51
|
Rubin S, Agrawal A, Stegmaier J, Krief S, Felsenthal N, Svorai J, Addadi Y, Villoutreix P, Stern T, Zelzer E. Application of 3D MAPs pipeline identifies the morphological sequence chondrocytes undergo and the regulatory role of GDF5 in this process. Nat Commun 2021; 12:5363. [PMID: 34508093 PMCID: PMC8433335 DOI: 10.1038/s41467-021-25714-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 08/19/2021] [Indexed: 02/08/2023] Open
Abstract
The activity of epiphyseal growth plates, which drives long bone elongation, depends on extensive changes in chondrocyte size and shape during differentiation. Here, we develop a pipeline called 3D Morphometric Analysis for Phenotypic significance (3D MAPs), which combines light-sheet microscopy, segmentation algorithms and 3D morphometric analysis to characterize morphogenetic cellular behaviors while maintaining the spatial context of the growth plate. Using 3D MAPs, we create a 3D image database of hundreds of thousands of chondrocytes. Analysis reveals broad repertoire of morphological changes, growth strategies and cell organizations during differentiation. Moreover, identifying a reduction in Smad 1/5/9 activity together with multiple abnormalities in cell growth, shape and organization provides an explanation for the shortening of Gdf5 KO tibias. Overall, our findings provide insight into the morphological sequence that chondrocytes undergo during differentiation and highlight the ability of 3D MAPs to uncover cellular mechanisms that may regulate this process.
Collapse
Affiliation(s)
- Sarah Rubin
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Ankit Agrawal
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Johannes Stegmaier
- Institute of Imaging and Computer Vision, RWTH Aachen University, Aachen, Germany
- Institute for Automation and Applied Informatics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Sharon Krief
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Neta Felsenthal
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Jonathan Svorai
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Yoseph Addadi
- Department of Life Science Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Paul Villoutreix
- LIS (UMR 7020), IBDM (UMR 7288), Turing Center For Living Systems, Aix-Marseille University, Marseille, France.
| | - Tomer Stern
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel.
| | - Elazar Zelzer
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
52
|
Li J, Geraldo LH, Dubrac A, Zarkada G, Eichmann A. Slit2-Robo Signaling Promotes Glomerular Vascularization and Nephron Development. J Am Soc Nephrol 2021; 32:2255-2272. [PMID: 34341180 PMCID: PMC8729857 DOI: 10.1681/asn.2020111640] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 04/22/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Kidney function requires continuous blood filtration by glomerular capillaries. Disruption of glomerular vascular development or maintenance contributes to the pathogenesis of kidney diseases, but the signaling events regulating renal endothelium development remain incompletely understood. Here, we discovered a novel role of Slit2-Robo signaling in glomerular vascularization. Slit2 is a secreted polypeptide that binds to transmembrane Robo receptors and regulates axon guidance as well as ureteric bud branching and angiogenesis. METHODS We performed Slit2-alkaline phosphatase binding to kidney cryosections from mice with or without tamoxifen-inducible Slit2 or Robo1 and -2 deletions, and we characterized the phenotypes using immunohistochemistry, electron microscopy, and functional intravenous dye perfusion analysis. RESULTS Only the glomerular endothelium, but no other renal endothelial compartment, responded to Slit2 in the developing kidney vasculature. Induced Slit2 gene deletion or Slit2 ligand trap at birth affected nephrogenesis and inhibited vascularization of developing glomeruli by reducing endothelial proliferation and migration, leading to defective cortical glomerular perfusion and abnormal podocyte differentiation. Global and endothelial-specific Robo deletion showed that both endothelial and epithelial Robo receptors contributed to glomerular vascularization. CONCLUSIONS Our study provides new insights into the signaling pathways involved in glomerular vascular development and identifies Slit2 as a potential tool to enhance glomerular angiogenesis.
Collapse
Affiliation(s)
- Jinyu Li
- Department of Cellular and Molecular Physiology, Yale University Medical School, New Haven, Connecticut
- Cardiovascular Research Center, Department of Internal Medicine, Yale University, New Haven, Connecticut
| | - Luiz Henrique Geraldo
- Cardiovascular Research Center, Department of Internal Medicine, Yale University, New Haven, Connecticut
- Université de Paris, Paris Cardiovascular Research Center, Institut National de la Santé et de la Recherche Médicale U907, Paris, France
| | - Alexandre Dubrac
- Cardiovascular Research Center, Department of Internal Medicine, Yale University, New Haven, Connecticut
| | - Georgia Zarkada
- Cardiovascular Research Center, Department of Internal Medicine, Yale University, New Haven, Connecticut
| | - Anne Eichmann
- Department of Cellular and Molecular Physiology, Yale University Medical School, New Haven, Connecticut
- Cardiovascular Research Center, Department of Internal Medicine, Yale University, New Haven, Connecticut
- Université de Paris, Paris Cardiovascular Research Center, Institut National de la Santé et de la Recherche Médicale U907, Paris, France
| |
Collapse
|
53
|
Wen-Jin C, Xiu-Wu P, Jian C, Da X, Jia-Xin C, Wei-Jie C, Lin-Hui W, Xin-Gang C. Study of cellular heterogeneity and differential dynamics of autophagy in human embryonic kidney development by single-cell RNA sequencing. Cancer Cell Int 2021; 21:460. [PMID: 34461918 PMCID: PMC8404318 DOI: 10.1186/s12935-021-02154-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 08/16/2021] [Indexed: 11/17/2022] Open
Abstract
Background Autophagy is believed to participate in embryonic development, but whether the expression of autophagy-associated genes undergoes changes during the development of human embryonic kidneys remains unknown. Methods In this work, we identified 36,151 human renal cells from embryonic kidneys of 9–18 gestational weeks in 16 major clusters by single-cell RNA sequencing (scRNA-seq), and detected 1350 autophagy-related genes in all fetal renal cells. The abundance of each cell cluster in Wilms tumor samples from scRNA-seq and GDC TARGET WT datasets was detected by CIBERSORTx. R package Monocle 3 was used to determine differentiation trajectories. Cyclone tool of R package scran was applied to calculate the cell cycle scores. R package SCENIC was used to investigate the transcriptional regulons. The FindMarkers tool from Seurat was used to calculate DEGs. GSVA was used to perform gene set enrichment analyses. CellphoneDB was utilized to analyze intercellular communication. Results It was found that cells in the 13th gestational week showed the lowest transcriptional level in each cluster in all stages. Nephron progenitors could be divided into four subgroups with diverse levels of autophagy corresponding to different SIX2 expressions. SSBpod (podocyte precursors) could differentiate into four types of podocytes (Pod), and autophagy-related regulation was involved in this process. Pseudotime analysis showed that interstitial progenitor cells (IPCs) potentially possessed two primitive directions of differentiation to interstitial cells with different expressions of autophagy. It was found that NPCs, pretubular aggregates and interstitial cell clusters had high abundance in Wilms tumor as compared with para-tumor samples with active intercellular communication. Conclusions All these findings suggest that autophagy may be involved in the development and cellular heterogeneity of early human fetal kidneys. In addition, part of Wilms tumor cancer cells possess the characteristics of some fetal renal cell clusters. Graphical abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02154-w.
Collapse
Affiliation(s)
- Chen Wen-Jin
- Department of Urology, The Third Affiliated Hospital of Second Military Medical University, 700 North Moyu Road, Shanghai, 201805, China
| | - Pan Xiu-Wu
- Department of Urology, The Third Affiliated Hospital of Second Military Medical University, 700 North Moyu Road, Shanghai, 201805, China.,Department of Urology, Xinhua Hospital, Shanghai Jiaotong University, School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Chu Jian
- Department of Urology, The Third Affiliated Hospital of Second Military Medical University, 700 North Moyu Road, Shanghai, 201805, China.,Department of Urology, Gongli Hospital of Second Military Medical University, 219 Miaopu Road, Shanghai, 200135, China
| | - Xu Da
- Department of Urology, The Third Affiliated Hospital of Second Military Medical University, 700 North Moyu Road, Shanghai, 201805, China
| | - Chen Jia-Xin
- Department of Urology, The Third Affiliated Hospital of Second Military Medical University, 700 North Moyu Road, Shanghai, 201805, China
| | - Chen Wei-Jie
- Department of Urology, The Third Affiliated Hospital of Second Military Medical University, 700 North Moyu Road, Shanghai, 201805, China
| | - Wang Lin-Hui
- Department of Urology, Changzheng Hospital of Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, China.
| | - Cui Xin-Gang
- Department of Urology, The Third Affiliated Hospital of Second Military Medical University, 700 North Moyu Road, Shanghai, 201805, China. .,Department of Urology, Xinhua Hospital, Shanghai Jiaotong University, School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China.
| |
Collapse
|
54
|
Lindström NO, Sealfon R, Chen X, Parvez RK, Ransick A, De Sena Brandine G, Guo J, Hill B, Tran T, Kim AD, Zhou J, Tadych A, Watters A, Wong A, Lovero E, Grubbs BH, Thornton ME, McMahon JA, Smith AD, Ruffins SW, Armit C, Troyanskaya OG, McMahon AP. Spatial transcriptional mapping of the human nephrogenic program. Dev Cell 2021; 56:2381-2398.e6. [PMID: 34428401 PMCID: PMC8396064 DOI: 10.1016/j.devcel.2021.07.017] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 05/06/2021] [Accepted: 07/27/2021] [Indexed: 12/11/2022]
Abstract
Congenital abnormalities of the kidney and urinary tract are among the most common birth defects, affecting 3% of newborns. The human kidney forms around a million nephrons from a pool of nephron progenitors over a 30-week period of development. To establish a framework for human nephrogenesis, we spatially resolved a stereotypical process by which equipotent nephron progenitors generate a nephron anlage, then applied data-driven approaches to construct three-dimensional protein maps on anatomical models of the nephrogenic program. Single-cell RNA sequencing identified progenitor states, which were spatially mapped to the nephron anatomy, enabling the generation of functional gene networks predicting interactions within and between nephron cell types. Network mining identified known developmental disease genes and predicted targets of interest. The spatially resolved nephrogenic program made available through the Human Nephrogenesis Atlas (https://sckidney.flatironinstitute.org/) will facilitate an understanding of kidney development and disease and enhance efforts to generate new kidney structures.
Collapse
Affiliation(s)
- Nils O Lindström
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Rachel Sealfon
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, NY, USA; Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Xi Chen
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, NY, USA; Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Riana K Parvez
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Andrew Ransick
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Guilherme De Sena Brandine
- Molecular and Computational Biology, Division of Biological Sciences, University of Southern, Los Angeles, CA 90089, USA
| | - Jinjin Guo
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Bill Hill
- MRC Human Genetics Unit, MRC Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Tracy Tran
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Albert D Kim
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jian Zhou
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, NY, USA; Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Alicja Tadych
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Aaron Watters
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, NY, USA
| | - Aaron Wong
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, NY, USA
| | - Elizabeth Lovero
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, NY, USA
| | - Brendan H Grubbs
- Maternal Fetal Medicine Division, Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Matthew E Thornton
- Maternal Fetal Medicine Division, Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jill A McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Andrew D Smith
- Molecular and Computational Biology, Division of Biological Sciences, University of Southern, Los Angeles, CA 90089, USA
| | - Seth W Ruffins
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Chris Armit
- MRC Human Genetics Unit, MRC Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK; BGI Hong Kong, 26/F, Kings Wing Plaza 2, 1 On Kwan Street, Shek Mun, NT, Hong Kong
| | - Olga G Troyanskaya
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, NY, USA; Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA; Department of Computer Science, Princeton University, Princeton, NJ, USA.
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
55
|
Frazier KS. The Impact of Functional and Structural Maturation of the Kidney on Susceptibility to Drug and Chemical Toxicity in Neonatal Rodents. Toxicol Pathol 2021; 49:1377-1388. [PMID: 34338059 DOI: 10.1177/01926233211035683] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Drug responses are often unpredictable in juvenile animal toxicity studies; hence, optimizing dosages is challenging. Renal functional differences based on age of development will often result in vastly different toxicologic responses. Developmental changes in renal function can alter plasma clearance of compounds with extensive renal elimination. Absorption, distribution, metabolism, and excretion of drugs vary depending on animal age and kidney maturation. Toxicity can result in malformations or renal degeneration. Although renal morphologic development in humans generally occurs in utero, maximal levels of tubular secretion, acid-base equilibrium, concentrating ability, or glomerular filtration rate (GFR) are reached postnatally in humans and animals and subject to drug effects. Maturation of renal metabolism and transporters occurs postnatally and plays a critical role in detoxification and excretion. Maturation times must be considered when designing juvenile toxicity studies and may require cohorts of animals of specific ages to achieve optimal dosing schemes and toxicokinetics. In recent years, critical end points and windows of susceptibility have been established comparatively between species to better model pharmacokinetics and understand pediatric nephrotoxicity. There are examples of agents where toxicity is enhanced in neonates, others where it is diminished, and others where rat nephrotoxicity is expressed as juvenile toxicity, but in humans as gestational toxicity.
Collapse
|
56
|
Tortelote GG, Colón-Leyva M, Saifudeen Z. Metabolic programming of nephron progenitor cell fate. Pediatr Nephrol 2021; 36:2155-2164. [PMID: 33089379 PMCID: PMC10734399 DOI: 10.1007/s00467-020-04752-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/30/2020] [Accepted: 08/31/2020] [Indexed: 11/28/2022]
Abstract
Metabolic pathways are one of the first responses at the cellular level to maternal/fetal interface stressors. Studies have revealed the previously unrecognized contributions of intermediary metabolism to developmental programs. Here, we provide an overview of cellular metabolic pathways and the cues that modulate metabolic states. We discuss the developmental and physiological implications of metabolic reprogramming and the key role of metabolites in epigenetic and epiproteomic modifications during embryonic development and with respect to kidney development and nephrogenesis.
Collapse
Affiliation(s)
- Giovane G Tortelote
- Department of Pediatrics, Tulane University School of Medicine, 1430 Tulane Avenue SL37, Room 5534, New Orleans, LA, 70112, USA
| | - Mariel Colón-Leyva
- Department of Pediatrics, Tulane University School of Medicine, 1430 Tulane Avenue SL37, Room 5534, New Orleans, LA, 70112, USA
| | - Zubaida Saifudeen
- Department of Pediatrics, Tulane University School of Medicine, 1430 Tulane Avenue SL37, Room 5534, New Orleans, LA, 70112, USA.
| |
Collapse
|
57
|
Makayes Y, Resnick E, Hinden L, Aizenshtein E, Shlomi T, Kopan R, Nechama M, Volovelsky O. Increasing mTORC1 Pathway Activity or Methionine Supplementation during Pregnancy Reverses the Negative Effect of Maternal Malnutrition on the Developing Kidney. J Am Soc Nephrol 2021; 32:1898-1912. [PMID: 33958489 PMCID: PMC8455268 DOI: 10.1681/asn.2020091321] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 03/01/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Low nephron number at birth is associated with a high risk of CKD in adulthood because nephrogenesis is completed in utero. Poor intrauterine environment impairs nephron endowment via an undefined molecular mechanism. A calorie-restricted diet (CRD) mouse model examined the effect of malnutrition during pregnancy on nephron progenitor cells (NPCs). METHODS Daily caloric intake was reduced by 30% during pregnancy. mRNA expression, the cell cycle, and metabolic activity were evaluated in sorted Six2 NPCs. The results were validated using transgenic mice, oral nutrient supplementation, and organ cultures. RESULTS Maternal CRD is associated with low nephron number in offspring, compromising kidney function at an older age. RNA-seq identified cell cycle regulators and the mTORC1 pathway, among other pathways, that maternal malnutrition in NPCs modifies. Metabolomics analysis of NPCs singled out the methionine pathway as crucial for NPC proliferation and maintenance. Methionine deprivation reduced NPC proliferation and lowered NPC number per tip in embryonic kidney cultures, with rescue from methionine metabolite supplementation. Importantly, in vivo, the negative effect of caloric restriction on nephrogenesis was prevented by adding methionine to the otherwise restricted diet during pregnancy or by removing one Tsc1 allele in NPCs. CONCLUSIONS These findings show that mTORC1 signaling and methionine metabolism are central to the cellular and metabolic effects of malnutrition during pregnancy on NPCs, contributing to nephrogenesis and later, to kidney health in adulthood.
Collapse
Affiliation(s)
- Yaniv Makayes
- Pediatric Nephrology Unit and Research Lab, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Elad Resnick
- Pediatric Nephrology Unit and Research Lab, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Liad Hinden
- Faculty of Medicine, School of Pharmacy, Institute for Drug Research, The Hebrew University, Jerusalem, Israel
| | | | | | - Raphael Kopan
- Division of Developmental Biology, Department of Pediatrics, University of Cincinnati College of Medicine and Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Morris Nechama
- Pediatric Nephrology Unit and Research Lab, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
- Wohl’s Translation Research Institute at Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Oded Volovelsky
- Pediatric Nephrology Unit and Research Lab, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
- Wohl’s Translation Research Institute at Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
58
|
Lang C, Conrad L, Iber D. Organ-Specific Branching Morphogenesis. Front Cell Dev Biol 2021; 9:671402. [PMID: 34150767 PMCID: PMC8212048 DOI: 10.3389/fcell.2021.671402] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 05/06/2021] [Indexed: 01/09/2023] Open
Abstract
A common developmental process, called branching morphogenesis, generates the epithelial trees in a variety of organs, including the lungs, kidneys, and glands. How branching morphogenesis can create epithelial architectures of very different shapes and functions remains elusive. In this review, we compare branching morphogenesis and its regulation in lungs and kidneys and discuss the role of signaling pathways, the mesenchyme, the extracellular matrix, and the cytoskeleton as potential organ-specific determinants of branch position, orientation, and shape. Identifying the determinants of branch and organ shape and their adaptation in different organs may reveal how a highly conserved developmental process can be adapted to different structural and functional frameworks and should provide important insights into epithelial morphogenesis and developmental disorders.
Collapse
Affiliation(s)
- Christine Lang
- Department of Biosystems, Science and Engineering, ETH Zürich, Basel, Switzerland.,Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Lisa Conrad
- Department of Biosystems, Science and Engineering, ETH Zürich, Basel, Switzerland.,Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Dagmar Iber
- Department of Biosystems, Science and Engineering, ETH Zürich, Basel, Switzerland.,Swiss Institute of Bioinformatics, Basel, Switzerland
| |
Collapse
|
59
|
Puelles VG, Combes AN, Bertram JF. Clearly imaging and quantifying the kidney in 3D. Kidney Int 2021; 100:780-786. [PMID: 34089762 DOI: 10.1016/j.kint.2021.04.042] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 04/09/2021] [Accepted: 04/14/2021] [Indexed: 12/16/2022]
Abstract
For decades, measurements of kidney microanatomy using 2-dimensional sections has provided us with a detailed knowledge of kidney morphology under physiological and pathological conditions. However, the rapid development of tissue clearing methods in recent years, in combination with the development of novel 3-dimensional imaging modalities have provided new insights into kidney structure and function. This review article describes a range of novel insights into kidney development and disease obtained recently using these new methodological approaches. For example, in the developing kidney these approaches have provided new understandings of ureteric branching morphogenesis, nephron progenitor cell proliferation and commitment, interactions between ureteric tip cells and nephron progenitor cells, and the establishment of nephron segmentation. In whole adult mouse kidneys, tissue clearing combined with light sheet microscopy can image and quantify the total number of glomeruli, a major breakthrough in the field. Similar approaches have provided new insights into the structure of the renal vasculature and innervation, tubulointerstitial remodeling, podocyte loss and hypertrophy, cyst formation, the evolution of cellular crescents, and the structure of the glomerular filtration barrier. Many more advances in the understanding of kidney biology and pathology can be expected as additional clearing and imaging techniques are developed and adopted by more investigators.
Collapse
Affiliation(s)
- Victor G Puelles
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Anatomy and Developmental Biology, and Stem Cells and Development Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Alexander N Combes
- Department of Anatomy and Developmental Biology, and Stem Cells and Development Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - John F Bertram
- Department of Anatomy and Developmental Biology, and Stem Cells and Development Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia.
| |
Collapse
|
60
|
Little MH. Returning to kidney development to deliver synthetic kidneys. Dev Biol 2021; 474:22-36. [PMID: 33333068 PMCID: PMC8052282 DOI: 10.1016/j.ydbio.2020.12.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 12/09/2020] [Indexed: 12/27/2022]
Abstract
There is no doubt that the development of transplantable synthetic kidneys could improve the outcome for the many millions of people worldwide suffering from chronic kidney disease. Substantial progress has been made in the last 6 years in the generation of kidney tissue from stem cells. However, the limited scale, incomplete cellular complexity and functional immaturity of such structures suggests we are some way from this goal. While developmental biology has successfully guided advances to date, these human kidney models are limited in their capacity for ongoing nephrogenesis and lack corticomedullary definition, a unified vasculature and a coordinated exit path for urinary filtrate. This review will reassess our developmental understanding of how the mammalian embryo manages to create kidneys, how this has informed our progress to date and how both engineering and developmental biology can continue to guide us towards a synthetic kidney.
Collapse
Affiliation(s)
- Melissa H Little
- Murdoch Children's Research Institute, Flemington Rd, Parkville, VIC, Australia; Department of Paediatrics, The University of Melbourne, VIC, Australia; Department of Anatomy and Neuroscience, The University of Melbourne, VIC, Australia.
| |
Collapse
|
61
|
Li H, Kurtzeborn K, Kupari J, Gui Y, Siefker E, Lu B, Mätlik K, Olfat S, Montaño-Rodríguez AR, Huh SH, Costantini F, Andressoo JO, Kuure S. Postnatal prolongation of mammalian nephrogenesis by excess fetal GDNF. Development 2021; 148:268366. [PMID: 34032268 PMCID: PMC8180252 DOI: 10.1242/dev.197475] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 04/26/2021] [Indexed: 01/21/2023]
Abstract
Nephron endowment, defined during the fetal period, dictates renal and related cardiovascular health throughout life. We show here that, despite its negative effects on kidney growth, genetic increase of GDNF prolongs the nephrogenic program beyond its normal cessation. Multi-stage mechanistic analysis revealed that excess GDNF maintains nephron progenitors and nephrogenesis through increased expression of its secreted targets and augmented WNT signaling, leading to a two-part effect on nephron progenitor maintenance. Abnormally high GDNF in embryonic kidneys upregulates its known targets but also Wnt9b and Axin2, with concomitant deceleration of nephron progenitor proliferation. Decline of GDNF levels in postnatal kidneys normalizes the ureteric bud and creates a permissive environment for continuation of the nephrogenic program, as demonstrated by morphologically and molecularly normal postnatal nephron progenitor self-renewal and differentiation. These results establish that excess GDNF has a bi-phasic effect on nephron progenitors in mice, which can faithfully respond to GDNF dosage manipulation during the fetal and postnatal period. Our results suggest that sensing the signaling activity level is an important mechanism through which GDNF and other molecules contribute to nephron progenitor lifespan specification. Summary: Dosage of neurotropic factor GDNF regulates nephron progenitors and in utero growth factor augmentation can extend postnatal lifespan and differentiation of nephron progenitors.
Collapse
Affiliation(s)
- Hao Li
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland.,Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland
| | - Kristen Kurtzeborn
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland.,Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland
| | - Jussi Kupari
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Yujuan Gui
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Edward Siefker
- Department of Developmental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68105, USA
| | - Benson Lu
- Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032, USA
| | - Kärt Mätlik
- Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland.,Department of Pharmacology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Soophie Olfat
- Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland.,Department of Pharmacology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Ana R Montaño-Rodríguez
- Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland.,Department of Pharmacology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Sung-Ho Huh
- Department of Developmental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68105, USA
| | - Franklin Costantini
- Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032, USA
| | - Jaan-Olle Andressoo
- Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland.,Department of Pharmacology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland.,Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Satu Kuure
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland.,GM-unit, Laboratory Animal Centre, Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland
| |
Collapse
|
62
|
Affiliation(s)
- Ian M Smyth
- Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Melbourne, Australia
| |
Collapse
|
63
|
Packard A, Klein WH, Costantini F. Ret signaling in ureteric bud epithelial cells controls cell movements, cell clustering and bud formation. Development 2021; 148:261695. [PMID: 33914865 DOI: 10.1242/dev.199386] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/31/2021] [Indexed: 11/20/2022]
Abstract
Ret signaling promotes branching morphogenesis during kidney development, but the underlying cellular mechanisms remain unclear. While Ret-expressing progenitor cells proliferate at the ureteric bud tips, some of these cells exit the tips to generate the elongating collecting ducts, and in the process turn off Ret. Genetic ablation of Ret in tip cells promotes their exit, suggesting that Ret is required for cell rearrangements that maintain the tip compartments. Here, we examine the behaviors of ureteric bud cells that are genetically forced to maintain Ret expression. These cells move to the nascent tips, and remain there during many cycles of branching; this tip-seeking behavior may require positional signals from the mesenchyme, as it occurs in whole kidneys but not in epithelial ureteric bud organoids. In organoids, cells forced to express Ret display a striking self-organizing behavior, attracting each other to form dense clusters within the epithelium, which then evaginate to form new buds. The ability of forced Ret expression to promote these events suggests that similar Ret-dependent cell behaviors play an important role in normal branching morphogenesis.
Collapse
Affiliation(s)
- Adam Packard
- Department of Genetics and Development, Columbia University, New York, NY 10032, USA
| | - William H Klein
- Department of Genetics and Development, Columbia University, New York, NY 10032, USA.,Department of Systems Biology, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Frank Costantini
- Department of Genetics and Development, Columbia University, New York, NY 10032, USA
| |
Collapse
|
64
|
Shimizu T, Yamagata K, Osafune K. Kidney organoids: Research in developmental biology and emerging applications. Dev Growth Differ 2021; 63:166-177. [PMID: 33569792 DOI: 10.1111/dgd.12714] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 12/11/2022]
Abstract
Kidney organoids generated from human pluripotent stem cells (hPSCs) have drastically changed the field of stem cell research on human kidneys within a few years. They are self-organizing multicellular structures that contain nephron components such as glomeruli and renal tubules in most cases, but hPSC-derived ureteric buds, the progenitors of collecting ducts and ureters, can also form three-dimensional organoids. Today's challenges facing human kidney organoids are further maturation and anatomical integrity in order to achieve a complete model of the developing kidneys and ultimately a complete adult organ. Since chronic kidney disease (CKD) and impaired kidney function are an increasing burden on public health worldwide, there is an urgent need to develop effective treatments for various renal conditions. In this regard, hPSC-derived kidney organoids may impact medicine by providing new translational approaches. The unique ability of kidney organoids derived from disease-specific hPSCs to reproduce human diseases caused by genetic alterations may help provide the next generation of kidney disease models. Recent advances in the field of kidney organoid research have been generally accompanied by progress in developmental biology and other technological breakthroughs. In this review, we consider the current trends in kidney organoid technology, especially focusing on the relationship to the study of human kidney development, and discuss the remaining hurdles and prospects in regenerating human kidney structures beyond organoids.
Collapse
Affiliation(s)
- Tatsuya Shimizu
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan.,Department of Nephrology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Kunihiro Yamagata
- Department of Nephrology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Kenji Osafune
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| |
Collapse
|
65
|
Abstract
The kidney plays an integral role in filtering the blood-removing metabolic by-products from the body and regulating blood pressure. This requires the establishment of large numbers of efficient and specialized blood filtering units (nephrons) that incorporate a system for vascular exchange and nutrient reabsorption as well as a collecting duct system to remove waste (urine) from the body. Kidney development is a dynamic process which generates these structures through a delicately balanced program of self-renewal and commitment of nephron progenitor cells that inhabit a constantly evolving cellular niche at the tips of a branching ureteric "tree." The former cells build the nephrons and the latter the collecting duct system. Maintaining these processes across fetal development is critical for establishing the normal "endowment" of nephrons in the kidney and perturbations to this process are associated both with mutations in integral genes and with alterations to the fetal environment.
Collapse
Affiliation(s)
- Ian M Smyth
- Department of Anatomy and Developmental Biology, Department of Biochemistry and Molecular Biology, Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
66
|
Papakrivopoulou E, Jafree DJ, Dean CH, Long DA. The Biological Significance and Implications of Planar Cell Polarity for Nephrology. Front Physiol 2021; 12:599529. [PMID: 33716764 PMCID: PMC7952641 DOI: 10.3389/fphys.2021.599529] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 02/01/2021] [Indexed: 11/13/2022] Open
Abstract
The orientation of cells in two-dimensional and three-dimensional space underpins how the kidney develops and responds to disease. The process by which cells orientate themselves within the plane of a tissue is termed planar cell polarity. In this Review, we discuss how planar cell polarity and the proteins that underpin it govern kidney organogenesis and pathology. The importance of planar cell polarity and its constituent proteins in multiple facets of kidney development is emphasised, including ureteric bud branching, tubular morphogenesis and nephron maturation. An overview is given of the relevance of planar cell polarity and its proteins for inherited human renal diseases, including congenital malformations with unknown aetiology and polycystic kidney disease. Finally, recent work is described outlining the influence of planar cell polarity proteins on glomerular diseases and highlight how this fundamental pathway could yield a new treatment paradigm for nephrology.
Collapse
Affiliation(s)
- Eugenia Papakrivopoulou
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, United Kingdom.,Department of Internal Medicine and Nephrology, Clinique Saint Jean, Brussels, Belgium
| | - Daniyal J Jafree
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, United Kingdom.,UCL MB/Ph.D. Programme, Faculty of Medical Science, University College London, London, United Kingdom
| | - Charlotte H Dean
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - David A Long
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| |
Collapse
|
67
|
Cai W, Wang Y, Zhang J, Zhang H, Luo T. Multi-scale simulation of early kidney branching morphogenesis. Phys Biol 2021; 18:026005. [PMID: 33395673 DOI: 10.1088/1478-3975/abd844] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
An important feature of the branch morphogenesis during kidney development is the termination of the tips on the outer surface of a kidney. This feature requires the avoidance of the intersection between the tips and existing ducts inside the kidney. Here, we started from a continuous model and implemented the coarse grained rules into a fast and discrete simulations. The ligand-receptor-based Turing mechanism suggests a repulsion that decreases exponentially with distance between interacting branches, preventing the intersection between neighboring branches. We considered this repulsive effect in numerical simulations and successfully reproduce the key features of the experimentally observed branch morphology for an E15.5 kidney. We examine the similarity of several geometrical parameters between the simulation results and experimental observations. The good agreement between the simulations and experiments suggests that the concentration decay caused by the absorption of glial cell line derived neurotrophic factor might be the key factor to affect the geometry in early kidney development.
Collapse
Affiliation(s)
- Wenran Cai
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, People's Republic of China
| | | | | | | | | |
Collapse
|
68
|
Li H, Hohenstein P, Kuure S. Embryonic Kidney Development, Stem Cells and the Origin of Wilms Tumor. Genes (Basel) 2021; 12:genes12020318. [PMID: 33672414 PMCID: PMC7926385 DOI: 10.3390/genes12020318] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/17/2021] [Accepted: 02/18/2021] [Indexed: 12/23/2022] Open
Abstract
The adult mammalian kidney is a poorly regenerating organ that lacks the stem cells that could replenish functional homeostasis similarly to, e.g., skin or the hematopoietic system. Unlike a mature kidney, the embryonic kidney hosts at least three types of lineage-specific stem cells that give rise to (a) a ureter and collecting duct system, (b) nephrons, and (c) mesangial cells together with connective tissue of the stroma. Extensive interest has been raised towards these embryonic progenitor cells, which are normally lost before birth in humans but remain part of the undifferentiated nephrogenic rests in the pediatric renal cancer Wilms tumor. Here, we discuss the current understanding of kidney-specific embryonic progenitor regulation in the innate environment of the developing kidney and the types of disruptions in their balanced regulation that lead to the formation of Wilms tumor.
Collapse
Affiliation(s)
- Hao Li
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, FIN-00014 Helsinki, Finland;
| | - Peter Hohenstein
- Department of Human Genetics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
| | - Satu Kuure
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, FIN-00014 Helsinki, Finland;
- GM-Unit, Laboratory Animal Center, Helsinki Institute of Life Science, University of Helsinki, FIN-00014 Helsinki, Finland
- Correspondence: ; Tel.: +358-2941-59395
| |
Collapse
|
69
|
Guo Q, Kim A, Li B, Ransick A, Bugacov H, Chen X, Lindström N, Brown A, Oxburgh L, Ren B, McMahon AP. A β-catenin-driven switch in TCF/LEF transcription factor binding to DNA target sites promotes commitment of mammalian nephron progenitor cells. eLife 2021; 10:64444. [PMID: 33587034 PMCID: PMC7924951 DOI: 10.7554/elife.64444] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 02/14/2021] [Indexed: 12/30/2022] Open
Abstract
The canonical Wnt pathway transcriptional co-activator β-catenin regulates self-renewal and differentiation of mammalian nephron progenitor cells (NPCs). We modulated β-catenin levels in NPC cultures using the GSK3 inhibitor CHIR99021 (CHIR) to examine opposing developmental actions of β-catenin. Low CHIR-mediated maintenance and expansion of NPCs are independent of direct engagement of TCF/LEF/β-catenin transcriptional complexes at low CHIR-dependent cell-cycle targets. In contrast, in high CHIR, TCF7/LEF1/β-catenin complexes replaced TCF7L1/TCF7L2 binding on enhancers of differentiation-promoting target genes. Chromosome confirmation studies showed pre-established promoter–enhancer connections to these target genes in NPCs. High CHIR-associated de novo looping was observed in positive transcriptional feedback regulation to the canonical Wnt pathway. Thus, β-catenin’s direct transcriptional role is restricted to the induction of NPCs, where rising β-catenin levels switch inhibitory TCF7L1/TCF7L2 complexes to activating LEF1/TCF7 complexes at primed gene targets poised for rapid initiation of a nephrogenic program.
Collapse
Affiliation(s)
- Qiuyu Guo
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, United States
| | - Albert Kim
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, United States
| | - Bin Li
- The Rogosin Institute, New York, United States
| | - Andrew Ransick
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, United States
| | - Helena Bugacov
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, United States
| | - Xi Chen
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, United States
| | - Nils Lindström
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, United States
| | - Aaron Brown
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, United States
| | | | - Bing Ren
- Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, Institute of Genomic Medicine, Moores Cancer Center, University of California San Diego, San Diego, United States
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, United States
| |
Collapse
|
70
|
Meng P, Zhu M, Ling X, Zhou L. Wnt signaling in kidney: the initiator or terminator? J Mol Med (Berl) 2020; 98:1511-1523. [PMID: 32939578 PMCID: PMC7591426 DOI: 10.1007/s00109-020-01978-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 08/14/2020] [Accepted: 09/08/2020] [Indexed: 12/14/2022]
Abstract
The kidney is a key organ in the human body that excretes toxins and sustains the water-electrolyte balance. During embryonic development and disease progression, the kidney undergoes enormous changes in macrostructure, accompanied by a variety of microstructural histological changes, such as glomerular formation and sclerosis, tubule elongation and atrophy, interstitial establishment, and fibrosis progression. All of these rely on the frequent occurrence of cell death and growth. Notably, to overcome disease, some cells regenerate through self-repair or progenitor cell differentiation. However, the signaling mechanisms underlying kidney development and regeneration have not been elucidated. Recently, Wnt signaling has been noted to play an important role. Although it is a well-known developmental signal, the role of Wnt signaling in kidney development and regeneration is not well recognized. In this review, we review the role of Wnt signaling in kidney embryonic development, tissue repair, cell division, and progenitor cell differentiation after injury. Moreover, we briefly highlight advances in our understanding of the pathogenic mechanisms of Wnt signaling in mediating cellular senescence in kidney parenchymal and stem cells, an irreversible arrest of cell proliferation blocking tissue repair and regeneration. We also highlight the therapeutic targets of Wnt signaling in kidney diseases and provide important clues for clinical strategies.
Collapse
Affiliation(s)
- Ping Meng
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, China
- Department of Nephrology, Huadu District People's Hospital, Southern Medical University, Guangzhou, China
| | - Mingsheng Zhu
- Department of Nephrology, The People's Hospital of Gaozhou, Maoming, China
| | - Xian Ling
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, China
| | - Lili Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, China.
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China.
| |
Collapse
|
71
|
Bednarz A, Lipiński P, Starzyński RR, Tomczyk M, Kraszewska I, Herman S, Kowalski K, Gruca E, Jończy A, Mazgaj R, Szudzik M, Rajfur Z, Baster Z, Józkowicz A, Lenartowicz M. Exacerbation of Neonatal Hemolysis and Impaired Renal Iron Handling in Heme Oxygenase 1-Deficient Mice. Int J Mol Sci 2020; 21:ijms21207754. [PMID: 33092142 PMCID: PMC7589678 DOI: 10.3390/ijms21207754] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/15/2020] [Accepted: 10/16/2020] [Indexed: 01/03/2023] Open
Abstract
In most mammals, neonatal intravascular hemolysis is a benign and moderate disorder that usually does not lead to anemia. During the neonatal period, kidneys play a key role in detoxification and recirculation of iron species released from red blood cells (RBC) and filtered out by glomeruli to the primary urine. Activity of heme oxygenase 1 (HO1), a heme-degrading enzyme localized in epithelial cells of proximal tubules, seems to be of critical importance for both processes. We show that, in HO1 knockout mouse newborns, hemolysis was prolonged despite a transient state and exacerbated, which led to temporal deterioration of RBC status. In neonates lacking HO1, functioning of renal molecular machinery responsible for iron reabsorption from the primary urine (megalin/cubilin complex) and its transfer to the blood (ferroportin) was either shifted in time or impaired, respectively. Those abnormalities resulted in iron loss from the body (excreted in urine) and in iron retention in the renal epithelium. We postulate that, as a consequence of these abnormalities, a tight systemic iron balance of HO1 knockout neonates may be temporarily affected.
Collapse
Affiliation(s)
- Aleksandra Bednarz
- Department of Genetics and Evolution, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387 Kraków, Poland; (A.B.); (S.H.); (K.K.); (E.G.)
| | - Paweł Lipiński
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Jastrzębiec, 05-552 Magdalenka, Poland; (P.L.); (R.R.S.); (A.J.); (R.M.); (M.S.)
| | - Rafał R. Starzyński
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Jastrzębiec, 05-552 Magdalenka, Poland; (P.L.); (R.R.S.); (A.J.); (R.M.); (M.S.)
| | - Mateusz Tomczyk
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; (M.T.); (I.K.); (A.J.)
| | - Izabela Kraszewska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; (M.T.); (I.K.); (A.J.)
| | - Sylwia Herman
- Department of Genetics and Evolution, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387 Kraków, Poland; (A.B.); (S.H.); (K.K.); (E.G.)
| | - Kacper Kowalski
- Department of Genetics and Evolution, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387 Kraków, Poland; (A.B.); (S.H.); (K.K.); (E.G.)
| | - Ewelina Gruca
- Department of Genetics and Evolution, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387 Kraków, Poland; (A.B.); (S.H.); (K.K.); (E.G.)
| | - Aneta Jończy
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Jastrzębiec, 05-552 Magdalenka, Poland; (P.L.); (R.R.S.); (A.J.); (R.M.); (M.S.)
| | - Rafał Mazgaj
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Jastrzębiec, 05-552 Magdalenka, Poland; (P.L.); (R.R.S.); (A.J.); (R.M.); (M.S.)
| | - Mateusz Szudzik
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Jastrzębiec, 05-552 Magdalenka, Poland; (P.L.); (R.R.S.); (A.J.); (R.M.); (M.S.)
| | - Zenon Rajfur
- Department of Molecular and Interfacial Biophysics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Łojasiewicza 11, 30-348 Kraków, Poland; (Z.R.); (Z.B.)
| | - Zbigniew Baster
- Department of Molecular and Interfacial Biophysics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Łojasiewicza 11, 30-348 Kraków, Poland; (Z.R.); (Z.B.)
| | - Alicja Józkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland; (M.T.); (I.K.); (A.J.)
| | - Małgorzata Lenartowicz
- Department of Genetics and Evolution, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387 Kraków, Poland; (A.B.); (S.H.); (K.K.); (E.G.)
- Correspondence:
| |
Collapse
|
72
|
Yu W, Marshall WF, Metzger RJ, Brakeman PR, Morsut L, Lim W, Mostov KE. Simple Rules Determine Distinct Patterns of Branching Morphogenesis. Cell Syst 2020; 9:221-227. [PMID: 31557453 DOI: 10.1016/j.cels.2019.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 05/09/2019] [Accepted: 07/31/2019] [Indexed: 10/25/2022]
Abstract
Many metazoan organs are comprised of branching trees of epithelial tubes; how patterning occurs in these trees is a fundamental problem of development. Commonly, branch tips fill the volume of the organ approximately uniformly, e.g., in mammalian lung, airway branch tips are dispersed roughly uniformly throughout the volume of the lung. In contrast, in the developing metanephric kidney, the tips of the ureteric bud tree are located close to the outer surface of the kidney rather than filling the kidney. Here, we describe a simple alteration in the branching rules that accounts for the difference between the kidney pattern that leads to tips near the organ surface versus previously known patterns that lead to the branch tips being dispersed throughout the organ. We further use a simple toy model to deduce from first principles how this rule change accounts for the differences in the two types of trees.
Collapse
Affiliation(s)
- Wei Yu
- Departments of Anatomy, University of California San Francisco, San Francisco, CA, USA; Cell and Molecular Pharmacology, University of California San Francisco, San Francisco, CA, USA
| | - Wallace F Marshall
- Center for Cellular Construction, University of California San Francisco, San Francisco, CA, USA; Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
| | - Ross J Metzger
- Departments of Anatomy, University of California San Francisco, San Francisco, CA, USA; Department of Pediatrics (Cardiology), Stanford University School of Medicine, Stanford, CA, USA
| | - Paul R Brakeman
- Department of Pediatrics (Nephrology), University of California San Francisco, San Francisco, CA, USA
| | - Leonardo Morsut
- Cell and Molecular Pharmacology, University of California San Francisco, San Francisco, CA, USA
| | - Wendell Lim
- Cell and Molecular Pharmacology, University of California San Francisco, San Francisco, CA, USA; Center for Cellular Construction, University of California San Francisco, San Francisco, CA, USA
| | - Keith E Mostov
- Departments of Anatomy, University of California San Francisco, San Francisco, CA, USA; Center for Cellular Construction, University of California San Francisco, San Francisco, CA, USA; Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
73
|
El Andalousi J, Khairallah H, Zhuang Y, Ryan AK, Gupta IR. Role of Claudins in Renal Branching Morphogenesis. Physiol Rep 2020; 8:e14492. [PMID: 32975899 PMCID: PMC7518295 DOI: 10.14814/phy2.14492] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 05/25/2020] [Accepted: 05/26/2020] [Indexed: 11/26/2022] Open
Abstract
Claudins are a family of tight junction proteins that are expressed during mouse kidney development. They regulate paracellular transport of solutes along the nephron and contribute to the final composition of the urinary filtrate. To understand their roles during development, we used a protein reagent, a truncated version of the Clostridium perfringens enterotoxin (C-CPE), to specifically remove a subset of claudin family members from mouse embryonic kidney explants at embryonic day 12. We observed that treatment with C-CPE decreased the number and the complexity of ureteric bud tips that formed: there were more single and less bifid ureteric bud tips when compared to control-treated explants. In addition, C-CPE-treated explants exhibited ureteric bud tips with larger lumens when compared to control explants (p < .05). Immunofluorescent analysis revealed decreased expression and localization of Claudin-3, -4, -6, and -8 to tight junctions of ureteric bud tips following treatment with C-CPE. Interestingly, Claudin-7 showed higher expression in the basolateral membrane of the ureteric bud lineage and poor localization to the tight junctions of the ureteric bud lineage both in controls and in C-CPE-treated explants. Taken together, it appears that claudin proteins may play a role in ureteric bud branching morphogenesis through changes in lumen formation that may affect the efficiency by which ureteric buds emerge and branch.
Collapse
Affiliation(s)
- Jasmine El Andalousi
- Research Institute of McGill University Health CentreMontreal Children's HospitalMontréalQuébecCanada
| | - Halim Khairallah
- Department of Human GeneticsMcGill UniversityMontréalQuébecCanada
| | - Yuan Zhuang
- Department of Human GeneticsMcGill UniversityMontréalQuébecCanada
| | - Aimee K. Ryan
- Department of Human GeneticsMcGill UniversityMontréalQuébecCanada
- Department of PediatricsMontreal Children's HospitalMcGill UniversityMontréalQuébecCanada
| | - Indra R. Gupta
- Department of Human GeneticsMcGill UniversityMontréalQuébecCanada
- Department of PediatricsMontreal Children's HospitalMcGill UniversityMontréalQuébecCanada
| |
Collapse
|
74
|
Short KM, Smyth IM. Branching morphogenesis as a driver of renal development. Anat Rec (Hoboken) 2020; 303:2578-2587. [PMID: 32790143 DOI: 10.1002/ar.24486] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/17/2019] [Accepted: 11/01/2019] [Indexed: 12/16/2022]
Abstract
Branching morphogenesis is an integral developmental mechanism central to the formation of a range of organs including the kidney, lung, pancreas and mammary gland. The ramified networks of epithelial tubules it establishes are critical for the processes of secretion, excretion and exchange mediated by these tissues. In the kidney, branching serves to establish the collecting duct system that transports urine from the nephrons into the renal pelvis, ureter and finally the bladder. Generally speaking, the formation of these networks in different organs begins with the specification and differentiation of simple bud-like organ anlage, which then undergo a process of elaboration, typically by bifurcation. This process is often governed by the interaction of progenitor cells at the tips of the epithelia with neighboring mesenchymal cell populations which direct the branching process and which often themselves differentiate to form part of the adult organ. In the kidney, the tips of ureteric bud elaborate through a dynamic cell signaling relationship with overlying nephron progenitor cell populations. These cells sequentially commit to differentiation and the resulting nephrons reintegrate with the ureteric epithelium as development progresses. This review will describe recent advances in understanding the how the elaboration of the ureteric bud is patterned and consider the extent to which this process is shared with other organs.
Collapse
Affiliation(s)
- Kieran M Short
- Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Ian M Smyth
- Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
75
|
Rens EG, Zeegers MT, Rabbers I, Szabó A, Merks RMH. Autocrine inhibition of cell motility can drive epithelial branching morphogenesis in the absence of growth. Philos Trans R Soc Lond B Biol Sci 2020; 375:20190386. [PMID: 32713299 DOI: 10.1098/rstb.2019.0386] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Epithelial branching morphogenesis drives the development of organs such as the lung, salivary gland, kidney and the mammary gland. It involves cell proliferation, cell differentiation and cell migration. An elaborate network of chemical and mechanical signals between the epithelium and the surrounding mesenchymal tissues regulates the formation and growth of branching organs. Surprisingly, when cultured in isolation from mesenchymal tissues, many epithelial tissues retain the ability to exhibit branching morphogenesis even in the absence of proliferation. In this work, we propose a simple, experimentally plausible mechanism that can drive branching morphogenesis in the absence of proliferation and cross-talk with the surrounding mesenchymal tissue. The assumptions of our mathematical model derive from in vitro observations of the behaviour of mammary epithelial cells. These data show that autocrine secretion of the growth factor TGF[Formula: see text]1 inhibits the formation of cell protrusions, leading to curvature-dependent inhibition of sprouting. Our hybrid cellular Potts and partial-differential equation model correctly reproduces the experimentally observed tissue-geometry-dependent determination of the sites of branching, and it suffices for the formation of self-avoiding branching structures in the absence and also in the presence of cell proliferation. This article is part of the theme issue 'Multi-scale analysis and modelling of collective migration in biological systems'.
Collapse
Affiliation(s)
- Elisabeth G Rens
- Centrum Wiskunde and Informatica, Amsterdam, The Netherlands.,Mathematical Institute, Leiden University, Leiden, The Netherlands
| | - Mathé T Zeegers
- Centrum Wiskunde and Informatica, Amsterdam, The Netherlands
| | - Iraes Rabbers
- Centrum Wiskunde and Informatica, Amsterdam, The Netherlands
| | - András Szabó
- Centrum Wiskunde and Informatica, Amsterdam, The Netherlands
| | - Roeland M H Merks
- Centrum Wiskunde and Informatica, Amsterdam, The Netherlands.,Mathematical Institute, Leiden University, Leiden, The Netherlands
| |
Collapse
|
76
|
Liu H, Hilliard S, Kelly E, Chen CH, Saifudeen Z, El-Dahr SS. The polycomb proteins EZH1 and EZH2 co-regulate chromatin accessibility and nephron progenitor cell lifespan in mice. J Biol Chem 2020; 295:11542-11558. [PMID: 32554463 DOI: 10.1074/jbc.ra120.013348] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 06/09/2020] [Indexed: 01/20/2023] Open
Abstract
SIX2 (SIX homeobox 2)-positive nephron progenitor cells (NPCs) give rise to all epithelial cell types of the nephron, the filtering unit of the kidney. NPCs have a limited lifespan and are depleted near the time of birth. Epigenetic factors are implicated in the maintenance of organ-restricted progenitors such as NPCs, but the chromatin-based mechanisms are incompletely understood. Here, using a combination of gene targeting, chromatin profiling, and single-cell RNA analysis, we examined the role of the murine histone 3 Lys-27 (H3K27) methyltransferases EZH1 (enhancer of zeste 1) and EZH2 in NPC maintenance. We found that EZH2 expression correlates with NPC growth potential and that EZH2 is the dominant H3K27 methyltransferase in NPCs and epithelial descendants. Surprisingly, NPCs lacking H3K27 trimethylation maintained their progenitor state but cycled slowly, leading to a smaller NPC pool and formation of fewer nephrons. Unlike Ezh2 loss of function, dual inactivation of Ezh1 and Ezh2 triggered overexpression of the transcriptional repressor Hes-related family BHLH transcription factor with YRPW motif 1 (Hey1), down-regulation of Six2, and unscheduled activation of Wnt4-driven differentiation, resulting in early termination of nephrogenesis and severe renal dysgenesis. Double-mutant NPCs also overexpressed the SIX family member Six1 However, in this context, SIX1 failed to maintain NPC stemness. At the chromatin level, EZH1 and EZH2 restricted accessibility to AP-1-binding motifs, and their absence promoted a regulatory landscape akin to differentiated and nonlineage cells. We conclude that EZH2 is required for NPC renewal potential and that tempering of the differentiation program requires cooperation of both EZH1 and EZH2.
Collapse
Affiliation(s)
- Hongbing Liu
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Sylvia Hilliard
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Elizabeth Kelly
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, Louisiana, USA.,Department of Obstetrics & Gynecology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Chao-Hui Chen
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Zubaida Saifudeen
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Samir S El-Dahr
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, Louisiana, USA
| |
Collapse
|
77
|
Abstract
ABSTRACT
Over the past 5 years, several studies have begun to uncover the links between the classical signal transduction pathways and the physical mechanisms that are used to sculpt branched tissues. These advances have been made, in part, thanks to innovations in live imaging and reporter animals. With modern research tools, our conceptual models of branching morphogenesis are rapidly evolving, and the differences in branching mechanisms between each organ are becoming increasingly apparent. Here, we highlight four branched epithelia that develop at different spatial scales, within different surrounding tissues and via divergent physical mechanisms. Each of these organs has evolved to employ unique branching strategies to achieve a specialized final architecture.
Collapse
Affiliation(s)
- Katharine Goodwin
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Celeste M. Nelson
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
78
|
Abstract
Human kidney tissue can now be generated via the directed differentiation of human pluripotent stem cells. This advance is anticipated to facilitate the modeling of human kidney diseases, provide platforms for nephrotoxicity screening, enable cellular therapy, and potentially generate tissue for renal replacement. All such applications will rely upon the accuracy and reliability of the model and the capacity for stem cell-derived kidney tissue to recapitulate both normal and diseased states. In this review, we discuss the models available, how well they recapitulate the human kidney, and how far we are from application of these cells for use in cellular therapies.
Collapse
Affiliation(s)
- Melissa H Little
- Murdoch Children's Research Institute, Parkville, Victoria 3052, Australia; .,Department of Paediatrics, University of Melbourne, Victoria 3010, Australia.,Department of Anatomy and Neuroscience, University of Melbourne, Victoria 3010, Australia
| | - Lorna J Hale
- Murdoch Children's Research Institute, Parkville, Victoria 3052, Australia;
| | - Sara E Howden
- Murdoch Children's Research Institute, Parkville, Victoria 3052, Australia; .,Department of Paediatrics, University of Melbourne, Victoria 3010, Australia
| | - Santhosh V Kumar
- Murdoch Children's Research Institute, Parkville, Victoria 3052, Australia; .,Department of Paediatrics, University of Melbourne, Victoria 3010, Australia
| |
Collapse
|
79
|
Bueters R, Bael A, Gasthuys E, Chen C, Schreuder MF, Frazier KS. Ontogeny and Cross-species Comparison of Pathways Involved in Drug Absorption, Distribution, Metabolism, and Excretion in Neonates (Review): Kidney. Drug Metab Dispos 2020; 48:353-367. [PMID: 32114509 DOI: 10.1124/dmd.119.089755] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 02/04/2020] [Indexed: 02/13/2025] Open
Abstract
The kidneys play an important role in many processes, including urine formation, water conservation, acid-base equilibrium, and elimination of waste. The anatomic and functional development of the kidney has different maturation time points in humans versus animals, with critical differences between species in maturation before and after birth. Absorption, distribution, metabolism, and excretion (ADME) of drugs vary depending on age and maturation, which will lead to differences in toxicity and efficacy. When neonate/juvenile laboratory animal studies are designed, a thorough knowledge of the differences in kidney development between newborns/children and laboratory animals is essential. The human and laboratory animal data must be combined to obtain a more complete picture of the development in the kidneys around the neonatal period and the complexity of ADME in newborns and children. This review examines the ontogeny and cross-species differences in ADME processes in the developing kidney in preterm and term laboratory animals and children. It provides an overview of insights into ADME functionality in the kidney by identifying what is currently known and which gaps still exist. Currently important renal function properties such as glomerular filtration rate, renal blood flow, and ability to concentrate are generally well known, while detailed knowledge about transporter and metabolism maturation is growing but is still lacking. Preclinical data in those properties is limited to rodents and generally covers only the expression levels of transporter or enzyme-encoding genes. More knowledge on a functional level is needed to predict the kinetics and toxicity in neonate/juvenile toxicity and efficacy studies. SIGNIFICANCE STATEMENT: This review provides insight in cross-species developmental differences of absorption, distribution, metabolism, and excretion properties in the kidney, which should be considered in neonate/juvenile study interpretation, hypotheses generation, and experimental design.
Collapse
Affiliation(s)
- Ruud Bueters
- Janssen Research & Development, a division of Janssen Pharmaceutica NV, Division of Discovery, Product Development & Supply, Department of Nonclinical Safety, Beerse, Belgium (R.B.); ZNA Queen Paola Children's Hospital, Department of Pediatric Nephrology, Antwerp, Belgium (A.B.); Department of Paediatrics, Faculty of Medicine, Ghent University, Gent, Belgium (E.G.); Health and Environmental Sciences Institute, Washington, DC (C.C.); Radboudumc Amalia Children's Hospital, Department of Pediatric Nephrology, Nijmegen, The Netherlands (M.F.S.); and GlaxoSmithKline, Collegeville, Pennsylvania (K.S.F.)
| | - An Bael
- Janssen Research & Development, a division of Janssen Pharmaceutica NV, Division of Discovery, Product Development & Supply, Department of Nonclinical Safety, Beerse, Belgium (R.B.); ZNA Queen Paola Children's Hospital, Department of Pediatric Nephrology, Antwerp, Belgium (A.B.); Department of Paediatrics, Faculty of Medicine, Ghent University, Gent, Belgium (E.G.); Health and Environmental Sciences Institute, Washington, DC (C.C.); Radboudumc Amalia Children's Hospital, Department of Pediatric Nephrology, Nijmegen, The Netherlands (M.F.S.); and GlaxoSmithKline, Collegeville, Pennsylvania (K.S.F.)
| | - Elke Gasthuys
- Janssen Research & Development, a division of Janssen Pharmaceutica NV, Division of Discovery, Product Development & Supply, Department of Nonclinical Safety, Beerse, Belgium (R.B.); ZNA Queen Paola Children's Hospital, Department of Pediatric Nephrology, Antwerp, Belgium (A.B.); Department of Paediatrics, Faculty of Medicine, Ghent University, Gent, Belgium (E.G.); Health and Environmental Sciences Institute, Washington, DC (C.C.); Radboudumc Amalia Children's Hospital, Department of Pediatric Nephrology, Nijmegen, The Netherlands (M.F.S.); and GlaxoSmithKline, Collegeville, Pennsylvania (K.S.F.)
| | - Connie Chen
- Janssen Research & Development, a division of Janssen Pharmaceutica NV, Division of Discovery, Product Development & Supply, Department of Nonclinical Safety, Beerse, Belgium (R.B.); ZNA Queen Paola Children's Hospital, Department of Pediatric Nephrology, Antwerp, Belgium (A.B.); Department of Paediatrics, Faculty of Medicine, Ghent University, Gent, Belgium (E.G.); Health and Environmental Sciences Institute, Washington, DC (C.C.); Radboudumc Amalia Children's Hospital, Department of Pediatric Nephrology, Nijmegen, The Netherlands (M.F.S.); and GlaxoSmithKline, Collegeville, Pennsylvania (K.S.F.)
| | - Michiel F Schreuder
- Janssen Research & Development, a division of Janssen Pharmaceutica NV, Division of Discovery, Product Development & Supply, Department of Nonclinical Safety, Beerse, Belgium (R.B.); ZNA Queen Paola Children's Hospital, Department of Pediatric Nephrology, Antwerp, Belgium (A.B.); Department of Paediatrics, Faculty of Medicine, Ghent University, Gent, Belgium (E.G.); Health and Environmental Sciences Institute, Washington, DC (C.C.); Radboudumc Amalia Children's Hospital, Department of Pediatric Nephrology, Nijmegen, The Netherlands (M.F.S.); and GlaxoSmithKline, Collegeville, Pennsylvania (K.S.F.)
| | - Kendall S Frazier
- Janssen Research & Development, a division of Janssen Pharmaceutica NV, Division of Discovery, Product Development & Supply, Department of Nonclinical Safety, Beerse, Belgium (R.B.); ZNA Queen Paola Children's Hospital, Department of Pediatric Nephrology, Antwerp, Belgium (A.B.); Department of Paediatrics, Faculty of Medicine, Ghent University, Gent, Belgium (E.G.); Health and Environmental Sciences Institute, Washington, DC (C.C.); Radboudumc Amalia Children's Hospital, Department of Pediatric Nephrology, Nijmegen, The Netherlands (M.F.S.); and GlaxoSmithKline, Collegeville, Pennsylvania (K.S.F.)
| |
Collapse
|
80
|
Jafree DJ, Long DA. Beyond a Passive Conduit: Implications of Lymphatic Biology for Kidney Diseases. J Am Soc Nephrol 2020; 31:1178-1190. [PMID: 32295825 DOI: 10.1681/asn.2019121320] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The kidney contains a network of lymphatic vessels that clear fluid, small molecules, and cells from the renal interstitium. Through modulating immune responses and via crosstalk with surrounding renal cells, lymphatic vessels have been implicated in the progression and maintenance of kidney disease. In this Review, we provide an overview of the development, structure, and function of lymphatic vessels in the healthy adult kidney. We then highlight the contributions of lymphatic vessels to multiple forms of renal pathology, emphasizing CKD, transplant rejection, and polycystic kidney disease and discuss strategies to target renal lymphatics using genetic and pharmacologic approaches. Overall, we argue the case for lymphatics playing a fundamental role in renal physiology and pathology and treatments modulating these vessels having therapeutic potential across the spectrum of kidney disease.
Collapse
Affiliation(s)
- Daniyal J Jafree
- Developmental Biology and Cancer Programme, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom.,MB/PhD Programme, Faculty of Medical Sciences, University College London, London, United Kingdom
| | - David A Long
- Developmental Biology and Cancer Programme, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| |
Collapse
|
81
|
Hemker SL, Cerqueira DM, Bodnar AJ, Cargill KR, Clugston A, Anslow MJ, Sims-Lucas S, Kostka D, Ho J. Deletion of hypoxia-responsive microRNA-210 results in a sex-specific decrease in nephron number. FASEB J 2020; 34:5782-5799. [PMID: 32141129 DOI: 10.1096/fj.201902767r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 02/06/2020] [Accepted: 02/19/2020] [Indexed: 12/25/2022]
Abstract
Low nephron number results in an increased risk of developing hypertension and chronic kidney disease. Intrauterine growth restriction is associated with a nephron deficit in humans, and is commonly caused by placental insufficiency, which results in fetal hypoxia. The underlying mechanisms by which hypoxia impacts kidney development are poorly understood. microRNA-210 is the most consistently induced microRNA in hypoxia and is known to promote cell survival in a hypoxic environment. In this study, the role of microRNA-210 in kidney development was evaluated using a global microRNA-210 knockout mouse. A male-specific 35% nephron deficit in microRNA-210 knockout mice was observed. Wnt/β-catenin signaling, a pathway crucial for nephron differentiation, was misregulated in male kidneys with increased expression of the canonical Wnt target lymphoid enhancer binding factor 1. This coincided with increased expression of caspase-8-associated protein 2, a known microRNA-210 target and apoptosis signal transducer. Together, these data are consistent with a sex-specific requirement for microRNA-210 in kidney development.
Collapse
Affiliation(s)
- Shelby L Hemker
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Rangos Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Débora M Cerqueira
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Rangos Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Andrew J Bodnar
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Rangos Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Kasey R Cargill
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Rangos Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Andrew Clugston
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Rangos Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA.,Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Melissa J Anslow
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Rangos Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Sunder Sims-Lucas
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Rangos Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Dennis Kostka
- Rangos Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA.,Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jacqueline Ho
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Rangos Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
82
|
Rutledge EA, Lindström NO, Michos O, McMahon AP. Genetic manipulation of ureteric bud tip progenitors in the mammalian kidney through an Adamts18 enhancer driven tet-on inducible system. Dev Biol 2020; 458:164-176. [PMID: 31734175 PMCID: PMC6995766 DOI: 10.1016/j.ydbio.2019.11.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 11/13/2019] [Accepted: 11/13/2019] [Indexed: 11/22/2022]
Abstract
The ureteric epithelial progenitor (UEP) population within the embryonic kidney generates the arborized epithelial network of the kidney's collecting system and plays a critical role in the expansion and induction of the surrounding nephron progenitor pool. Adamts18 shows UEP- restricted expression in the kidney and progenitor tip-restricted expression in several other organs undergoing branching epithelial growth. Adamts18 is encoded by 23 exons. Genetic removal of genomic sequence spanning exons 1 to 3 led to a specific loss of Adamts18 expression in UEPs, suggesting this region may encode a UEP-specific enhancer. Intron 2 (3 kb) was shown to have enhancer activity driving expression of the doxycycline inducible tet-on transcriptional regulator (rtTA) in an Adamts18en-rtTA transgenic mouse strain. Crossing Adamts18en-rtTA mice to a doxycycline dependent GFP reporter mouse enabled the live imaging of embryonic kidney explants. This facilitated the analysis of ureteric epithelial branching events at the cellular level. Ablation of UEPs at the initiation of ureteric bud outgrowth through the doxycycline-mediated induction of Diphtheria Toxin A (DTA) generated a range of phenotypes from complete kidneys agenesis, to duplex kidneys with double ureters. The latter outcome points to the potential of regulative processes to restore UEPs. In contrast, overexpression of YAP prior to ureteric bud outgrowth led to a complete failure of kidney development. Elevating YAP levels at later stages retarded branching growth. A similar phenotype was observed with the overexpression of MYC within the branch-tip localized UEP population. These experiments showcase the utility of the Adamts18en-rtTA transgenic model to the investigation of cellular and molecular events specific to branch tip progenitors within the mammalian kidney complementing existing CRE-dependent genetic tools. Further, the illustrative examples point to areas where new insight may be gained into the regulation of UEP programs.
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, CA, 90089, USA
| | - Nils O Lindström
- 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, CA, 90089, USA
| | - Odysse Michos
- Department of Biosystems, Science and Engineering (D-BSSE), ETH Zurich, Basel, 4058, Switzerland
| | - 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, CA, 90089, USA.
| |
Collapse
|
83
|
Lau RWK, Al‐Rubaie A, Saini S, Wise AF, Ricardo SD. Percutaneous intrarenal transplantation of differentiated induced pluripotent stem cells into newborn mice. Anat Rec (Hoboken) 2020; 303:2603-2612. [DOI: 10.1002/ar.24371] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 11/22/2019] [Accepted: 12/07/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Ricky W. K. Lau
- Department of Anatomy and Developmental BiologyBiomedical Discovery Institute, Monash University Clayton Victoria Australia
| | - Ali Al‐Rubaie
- Department of Anatomy and Developmental BiologyBiomedical Discovery Institute, Monash University Clayton Victoria Australia
| | - Sheetal Saini
- Department of Anatomy and Developmental BiologyBiomedical Discovery Institute, Monash University Clayton Victoria Australia
| | - Andrea F. Wise
- Department of Anatomy and Developmental BiologyBiomedical Discovery Institute, Monash University Clayton Victoria Australia
| | - Sharon D. Ricardo
- Department of Anatomy and Developmental BiologyBiomedical Discovery Institute, Monash University Clayton Victoria Australia
| |
Collapse
|
84
|
Boivin FJ, Schmidt-Ott KM. Functional roles of Grainyhead-like transcription factors in renal development and disease. Pediatr Nephrol 2020; 35:181-190. [PMID: 30554362 DOI: 10.1007/s00467-018-4171-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 11/07/2018] [Accepted: 12/06/2018] [Indexed: 12/11/2022]
Abstract
Proper renal function relies on the tightly regulated development of nephrons and collecting ducts. This process, known as tubulogenesis, involves dynamic cellular and molecular changes that instruct cells to form highly organized tubes of epithelial cells which compartmentalize the renal interstitium and tubular lumen via assembly of a selective barrier. The integrity and diversity of the various renal epithelia is achieved via formation of intercellular protein complexes along the apical-basal axis of the epithelial cells. In recent years, the evolutionarily conserved family of Grainyhead-like (GRHL) transcription factors which encompasses three mammalian family members (Grainyhead-like 1, 2, 3) has emerged as a group of critical regulators for organ development, epithelial differentiation, and barrier formation. Evidence from transgenic animal models supports the presence of Grainyhead-like-dependent transcriptional mechanisms that promote formation and maintenance of epithelial barriers in the kidney. In this review, we highlight different Grhl-dependent mechanisms that modulate epithelial differentiation in the kidney. Additionally, we discuss how disruptions in these mechanisms result in impaired renal function later in life.
Collapse
Affiliation(s)
- Felix J Boivin
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Strasse 10, 13125, Berlin, Germany
| | - Kai M Schmidt-Ott
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Strasse 10, 13125, Berlin, Germany. .,Department of Nephrology, Charité Medical University, Berlin, Germany.
| |
Collapse
|
85
|
Modulation of polycystic kidney disease by non-coding RNAs. Cell Signal 2020; 71:109548. [PMID: 31982550 DOI: 10.1016/j.cellsig.2020.109548] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/22/2020] [Accepted: 01/23/2020] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW microRNAs (miRNAs) are a class of small, evolutionarily conserved, non-coding RNAs (ncRNAs) that function as inhibitors of post-transcriptional mRNA expression. They are implicated in the pathogenesis of numerous diseases, including many common kidney conditions. In this review, we focus on how miRNAs impact autosomal dominant polycystic kidney disease (ADPKD) progression. We also discuss the feasibility of the emerging novel antisense oligonucleotides (ASOs) drug class, which includes anti-miRNA drugs, for the treatment of ADPKD. RECENT FINDINGS Aberrant miRNA expression is observed in multiple PKD murine models and human ADPKD samples. Gain and loss-of-function studies have directly linked dysregulated miRNA activity to kidney cyst growth. The most comprehensively studied miRNA in PKD is the miR-17 family, which promotes PKD progression through the rewiring of cyst metabolism and by directly inhibiting PKD1 and PKD2 expression. This discovery has led to the development of an anti-miR-17 drug for ADPKD treatment. Other miRNAs such as miR-21, miR-193, and miR-214 are also known to regulate cyst growth by modulating cyst epithelial apoptosis, proliferation, and interstitial inflammation. SUMMARY miRNAs have emerged as novel pathogenic regulators of ADPKD progression. Anti-miR-based drugs represent a new therapeutic modality to treat ADPKD patients.
Collapse
|
86
|
Abstract
Congenital abnormalities of the kidney and urinary tract (CAKUT) are a highly diverse group of diseases that together belong to the most common abnormalities detected in the new-born child. Consistent with this diversity, CAKUT are caused by mutations in a large number of genes and present a wide spectrum of phenotypes. In this review, we will focus on duplex kidneys, a relatively frequent form of CAKUT that is often asymptomatic but predisposes to vesicoureteral reflux and hydronephrosis. We will summarise the molecular programs responsible for ureter induction, review the genes that have been identified as risk factors in duplex kidney formation and discuss molecular and cellular mechanisms that may lead to this malformation.
Collapse
Affiliation(s)
- Vladimir M Kozlov
- iBV, Institut de Biologie Valrose, Equipe Labellisée Ligue Contre le Cancer, Université Cote d'Azur, Centre de Biochimie, UFR Sciences, Parc Valrose, Nice Cedex 2, 06108, France
| | - Andreas Schedl
- iBV, Institut de Biologie Valrose, Equipe Labellisée Ligue Contre le Cancer, Université Cote d'Azur, Centre de Biochimie, UFR Sciences, Parc Valrose, Nice Cedex 2, 06108, France
| |
Collapse
|
87
|
Kuure S, Sariola H. Mouse Models of Congenital Kidney Anomalies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1236:109-136. [PMID: 32304071 DOI: 10.1007/978-981-15-2389-2_5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Congenital anomalies of the kidney and urinary tract (CAKUT) are common birth defects, which cause the majority of chronic kidney diseases in children. CAKUT covers a wide range of malformations that derive from deficiencies in embryonic kidney and lower urinary tract development, including renal aplasia, hypodysplasia, hypoplasia, ectopia, and different forms of ureter abnormalities. The majority of the genetic causes of CAKUT remain unknown. Research on mutant mice has identified multiple genes that critically regulate renal differentiation. The data generated from this research have served as an excellent resource to identify the genetic bases of human kidney defects and have led to significantly improved diagnostics. Furthermore, genetic data from human CAKUT studies have also revealed novel genes regulating kidney differentiation.
Collapse
Affiliation(s)
- Satu Kuure
- GM-Unit, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland. .,Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland. .,Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Hannu Sariola
- Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Paediatric Pathology, HUSLAB, Helsinki University Central Hospital, Helsinki, Finland
| |
Collapse
|
88
|
Jafree DJ, Moulding D, Kolatsi-Joannou M, Perretta Tejedor N, Price KL, Milmoe NJ, Walsh CL, Correra RM, Winyard PJ, Harris PC, Ruhrberg C, Walker-Samuel S, Riley PR, Woolf AS, Scambler PJ, Long DA. Spatiotemporal dynamics and heterogeneity of renal lymphatics in mammalian development and cystic kidney disease. eLife 2019; 8:48183. [PMID: 31808745 PMCID: PMC6948954 DOI: 10.7554/elife.48183] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 11/30/2019] [Indexed: 12/11/2022] Open
Abstract
Heterogeneity of lymphatic vessels during embryogenesis is critical for organ-specific lymphatic function. Little is known about lymphatics in the developing kidney, despite their established roles in pathology of the mature organ. We performed three-dimensional imaging to characterize lymphatic vessel formation in the mammalian embryonic kidney at single-cell resolution. In mouse, we visually and quantitatively assessed the development of kidney lymphatic vessels, remodeling from a ring-like anastomosis under the nascent renal pelvis; a site of VEGF-C expression, to form a patent vascular plexus. We identified a heterogenous population of lymphatic endothelial cell clusters in mouse and human embryonic kidneys. Exogenous VEGF-C expanded the lymphatic population in explanted mouse embryonic kidneys. Finally, we characterized complex kidney lymphatic abnormalities in a genetic mouse model of polycystic kidney disease. Our study provides novel insights into the development of kidney lymphatic vasculature; a system which likely has fundamental roles in renal development, physiology and disease. In most organs in the body, fluid tends to build up in the spaces between cells, especially if the organs become inflamed. Each organ has a ‘waste disposal system’; a set of specialized tubes called lymphatic vessels, to clear away this excess fluid and keep a check on inflammation. Defects in these tubes have been linked to a wide range of diseases including heart attacks, obesity, dementia and cancer. The kidneys are responsible for filtering blood and balancing many of the body’s chemical processes. Polycystic kidney disease (PKD) is the most common genetic kidney disorder and it results in cysts filled with fluid building up in the kidney. The growth of cysts in PKD may be due to a problem with the lymphatic vessels. However, compared to other organs, how lymphatic vessels first form within the kidney and what they do is not well understood. Now, Jafree et al. have used three-dimensional imaging to study how lymphatic vessels form in the kidneys of mice and humans. The experiments showed that lymphatic vessels first appear when mouse kidneys are about half developed, and start to grow rapidly when the kidneys are thought to begin filtering blood. Clusters of cells that may help lymphatic vessels to grow were also found hidden deep within the kidneys of mouse embryos. Treating the kidneys with a factor that stimulates the growth of lymphatic vessels increased the numbers of these clusters. Jafree et al. found similar clusters of cells in human kidneys, suggesting that lymphatic vessels in the kidneys of different mammals may develop in the same way. Further experiments showed that the lymphatic vessels of kidneys in mice with PKD become distorted early on in the disease, when cysts are still small and before the mice develop symptoms. In the future, identifying drugs that target kidney lymphatic vessels may lead to more effective treatments for patients with PKD and other kidney diseases.
Collapse
Affiliation(s)
- Daniyal J Jafree
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom.,MB/PhD Programme, Faculty of Medical Sciences, University College London, London, United Kingdom
| | - Dale Moulding
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Maria Kolatsi-Joannou
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Nuria Perretta Tejedor
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Karen L Price
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Natalie J Milmoe
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Claire L Walsh
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, United Kingdom
| | - Rosa Maria Correra
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Paul Jd Winyard
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Peter C Harris
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, United States
| | - Christiana Ruhrberg
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Simon Walker-Samuel
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, United Kingdom
| | - Paul R Riley
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Adrian S Woolf
- School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom.,Royal Manchester Children's Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Peter J Scambler
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - David A Long
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| |
Collapse
|
89
|
Coudert Y, Harris S, Charrier B. Design Principles of Branching Morphogenesis in Filamentous Organisms. Curr Biol 2019; 29:R1149-R1162. [PMID: 31689405 DOI: 10.1016/j.cub.2019.09.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The radiation of life on Earth was accompanied by the diversification of multicellular body plans in the eukaryotic kingdoms Animalia, Plantae, Fungi and Chromista. Branching forms are ubiquitous in nature and evolved repeatedly in the above lineages. The developmental and genetic basis of branch formation is well studied in the three-dimensional shoot and root systems of land plants, and in animal organs such as the lung, kidney, mammary gland, vasculature, etc. Notably, recent thought-provoking studies combining experimental analysis and computational modeling of branching patterns in whole animal organs have identified global patterning rules and proposed unifying principles of branching morphogenesis. Filamentous branching forms represent one of the simplest expressions of the multicellular body plan and constitute a key step in the evolution of morphological complexity. Similarities between simple and complex branching forms distantly related in evolution are compelling, raising the question whether shared mechanisms underlie their development. Here, we focus on filamentous branching organisms that represent major study models from three distinct eukaryotic kingdoms, including the moss Physcomitrella patens (Plantae), the brown alga Ectocarpus sp. (Chromista), and the ascomycetes Neurospora crassa and Aspergillus nidulans (Fungi), and bring to light developmental regulatory mechanisms and design principles common to these lineages. Throughout the review we explore how the regulatory mechanisms of branching morphogenesis identified in other models, and in particular animal organs, may inform our thinking on filamentous systems and thereby advance our understanding of the diverse strategies deployed across the eukaryotic tree of life to evolve similar forms.
Collapse
Affiliation(s)
- Yoan Coudert
- Laboratoire Reproduction et Développement des Plantes, Université de Lyon, ENS de Lyon, UCB Lyon 1, CNRS, INRA, INRIA, Lyon 69007, France.
| | - Steven Harris
- University of Manitoba, Department of Biological Sciences, Winnipeg, MB, Canada; Center for Plant Science Innovation and Department of Plant Pathology, University of Nebraska, Lincoln, NE, USA
| | - Bénédicte Charrier
- CNRS, Sorbonne Université, Laboratoire de Biologie Intégrative des Modèles Marins LBI2M, Station Biologique de Roscoff, Roscoff 29680, France
| |
Collapse
|
90
|
Cerqueira DM, Hemker SL, Bodnar AJ, Ortiz DM, Oladipupo FO, Mukherjee E, Gong Z, Appolonia C, Muzumdar R, Sims-Lucas S, Ho J. In utero exposure to maternal diabetes impairs nephron progenitor differentiation. Am J Physiol Renal Physiol 2019; 317:F1318-F1330. [PMID: 31509011 PMCID: PMC6879946 DOI: 10.1152/ajprenal.00204.2019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 08/23/2019] [Accepted: 09/03/2019] [Indexed: 01/08/2023] Open
Abstract
The incidence of diabetes mellitus has significantly increased among women of childbearing age, and it has been shown that prenatal exposure to maternal diabetes increases the risk of associated congenital anomalies of the kidney. Congenital anomalies of the kidney are among the leading causes of chronic kidney disease in children. To better understand the effect of maternal diabetes on kidney development, we analyzed wild-type offspring (DM_Exp) of diabetic Ins2+/C96Y mice (Akita mice). DM_Exp mice at postnatal day 34 have a reduction of ~20% in the total nephron number compared with controls, using the gold standard physical dissector/fractionator method. At the molecular level, the expression of the nephron progenitor markers sine oculis homeobox homolog 2 and Cited1 was increased in DM_Exp kidneys at postnatal day 2. Conversely, the number of early developing nephrons was diminished in DM_Exp kidneys. This was associated with decreased expression of the intracellular domain of Notch1 and the canonical Wnt target lymphoid enhancer binding factor 1. Together, these data suggest that the diabetic intrauterine environment impairs the differentiation of nephron progenitors into nephrons, possibly by perturbing the Notch and Wnt/β-catenin signaling pathways.
Collapse
Affiliation(s)
- Débora M Cerqueira
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Rangos Research Center, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
| | - Shelby L Hemker
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Rangos Research Center, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
| | - Andrew J Bodnar
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Rangos Research Center, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
| | - Daniella M Ortiz
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Rangos Research Center, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
| | - Favour O Oladipupo
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Rangos Research Center, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
| | - Elina Mukherjee
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Rangos Research Center, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
| | - Zhenwei Gong
- Rangos Research Center, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
- Division of Endocrinology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Corynn Appolonia
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Rangos Research Center, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
| | - Radhika Muzumdar
- Rangos Research Center, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
- Division of Endocrinology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Sunder Sims-Lucas
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Rangos Research Center, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jacqueline Ho
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Rangos Research Center, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
91
|
Abstract
There are now many reports of human kidney organoids generated via the directed differentiation of human pluripotent stem cells (PSCs) based on an existing understanding of mammalian kidney organogenesis. Such kidney organoids potentially represent tractable tools for the study of normal human development and disease with improvements in scale, structure, and functional maturation potentially providing future options for renal regeneration. The utility of such organotypic models, however, will ultimately be determined by their developmental accuracy. While initially inferred from mouse models, recent transcriptional analyses of human fetal kidney have provided greater insight into nephrogenesis. In this review, we discuss how well human kidney organoids model the human fetal kidney and how the remaining differences challenge their utility.
Collapse
Affiliation(s)
- Melissa H Little
- Murdoch Children's Research Institute, Parkville, Victoria 3052, Australia
- Department of Anatomy and Neuroscience, The University of Melbourne, Victoria 3052, Australia
- Department of Paediatrics, The University of Melbourne, Victoria 3052, Australia
| | - Alexander N Combes
- Murdoch Children's Research Institute, Parkville, Victoria 3052, Australia
- Department of Anatomy and Neuroscience, The University of Melbourne, Victoria 3052, Australia
| |
Collapse
|
92
|
Multiscale dynamics of branching morphogenesis. Curr Opin Cell Biol 2019; 60:99-105. [DOI: 10.1016/j.ceb.2019.04.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 04/09/2019] [Accepted: 04/16/2019] [Indexed: 12/18/2022]
|
93
|
Elmore SA, Kavari SL, Hoenerhoff MJ, Mahler B, Scott BE, Yabe K, Seely JC. Histology Atlas of the Developing Mouse Urinary System With Emphasis on Prenatal Days E10.5-E18.5. Toxicol Pathol 2019; 47:865-886. [PMID: 31599209 PMCID: PMC6814567 DOI: 10.1177/0192623319873871] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Congenital abnormalities of the urinary tract are some of the most common human developmental abnormalities. Several genetically engineered mouse models have been developed to mimic these abnormalities and aim to better understand the molecular mechanisms of disease. This atlas has been developed as an aid to pathologists and other biomedical scientists for identification of abnormalities in the developing murine urinary tract by cataloguing normal structures at each stage of development. Hematoxylin and eosin- and immunohistochemical-stained sections are provided, with a focus on E10.5-E18.5, as well as a brief discussion of postnatal events in urinary tract development. A section on abnormalities in the development of the urinary tract is also provided, and molecular mechanisms are presented as supplementary material. Additionally, overviews of the 2 key processes of kidney development, branching morphogenesis and nephrogenesis, are provided to aid in the understanding of the complex organogenesis of the kidney. One of the key findings of this atlas is the histological identification of the ureteric bud at E10.5, as previous literature has provided conflicting reports on the initial point of budding. Furthermore, attention is paid to points where murine development is significantly distinct from human development, namely, in the cessation of nephrogenesis.
Collapse
Affiliation(s)
- Susan A Elmore
- Cellular and Molecular Pathology Branch, National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, NC, USA
| | - Sanam L Kavari
- Cellular and Molecular Pathology Branch, National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, NC, USA
| | - Mark J Hoenerhoff
- In Vivo Animal Core, Unit for Laboratory Animal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Beth Mahler
- Experimental Pathology Laboratories, Inc, Research Triangle Park, NC, USA
| | | | - Koichi Yabe
- Pharmacovigilance Department, Daiichi Sankyo Co, Ltd, Tokyo, Japan
| | - John C Seely
- Experimental Pathology Laboratories, Inc, Research Triangle Park, NC, USA
| |
Collapse
|
94
|
Myllymäki SM, Mikkola ML. Inductive signals in branching morphogenesis - lessons from mammary and salivary glands. Curr Opin Cell Biol 2019; 61:72-78. [PMID: 31387017 DOI: 10.1016/j.ceb.2019.07.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 06/28/2019] [Accepted: 07/02/2019] [Indexed: 12/30/2022]
Abstract
Branching morphogenesis is a fundamental developmental program that generates large epithelial surfaces in a limited three-dimensional space. It is regulated by inductive tissue interactions whose effects are mediated by soluble signaling molecules, and cell-cell and cell-extracellular matrix interactions. Here, we will review recent studies on inductive signaling interactions governing branching morphogenesis in light of phenotypes of mouse mutants and ex vivo organ culture studies with emphasis on developing mammary and salivary glands. We will highlight advances in understanding how cell fate decisions are intimately linked with branching morphogenesis. We will also discuss novel insights into the molecular control of cellular mechanisms driving the formation of these arborized ductal structures and reflect upon how distinct spatial patterns are generated.
Collapse
Affiliation(s)
- Satu-Marja Myllymäki
- Developmental Biology Program, Institute of Biotechnology, HiLIFE, P.O.B. 56, University of Helsinki, 00014 Helsinki, Finland.
| | - Marja L Mikkola
- Developmental Biology Program, Institute of Biotechnology, HiLIFE, P.O.B. 56, University of Helsinki, 00014 Helsinki, Finland.
| |
Collapse
|
95
|
Ferrè S, Igarashi P. New insights into the role of HNF-1β in kidney (patho)physiology. Pediatr Nephrol 2019; 34:1325-1335. [PMID: 29961928 PMCID: PMC6312759 DOI: 10.1007/s00467-018-3990-7] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 05/24/2018] [Accepted: 05/25/2018] [Indexed: 12/14/2022]
Abstract
Hepatocyte nuclear factor-1β (HNF-1β) is an essential transcription factor that regulates the development and function of epithelia in the kidney, liver, pancreas, and genitourinary tract. Humans who carry HNF1B mutations develop heterogeneous renal abnormalities, including multicystic dysplastic kidneys, glomerulocystic kidney disease, renal agenesis, renal hypoplasia, and renal interstitial fibrosis. In the embryonic kidney, HNF-1β is required for ureteric bud branching, initiation of nephrogenesis, and nephron segmentation. Ablation of mouse Hnf1b in nephron progenitors causes defective tubulogenesis, whereas later inactivation in elongating tubules leads to cyst formation due to downregulation of cystic disease genes, including Umod, Pkhd1, and Pkd2. In the adult kidney, HNF-1β controls the expression of genes required for intrarenal metabolism and solute transport by tubular epithelial cells. Tubular abnormalities observed in HNF-1β nephropathy include hyperuricemia with or without gout, hypokalemia, hypomagnesemia, and polyuria. Recent studies have identified novel post-transcriptional and post-translational regulatory mechanisms that control HNF-1β expression and activity, including the miRNA cluster miR17 ∼ 92 and the interacting proteins PCBD1 and zyxin. Further understanding of the molecular mechanisms upstream and downstream of HNF-1β may lead to the development of new therapeutic approaches in cystic kidney disease and other HNF1B-related renal diseases.
Collapse
Affiliation(s)
- Silvia Ferrè
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, Texas, USA,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Peter Igarashi
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA. .,Department of Medicine, University of Minnesota Medical School, 420 Delaware St. SE, MMC 194, Minneapolis, MN, 55455, USA.
| |
Collapse
|
96
|
Francipane MG, Han B, Oxburgh L, Sims-Lucas S, Li Z, Lagasse E. Kidney-in-a-lymph node: A novel organogenesis assay to model human renal development and test nephron progenitor cell fates. J Tissue Eng Regen Med 2019; 13:1724-1731. [PMID: 31267702 DOI: 10.1002/term.2924] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 04/18/2019] [Accepted: 06/30/2019] [Indexed: 12/12/2022]
Abstract
Stem cell-derived organoids are emerging as sophisticated models for studying development and disease and as potential sources for developing organ substitutes. Unfortunately, although organoids containing renal structures have been generated from mouse and human pluripotent stem cells, there are still critical unanswered questions that are difficult to attain via in vitro systems, including whether these nonvascularized organoids have a stable and physiologically relevant phenotype or whether a suitable transplantation site for long-term in vivo studies can be identified. Even orthotopic engraftment of organoid cultures in the adult does not provide an environment conducive to vascularization and functional differentiation. Previously, we showed that the lymph node offers an alternative transplantation site where mouse metanephroi can differentiate into mature renal structures with excretory, homeostatic, and endocrine functions. Here, we show that the lymph node lends itself well as a niche to also grow human primary kidney rudiments and can additionally be viewed as a platform to interrogate emerging renal organoid cultures. Our study has a wide-ranging impact for tissue engineering approaches to rebuild functional tissues in vivo including-but not limited to-the kidney.
Collapse
Affiliation(s)
- Maria Giovanna Francipane
- McGowan Institute for Regenerative Medicine and Pathology Department, University of Pittsburgh, Pittsburgh, Pennsylvania.,Ri.MED Foundation, Palermo, Italy
| | - Bing Han
- McGowan Institute for Regenerative Medicine and Pathology Department, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Leif Oxburgh
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine
| | - Sunder Sims-Lucas
- Rangos Research Center, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania
| | - Zhongwei Li
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Eric Lagasse
- McGowan Institute for Regenerative Medicine and Pathology Department, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
97
|
Combes AN, Phipson B, Lawlor KT, Dorison A, Patrick R, Zappia L, Harvey RP, Oshlack A, Little MH. Single cell analysis of the developing mouse kidney provides deeper insight into marker gene expression and ligand-receptor crosstalk. Development 2019; 146:dev.178673. [PMID: 31118232 DOI: 10.1242/dev.178673] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 05/07/2019] [Indexed: 12/12/2022]
Abstract
Recent advances in the generation of kidney organoids and the culture of primary nephron progenitors from mouse and human have been based on knowledge of the molecular basis of kidney development in mice. Although gene expression during kidney development has been intensely investigated, single cell profiling provides new opportunities to further subsect component cell types and the signalling networks at play. Here, we describe the generation and analysis of 6732 single cell transcriptomes from the fetal mouse kidney [embryonic day (E)18.5] and 7853 sorted nephron progenitor cells (E14.5). These datasets provide improved resolution of cell types and specific markers, including subdivision of the renal stroma and heterogeneity within the nephron progenitor population. Ligand-receptor interaction and pathway analysis reveals novel crosstalk between cellular compartments and associates new pathways with differentiation of nephron and ureteric epithelium cell types. We identify transcriptional congruence between the distal nephron and ureteric epithelium, showing that most markers previously used to identify ureteric epithelium are not specific. Together, this work improves our understanding of metanephric kidney development and provides a template to guide the regeneration of renal tissue.
Collapse
Affiliation(s)
- Alexander N Combes
- Department of Anatomy & Neuroscience, University of Melbourne, Parkville, Victoria 3010, Australia .,Cell Biology, Murdoch Children's Research Institute, Flemington Rd, Parkville, Victoria 3052, Australia
| | - Belinda Phipson
- Cell Biology, Murdoch Children's Research Institute, Flemington Rd, Parkville, Victoria 3052, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Kynan T Lawlor
- Cell Biology, Murdoch Children's Research Institute, Flemington Rd, Parkville, Victoria 3052, Australia
| | - Aude Dorison
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales 2010, Australia
| | - Ralph Patrick
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales 2010, Australia.,St. Vincent's Clinical School, University of New South Wales, Kensington, New South Wales 2033, Australia
| | - Luke Zappia
- Cell Biology, Murdoch Children's Research Institute, Flemington Rd, Parkville, Victoria 3052, Australia.,School of Biosciences, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Richard P Harvey
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales 2010, Australia.,St. Vincent's Clinical School, University of New South Wales, Kensington, New South Wales 2033, Australia.,School of Biotechnology and Biomolecular Science, University of New South Wales, Kensington, New South Wales 2010, Australia
| | - Alicia Oshlack
- Department of Anatomy & Neuroscience, University of Melbourne, Parkville, Victoria 3010, Australia.,School of Biosciences, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Melissa H Little
- Department of Anatomy & Neuroscience, University of Melbourne, Parkville, Victoria 3010, Australia .,Cell Biology, Murdoch Children's Research Institute, Flemington Rd, Parkville, Victoria 3052, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
98
|
Hilliard S, Song R, Liu H, Chen CH, Li Y, Baddoo M, Flemington E, Wanek A, Kolls J, Saifudeen Z, El-Dahr SS. Defining the dynamic chromatin landscape of mouse nephron progenitors. Biol Open 2019; 8:bio.042754. [PMID: 31064740 PMCID: PMC6550063 DOI: 10.1242/bio.042754] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Six2+ cap mesenchyme cells, also called nephron progenitor cells (NPC), are precursors of all epithelial cell types of the nephron, the filtering unit of the kidney. Current evidence indicates that perinatal ‘old’ NPC have a greater tendency to exit the progenitor niche and differentiate into nascent nephrons than their embryonic ‘young’ counterpart. Understanding the underpinnings of NPC development may offer insights to rejuvenate old NPC and expand the progenitor pool. Here, we compared the chromatin landscape of young and old NPC and found common features reflecting their shared lineage but also intrinsic differences in chromatin accessibility and enhancer landscape supporting the view that old NPC are epigenetically poised for differentiation. Annotation of open chromatin regions and active enhancers uncovered the transcription factor Bach2 as a potential link between the pro-renewal MAPK/AP1 and pro-differentiation Six2/b-catenin pathways that might be of critical importance in regulation of NPC fate. Our data provide the first glimpse of the dynamic chromatin landscape of NPC and serve as a platform for future studies of the impact of genetic or environmental perturbations on the epigenome of NPC. Summary: An investigation of the chromatin landscape of mouse nephron progenitors across their life span supports the view that old nephron progenitors are epigenetically poised for differentiation.
Collapse
Affiliation(s)
- Sylvia Hilliard
- Department of Pediatrics, Section of Pediatric Nephrology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Renfang Song
- Department of Pediatrics, Section of Pediatric Nephrology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Hongbing Liu
- Department of Pediatrics, Section of Pediatric Nephrology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Chao-Hui Chen
- Department of Pediatrics, Section of Pediatric Nephrology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Yuwen Li
- Department of Pediatrics, Section of Pediatric Nephrology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Melody Baddoo
- Department of Pathology & Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Erik Flemington
- Department of Pathology & Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Alanna Wanek
- Departments of Pediatrics & Medicine, Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Jay Kolls
- Departments of Pediatrics & Medicine, Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Zubaida Saifudeen
- Department of Pediatrics, Section of Pediatric Nephrology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Samir S El-Dahr
- Department of Pediatrics, Section of Pediatric Nephrology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
99
|
Kurtzeborn K, Kwon HN, Kuure S. MAPK/ERK Signaling in Regulation of Renal Differentiation. Int J Mol Sci 2019; 20:E1779. [PMID: 30974877 PMCID: PMC6479953 DOI: 10.3390/ijms20071779] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 04/04/2019] [Accepted: 04/08/2019] [Indexed: 12/20/2022] Open
Abstract
Congenital anomalies of the kidney and urinary tract (CAKUT) are common birth defects derived from abnormalities in renal differentiation during embryogenesis. CAKUT is the major cause of end-stage renal disease and chronic kidney diseases in children, but its genetic causes remain largely unresolved. Here we discuss advances in the understanding of how mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) activity contributes to the regulation of ureteric bud branching morphogenesis, which dictates the final size, shape, and nephron number of the kidney. Recent studies also demonstrate that the MAPK/ERK pathway is directly involved in nephrogenesis, regulating both the maintenance and differentiation of the nephrogenic mesenchyme. Interestingly, aberrant MAPK/ERK signaling is linked to many cancers, and recent studies suggest it also plays a role in the most common pediatric renal cancer, Wilms' tumor.
Collapse
Affiliation(s)
- Kristen Kurtzeborn
- Helsinki Institute of Life Science, University of Helsinki, FIN-00014 Helsinki, Finland.
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, FIN-00014 Helsinki, Finland.
| | - Hyuk Nam Kwon
- Helsinki Institute of Life Science, University of Helsinki, FIN-00014 Helsinki, Finland.
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, FIN-00014 Helsinki, Finland.
| | - Satu Kuure
- Helsinki Institute of Life Science, University of Helsinki, FIN-00014 Helsinki, Finland.
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, FIN-00014 Helsinki, Finland.
- GM-unit, Laboratory Animal Center, Helsinki Institute of Life Science, University of Helsinki, FIN-00014 Helsinki, Finland.
| |
Collapse
|
100
|
Blackburn ATM, Miller RK. Modeling congenital kidney diseases in Xenopus laevis. Dis Model Mech 2019; 12:12/4/dmm038604. [PMID: 30967415 PMCID: PMC6505484 DOI: 10.1242/dmm.038604] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Congenital anomalies of the kidney and urinary tract (CAKUT) occur in ∼1/500 live births and are a leading cause of pediatric kidney failure. With an average wait time of 3-5 years for a kidney transplant, the need is high for the development of new strategies aimed at reducing the incidence of CAKUT and preserving renal function. Next-generation sequencing has uncovered a significant number of putative causal genes, but a simple and efficient model system to examine the function of CAKUT genes is needed. Xenopus laevis (frog) embryos are well-suited to model congenital kidney diseases and to explore the mechanisms that cause these developmental defects. Xenopus has many advantages for studying the kidney: the embryos develop externally and are easily manipulated with microinjections, they have a functional kidney in ∼2 days, and 79% of identified human disease genes have a verified ortholog in Xenopus. This facilitates high-throughput screening of candidate CAKUT-causing genes. In this Review, we present the similarities between Xenopus and mammalian kidneys, highlight studies of CAKUT-causing genes in Xenopus and describe how common kidney diseases have been modeled successfully in this model organism. Additionally, we discuss several molecular pathways associated with kidney disease that have been studied in Xenopus and demonstrate why it is a useful model for studying human kidney diseases. Summary: Understanding how congenital kidney diseases arise is imperative to their treatment. Using Xenopus as a model will aid in elucidating kidney development and congenital kidney diseases.
Collapse
Affiliation(s)
- Alexandria T M Blackburn
- Pediatric Research Center, Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center, Houston, TX 77030, USA.,The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Program in Genetics and Epigenetics, Houston, TX 77030, USA
| | - Rachel K Miller
- Pediatric Research Center, Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center, Houston, TX 77030, USA .,The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Program in Genetics and Epigenetics, Houston, TX 77030, USA.,The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Program in Biochemistry and Cell Biology Houston, Houston, TX 77030, USA.,Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|