidney branching morphogenesis. J Math Biol 2018; 76:1673-1697. [PMID: 29392399 PMCID: PMC5906521 DOI: 10.1007/s00285-018-1208-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 01/02/2018] [Indexed: 12/11/2022]
Abstract
The adult mammalian kidney has a complex, highly-branched collecting duct epithelium that arises as a ureteric bud sidebranch from an epithelial tube known as the nephric duct. Subsequent branching of the ureteric bud to form the collecting duct tree is regulated by subcellular interactions between the epithelium and a population of mesenchymal cells that surround the tips of outgrowing branches. The mesenchymal cells produce glial cell-line derived neurotrophic factor (GDNF), that binds with RET receptors on the surface of the epithelial cells to stimulate several subcellular pathways in the epithelium. Such interactions are known to be a prerequisite for normal branching development, although competing theories exist for their role in morphogenesis. Here we introduce the first agent-based model of ex vivo kidney uretic branching. Through comparison with experimental data, we show that growth factor-regulated growth mechanisms can explain early epithelial cell branching, but only if epithelial cell division depends in a switch-like way on the local growth factor concentration; cell division occurring only if the driving growth factor level exceeds a threshold. We also show how a recently-developed method, "Approximate Approximate Bayesian Computation", can be used to infer key model parameters, and reveal the dependency between the parameters controlling a growth factor-dependent growth switch. These results are consistent with a requirement for signals controlling proliferation and chemotaxis, both of which are previously identified roles for GDNF.
Collapse
Affiliation(s)
- Ben Lambert
- Department of Zoology, University of Oxford, Oxford, UK.
| | - Adam L MacLean
- Mathematical Institute, University of Oxford, Andrew Wiles Building, Woodstock Road, Oxford, UK
- Department of Mathematics, University of California, Irvine, Irvine, CA, USA
| | - Alexander G Fletcher
- School of Mathematics and Statistics, University of Sheffield, Hicks Building, Hounsfield Road, Sheffield, S3 7RH, UK
- Bateson Centre, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK
| | - Alexander N Combes
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, VIC, 3010, Australia
- Murdoch Childrens Research Institute, Flemington Rd, Parkville, Melbourne, VIC, 3052, Australia
| | - Melissa H Little
- Murdoch Childrens Research Institute, Flemington Rd, Parkville, Melbourne, VIC, 3052, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Helen M Byrne
- Mathematical Institute, University of Oxford, Andrew Wiles Building, Woodstock Road, Oxford, UK
| |
Collapse
|
126
|
Hamartin regulates cessation of mouse nephrogenesis independently of Mtor. Proc Natl Acad Sci U S A 2018; 115:5998-6003. [PMID: 29784808 DOI: 10.1073/pnas.1712955115] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
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.
Collapse
|
127
|
Vue Z, Gonzalez G, Stewart CA, Mehra S, Behringer RR. Volumetric imaging of the developing prepubertal mouse uterine epithelium using light sheet microscopy. Mol Reprod Dev 2018. [PMID: 29543367 DOI: 10.1002/mrd.22973] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Endometrial or uterine glands secrete substances essential for uterine receptivity to the embryo, implantation, conceptus survival, and growth. Adenogenesis is the process of gland formation within the stroma of the uterus. In the mouse, uterine gland formation initiates at postnatal day (P) 5. Uterine gland morphology is poorly understood because it is primarily based on two-dimensional (2D) histology. To more fully describe uterine gland morphogenesis, we generated three-dimensional (3D) models of postnatal uterine glands from P0 to P21, based on volumetric imaging using light sheet microscopy. At birth (P0), there were no glands. At P8, we found bud- and teardrop-shaped epithelial invaginations. By P11, the forming glands were elongated epithelial tubes. By P21, the elongated tubes had a sinuous morphology. These morphologies are homogeneously distributed along the anterior-posterior axis of the uterus. To facilitate uterine gland analyses, we propose a novel 3D staging system of uterine gland morphology during development in the prepubertal mouse. We define five uterine gland stages: Stage 1: bud; Stage 2: teardrop; Stage 3: elongated; Stage 4: sinuous; and Stage 5: primary branches. This staging system provides a standardized key to assess and quantify prepubertal uterine gland morphology that can be used for studies of uterine gland development and pathology. In addition, our studies suggest that gland formation initiation occurs during P8 and P11. However, between P11 and P21 gland formation initiation stops and all glands elongate and become sinuous. We also found that the mesometrial epithelium develops a unique morphology we term the uterine rail.
Collapse
Affiliation(s)
- Zer Vue
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas.,Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gabriel Gonzalez
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - C Allison Stewart
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shyamin Mehra
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Richard R Behringer
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas.,Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
128
|
Zhang LY, Lin P, Pan J, Ma Y, Wei Z, Jiang L, Wang L, Song Y, Wang Y, Zhang Z, Jin K, Wang Q, Yang GY. CLARITY for High-resolution Imaging and Quantification of Vasculature in the Whole Mouse Brain. Aging Dis 2018; 9:262-272. [PMID: 29896415 PMCID: PMC5963347 DOI: 10.14336/ad.2017.0613] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 06/13/2017] [Indexed: 12/11/2022] Open
Abstract
Elucidating the normal structure and distribution of cerebral vascular system is fundamental for understanding its function. However, studies on visualization and whole-brain quantification of vasculature with cellular resolution are limited. Here, we explored the structure of vasculature at the whole-brain level using the newly developed CLARITY technique. Adult male C57BL/6J mice undergoing transient middle cerebral artery occlusion and Tie2-RFP transgenic mice were used. Whole mouse brains were extracted for CLARITY processing. Immunostaining was performed to label vessels. Customized MATLAB code was used for image processing and quantification. Three-dimensional images were visualized using the Vaa3D software. Our results showed that whole mouse brain became transparent using the CLARITY method. Three-dimensional imaging and visualization of vasculature were achieved at the whole-brain level with a 1-μm voxel resolution. The quantitative results showed that the fractional vascular volume was 0.018 ± 0.004 mm3 per mm3, the normalized vascular length was 0.44 ± 0.04 m per mm3, and the mean diameter of the microvessels was 4.25 ± 0.08 μm. Furthermore, a decrease in the fractional vascular volume and a decrease in the normalized vascular length were found in the penumbra of ischemic mice compared to controls (p < 0.05). In conclusion, CLARITY provides a novel approach for mapping vasculature in the whole mouse brain at cellular resolution. CLARITY-optimized algorithms facilitate the assessment of structural change in vasculature after brain injury.
Collapse
Affiliation(s)
- Lin-Yuan Zhang
- 1Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pan Lin
- 2Medical Image Computing Lab and
| | - Jiaji Pan
- 3Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yuanyuan Ma
- 1Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenyu Wei
- 4Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201999, China
| | - Lu Jiang
- 3Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Liping Wang
- 1Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yaying Song
- 1Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongting Wang
- 3Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Zhijun Zhang
- 3Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Kunlin Jin
- 5Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, TX76107, USA
| | | | - Guo-Yuan Yang
- 1Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,3Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| |
Collapse
|
129
|
Magella B, Mahoney R, Adam M, Potter SS. Reduced Abd-B Hox function during kidney development results in lineage infidelity. Dev Biol 2018; 438:84-93. [PMID: 29596840 DOI: 10.1016/j.ydbio.2018.03.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 02/28/2018] [Accepted: 03/21/2018] [Indexed: 02/05/2023]
Abstract
Hox genes can function as key drivers of segment identity, with Hox mutations in Drosophila often resulting in dramatic homeotic transformations. In addition, however, they can serve other essential functions. In mammals, the study of Hox gene roles in development is complicated by the presence of four Hox clusters with a total of 39 genes showing extensive functional overlap. In this study, in order to better understand shared core Hox functions, we examined kidney development in mice with frameshift mutations of multiple Abd-B type Hox genes. The resulting phenotypes included dramatically reduced branching morphogenesis of the ureteric bud, premature depletion of nephron progenitors and abnormal development of the stromal compartment. Most unexpected, however, we also observed a cellular level lineage infidelity in nephron segments. Scattered cells within the proximal tubules, for example, expressed genes normally expressed only in collecting ducts. Multiple combinations of inappropriate nephron segment specific marker expression were found. In some cases, cells within a tubule showed incorrect identity, while in other cases cells showed ambiguous character, with simultaneous expression of genes associated with more than one nephron segment. These results give evidence that Hox genes have an overlapping core function at the cellular level in driving and/or maintaining correct differentiation decisions.
Collapse
Affiliation(s)
- Bliss Magella
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States
| | - Robert Mahoney
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States
| | - Mike Adam
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States
| | - S Steven Potter
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States.
| |
Collapse
|
130
|
Ramsawhook A, Ruzov A, Coyle B. Wilms' Tumor Protein 1 and Enzymatic Oxidation of 5-Methylcytosine in Brain Tumors: Potential Perspectives. Front Cell Dev Biol 2018; 6:26. [PMID: 29623275 PMCID: PMC5874295 DOI: 10.3389/fcell.2018.00026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 03/05/2018] [Indexed: 12/24/2022] Open
Abstract
The patterns of 5-methylcytosine (5mC) and its oxidized derivatives, 5-hydroxymethylcytosine, 5-formylcytosine, and 5-carboxylcytosine (5caC) are reportedly altered in a range of cancers. Likewise, Wilms' Tumor protein 1 (WT1), a transcription factor essential for urogenital, epicardium, and kidney development exhibits aberrant expression in multiple tumors. Interestingly, WT1 directly interacts with TET proteins that catalyze the enzymatic oxidation of 5mC and exhibits high affinity for 5caC-containing DNA substrates in vitro. Here we review recent developments in the fields of Tet-dependent 5mC oxidation and WT1 biology and explore potential perspectives for studying the interplay between TETs and WT1 in brain tumors.
Collapse
Affiliation(s)
- Ashley Ramsawhook
- Wolfson Centre for Stem Cells, Tissue Engineering and Modelling (STEM), Centre for Biomolecular Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Alexey Ruzov
- Wolfson Centre for Stem Cells, Tissue Engineering and Modelling (STEM), Centre for Biomolecular Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Beth Coyle
- Children's Brain Tumour Research Centre, Medical School, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
131
|
Lindström NO, Tran T, Guo J, Rutledge E, Parvez RK, Thornton ME, Grubbs B, McMahon JA, McMahon AP. Conserved and Divergent Molecular and Anatomic Features of Human and Mouse Nephron Patterning. J Am Soc Nephrol 2018; 29:825-840. [PMID: 29449451 PMCID: PMC5827611 DOI: 10.1681/asn.2017091036] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 11/27/2017] [Indexed: 11/03/2022] Open
Abstract
The nephron is the functional unit of the kidney, but the mechanism of nephron formation during human development is unclear. We conducted a detailed analysis of nephron development in humans and mice by immunolabeling, and we compared human and mouse nephron patterning to describe conserved and divergent features. We created protein localization maps that highlight the emerging patterns along the proximal-distal axis of the developing nephron and benchmark expectations for localization of functionally important transcription factors, which revealed unanticipated cellular diversity. Moreover, we identified a novel nephron subdomain marked by Wnt4 expression that we fate-mapped to the proximal mature nephron. Significant conservation was observed between human and mouse patterning. We also determined the time at which markers for mature nephron cell types first emerge-critical data for the renal organoid field. These findings have conceptual implications for the evolutionary processes driving the diversity of mammalian organ systems. Furthermore, these findings provide practical insights beyond those gained with mouse and rat models that will guide in vitro efforts to harness the developmental programs necessary to build human kidney structures.
Collapse
Affiliation(s)
| | - Tracy Tran
- Department of Stem Cell Biology and Regenerative Medicine and
| | - Jinjin Guo
- Department of Stem Cell Biology and Regenerative Medicine and
| | | | - Riana K Parvez
- Department of Stem Cell Biology and Regenerative Medicine and
| | - Matthew E Thornton
- Maternal Fetal Medicine Division, Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Brendan Grubbs
- Maternal Fetal Medicine Division, Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Jill A McMahon
- Department of Stem Cell Biology and Regenerative Medicine and
| | | |
Collapse
|
132
|
Lindström NO, McMahon JA, Guo J, Tran T, Guo Q, Rutledge E, Parvez RK, Saribekyan G, Schuler RE, Liao C, Kim AD, Abdelhalim A, Ruffins SW, Thornton ME, Baskin L, Grubbs B, Kesselman C, McMahon AP. Conserved and Divergent Features of Human and Mouse Kidney Organogenesis. J Am Soc Nephrol 2018; 29:785-805. [PMID: 29449453 DOI: 10.1681/asn.2017080887] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 11/27/2017] [Indexed: 01/22/2023] Open
Abstract
Human kidney function is underpinned by approximately 1,000,000 nephrons, although the number varies substantially, and low nephron number is linked to disease. Human kidney development initiates around 4 weeks of gestation and ends around 34-37 weeks of gestation. Over this period, a reiterative inductive process establishes the nephron complement. Studies have provided insightful anatomic descriptions of human kidney development, but the limited histologic views are not readily accessible to a broad audience. In this first paper in a series providing comprehensive insight into human kidney formation, we examined human kidney development in 135 anonymously donated human kidney specimens. We documented kidney development at a macroscopic and cellular level through histologic analysis, RNA in situ hybridization, immunofluorescence studies, and transcriptional profiling, contrasting human development (4-23 weeks) with mouse development at selected stages (embryonic day 15.5 and postnatal day 2). The high-resolution histologic interactive atlas of human kidney organogenesis generated can be viewed at the GUDMAP database (www.gudmap.org) together with three-dimensional reconstructions of key components of the data herein. At the anatomic level, human and mouse kidney development differ in timing, scale, and global features such as lobe formation and progenitor niche organization. The data also highlight differences in molecular and cellular features, including the expression and cellular distribution of anchor gene markers used to identify key cell types in mouse kidney studies. These data will facilitate and inform in vitro efforts to generate human kidney structures and comparative functional analyses across mammalian species.
Collapse
Affiliation(s)
- Nils O Lindström
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
| | - Jill A McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
| | - Jinjin Guo
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
| | - Tracy Tran
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
| | - Qiuyu Guo
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
| | - Elisabeth Rutledge
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
| | - Riana K Parvez
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
| | - Gohar Saribekyan
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
| | | | - Christopher Liao
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
| | - Albert D Kim
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
| | - Ahmed Abdelhalim
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
| | - Seth W Ruffins
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
| | - Matthew E Thornton
- Maternal Fetal Medicine Division, University of Southern California, Los Angeles, California; and
| | - Laurence Baskin
- Department of Urology and Pediatrics, University of California San Francisco, San Francisco, California
| | - Brendan Grubbs
- Maternal Fetal Medicine Division, University of Southern California, Los Angeles, California; and
| | - Carl Kesselman
- Information Sciences Institute, Viterbi School of Engineering.,Epstein Department of Industrial and Systems Engineering and Information Sciences Institute, Viterbi School of Engineering and Department of Preventive Medicine, Keck School of Medicine, and
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine,
| |
Collapse
|
133
|
Lindström NO, Guo J, Kim AD, Tran T, Guo Q, De Sena Brandine G, Ransick A, Parvez RK, Thornton ME, Baskin L, Grubbs B, McMahon JA, Smith AD, McMahon AP. Conserved and Divergent Features of Mesenchymal Progenitor Cell Types within the Cortical Nephrogenic Niche of the Human and Mouse Kidney. J Am Soc Nephrol 2018; 29:806-824. [PMID: 29449449 DOI: 10.1681/asn.2017080890] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 11/27/2017] [Indexed: 01/12/2023] Open
Abstract
Cellular interactions among nephron, interstitial, and collecting duct progenitors drive mammalian kidney development. In mice, Six2+ nephron progenitor cells (NPCs) and Foxd1+ interstitial progenitor cells (IPCs) form largely distinct lineage compartments at the onset of metanephric kidney development. Here, we used the method for analyzing RNA following intracellular sorting (MARIS) approach, single-cell transcriptional profiling, in situ hybridization, and immunolabeling to characterize the presumptive NPC and IPC compartments of the developing human kidney. As in mice, each progenitor population adopts a stereotypical arrangement in the human nephron-forming niche: NPCs capped outgrowing ureteric branch tips, whereas IPCs were sandwiched between the NPCs and the renal capsule. Unlike mouse NPCs, human NPCs displayed a transcriptional profile that overlapped substantially with the IPC transcriptional profile, and key IPC determinants, including FOXD1, were readily detected within SIX2+ NPCs. Comparative gene expression profiling in human and mouse Six2/SIX2+ NPCs showed broad agreement between the species but also identified species-biased expression of some genes. Notably, some human NPC-enriched genes, including DAPL1 and COL9A2, are linked to human renal disease. We further explored the cellular diversity of mesenchymal cell types in the human nephrogenic niche through single-cell transcriptional profiling. Data analysis stratified NPCs into two main subpopulations and identified a third group of differentiating cells. These findings were confirmed by section in situ hybridization with novel human NPC markers predicted through the single-cell studies. This study provides a benchmark for the mesenchymal progenitors in the human nephrogenic niche and highlights species-variability in kidney developmental programs.
Collapse
Affiliation(s)
- Nils O Lindström
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
| | - Jinjin Guo
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
| | - Albert D Kim
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
| | - Tracy Tran
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
| | - Qiuyu Guo
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
| | | | - Andrew Ransick
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
| | - Riana K Parvez
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
| | - Matthew E Thornton
- Maternal Fetal Medicine Division, University of Southern California, Los Angeles, California; and
| | - Laurence Baskin
- Department of Urology and Pediatrics, University of California San Francisco, San Francisco, California
| | - Brendan Grubbs
- Maternal Fetal Medicine Division, University of Southern California, Los Angeles, California; and
| | - Jill A McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
| | - Andrew D Smith
- Molecular and Computational Biology, Department of Biological Sciences, and
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine,
| |
Collapse
|
134
|
Wang H, Zhang C, Wang X, Lian Y, Guo B, Han M, Zhang X, Zhu X, Xu S, Guo Z, Bi Y, Shen Q, Wang X, Liu J, Zhuang Y, Ni T, Xu H, Wu X. Disruption of Gen1 Causes Congenital Anomalies of the Kidney and Urinary Tract in Mice. Int J Biol Sci 2018; 14:10-20. [PMID: 29483821 PMCID: PMC5821045 DOI: 10.7150/ijbs.22768] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 10/02/2017] [Indexed: 01/20/2023] Open
Abstract
Congenital anomalies of the kidney and urinary tract (CAKUT) are among the most common developmental defects in humans. Despite of several known CAKUT-related loci (HNF1B, PAX2, EYA1, etc.), the genetic etiology of CAKUT remains to be elucidated for most patients. In this study, we report that disruption of the Holliday Junction resolvase gene Gen1 leads to renal agenesis, duplex kidney, hydronephrosis, and vesicoureteral reflux (VUR) in mice. GEN1 interacts with SIX1 and enhances the transcriptional activity of SIX1/EYA1, a key regulatory complex of the GDNF morphogen. Gen1 mutation impairs Grem1 and Gdnf expression, resulting in excessive ureteric bud formation and defective ureteric bud branching during early kidney development. These results revealed an unidentified role of GEN1 in kidney development and suggested its contribution to CAKUT.
Collapse
Affiliation(s)
- Herui Wang
- State Key Laboratory of Genetic Engineering and National Center for International Research of Development and Disease, Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200433, China.,Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Children's Hospital of Fudan University, Shanghai 201102, China.,Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Chi Zhang
- State Key Laboratory of Genetic Engineering and National Center for International Research of Development and Disease, Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200433, China.,Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Children's Hospital of Fudan University, Shanghai 201102, China.,Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| | - Xiaowen Wang
- Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Children's Hospital of Fudan University, Shanghai 201102, China.,Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430000, China
| | - Yaru Lian
- State Key Laboratory of Genetic Engineering and National Center for International Research of Development and Disease, Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200433, China.,MOE Key Laboratory of Contemporary Anthropology, Fudan University, Shanghai 200433, China
| | - Bin Guo
- State Key Laboratory of Genetic Engineering and National Center for International Research of Development and Disease, Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Miao Han
- State Key Laboratory of Genetic Engineering and National Center for International Research of Development and Disease, Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200433, China.,MOE Key Laboratory of Contemporary Anthropology, Fudan University, Shanghai 200433, China
| | - Xiaoe Zhang
- Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Xiaoting Zhu
- State Key Laboratory of Genetic Engineering and National Center for International Research of Development and Disease, Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Sixian Xu
- State Key Laboratory of Genetic Engineering and National Center for International Research of Development and Disease, Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Zengli Guo
- State Key Laboratory of Genetic Engineering and National Center for International Research of Development and Disease, Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Yunli Bi
- Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Qian Shen
- Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Xiang Wang
- Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Jiaojiao Liu
- Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Yuan Zhuang
- State Key Laboratory of Genetic Engineering and National Center for International Research of Development and Disease, Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200433, China.,Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA
| | - Ting Ni
- State Key Laboratory of Genetic Engineering and National Center for International Research of Development and Disease, Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200433, China.,MOE Key Laboratory of Contemporary Anthropology, Fudan University, Shanghai 200433, China
| | - Hong Xu
- Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Xiaohui Wu
- State Key Laboratory of Genetic Engineering and National Center for International Research of Development and Disease, Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200433, China.,Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Children's Hospital of Fudan University, Shanghai 201102, China
| |
Collapse
|
135
|
Phelep A, Laouari D, Bharti K, Burtin M, Tammaccaro S, Garbay S, Nguyen C, Vasseur F, Blanc T, Berissi S, Langa-Vives F, Fischer E, Druilhe A, Arnheiter H, Friedlander G, Pontoglio M, Terzi F. MITF - A controls branching morphogenesis and nephron endowment. PLoS Genet 2017; 13:e1007093. [PMID: 29240767 PMCID: PMC5746285 DOI: 10.1371/journal.pgen.1007093] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 12/28/2017] [Accepted: 11/01/2017] [Indexed: 12/31/2022] Open
Abstract
Congenital nephron number varies widely in the human population and individuals with low nephron number are at risk of developing hypertension and chronic kidney disease. The development of the kidney occurs via an orchestrated morphogenetic process where metanephric mesenchyme and ureteric bud reciprocally interact to induce nephron formation. The genetic networks that modulate the extent of this process and set the final nephron number are mostly unknown. Here, we identified a specific isoform of MITF (MITF-A), a bHLH-Zip transcription factor, as a novel regulator of the final nephron number. We showed that overexpression of MITF-A leads to a substantial increase of nephron number and bigger kidneys, whereas Mitfa deficiency results in reduced nephron number. Furthermore, we demonstrated that MITF-A triggers ureteric bud branching, a phenotype that is associated with increased ureteric bud cell proliferation. Molecular studies associated with an in silico analyses revealed that amongst the putative MITF-A targets, Ret was significantly modulated by MITF-A. Consistent with the key role of this network in kidney morphogenesis, Ret heterozygosis prevented the increase of nephron number in mice overexpressing MITF-A. Collectively, these results uncover a novel transcriptional network that controls branching morphogenesis during kidney development and identifies one of the first modifier genes of nephron endowment. The number of nephrons, the functional unit of kidney, varies widely among humans. Indeed, it has been shown that kidneys may contain from 0.3 to more than 2 million of nephrons. Nephrons are formed during development via a coordinated morphogenetic program in which the metanephric mesenchyme reciprocally and recursively interacts with the ureteric bud. The fine-tuning of this cross-talk determines the final number of nephrons. Strong evidence indicates that suboptimal nephron endowment is associated with an increased risk of hypertension and chronic kidney disease, a major healthcare burden. Indeed, chronic kidney disease is characterized by the progressive decline of renal function towards end stage renal disease, which occurs once a critical number of nephrons has been lost. Elucidating the molecular mechanisms that control nephron endowment is, therefore, a critical issue for public health. However, little is known about the factors that determine the final number of nephrons in the healthy population. Our data showed that nephron endowment is genetically predetermined and identified Mitfa, a bHLH transcription factor, as one of the first modifiers of nephron formation during kidney development. By generating an allelic series of transgenic mice expressing different levels of MITF-A, we discovered that MITF-A promotes final nephron endowment. In addition, we elucidated the molecular mechanisms by which MITF-A promotes nephron formation and identified RET as one of the critical effectors.
Collapse
Affiliation(s)
- Aurélie Phelep
- INSERM U1151-CNRS UMR 8253, Université Paris Descartes, Institut Necker Enfants Malades, Département « Croissance et Signalisation », Hôpital Necker Enfants Malades, Paris, France
| | - Denise Laouari
- INSERM U1151-CNRS UMR 8253, Université Paris Descartes, Institut Necker Enfants Malades, Département « Croissance et Signalisation », Hôpital Necker Enfants Malades, Paris, France
| | - Kapil Bharti
- Unit on Ocular and Stem Cells Translational Research National Eye Institute, National Institutes of Health, Bethesda, MD, United States of America
| | - Martine Burtin
- INSERM U1151-CNRS UMR 8253, Université Paris Descartes, Institut Necker Enfants Malades, Département « Croissance et Signalisation », Hôpital Necker Enfants Malades, Paris, France
| | - Salvina Tammaccaro
- INSERM U1016-CNRS UMR 8104, Université Paris Descartes, Institut Cochin, Paris, France
| | - Serge Garbay
- INSERM U1016-CNRS UMR 8104, Université Paris Descartes, Institut Cochin, Paris, France
| | - Clément Nguyen
- INSERM U1151-CNRS UMR 8253, Université Paris Descartes, Institut Necker Enfants Malades, Département « Croissance et Signalisation », Hôpital Necker Enfants Malades, Paris, France
| | - Florence Vasseur
- INSERM U1151-CNRS UMR 8253, Université Paris Descartes, Institut Necker Enfants Malades, Département « Croissance et Signalisation », Hôpital Necker Enfants Malades, Paris, France
| | - Thomas Blanc
- INSERM U1151-CNRS UMR 8253, Université Paris Descartes, Institut Necker Enfants Malades, Département « Croissance et Signalisation », Hôpital Necker Enfants Malades, Paris, France
| | - Sophie Berissi
- INSERM U1151-CNRS UMR 8253, Université Paris Descartes, Institut Necker Enfants Malades, Département « Croissance et Signalisation », Hôpital Necker Enfants Malades, Paris, France
| | | | - Evelyne Fischer
- INSERM U1016-CNRS UMR 8104, Université Paris Descartes, Institut Cochin, Paris, France
| | - Anne Druilhe
- INSERM U1151-CNRS UMR 8253, Université Paris Descartes, Institut Necker Enfants Malades, Département « Croissance et Signalisation », Hôpital Necker Enfants Malades, Paris, France
| | - Heinz Arnheiter
- Scientist Emeritus, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 10 Center Drive, Bethesda, MD, United States of America
| | - Gerard Friedlander
- INSERM U1151-CNRS UMR 8253, Université Paris Descartes, Institut Necker Enfants Malades, Département « Croissance et Signalisation », Hôpital Necker Enfants Malades, Paris, France
| | - Marco Pontoglio
- INSERM U1016-CNRS UMR 8104, Université Paris Descartes, Institut Cochin, Paris, France
| | - Fabiola Terzi
- INSERM U1151-CNRS UMR 8253, Université Paris Descartes, Institut Necker Enfants Malades, Département « Croissance et Signalisation », Hôpital Necker Enfants Malades, Paris, France
| |
Collapse
|
136
|
Combes AN, Wilson S, Phipson B, Binnie BB, Ju A, Lawlor KT, Cebrian C, Walton SL, Smyth IM, Moritz KM, Kopan R, Oshlack A, Little MH. Haploinsufficiency for the Six2 gene increases nephron progenitor proliferation promoting branching and nephron number. Kidney Int 2017; 93:589-598. [PMID: 29217079 DOI: 10.1016/j.kint.2017.09.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 09/03/2017] [Accepted: 09/07/2017] [Indexed: 01/05/2023]
Abstract
The regulation of final nephron number in the kidney is poorly understood. Cessation of nephron formation occurs when the self-renewing nephron progenitor population commits to differentiation. Transcription factors within this progenitor population, such as SIX2, are assumed to control expression of genes promoting self-renewal such that homozygous Six2 deletion results in premature commitment and an early halt to kidney development. In contrast, Six2 heterozygotes were assumed to be unaffected. Using quantitative morphometry, we found a paradoxical 18% increase in ureteric branching and final nephron number in Six2 heterozygotes, despite evidence for reduced levels of SIX2 protein and transcript. This was accompanied by a clear shift in nephron progenitor identity with a distinct subset of downregulated progenitor genes such as Cited1 and Meox1 while other genes were unaffected. The net result was an increase in nephron progenitor proliferation, as assessed by elevated EdU (5-ethynyl-2'-deoxyuridine) labeling, an increase in MYC protein, and transcriptional upregulation of MYC target genes. Heterozygosity for Six2 on an Fgf20-/- background resulted in premature differentiation of the progenitor population, confirming that progenitor regulation is compromised in Six2 heterozygotes. Overall, our studies reveal a unique dose response of nephron progenitors to the level of SIX2 protein in which the role of SIX2 in progenitor proliferation versus self-renewal is separable.
Collapse
Affiliation(s)
- Alexander N Combes
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, Australia; Murdoch Children's Research Institute, Parkville, Victoria, Australia.
| | - Sean Wilson
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Belinda Phipson
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Brandon B Binnie
- Division of Cell Biology and Molecular Medicine, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Adler Ju
- Division of Cell Biology and Molecular Medicine, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Kynan T Lawlor
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Cristina Cebrian
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Sarah L Walton
- School of Biomedical Sciences and Centre for Children's Health Research, The University of Queensland, Brisbane, Queensland, Australia
| | - Ian M Smyth
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Melbourne, Australia; Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia; Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Karen M Moritz
- School of Biomedical Sciences and Centre for Children's Health Research, The University of Queensland, Brisbane, Queensland, Australia
| | - Raphael Kopan
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Alicia Oshlack
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Melissa H Little
- Murdoch Children's Research Institute, Parkville, Victoria, Australia; Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
137
|
Yamanaka S, Tajiri S, Fujimoto T, Matsumoto K, Fukunaga S, Kim BS, Okano HJ, Yokoo T. Generation of interspecies limited chimeric nephrons using a conditional nephron progenitor cell replacement system. Nat Commun 2017; 8:1719. [PMID: 29170512 PMCID: PMC5701015 DOI: 10.1038/s41467-017-01922-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 10/25/2017] [Indexed: 01/08/2023] Open
Abstract
Animal fetuses and embryos may have applications in the generation of human organs. Progenitor cells may be an appropriate cell source for regenerative organs because of their safety and availability. However, regenerative organs derived from exogenous lineage progenitors in developing animal fetuses have not yet been obtained. Here, we established a combination system through which donor cells could be precisely injected into the nephrogenic zone and native nephron progenitor cells (NPCs) could be eliminated in a time- and tissue-specific manner. We successfully achieved removal of Six2+ NPCs within the nephrogenic niche and complete replacement of transplanted NPCs with donor cells. These NPCs developed into mature glomeruli and renal tubules, and blood flow was observed following transplantation in vivo. Furthermore, this artificial nephron could be obtained using NPCs from different species. Thus, this technique enables in vivo differentiation from progenitor cells into nephrons, providing insights into nephrogenesis and organ regeneration. The transplantation of tissue-specific progenitor cells may be an approach in organ regeneration. Here the authors show that the nephron progenitor population of a developing mouse kidney, when ablated, can be replaced by exogenously supplied rat nephron progenitors, generating interspecies nephrons.
Collapse
Affiliation(s)
- S Yamanaka
- Division of Nephrology and Hypertension, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, 1058461, Japan
| | - S Tajiri
- Division of Nephrology and Hypertension, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, 1058461, Japan.,Division of Regenerative Medicine, Jikei University School of Medicine, Tokyo, 1058461, Japan
| | - T Fujimoto
- Division of Nephrology and Hypertension, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, 1058461, Japan.,Division of Regenerative Medicine, Jikei University School of Medicine, Tokyo, 1058461, Japan
| | - K Matsumoto
- Division of Nephrology and Hypertension, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, 1058461, Japan
| | - S Fukunaga
- Division of Nephrology and Hypertension, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, 1058461, Japan.,Department of Internal Medicine IV, Shimane University, Izumo, Shimane, 6938501, Japan
| | - B S Kim
- Division of Regenerative Medicine, Jikei University School of Medicine, Tokyo, 1058461, Japan.,Department of Urology, Kyungpook National University School of Medicine, Daegu, 41944, Korea
| | - H J Okano
- Division of Regenerative Medicine, Jikei University School of Medicine, Tokyo, 1058461, Japan
| | - T Yokoo
- Division of Nephrology and Hypertension, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, 1058461, Japan.
| |
Collapse
|
138
|
Desgrange A, Heliot C, Skovorodkin I, Akram SU, Heikkilä J, Ronkainen VP, Miinalainen I, Vainio SJ, Cereghini S. HNF1B controls epithelial organization and cell polarity during ureteric bud branching and collecting duct morphogenesis. Development 2017; 144:4704-4719. [PMID: 29158444 DOI: 10.1242/dev.154336] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 11/03/2017] [Indexed: 12/16/2022]
Abstract
Kidney development depends crucially on proper ureteric bud branching giving rise to the entire collecting duct system. The transcription factor HNF1B is required for the early steps of ureteric bud branching, yet the molecular and cellular events regulated by HNF1B are poorly understood. We report that specific removal of Hnf1b from the ureteric bud leads to defective cell-cell contacts and apicobasal polarity during the early branching events. High-resolution ex vivo imaging combined with a membranous fluorescent reporter strategy show decreased mutant cell rearrangements during mitosis-associated cell dispersal and severe epithelial disorganization. Molecular analysis reveals downregulation of Gdnf-Ret pathway components and suggests that HNF1B acts both upstream and downstream of Ret signaling by directly regulating Gfra1 and Etv5 Subsequently, Hnf1b deletion leads to massively mispatterned ureteric tree network, defective collecting duct differentiation and disrupted tissue architecture, which leads to cystogenesis. Consistently, mRNA-seq analysis shows that the most impacted genes encode intrinsic cell-membrane components with transporter activity. Our study uncovers a fundamental and recurring role of HNF1B in epithelial organization during early ureteric bud branching and in further patterning and differentiation of the collecting duct system in mouse.
Collapse
Affiliation(s)
- Audrey Desgrange
- Sorbonne Universités, UPMC Université Paris 06, IBPS - UMR7622, F-75005 Paris, France .,CNRS, UMR7622, Institut de Biologie Paris-Seine (IBPS) - Developmental Biology Laboratory, F-75005 Paris, France
| | - Claire Heliot
- Sorbonne Universités, UPMC Université Paris 06, IBPS - UMR7622, F-75005 Paris, France.,CNRS, UMR7622, Institut de Biologie Paris-Seine (IBPS) - Developmental Biology Laboratory, F-75005 Paris, France
| | - Ilya Skovorodkin
- Faculty of Biochemistry and Molecular Medicine, Biocenter, University of Oulu; Laboratory of Developmental Biology, Biocenter Oulu and InfoTech, Department of Medical Biochemistry and Molecular Medicine, Oulu Center for Cell Matrix Research, 90220 Oulu, Finland
| | - Saad U Akram
- Center for Machine Vision Research and Signal Analysis (CMVS), University of Oulu, FIN-90014, Oulu, Finland
| | - Janne Heikkilä
- Center for Machine Vision Research and Signal Analysis (CMVS), University of Oulu, FIN-90014, Oulu, Finland
| | | | | | - Seppo J Vainio
- Faculty of Biochemistry and Molecular Medicine, Biocenter, University of Oulu; Laboratory of Developmental Biology, Biocenter Oulu and InfoTech, Department of Medical Biochemistry and Molecular Medicine, Oulu Center for Cell Matrix Research, 90220 Oulu, Finland
| | - Silvia Cereghini
- Sorbonne Universités, UPMC Université Paris 06, IBPS - UMR7622, F-75005 Paris, France .,CNRS, UMR7622, Institut de Biologie Paris-Seine (IBPS) - Developmental Biology Laboratory, F-75005 Paris, France
| |
Collapse
|
139
|
Minuth W. Reading First Coordinates from the Nephrogenic Zone in Human Fetal Kidney. Nephron Clin Pract 2017; 138:137-146. [DOI: 10.1159/000481441] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 09/11/2017] [Indexed: 01/15/2023] Open
|
140
|
Lefevre JG, Short KM, Lamberton TO, Michos O, Graf D, Smyth IM, Hamilton NA. Branching morphogenesis in the developing kidney is governed by rules that pattern the ureteric tree. Development 2017; 144:4377-4385. [PMID: 29038307 DOI: 10.1242/dev.153874] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 10/05/2017] [Indexed: 12/23/2022]
Abstract
Metanephric kidney development is orchestrated by the iterative branching morphogenesis of the ureteric bud. We describe an underlying patterning associated with the ramification of this structure and show that this pattern is conserved between developing kidneys, in different parts of the organ and across developmental time. This regularity is associated with a highly reproducible branching asymmetry that is consistent with locally operative growth mechanisms. We then develop a class of tip state models to represent elaboration of the ureteric tree and describe rules for 'half-delay' branching morphogenesis that describe almost perfectly the patterning of this structure. Spatial analysis suggests that the observed asymmetry may arise from mutual suppression of bifurcation, but not extension, between the growing ureteric tips, and demonstrates that disruption of patterning occurs in mouse mutants in which the distribution of tips on the surface of the kidney is altered. These findings demonstrate that kidney development occurs by way of a highly conserved reiterative pattern of asymmetric bifurcation that is governed by intrinsic and locally operative mechanisms.
Collapse
Affiliation(s)
- James G Lefevre
- Division of Genomics and Development of Disease, Institute for Molecular Biosciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Kieran M Short
- Biomedicine Discovery Institute, Monash University, Clayton, Melbourne, Victoria 3800, Australia.,Department of Anatomy and Developmental Biology, Monash University, Clayton, Melbourne, Victoria 3800, Australia
| | - Timothy O Lamberton
- Division of Genomics and Development of Disease, Institute for Molecular Biosciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Odyssé Michos
- Department of Biosystems, Science and Engineering (D-BSSE), ETH Zurich, Basel 4058, Switzerland
| | - Daniel Graf
- School of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 1C9, Canada
| | - Ian M Smyth
- Biomedicine Discovery Institute, Monash University, Clayton, Melbourne, Victoria 3800, Australia .,Department of Anatomy and Developmental Biology, Monash University, Clayton, Melbourne, Victoria 3800, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Clayton, Melbourne, Victoria 3800, Australia
| | - Nicholas A Hamilton
- Division of Genomics and Development of Disease, Institute for Molecular Biosciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
141
|
Munro DAD, Hohenstein P, Coate TM, Davies JA. Refuting the hypothesis that semaphorin-3f/neuropilin-2 exclude blood vessels from the cap mesenchyme in the developing kidney. Dev Dyn 2017; 246:1047-1056. [PMID: 28929539 DOI: 10.1002/dvdy.24592] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 08/16/2017] [Accepted: 09/16/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND During murine kidney development, new cortical blood vessels form and pattern in cycles that coincide with cycles of collecting duct branching and the accompanying splitting of the cap mesenchyme (nephron progenitor cell populations that "cap" collecting duct ends). At no point in the patterning cycle do blood vessels enter the cap mesenchyme. We hypothesized that the exclusion of blood vessels from the cap mesenchyme may be controlled, at least in part, by an anti-angiogenic signal expressed by the cap mesenchyme cells. RESULTS We show that semaphorin-3f (Sema3f), a known anti-angiogenic factor, is expressed in cap mesenchymal cells and its receptor, neuropilin-2 (Nrp2), is expressed by newly forming blood vessels in the cortex of the developing kidney. We hypothesized that Sema3f/Nrp2 signaling excludes vessels from the cap mesenchyme. Genetic ablation of Sema3f and of Nrp2, however, failed to result in vessels invading the cap mesenchyme. CONCLUSIONS Despite complementary expression patterns, our data suggest that Sema3f and Nrp2 are dispensable for the exclusion of vessels from the cap mesenchyme during kidney development. These results should provoke additional experiments to ascertain the biological significance of Sema3f/Nrp2 expression in the developing kidney. Developmental Dynamics 246:1047-1056, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- David A D Munro
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
| | - Peter Hohenstein
- The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Thomas M Coate
- Georgetown University, Department of Biology, Washington, DC
| | - Jamie A Davies
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
142
|
Hannezo E, Scheele CLGJ, Moad M, Drogo N, Heer R, Sampogna RV, van Rheenen J, Simons BD. A Unifying Theory of Branching Morphogenesis. Cell 2017; 171:242-255.e27. [PMID: 28938116 PMCID: PMC5610190 DOI: 10.1016/j.cell.2017.08.026] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 06/20/2017] [Accepted: 08/15/2017] [Indexed: 11/23/2022]
Abstract
The morphogenesis of branched organs remains a subject of abiding interest. Although much is known about the underlying signaling pathways, it remains unclear how macroscopic features of branched organs, including their size, network topology, and spatial patterning, are encoded. Here, we show that, in mouse mammary gland, kidney, and human prostate, these features can be explained quantitatively within a single unifying framework of branching and annihilating random walks. Based on quantitative analyses of large-scale organ reconstructions and proliferation kinetics measurements, we propose that morphogenesis follows from the proliferative activity of equipotent tips that stochastically branch and randomly explore their environment but compete neutrally for space, becoming proliferatively inactive when in proximity with neighboring ducts. These results show that complex branched epithelial structures develop as a self-organized process, reliant upon a strikingly simple but generic rule, without recourse to a rigid and deterministic sequence of genetically programmed events. Branching morphogenesis follows conserved statistical rules in multiple organs Ductal tips grow and branch as default state and stop dividing in high-density regions Model reproduces quantitatively organ properties in a parameter-free manner Shows that complex organ formation proceeds in a stochastic, self-organized manner
Collapse
Affiliation(s)
- Edouard Hannezo
- Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge CB3 0HE, UK; The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK; The Wellcome Trust/Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge CB2 1QN, UK.
| | - Colinda L G J Scheele
- Cancer Genomics Netherlands, Hubrecht Institute-KNAW and University Medical Centre Utrecht, Utrecht 3584CT, the Netherlands
| | - Mohammad Moad
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne NE2 4AD, UK
| | - Nicholas Drogo
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14627, USA
| | - Rakesh Heer
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne NE2 4AD, UK
| | - Rosemary V Sampogna
- Division of Nephrology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Jacco van Rheenen
- Cancer Genomics Netherlands, Hubrecht Institute-KNAW and University Medical Centre Utrecht, Utrecht 3584CT, the Netherlands.
| | - Benjamin D Simons
- Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge CB3 0HE, UK; The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK; The Wellcome Trust/Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge CB2 1QN, UK.
| |
Collapse
|
143
|
Barnett C, Nnoli O, Abdulmahdi W, Nesi L, Shen M, Zullo JA, Payne DL, Azar T, Dwivedi P, Syed K, Gromis J, Lipphardt M, Jules E, Maranda EL, Patel A, Rabadi MM, Ratliff BB. Low birth weight is associated with impaired murine kidney development and function. Pediatr Res 2017; 82:340-348. [PMID: 28419086 DOI: 10.1038/pr.2017.53] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 02/19/2017] [Indexed: 11/08/2022]
Abstract
BackgroundLow birth weight (LBW) neonates have impaired kidney development that leaves them susceptible to kidney disease and hypertension during adulthood. The study here identifies events that blunt nephrogenesis and kidney development in the murine LBW neonate.MethodsWe examined survival, kidney development, GFR, gene expression, and cyto-/chemokines in the LBW offspring of malnourished (caloric and protein-restricted) pregnant mice.ResultsMalnourished pregnant mothers gave birth to LBW neonates that had 40% reduced body weight and 54% decreased survival. Renal blood perfusion was reduced by 37%, whereas kidney volume and GFR were diminished in the LBW neonate. During gestation, the LBW neonatal kidney had 2.2-fold increased apoptosis, 76% decreased SIX2+ progenitor cells, downregulation of mesenchymal-to-epithelial signaling factors Wnt9b and Fgf8, 64% less renal vesicle formation, and 32% fewer nephrons than controls. At birth, increased plasma levels of IL-1β, IL-6, IL-12(p70), and granulocyte-macrophage colony-stimulating factor in the LBW neonate reduced SIX2+ progenitor cells.ConclusionIncreased pro-inflammatory cytokines in the LBW neonate decrease SIX2+ stem cells in the developing kidney. Reduced renal stem cells (along with the decreased mesenchymal-to-epithelial signaling) blunt renal vesicle generation, nephron formation, and kidney development. Subsequently, the mouse LBW neonate has reduced glomeruli volume, renal perfusion, and GFR.
Collapse
Affiliation(s)
- Christina Barnett
- Department of Medicine, Renal Research Institute, New York Medical College, Valhalla, New York
| | - Oluwadara Nnoli
- Department of Physiology, Renal Research Institute, New York Medical College, Valhalla, New York
| | - Wasan Abdulmahdi
- Department of Physiology, Renal Research Institute, New York Medical College, Valhalla, New York
| | - Lauren Nesi
- Department of Medicine, Renal Research Institute, New York Medical College, Valhalla, New York
| | - Michael Shen
- Department of Medicine, Renal Research Institute, New York Medical College, Valhalla, New York
| | - Joseph A Zullo
- Department of Physiology, Renal Research Institute, New York Medical College, Valhalla, New York
| | - David L Payne
- Department of Medicine, Renal Research Institute, New York Medical College, Valhalla, New York
| | - Tala Azar
- Department of Medicine, Renal Research Institute, New York Medical College, Valhalla, New York
| | - Parth Dwivedi
- Department of Physiology, Renal Research Institute, New York Medical College, Valhalla, New York
| | - Kunzah Syed
- Department of Medicine, Renal Research Institute, New York Medical College, Valhalla, New York
| | - Jonathan Gromis
- Department of Medicine, Renal Research Institute, New York Medical College, Valhalla, New York
| | - Mark Lipphardt
- Department of Medicine, Renal Research Institute, New York Medical College, Valhalla, New York
| | - Edson Jules
- Department of Medicine, Renal Research Institute, New York Medical College, Valhalla, New York
| | - Eric L Maranda
- Department of Dermatology and Cutaneous Surgery, Miami University, Miami, Florida
| | - Amy Patel
- Department of Medicine, Renal Research Institute, New York Medical College, Valhalla, New York
| | - May M Rabadi
- Department of Anesthesiology, College of Physicians and Surgeons of Columbia University, New York, New York
| | - Brian B Ratliff
- Department of Medicine, Renal Research Institute, New York Medical College, Valhalla, New York
| |
Collapse
|
144
|
Basta JM, Robbins L, Denner DR, Kolar GR, Rauchman M. A Sall1-NuRD interaction regulates multipotent nephron progenitors and is required for loop of Henle formation. Development 2017; 144:3080-3094. [PMID: 28760814 DOI: 10.1242/dev.148692] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 07/24/2017] [Indexed: 01/03/2023]
Abstract
The formation of the proper number of nephrons requires a tightly regulated balance between renal progenitor cell self-renewal and differentiation. The molecular pathways that regulate the transition from renal progenitor to renal vesicle are not well understood. Here, we show that Sall1interacts with the nucleosome remodeling and deacetylase complex (NuRD) to inhibit premature differentiation of nephron progenitor cells. Disruption of Sall1-NuRD in vivo in knock-in mice (ΔSRM) resulted in accelerated differentiation of nephron progenitors and bilateral renal hypoplasia. Transcriptional profiling of mutant kidneys revealed a striking pattern in which genes of the glomerular and proximal tubule lineages were either unchanged or upregulated, and those in the loop of Henle and distal tubule lineages were downregulated. These global changes in gene expression were accompanied by a significant decrease in THP-, NKCC2- and AQP1-positive loop of Henle nephron segments in mutant ΔSRM kidneys. These findings highlight an important function of Sall1-NuRD interaction in the regulation of Six2-positive multipotent renal progenitor cells and formation of the loop of Henle.
Collapse
Affiliation(s)
- Jeannine M Basta
- Department of Internal Medicine, Saint Louis University, St Louis, MO 63104, USA
| | - Lynn Robbins
- Department of Internal Medicine, Saint Louis University, St Louis, MO 63104, USA
| | - Darcy R Denner
- Department of Biochemistry and Molecular Biology, Saint Louis University, St Louis, MO 63104, USA
| | - Grant R Kolar
- Department of Pathology, Saint Louis University, St Louis, MO 63104, USA
| | - Michael Rauchman
- Department of Internal Medicine, Saint Louis University, St Louis, MO 63104, USA .,Department of Biochemistry and Molecular Biology, Saint Louis University, St Louis, MO 63104, USA.,VA Saint Louis Health Care System, John Cochran Division, St Louis, MO 63106, USA
| |
Collapse
|
145
|
Liu J, Edgington-Giordano F, Dugas C, Abrams A, Katakam P, Satou R, Saifudeen Z. Regulation of Nephron Progenitor Cell Self-Renewal by Intermediary Metabolism. J Am Soc Nephrol 2017; 28:3323-3335. [PMID: 28754792 DOI: 10.1681/asn.2016111246] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Accepted: 06/09/2017] [Indexed: 12/21/2022] Open
Abstract
Nephron progenitor cells (NPCs) show an age-dependent capacity to balance self-renewal with differentiation. Older NPCs (postnatal day 0) exit the progenitor niche at a higher rate than younger (embryonic day 13.5) NPCs do. This behavior is reflected in the transcript profiles of young and old NPCs. Bioenergetic pathways have emerged as important regulators of stem cell fate. Here, we investigated the mechanisms underlying this regulation in murine NPCs. Upon isolation and culture in NPC renewal medium, younger NPCs displayed a higher glycolysis rate than older NPCs. Inhibition of glycolysis enhanced nephrogenesis in cultured embryonic kidneys, without increasing ureteric tree branching, and promoted mesenchymal-to-epithelial transition in cultured isolated metanephric mesenchyme. Cotreatment with a canonical Wnt signaling inhibitor attenuated but did not entirely block the increase in nephrogenesis observed after glycolysis inhibition. Furthermore, inhibition of the phosphatidylinositol 3-kinase/Akt self-renewal signaling pathway or stimulation of differentiation pathways in the NPC decreased glycolytic flux. Our findings suggest that glycolysis is a pivotal, cell-intrinsic determinant of NPC fate, with a high glycolytic flux supporting self-renewal and inhibition of glycolysis stimulating differentiation.
Collapse
Affiliation(s)
- Jiao Liu
- Department of Pediatrics, Section of Nephrology
| | | | | | - Anna Abrams
- Department of Pediatrics, Section of Nephrology
| | - Prasad Katakam
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana
| | | | | |
Collapse
|
146
|
Rutledge EA, Benazet JD, McMahon AP. Cellular heterogeneity in the ureteric progenitor niche and distinct profiles of branching morphogenesis in organ development. Development 2017; 144:3177-3188. [PMID: 28705898 DOI: 10.1242/dev.149112] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 07/10/2017] [Indexed: 12/26/2022]
Abstract
Branching morphogenesis creates arborized epithelial networks. In the mammalian kidney, an epithelial progenitor pool at ureteric branch tips (UBTs) creates the urine-transporting collecting system. Using region-specific mouse reporter strains, we performed an RNA-seq screen, identifying tip- and stalk-enriched gene sets in the developing collecting duct system. Detailed in situ hybridization studies of tip-enriched predictions identified UBT-enriched gene sets conserved between the mouse and human kidney. Comparative spatial analysis of their UBT niche expression highlighted distinct patterns of gene expression revealing novel molecular heterogeneity within the UBT progenitor population. To identify kidney-specific and shared programs of branching morphogenesis, comparative expression studies on the developing mouse lung were combined with in silico analysis of the developing mouse salivary gland. These studies highlight a shared gene set with multi-organ tip enrichment and a gene set specific to UBTs. This comprehensive analysis extends our current understanding of the ureteric branch tip niche.
Collapse
Affiliation(s)
- Elisabeth A Rutledge
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, W.M. Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Jean-Denis Benazet
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, W.M. Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA.,Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, CA 94143, USA
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, W.M. Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
147
|
Cycles of vascular plexus formation within the nephrogenic zone of the developing mouse kidney. Sci Rep 2017; 7:3273. [PMID: 28607473 PMCID: PMC5468301 DOI: 10.1038/s41598-017-03808-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 05/04/2017] [Indexed: 12/25/2022] Open
Abstract
The renal vasculature is required for blood filtration, blood pressure regulation, and pH maintenance, as well as other specialised kidney functions. Yet, despite its importance, many aspects of its development are poorly understood. To provide a detailed spatiotemporal analysis of kidney vascularisation, we collected images of embryonic mouse kidneys at various developmental time-points. Here we describe the first stages of kidney vascularisation and demonstrate that polygonal networks of vessels (endothelial plexuses) form in cycles at the periphery of the kidney. We show that kidney vascularisation initiates at E11, when vessels connected to the embryonic circulation form a ring around the ureteric bud. From E13.5, endothelial plexuses organise around populations of cap mesenchymal and ureteric bud cells in a cyclical, predictable manner. Specifically, as the ureteric bud bifurcates, endothelia form across the bifurcation site as the cap mesenchyme splits. The plexuses are vascular, carry erythrocytes, are enclosed within a basement membrane, and can always be traced back to the renal artery. Our results are a major step towards understanding how the global architecture of the renal vasculature is achieved.
Collapse
|
148
|
Chevalier RL. Evolutionary Nephrology. Kidney Int Rep 2017; 2:302-317. [PMID: 28845468 PMCID: PMC5568830 DOI: 10.1016/j.ekir.2017.01.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 01/11/2017] [Accepted: 01/25/2017] [Indexed: 01/05/2023] Open
Abstract
Progressive kidney disease follows nephron loss, hyperfiltration, and incomplete repair, a process described as "maladaptive." In the past 20 years, a new discipline has emerged that expands research horizons: evolutionary medicine. In contrast to physiologic (homeostatic) adaptation, evolutionary adaptation is the result of reproductive success that reflects natural selection. Evolutionary explanations for physiologically maladaptive responses can emerge from mismatch of the phenotype with environment or evolutionary tradeoffs. Evolutionary adaptation to a terrestrial environment resulted in a vulnerable energy-consuming renal tubule and a hypoxic, hyperosmolar microenvironment. Natural selection favors successful energy investment strategy: energy is allocated to maintenance of nephron integrity through reproductive years, but this declines with increasing senescence after ~40 years of age. Risk factors for chronic kidney disease include restricted fetal growth or preterm birth (life history tradeoff resulting in fewer nephrons), evolutionary selection for APOL1 mutations (that provide resistance to trypanosome infection, a tradeoff), and modern life experience (Western diet mismatch leading to diabetes and hypertension). Current advances in genomics, epigenetics, and developmental biology have revealed proximate causes of kidney disease, but attempts to slow kidney disease remain elusive. Evolutionary medicine provides a complementary approach by addressing ultimate causes of kidney disease. Marked variation in nephron number at birth, nephron heterogeneity, and changing susceptibility to kidney injury throughout life history are the result of evolutionary processes. Combined application of molecular genetics, evolutionary developmental biology (evo-devo), developmental programming and life history theory may yield new strategies for prevention and treatment of chronic kidney disease.
Collapse
Affiliation(s)
- Robert L. Chevalier
- Department of Pediatrics, The University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
149
|
CLARITY reveals dynamics of ovarian follicular architecture and vasculature in three-dimensions. Sci Rep 2017; 7:44810. [PMID: 28333125 PMCID: PMC5363086 DOI: 10.1038/srep44810] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 02/15/2017] [Indexed: 12/11/2022] Open
Abstract
Optimal distribution of heterogeneous organelles and cell types within an organ is essential for physiological processes. Unique for the ovary, hormonally regulated folliculogenesis, ovulation, luteal formation/regression and associated vasculature changes lead to tissue remodeling during each reproductive cycle. Using the CLARITY approach and marker immunostaining, we identified individual follicles and corpora lutea in intact ovaries. Monitoring lifetime changes in follicle populations showed age-dependent decreases in total follicles and percentages of advanced follicles. Follicle development from primordial to preovulatory stage was characterized by 3 × 105-fold increases in volume, decreases in roundness, and decreased clustering of same stage follicles. Construction of follicle-vasculature relationship maps indicated age- and gonadotropin-dependent increases in vasculature and branching surrounding follicles. Heterozygous mutant mice with deletion of hypoxia-response element in the vascular endothelial growth factor A (VEGFA) promoter showed defective ovarian vasculature and decreased ovulatory responses. Unilateral intrabursal injection of axitinib, an inhibitor of VEGF receptors, retarded neo-angiogenesis that was associated with defective ovulation in treated ovaries. Our approach uncovers unique features of ovarian architecture and essential roles of vasculature in organizing follicles to allow future studies on normal and diseased human ovaries. Similar approaches could also reveal roles of neo-angiogenesis during embryonic development and tumorigenesis.
Collapse
|
150
|
Wang S, Sekiguchi R, Daley WP, Yamada KM. Patterned cell and matrix dynamics in branching morphogenesis. J Cell Biol 2017; 216:559-570. [PMID: 28174204 PMCID: PMC5350520 DOI: 10.1083/jcb.201610048] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 12/05/2016] [Accepted: 12/21/2016] [Indexed: 12/16/2022] Open
Abstract
Many embryonic organs undergo branching morphogenesis to maximize their functional epithelial surface area. Branching morphogenesis requires the coordinated interplay of multiple types of cells with the extracellular matrix (ECM). During branching morphogenesis, new branches form by "budding" or "clefting." Cell migration, proliferation, rearrangement, deformation, and ECM dynamics have varied roles in driving budding versus clefting in different organs. Elongation of the newly formed branch and final maturation of the tip involve cellular mechanisms that include cell elongation, intercalation, convergent extension, proliferation, and differentiation. New methodologies such as high-resolution live imaging, tension sensors, and force-mapping techniques are providing exciting new opportunities for future research into branching morphogenesis.
Collapse
Affiliation(s)
- Shaohe Wang
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
| | - Rei Sekiguchi
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
| | - William P Daley
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
| | - Kenneth M Yamada
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|