51
|
Namestnikov M, Pleniceanu O, Dekel B. Mixing Cells for Vascularized Kidney Regeneration. Cells 2021; 10:1119. [PMID: 34066487 PMCID: PMC8148539 DOI: 10.3390/cells10051119] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/26/2021] [Accepted: 05/01/2021] [Indexed: 02/06/2023] Open
Abstract
The worldwide rise in prevalence of chronic kidney disease (CKD) demands innovative bio-medical solutions for millions of kidney patients. Kidney regenerative medicine aims to replenish tissue which is lost due to a common pathological pathway of fibrosis/inflammation and rejuvenate remaining tissue to maintain sufficient kidney function. To this end, cellular therapy strategies devised so far utilize kidney tissue-forming cells (KTFCs) from various cell sources, fetal, adult, and pluripotent stem-cells (PSCs). However, to increase engraftment and potency of the transplanted cells in a harsh hypoxic diseased environment, it is of importance to co-transplant KTFCs with vessel forming cells (VFCs). VFCs, consisting of endothelial cells (ECs) and mesenchymal stem-cells (MSCs), synergize to generate stable blood vessels, facilitating the vascularization of self-organizing KTFCs into renovascular units. In this paper, we review the different sources of KTFCs and VFCs which can be mixed, and report recent advances made in the field of kidney regeneration with emphasis on generation of vascularized kidney tissue by cell transplantation.
Collapse
Affiliation(s)
- Michael Namestnikov
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel Hashomer, Ramat Gan 52621, Israel;
- Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel;
- ediatric Nephrology Division, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel Hashomer, Ramat Gan 52621, Israel;
| | - Oren Pleniceanu
- Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel;
- The Kidney Research Lab, Institute of Nephrology and Hypertension, Sheba Medical Center, Tel Hashomer, Ramat Gan 52621, Israel
| | - Benjamin Dekel
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel Hashomer, Ramat Gan 52621, Israel;
- Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel;
| |
Collapse
|
52
|
Tsujimoto H, Kasahara T, Sueta SI, Araoka T, Sakamoto S, Okada C, Mae SI, Nakajima T, Okamoto N, Taura D, Nasu M, Shimizu T, Ryosaka M, Li Z, Sone M, Ikeya M, Watanabe A, Osafune K. A Modular Differentiation System Maps Multiple Human Kidney Lineages from Pluripotent Stem Cells. Cell Rep 2021; 31:107476. [PMID: 32268094 DOI: 10.1016/j.celrep.2020.03.040] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 01/17/2020] [Accepted: 03/13/2020] [Indexed: 02/08/2023] Open
Abstract
Recent studies using human pluripotent stem cells (hPSCs) have developed protocols to induce kidney-lineage cells and reconstruct kidney organoids. However, the separate generation of metanephric nephron progenitors (NPs), mesonephric NPs, and ureteric bud (UB) cells, which constitute embryonic kidneys, in in vitro differentiation culture systems has not been fully investigated. Here, we create a culture system in which these mesoderm-like cell types and paraxial and lateral plate mesoderm-like cells are separately generated from hPSCs. We recapitulate nephrogenic niches from separately induced metanephric NP-like and UB-like cells, which are subsequently differentiated into glomeruli, renal tubules, and collecting ducts in vitro and further vascularized in vivo. Our selective differentiation protocols should contribute to understanding the mechanisms underlying human kidney development and disease and also supply cell sources for regenerative therapies.
Collapse
Affiliation(s)
- Hiraku Tsujimoto
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Tomoko Kasahara
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Shin-Ichi Sueta
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Toshikazu Araoka
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Satoko Sakamoto
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Chihiro Okada
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan; Mitsubishi Space Software, 5-4-36 Tsukaguchi-honmachi, Amagasaki, Hyogo 661-0001, Japan
| | - Shin-Ichi Mae
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Taiki Nakajima
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Natsumi Okamoto
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Daisuke Taura
- Department of Medicine and Clinical Science, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Makoto Nasu
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Tatsuya Shimizu
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Makoto Ryosaka
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Zhongwei Li
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, 1333 San Pablo Street, MMR 618, Los Angeles, CA 90033, USA
| | - Masakatsu Sone
- Department of Medicine and Clinical Science, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Makoto Ikeya
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Akira Watanabe
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Kenji Osafune
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan.
| |
Collapse
|
53
|
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
|
54
|
Nishikawa M, Yanagawa N. Knockdown of Nuclear lncRNAs by Locked Nucleic Acid (LNA) Gapmers in Nephron Progenitor Cells. Methods Mol Biol 2021; 2161:29-36. [PMID: 32681503 DOI: 10.1007/978-1-0716-0680-3_3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Despite recent advance in our understanding on the role of long noncoding RNAs (lncRNAs), the function of the vast majority of lncRNAs remains poorly understood. To characterize the function of lncRNAs, knockdown studies are essential. However, the conventional silencing methods for mRNA, such as RNA interference (RNAi), may not be as efficient against lncRNAs, partly due to the mismatch of the localization of lncRNAs and RNAi machinery. To circumvent such limitation, a new technique has recently been developed, i.e., locked nucleic acid (LNA) gapmers. This system utilizes RNase H that distributes evenly in both nucleus and cytoplasm and is expected to knock down lncRNAs of interest more consistently regardless of their localization in the cell. In this chapter, we describe the procedure with tips to silence lncRNAs by LNA gapmers, by using mouse nephron progenitor cells as an example.
Collapse
Affiliation(s)
- Masaki Nishikawa
- School of Engineering, Chemical System Engineering, University of Tokyo, Tokyo, Japan.
| | - Norimoto Yanagawa
- Medical and Research Services, Greater Los Angeles Veterans Affairs Healthcare System at Sepulveda, North Hills, CA, USA.
- David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
55
|
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
|
56
|
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
|
57
|
The Renal Extracellular Matrix as a Supportive Scaffold for Kidney Tissue Engineering: Progress and Future Considerations. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1345:103-118. [PMID: 34582017 DOI: 10.1007/978-3-030-82735-9_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
During the past decades, diverse methods have been used toward renal tissue engineering in order to replace renal function. The goals of all these techniques included the recapitulation of renal filtration, re-absorptive, and secretary functions, and replacement of endocrine/metabolic activities. It is also imperative to develop a reliable, up scalable, and timely manufacturing process. Decellularization of the kidney with intact ECM is crucial for in-vivo compatibility and targeted clinical application. Contemporarily there is an increasing interest and research in the field of regenerative medicine including stem cell therapy and tissue bioengineering in search for new and reproducible sources of kidneys. In this chapter, we sought to determine the most effective method of renal decellularization and recellularization with emphasis on biologic composition and support of stem cell growth. Current barriers and limitations of bioengineered strategies will be also discussed, and strategies to overcome these are suggested.
Collapse
|
58
|
Chambers JM, Wingert RA. Advances in understanding vertebrate nephrogenesis. Tissue Barriers 2020; 8:1832844. [PMID: 33092489 PMCID: PMC7714473 DOI: 10.1080/21688370.2020.1832844] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 10/01/2020] [Accepted: 10/01/2020] [Indexed: 02/07/2023] Open
Abstract
The kidney is a complex organ that performs essential functions such as blood filtration and fluid homeostasis, among others. Recent years have heralded significant advancements in our knowledge of the mechanisms that control kidney formation. Here, we provide an overview of vertebrate renal development with a focus on nephrogenesis, the process of generating the epithelialized functional units of the kidney. These steps begin with intermediate mesoderm specification and proceed all the way to the terminally differentiated nephron cell, with many detailed stages in between. The establishment of nephron architecture with proper cellular barriers is vital throughout these processes. Continuously striving to gain further insights into nephrogenesis can ultimately lead to a better understanding and potential treatments for developmental maladies such as Congenital Anomalies of the Kidney and Urinary Tract (CAKUT).
Collapse
Affiliation(s)
- Joseph M. Chambers
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, University of Notre Dame, Notre Dame, IN, USA
| | - Rebecca A. Wingert
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, University of Notre Dame, Notre Dame, IN, USA
| |
Collapse
|
59
|
Basta JM, Singh AP, Robbins L, Stout L, Pherson M, Rauchman M. The core SWI/SNF catalytic subunit Brg1 regulates nephron progenitor cell proliferation and differentiation. Dev Biol 2020; 464:176-187. [PMID: 32504627 DOI: 10.1016/j.ydbio.2020.05.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 05/11/2020] [Accepted: 05/12/2020] [Indexed: 01/09/2023]
Abstract
Chromatin-remodeling complexes play critical roles in establishing gene expression patterns in response to developmental signals. How these epigenetic regulators determine the fate of progenitor cells during development of specific organs is not well understood. We found that genetic deletion of Brg1 (Smarca4), the core enzymatic protein in SWI/SNF, in nephron progenitor cells leads to severe renal hypoplasia. Nephron progenitor cells were depleted in Six2-Cre, Brg1flx/flx mice due to reduced cell proliferation. This defect in self-renewal, together with impaired differentiation resulted in a profound nephron deficit in Brg1 mutant kidneys. Sall1, a transcription factor that is required for expansion and maintenance of nephron progenitors, associates with SWI/SNF. Brg1 and Sall1 bind promoters of many progenitor cell genes and regulate expression of key targets that promote their proliferation.
Collapse
Affiliation(s)
- Jeannine M Basta
- John T. Milliken Department of Medicine, Division of Nephrology, Washington University School of Medicine, St. Louis, Mo 63110 USA
| | - Ajeet P Singh
- Division of Pediatric Hematology/Oncology, Departement of Pediatrics and Department of Biochemistry & Molecular Biology, Pennsylvania State University, Hershey, PA 17033 USA
| | - Lynn Robbins
- John T. Milliken Department of Medicine, Division of Nephrology, Washington University School of Medicine, St. Louis, Mo 63110 USA; VA St. Louis Health Care System, John Cochran Division, St. Louis, MO, 63106, USA
| | - Lisa Stout
- John T. Milliken Department of Medicine, Division of Nephrology, Washington University School of Medicine, St. Louis, Mo 63110 USA
| | - Michelle Pherson
- Department of Biochemistry & Molecular Biology, Saint Louis University, St. Louis, MO 63104 USA
| | - Michael Rauchman
- John T. Milliken Department of Medicine, Division of Nephrology, Washington University School of Medicine, St. Louis, Mo 63110 USA; VA St. Louis Health Care System, John Cochran Division, St. Louis, MO, 63106, USA; Deaprtememt of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110 USA.
| |
Collapse
|
60
|
Kumar Gupta A, Sarkar P, Wertheim JA, Pan X, Carroll TJ, Oxburgh L. Asynchronous mixing of kidney progenitor cells potentiates nephrogenesis in organoids. Commun Biol 2020; 3:231. [PMID: 32393756 PMCID: PMC7214420 DOI: 10.1038/s42003-020-0948-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 04/10/2020] [Indexed: 11/17/2022] Open
Abstract
A fundamental challenge in emulating kidney tissue formation through directed differentiation of human pluripotent stem cells is that kidney development is iterative, and to reproduce the asynchronous mix of differentiation states found in the fetal kidney we combined cells differentiated at different times in the same organoid. Asynchronous mixing promoted nephrogenesis, and heterochronic organoids were well vascularized when engrafted under the kidney capsule. Micro-CT and injection of a circulating vascular marker demonstrated that engrafted kidney tissue was connected to the systemic circulation by 2 weeks after engraftment. Proximal tubule glucose uptake was confirmed, but despite these promising measures of graft function, overgrowth of stromal cells prevented long-term study. We propose that this is a technical feature of the engraftment procedure rather than a specific shortcoming of the directed differentiation because kidney organoids derived from primary cells and whole embryonic kidneys develop similar stromal overgrowth when engrafted under the kidney capsule.
Collapse
Affiliation(s)
- Ashwani Kumar Gupta
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Jason A Wertheim
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL, USA
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
- Department of Surgery, Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Xinchao Pan
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Thomas J Carroll
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | |
Collapse
|
61
|
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
|
62
|
Vidal VP, Jian-Motamedi F, Rekima S, Gregoire EP, Szenker-Ravi E, Leushacke M, Reversade B, Chaboissier MC, Schedl A. R-spondin signalling is essential for the maintenance and differentiation of mouse nephron progenitors. eLife 2020; 9:53895. [PMID: 32324134 PMCID: PMC7228766 DOI: 10.7554/elife.53895] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Accepted: 04/23/2020] [Indexed: 02/06/2023] Open
Abstract
During kidney development, WNT/β-catenin signalling has to be tightly controlled to ensure proliferation and differentiation of nephron progenitor cells. Here, we show in mice that the signalling molecules RSPO1 and RSPO3 act in a functionally redundant manner to permit WNT/β-catenin signalling and their genetic deletion leads to a rapid decline of nephron progenitors. By contrast, tissue specific deletion in cap mesenchymal cells abolishes mesenchyme to epithelial transition (MET) that is linked to a loss of Bmp7 expression, absence of SMAD1/5 phosphorylation and a concomitant failure to activate Lef1, Fgf8 and Wnt4, thus explaining the observed phenotype on a molecular level. Surprisingly, the full knockout of LGR4/5/6, the cognate receptors of R-spondins, only mildly affects progenitor numbers, but does not interfere with MET. Taken together our data demonstrate key roles for R-spondins in permitting stem cell maintenance and differentiation and reveal Lgr-dependent and independent functions for these ligands during kidney formation.
Collapse
Affiliation(s)
- Valerie Pi Vidal
- Université Côte d'Azur, Inserm, CNRS, Institut de Biologie Valrose, Nice, France
| | - Fariba Jian-Motamedi
- Université Côte d'Azur, Inserm, CNRS, Institut de Biologie Valrose, Nice, France
| | - Samah Rekima
- Université Côte d'Azur, Inserm, CNRS, Institut de Biologie Valrose, Nice, France
| | - Elodie P Gregoire
- Université Côte d'Azur, Inserm, CNRS, Institut de Biologie Valrose, Nice, France
| | | | - Marc Leushacke
- Institute of Medical Biology, A*STAR, Singapore, Singapore
| | | | | | - Andreas Schedl
- Université Côte d'Azur, Inserm, CNRS, Institut de Biologie Valrose, Nice, France
| |
Collapse
|
63
|
Little MH, Lawlor KT. Recreating, expanding and using nephron progenitor populations. Nat Rev Nephrol 2020; 16:75-76. [PMID: 31811252 DOI: 10.1038/s41581-019-0238-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Melissa H Little
- Murdoch Children's Research Institute, Parkville, VIC, Australia. .,Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia. .,Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, Australia.
| | - Kynan T Lawlor
- Murdoch Children's Research Institute, Parkville, VIC, Australia
| |
Collapse
|
64
|
Huang B, Liu Z, Vonk A, Zeng Z, Li Z. Epigenetic regulation of kidney progenitor cells. Stem Cells Transl Med 2020; 9:655-660. [PMID: 32163228 PMCID: PMC7214665 DOI: 10.1002/sctm.19-0289] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 01/26/2020] [Accepted: 02/17/2020] [Indexed: 12/15/2022] Open
Abstract
The reciprocal interactions among the different embryonic kidney progenitor populations lay the basis for proper kidney organogenesis. During kidney development, three types of progenitor cells, including nephron progenitor cells, ureteric bud progenitor cells, and interstitial progenitor cells, generate the three major kidney structures—the nephrons, the collecting duct network, and the stroma, respectively. Epigenetic mechanisms are well recognized for playing important roles in organism development, in fine‐tuned control of physiological activities, and in responses to environment stimuli. Recently, evidence supporting the importance of epigenetic mechanisms underlying kidney organogenesis has emerged. In this perspective, we summarize the research progress and discuss the potential contribution of novel stem cell, organoid, and next‐generation sequencing tools in advancing this field in the future.
Collapse
Affiliation(s)
- Biao Huang
- Division of Nephrology and Hypertension, Department of Medicine and USC/UKRO Kidney Research Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Zhenqing Liu
- Division of Oral Biology and Medicine, School of Dentistry, UCLA, Los Angeles, California, USA
| | - Ariel Vonk
- Division of Nephrology and Hypertension, Department of Medicine and USC/UKRO Kidney Research Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Zipeng Zeng
- Division of Nephrology and Hypertension, Department of Medicine and USC/UKRO Kidney Research Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Zhongwei Li
- Division of Nephrology and Hypertension, Department of Medicine and USC/UKRO Kidney Research Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
65
|
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
|
66
|
Tsujimoto H, Araoka T, Nishi Y, Ohta A, Nakahata T, Osafune K. Small molecule TCS21311 can replace BMP7 and facilitate cell proliferation in in vitro expansion culture of nephron progenitor cells. Biochem Biophys Res Commun 2020; 558:231-238. [PMID: 32113685 DOI: 10.1016/j.bbrc.2020.02.130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 02/21/2020] [Indexed: 11/29/2022]
Abstract
Several groups have developed in vitro expansion cultures for mouse metanephric nephron progenitor cells (NPCs) using cocktails of small molecules and growth factors including BMP7. However, the detailed mechanisms by which BMP7 acts in the NPC expansion remain to be elucidated. Here, by performing chemical screening for BMP substitutes, we identified a small molecule, TCS21311, that can replace BMP7 and revealed a novel inhibitory role of BMP7 in JAK3-STAT3 signaling in NPC expansion culture. Further, we found that TCS21311 facilitates the proliferation of mouse embryonic NPCs and human induced pluripotent stem cell-derived NPCs when added to the expansion culture. These results will contribute to understanding the mechanisms of action of BMP7 in NPC proliferation in vitro and in vivo and to the stable supply of NPCs for regenerative therapy, disease modeling and drug discovery for kidney diseases.
Collapse
Affiliation(s)
- Hiraku Tsujimoto
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Toshikazu Araoka
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yohei Nishi
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Akira Ohta
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Tatsutoshi Nakahata
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Kenji Osafune
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| |
Collapse
|
67
|
Steichen C, Giraud S, Hauet T. Combining Kidney Organoids and Genome Editing Technologies for a Better Understanding of Physiopathological Mechanisms of Renal Diseases: State of the Art. Front Med (Lausanne) 2020; 7:10. [PMID: 32118002 PMCID: PMC7010937 DOI: 10.3389/fmed.2020.00010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 01/13/2020] [Indexed: 12/15/2022] Open
Abstract
Kidney organoids derived from pluripotent stem cells became a real alternative to the use of in vitro cellular models or in vivo animal models. Indeed, the comprehension of the key steps involved during kidney embryonic development led to the establishment of protocols enabling the differentiation of pluripotent stem cells into highly complex and organized structures, composed of various renal cell types. These organoids are linked with one major application based on iPSC technology advantage: the possibility to control iPSC genome, by selecting patients with specific disease or by genome editing tools such as CRISPR/Cas9 system. This allows the generation of kidney organoïds which recapitulate important physiopathological mechanisms such as cyst formation in renal polycystic disease for example. This review will focus on studies combining these both cutting edge technologies i.e., kidney organoid differentiation and genome editing and will describe what are the main advances performed in the comprehension of physiopathological mechanisms of renal diseases, as well as discuss remaining technical barriers and perspectives in the field.
Collapse
Affiliation(s)
- Clara Steichen
- INSERM U1082-IRTOMIT, Poitiers, France.,Université de Poitiers, Faculté de Médecine et de Pharmacie, Poitiers, France
| | - Sébastien Giraud
- INSERM U1082-IRTOMIT, Poitiers, France.,CHU Poitiers, Service de Biochimie, Poitiers, France
| | - Thierry Hauet
- INSERM U1082-IRTOMIT, Poitiers, France.,Université de Poitiers, Faculté de Médecine et de Pharmacie, Poitiers, France.,CHU Poitiers, Service de Biochimie, Poitiers, France
| |
Collapse
|
68
|
Nishikawa M, Sakai Y, Yanagawa N. Design and strategy for manufacturing kidney organoids. Biodes Manuf 2020. [DOI: 10.1007/s42242-020-00060-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
69
|
The FGF, TGFβ and WNT axis Modulate Self-renewal of Human SIX2 + Urine Derived Renal Progenitor Cells. Sci Rep 2020; 10:739. [PMID: 31959818 PMCID: PMC6970988 DOI: 10.1038/s41598-020-57723-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 12/31/2019] [Indexed: 12/22/2022] Open
Abstract
Human urine is a non-invasive source of renal stem cells with regeneration potential. Urine-derived renal progenitor cells were isolated from 10 individuals of both genders and distinct ages. These renal progenitors express pluripotency-associated proteins- TRA-1-60, TRA-1-81, SSEA4, C-KIT and CD133, as well as the renal stem cell markers -SIX2, CITED1, WT1, CD24 and CD106. The transcriptomes of all SIX2+ renal progenitors clustered together, and distinct from the human kidney biopsy-derived epithelial proximal cells (hREPCs). Stimulation of the urine-derived renal progenitor cells (UdRPCs) with the GSK3β-inhibitor (CHIR99021) induced differentiation. Transcriptome and KEGG pathway analysis revealed upregulation of WNT-associated genes- AXIN2, JUN and NKD1. Protein interaction network identified JUN- a downstream target of the WNT pathway in association with STAT3, ATF2 and MAPK1 as a putative negative regulator of self-renewal. Furthermore, like pluripotent stem cells, self-renewal is maintained by FGF2-driven TGFβ-SMAD2/3 pathway. The urine-derived renal progenitor cells and the data presented should lay the foundation for studying nephrogenesis in human.
Collapse
|
70
|
Glass NR, Takasako M, Er PX, Titmarsh DM, Hidalgo A, Wolvetang EJ, Little MH, Cooper-White JJ. Multivariate patterning of human pluripotent cells under perfusion reveals critical roles of induced paracrine factors in kidney organoid development. SCIENCE ADVANCES 2020; 6:eaaw2746. [PMID: 31934619 PMCID: PMC6949035 DOI: 10.1126/sciadv.aaw2746] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 10/30/2019] [Indexed: 06/10/2023]
Abstract
Creating complex multicellular kidney organoids from pluripotent stem cells shows great promise. Further improvements in differentiation outcomes, patterning, and maturation of specific cell types are, however, intrinsically limited by standard tissue culture approaches. We describe a novel full factorial microbioreactor array-based methodology to achieve rapid interrogation and optimization of this complex multicellular differentiation process in a facile manner. We successfully recapitulate early kidney tissue patterning events, exploring more than 1000 unique conditions in an unbiased and quantitative manner, and define new media combinations that achieve near-pure renal cell type specification. Single-cell resolution identification of distinct renal cell types within multilayered kidney organoids, coupled with multivariate analysis, defined the definitive roles of Wnt, fibroblast growth factor, and bone morphogenetic protein signaling in their specification, exposed retinoic acid as a minimal effector of nephron patterning, and highlighted critical contributions of induced paracrine factors on cell specification and patterning.
Collapse
Affiliation(s)
- Nick R. Glass
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Minoru Takasako
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia 4072, Australia
- Murdoch Children’s Research Institute, Flemington Rd., Parkville, VIC 3052, Australia
| | - Pei Xuan Er
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia 4072, Australia
- Murdoch Children’s Research Institute, Flemington Rd., Parkville, VIC 3052, Australia
| | - Drew M. Titmarsh
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Alejandro Hidalgo
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Ernst J. Wolvetang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, QLD 4072, Australia
- UQ Centre in Stem Cell and Regenerative Engineering, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Melissa H. Little
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia 4072, Australia
- Murdoch Children’s Research Institute, Flemington Rd., Parkville, VIC 3052, Australia
- Department of Pediatrics, University of Melbourne, Parkville, VIC 3052, Australia
| | - Justin J. Cooper-White
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, QLD 4072, Australia
- UQ Centre in Stem Cell and Regenerative Engineering, The University of Queensland, St. Lucia, QLD 4072, Australia
- Biomedical Manufacturing, Manufacturing Flagship, CSIRO, Clayton, VIC 3169, Australia
- School of Chemical Engineering, The University of Queensland, St. Lucia, QLD 4072, Australia
| |
Collapse
|
71
|
Kim AD, Lake BB, Chen S, Wu Y, Guo J, Parvez RK, Tran T, Thornton ME, Grubbs B, McMahon JA, Zhang K, McMahon AP. Cellular Recruitment by Podocyte-Derived Pro-migratory Factors in Assembly of the Human Renal Filter. iScience 2019; 20:402-414. [PMID: 31622881 PMCID: PMC6817668 DOI: 10.1016/j.isci.2019.09.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 08/21/2019] [Accepted: 09/23/2019] [Indexed: 12/20/2022] Open
Abstract
Analysis of kidney disease-causing genes and pathology resulting from systemic diseases highlight the importance of the kidney's filtering system, the renal corpuscles. To elucidate the developmental processes that establish the renal corpuscle, we performed single-nucleus droplet-based sequencing of the human fetal kidney. This enabled the identification of nephron, interstitial, and vascular cell types that together generate the renal corpuscles. Trajectory analysis identified transient developmental gene expression, predicting precursors or mature podocytes express FBLN2, BMP4, or NTN4, in conjunction with recruitment, differentiation, and modeling of vascular and mesangial cell types into a functional filter. In vitro studies provide evidence that these factors exhibit angiogenic or mesangial recruiting and inductive properties consistent with a key organizing role for podocyte precursors in kidney development. Together these studies define a spatiotemporal developmental program for the primary filtration unit of the human kidney and provide novel insights into cell interactions regulating co-assembly of constituent cell types.
Collapse
Affiliation(s)
- 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 90089, USA
| | - Blue B Lake
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Song Chen
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Yan Wu
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, 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 90089, 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 90089, USA
| | - Tracy Tran
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Matthew E Thornton
- Maternal Fetal Medicine Division, University of Southern California, Los Angeles, CA, USA
| | - Brendan Grubbs
- Maternal Fetal Medicine Division, 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 90089, USA
| | - Kun Zhang
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, 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 90089, USA.
| |
Collapse
|
72
|
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
|
73
|
Faraj R, Irizarry-Alfonzo A, Puri P. Molecular characterization of nephron progenitors and their early epithelial derivative structures in the nephrogenic zone of the canine fetal kidney. Eur J Histochem 2019; 63. [PMID: 31544449 PMCID: PMC6763752 DOI: 10.4081/ejh.2019.3049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 09/10/2019] [Indexed: 01/14/2023] Open
Abstract
Nephron progenitors (NPs) and nephrogenesis have been extensively studied in mice and humans and have provided insights into the mechanisms of renal development, disease and possibility of NP-based therapies. However, molecular features of NPs and their derivatives in the canine fetal kidney (CFK) remain unknown. This study was focused to characterize the expression of potential markers of canine NPs and their derivatives by immuno-fluorescence and western blot analysis. Transcription factors (TFs) SIX1 and SIX2, well-characterized human NP markers, were expressed in NPs surrounding the ureteric bud in the CFK. Canine NPs also expressed ITGA8 and NCAM1, surface markers previously used to isolate NPs from the mouse and human fetal kidneys. TF, PAX2 was detected in the ureteric bud, NPs and their derivative structures such as renal vesicle and S-shaped body. This study highlights the similarities in dog, mouse and human renal development and characterizes markers to identify canine NPs and their derivatives. These results will facilitate the isolation of canine NPs and their functional characterization to develop NP-based therapies for canine renal diseases.
Collapse
Affiliation(s)
- Rawah Faraj
- Department of Biomedical Sciences, College of Veterinary Medicine, Tuskegee University, Tuskegee.
| | | | | |
Collapse
|
74
|
Li Q, Cui S, Ma Q, Liu Y, Yu H, Geng G, Agborbesong E, Ren C, Wei K, Zhang Y, Yang J, Bai X, Cai G, Xie Y, Li X, Chen X. Disruption of Robo2-Baiap2 integrated signaling drives cystic disease. JCI Insight 2019; 4:127602. [PMID: 31534052 DOI: 10.1172/jci.insight.127602] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 08/21/2019] [Indexed: 11/17/2022] Open
Abstract
Hereditary renal cystic diseases are characterized by defects in primary cilia of renal tubular epithelial cells and abnormality of tubular epithelium, which ultimately result in the development of renal cysts. However, the mechanism leading from abnormality of the tubular epithelium to cystogenesis is not well understood. In this report, we demonstrate a critical role for Robo2 in regulating epithelial development, including ciliogenesis, polarization, and differentiation. We found that Robo2 deficiency results in cystic kidneys, and the cyst cells showed defective cilia and polarity defects in tubular epithelium. The cyst cells, less than terminally differentiated, continue to proliferate. We further established that Robo2 works with p53 as well as polarity and ciliary proteins (Par3, PKCς, ZO-2, and Claudin-2) to regulate these processes. Robo2 binds to Baiap2 (also known as IRSp53) through the IRSp53/MIM homology domain in renal epithelial cells. This binding allows Robo2 to phosphorylate MDM2 at Ser166 via Baiap2 and maintain p53 homeostasis. Disruption of the Robo2-Baiap2 complex causes MDM2 to be subjected to dephosphorylation, leading to a high level of active p53, and initiated p53-mediated cellular senescence via p21 and decreased the expression of ZO-1, ZO-2, PKCς, Par3, and Claudin-2 proteins, resulting in defects in epithelial development, including ciliogenesis, polarization, and differentiation. Importantly, double knockout of Robo2 and p53 rescued all the epithelial defects in kidneys compared with those in Robo2-knockout kidneys. Taken together, the present results demonstrate that Robo2 deficiency causes renal cystic disease, which is largely dependent on defective Robo2-Baiap2 integrated signaling in kidneys.
Collapse
Affiliation(s)
- Qinggang Li
- Department of Nephrology, Chinese PLA General Hospital, Medical School of Chinese PLA, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| | - Shaoyuan Cui
- Department of Nephrology, Chinese PLA General Hospital, Medical School of Chinese PLA, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| | - Qian Ma
- Department of Nephrology, Chinese PLA General Hospital, Medical School of Chinese PLA, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| | - Ying Liu
- Department of Nephrology, Chinese PLA General Hospital, Medical School of Chinese PLA, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| | - Hongyu Yu
- Department of Nephrology, Chinese PLA General Hospital, Medical School of Chinese PLA, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| | - GuangRui Geng
- Department of Nephrology, Chinese PLA General Hospital, Medical School of Chinese PLA, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| | - Ewud Agborbesong
- Department of Internal Medicine, Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Chongyu Ren
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Kai Wei
- Department of Nephrology, Chinese PLA General Hospital, Medical School of Chinese PLA, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| | - Yingjie Zhang
- Department of Nephrology, Chinese PLA General Hospital, Medical School of Chinese PLA, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| | - Jurong Yang
- Department of Urology, Third Affiliated Hospital of Chongqing Medical University (General Hospital), Chongqing, China
| | - Xueyuan Bai
- Department of Nephrology, Chinese PLA General Hospital, Medical School of Chinese PLA, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| | - Guangyan Cai
- Department of Nephrology, Chinese PLA General Hospital, Medical School of Chinese PLA, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| | - Yuansheng Xie
- Department of Nephrology, Chinese PLA General Hospital, Medical School of Chinese PLA, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| | - Xiaogang Li
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Xiangmei Chen
- Department of Nephrology, Chinese PLA General Hospital, Medical School of Chinese PLA, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| |
Collapse
|
75
|
Dapkunas A, Rantanen V, Gui Y, Lalowski M, Sainio K, Kuure S, Sariola H. Simple 3D culture of dissociated kidney mesenchyme mimics nephron progenitor niche and facilitates nephrogenesis Wnt-independently. Sci Rep 2019; 9:13433. [PMID: 31530822 PMCID: PMC6748995 DOI: 10.1038/s41598-019-49526-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 08/21/2019] [Indexed: 02/06/2023] Open
Abstract
Kidney mesenchyme (KM) and nephron progenitors (NPs) depend on WNT activity, and their culture in vitro requires extensive repertoire of recombinant proteins and chemicals. Here we established a robust, simple culture of mouse KM using a combination of 3D Matrigel and growth media supplemented with Fibroblast Growth Factor 2 (FGF2) and Src inhibitor PP2. This allows dissociated KM to spontaneously self-organize into spheres. To reassess the requirement of WNT activity in KM self-organization and NPs maintenance, cells were cultured with short pulse of high-dose GSK3β inhibitor BIO, on a constant low-dose or without BIO. Robust proliferation at 48 hours and differentiation at 1 week were observed in cultures with high BIO pulse. Importantly, dissociated KM cultured without BIO, similarly to that exposed to constant low dose of BIO, maintained NPs up to one week and spontaneously differentiated into nephron tubules at 3 weeks of culture. Our results show that KM is maintained and induced to differentiate in a simple culture system. They also imply that GSK3β/WNT-independent pathways contribute to the maintenance and induction of mouse KM. The robust and easy 3D culture enables further characterization of NPs, and may facilitate disease modeling when applied to human cells.
Collapse
Affiliation(s)
- Arvydas Dapkunas
- Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, FI-00014, Helsinki, Finland. .,Meilahti Clinical Proteomics Core Facility, Helsinki Institute of Life Science, University of Helsinki, FI-00014, Helsinki, Finland.
| | - Ville Rantanen
- Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, FI-00014, Helsinki, Finland.,Genome-Scale Biology Research Program, Faculty of Medicine, University of Helsinki, FI-00014, Helsinki, Finland
| | - Yujuan Gui
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, FI-00014, Helsinki, Finland
| | - Maciej Lalowski
- Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, FI-00014, Helsinki, Finland.,Meilahti Clinical Proteomics Core Facility, Helsinki Institute of Life Science, University of Helsinki, FI-00014, Helsinki, Finland
| | - Kirsi Sainio
- Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, FI-00014, Helsinki, Finland
| | - Satu Kuure
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, FI-00014, Helsinki, Finland.,GM-unit, Laboratory Animal Centre, Helsinki Institute of Life Science, University of Helsinki, FI-00014, Helsinki, Finland
| | - Hannu Sariola
- Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, FI-00014, Helsinki, Finland
| |
Collapse
|
76
|
Vanslambrouck JM, Wilson SB, Tan KS, Soo JYC, Scurr M, Spijker HS, Starks LT, Neilson A, Cui X, Jain S, Little MH, Howden SE. A Toolbox to Characterize Human Induced Pluripotent Stem Cell-Derived Kidney Cell Types and Organoids. J Am Soc Nephrol 2019; 30:1811-1823. [PMID: 31492807 DOI: 10.1681/asn.2019030303] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 07/25/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The generation of reporter lines for cell identity, lineage, and physiologic state has provided a powerful tool in advancing the dissection of mouse kidney morphogenesis at a molecular level. Although use of this approach is not an option for studying human development in vivo, its application in human induced pluripotent stem cells (iPSCs) is now feasible. METHODS We used CRISPR/Cas9 gene editing to generate ten fluorescence reporter iPSC lines designed to identify nephron progenitors, podocytes, proximal and distal nephron, and ureteric epithelium. Directed differentiation to kidney organoids was performed according to published protocols. Using immunofluorescence and live confocal microscopy, flow cytometry, and cell sorting techniques, we investigated organoid patterning and reporter expression characteristics. RESULTS Each iPSC reporter line formed well patterned kidney organoids. All reporter lines showed congruence of endogenous gene and protein expression, enabling isolation and characterization of kidney cell types of interest. We also demonstrated successful application of reporter lines for time-lapse imaging and mouse transplantation experiments. CONCLUSIONS We generated, validated, and applied a suite of fluorescence iPSC reporter lines for the study of morphogenesis within human kidney organoids. This fluorescent iPSC reporter toolbox enables the visualization and isolation of key populations in forming kidney organoids, facilitating a range of applications, including cellular isolation, time-lapse imaging, protocol optimization, and lineage-tracing approaches. These tools offer promise for enhancing our understanding of this model system and its correspondence with human kidney morphogenesis.
Collapse
Affiliation(s)
| | - Sean B Wilson
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Ker Sin Tan
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Joanne Y-C Soo
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Michelle Scurr
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - H Siebe Spijker
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | - Lakshi T Starks
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Amber Neilson
- Department of Genetics, Genome Engineering and iPSC Center and
| | - Xiaoxia Cui
- Department of Genetics, Genome Engineering and iPSC Center and
| | - Sanjay Jain
- Department of Medicine, Kidney Translational Research Center, Washington University School of Medicine, St. Louis, Missouri; and
| | - Melissa Helen Little
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia; .,Faculty of Medicine, Dentistry and Health Sciences, Department of Paediatrics and.,Department of Anatomy and Neuroscience, University of Melbourne, Victoria, Australia
| | - Sara E Howden
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Faculty of Medicine, Dentistry and Health Sciences, Department of Paediatrics and
| |
Collapse
|
77
|
Islam M, Nishinakamura R. How to rebuild the kidney: recent advances in kidney organoids. J Biochem 2019; 166:7-12. [PMID: 30847486 DOI: 10.1093/jb/mvz021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 03/06/2019] [Indexed: 01/31/2023] Open
Abstract
It is difficult to restore kidney function once it has become severely impaired. Although kidney transplantation is a curative therapy, donor numbers remain limited. Thus, the generation of kidney organoids (mainly comprising glomeruli and renal tubules) from multipotent stem cells represents an important advance in regenerative medicine of the kidney. Recently, a protocol that can generate the higher-order structure of the mouse embryonic kidney was reported. Kidney organoids are now being used for disease modelling, and may eventually be applicable for clinical transplantation. In this review, we summarize the recent advances in kidney organoid research, and discuss the issues to be resolved toward kidney reconstruction.
Collapse
Affiliation(s)
- Mazharul Islam
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Ryuichi Nishinakamura
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
78
|
Abstract
The worldwide increase in the number of patients with end-stage renal disease leads to a growing waiting list for kidney transplantation resulting from the scarcity of kidney donors. Therefore, alternative treatment options for patients with end-stage renal disease are being sought. In vitro differentiation of stem cells into renal tissue is a promising approach to repair nonfunctional kidney tissue. Impressive headway has been made in the use of stem cells with the use of adult renal progenitor cells, embryonic stem cells, and induced pluripotent stem cells for the development toward primitive kidney structures. Currently, efforts are directed at improving long-term maintenance and stability of the cells. This review aims to provide a comprehensive overview of the cell sources used for the generation of kidney cells and strategies used for transplantation in in vivo models. Furthermore, it provides a perspective on stability and safety during future clinical application of in vitro generated kidney cells.
Collapse
|
79
|
Abstract
Kidney organoids are regarded as important tools with which to study the development of the normal and diseased human kidney. Since the first reports of human pluripotent stem cell-derived kidney organoids 5 years ago, kidney organoids have been successfully used to model glomerular and tubular diseases. In parallel, advances in single-cell RNA sequencing have led to identification of a variety of cell types in the organoids, and have shown these to be similar to, but more immature than, human kidney cells in vivo. Protocols for the in vitro expansion of stem cell-derived nephron progenitor cells (NPCs), as well as those for the selective induction of specific lineages, especially glomerular podocytes, have also been reported. Although most current organoids are based on the induction of NPCs, an induction protocol for ureteric buds (collecting duct precursors) has also been developed, and approaches to generate more complex kidney structures may soon be possible. Maturation of organoids is a major challenge, and more detailed analysis of the developing kidney at a single cell level is needed. Eventually, organotypic kidney structures equipped with nephrons, collecting ducts, ureters, stroma and vascular flow are required to generate transplantable kidneys; such attempts are in progress.
Collapse
|
80
|
Activin Is Superior to BMP7 for Efficient Maintenance of Human iPSC-Derived Nephron Progenitors. Stem Cell Reports 2019; 13:322-337. [PMID: 31378669 PMCID: PMC6700502 DOI: 10.1016/j.stemcr.2019.07.003] [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] [Received: 08/17/2018] [Revised: 07/01/2019] [Accepted: 07/01/2019] [Indexed: 12/29/2022] Open
Abstract
Kidney formation is regulated by the balance between maintenance and differentiation of nephron progenitor cells (NPCs). Now that directed differentiation of NPCs from human induced pluripotent stem cells (iPSCs) can be achieved, maintenance and propagation of NPCs in vitro should be beneficial for regenerative medicine. Although WNT and FGF signals were previously shown to be essential for NPC propagation, the requirement for BMP/TGFβ signaling remains controversial. Here we reveal that activin has superior effects to BMP7 on maintenance efficiency of human iPSC-derived NPCs. Activin expanded ITGA8+/PDGFRA-/SIX2-GFP+ NPCs by 5-fold per week at 80%-90% efficiency, and the propagated cells possessed robust capacity for nephron formation both in vitro and in vivo. The expanded cells also maintained their nephron-forming potential after freezing. Furthermore, the protocol was applicable to multiple non-GFP-tagged iPSC lines. Thus, our activin-based protocol will be applicable to a variety of research fields including disease modeling and drug screening.
Collapse
|
81
|
Cargill K, Hemker SL, Clugston A, Murali A, Mukherjee E, Liu J, Bushnell D, Bodnar AJ, Saifudeen Z, Ho J, Bates CM, Kostka D, Goetzman ES, Sims-Lucas S. Von Hippel-Lindau Acts as a Metabolic Switch Controlling Nephron Progenitor Differentiation. J Am Soc Nephrol 2019; 30:1192-1205. [PMID: 31142573 PMCID: PMC6622426 DOI: 10.1681/asn.2018111170] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 04/01/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Nephron progenitors, the cell population that give rise to the functional unit of the kidney, are metabolically active and self-renew under glycolytic conditions. A switch from glycolysis to mitochondrial respiration drives these cells toward differentiation, but the mechanisms that control this switch are poorly defined. Studies have demonstrated that kidney formation is highly dependent on oxygen concentration, which is largely regulated by von Hippel-Lindau (VHL; a protein component of a ubiquitin ligase complex) and hypoxia-inducible factors (a family of transcription factors activated by hypoxia). METHODS To explore VHL as a regulator defining nephron progenitor self-renewal versus differentiation, we bred Six2-TGCtg mice with VHLlox/lox mice to generate mice with a conditional deletion of VHL from Six2+ nephron progenitors. We used histologic, immunofluorescence, RNA sequencing, and metabolic assays to characterize kidneys from these mice and controls during development and up to postnatal day 21. RESULTS By embryonic day 15.5, kidneys of nephron progenitor cell-specific VHL knockout mice begin to exhibit reduced maturation of nephron progenitors. Compared with controls, VHL knockout kidneys are smaller and developmentally delayed by postnatal day 1, and have about half the number of glomeruli at postnatal day 21. VHL knockout nephron progenitors also exhibit persistent Six2 and Wt1 expression, as well as decreased mitochondrial respiration and prolonged reliance on glycolysis. CONCLUSIONS Our findings identify a novel role for VHL in mediating nephron progenitor differentiation through metabolic regulation, and suggest that VHL is required for normal kidney development.
Collapse
Affiliation(s)
- Kasey Cargill
- Rangos Research Center, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Shelby L Hemker
- Rangos Research Center, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Andrew Clugston
- Rangos Research Center, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Developmental Biology and
| | - Anjana Murali
- Rangos Research Center, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Elina Mukherjee
- Rangos Research Center, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Jiao Liu
- Section of Pediatric Nephrology, Department of Pediatrics and
- The Hypertension and Renal Centers of Excellence, Tulane University Health Sciences Center, New Orleans, Louisiana
| | - Daniel Bushnell
- Rangos Research Center, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Andrew J Bodnar
- Rangos Research Center, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Zubaida Saifudeen
- Section of Pediatric Nephrology, Department of Pediatrics and
- The Hypertension and Renal Centers of Excellence, Tulane University Health Sciences Center, New Orleans, Louisiana
| | - Jacqueline Ho
- Rangos Research Center, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Carlton M Bates
- Rangos Research Center, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Dennis Kostka
- Rangos Research Center, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Developmental Biology and
| | - Eric S Goetzman
- Rangos Research Center, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Division of Medical Genetics, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Sunder Sims-Lucas
- Rangos Research Center, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania;
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
82
|
Cruz NM, Freedman BS. Differentiation of human kidney organoids from pluripotent stem cells. Methods Cell Biol 2019; 153:133-150. [PMID: 31395376 DOI: 10.1016/bs.mcb.2019.05.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
It is now possible to direct the differentiation of human pluripotent stem cells into three-dimensional nephron-like structures called kidney organoids. Organoids contain proximal and distal tubules as well as podocytes, in addition to a variety of other lineages such as endothelial cells. Organoid technology has great potential for kidney regeneration and has already been proven to be suitable for modeling kidney disease. However, the methodologies that are used for the generation of kidney organoids require expertise and can be daunting for the inexperienced. Here, we describe in detail a well-established and relatively simple method for the generation of human kidney organoids. We include notes on technical and design considerations for these experiments, and highlight key advantages and limitations of the system.
Collapse
Affiliation(s)
- Nelly M Cruz
- Department of Medicine, Division of Nephrology, University of Washington School of Medicine, Seattle, WA, United States; Kidney Research Institute, University of Washington School of Medicine, Seattle, WA, United States; Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, WA, United States; Department of Pathology, University of Washington School of Medicine, Seattle, WA, United States
| | - Benjamin S Freedman
- Department of Medicine, Division of Nephrology, University of Washington School of Medicine, Seattle, WA, United States; Kidney Research Institute, University of Washington School of Medicine, Seattle, WA, United States; Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, WA, United States; Department of Pathology, University of Washington School of Medicine, Seattle, WA, United States.
| |
Collapse
|
83
|
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
|
84
|
Abstract
This review focus on kidney organoids derived from pluripotent stem cells, which become a real alternative to the use of in vitro cellular models or in vivo animals models. The comprehension of the key steps involved during kidney embryonic development led to the establishment of protocols enabling the differentiation of pluripotent stem cells into kidney organoids that are highly complex and organized structures, composed of various renal cell types. These mini-organs are endowed with major applications: the possibility to control iPSC genome (by selecting patients with specific disease or by genome editing) allows the generation of kidney organoïds which recapitulate important physiopathological mechanisms such as cyste formation in renal polycystic disease. Kidney organoids can also be used in high-throughput screening to fasten the screening of nephrotoxic/therapeutic compounds. Finally, kidney organoids have a huge interest in the context of tissue repair, which remains for now a challenging goal linked with technological barriers that need still to be overcome.
Collapse
Affiliation(s)
- Clara Steichen
- Inserm U1082 - IRTOMIT (Ischémie reperfusion en transplantation d'organes mécanismes et innovations thérapeutiques), Poitiers, F-86000, France - Université de Poitiers, Faculté de médecine et de pharmacie, Poitiers, F-86000, France
| | - Sébastien Giraud
- Inserm U1082 - IRTOMIT (Ischémie reperfusion en transplantation d'organes mécanismes et innovations thérapeutiques), Poitiers, F-86000, France - CHU de Poitiers, service de biochimie, Poitiers, F-86000, France
| | - Thierry Hauet
- Inserm U1082 - IRTOMIT (Ischémie reperfusion en transplantation d'organes mécanismes et innovations thérapeutiques), Poitiers, F-86000, France - Université de Poitiers, Faculté de médecine et de pharmacie, Poitiers, F-86000, France - CHU de Poitiers, service de biochimie, Poitiers, F-86000, France
| |
Collapse
|
85
|
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
|
86
|
Gupta AK, Coburn JM, Davis-Knowlton J, Kimmerling E, Kaplan DL, Oxburgh L. Scaffolding kidney organoids on silk. J Tissue Eng Regen Med 2019; 13:812-822. [PMID: 30793851 PMCID: PMC6529243 DOI: 10.1002/term.2830] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 02/10/2019] [Accepted: 02/21/2019] [Indexed: 12/26/2022]
Abstract
End stage kidney disease affects hundreds of thousands of patients in the United States. The therapy of choice is kidney replacement, but availability of organs is limited, and alternative sources of tissue are needed. Generation of new kidney tissue in the laboratory has been made possible through pluripotent cell reprogramming and directed differentiation. In current procedures, aggregates of cells known as organoids are grown either submerged or at the air-liquid interface. These studies have demonstrated that kidney tissue can be generated from pluripotent stem cells, but they also identify limitations. The first is that perfusion of cell aggregates is limited, restricting the size to which they can be grown. The second is that aggregates lack the structural integrity required for convenient engraftment and suturing or adhesion to regions of kidney injury. In this study, we evaluated the capacity of silk to serve as a support for the growth and differentiation of kidney tissue from primary cells and from human induced pluripotent stem cells. We find that cells can differentiate to epithelia characteristic of the developing kidney on this material and that these structures are maintained following engraftment under the capsule of the adult kidney. Blood vessel investment can be promoted by the addition of vascular endothelial growth factor to the scaffold, but the proliferation of stromal cells within the graft presents a challenge, which will require some readjustment of cell growth and differentiation conditions. In summary, we find that silk can be used to support growth of stem cell derived kidney tissue.
Collapse
Affiliation(s)
| | | | - Jessica Davis-Knowlton
- Maine Medical Center Research Institute, Scarborough, ME 04074
- Tufts University School of Medicine, Boston, MA 02111
| | | | | | - Leif Oxburgh
- Maine Medical Center Research Institute, Scarborough, ME 04074
| |
Collapse
|
87
|
Mansoori-Moghadam Z, Totonchi M, Hesaraki M, Aghdami N, Baharvand H, Moghadasali R. Programming of ES cells and reprogramming of fibroblasts into renal lineage-like cells. Exp Cell Res 2019; 379:225-234. [PMID: 30981668 DOI: 10.1016/j.yexcr.2019.04.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 04/06/2019] [Accepted: 04/08/2019] [Indexed: 01/08/2023]
Abstract
This study aims to prepare intermediate mesoderm-like cells from mouse embryonic fibroblasts (MEFs). In the first step, intermediate mesoderm-like cells (IMLCs) and renal epithelial-like cells (RELCs) were extracted from mouse embryonic stem cells (mESCs) in a specified media that contained two small molecules, CHIR99021 and TTNPB, along with growth factors, FGF9and BMP7. Then, MEFs were directly converted into IM by genes for the pluripotency factors, which encode the transcription factors; Oct4, Sox2, Klf4, and c-Myc (OSKM). These unstable intermediate cells were quickly encouraged to form IM with the assistance of CHIR99021 and TTNPB. The results showed that exogenous expression of OSKM factors for four days was adequate to generate partially reprogrammed cells (SSEA1+/Nanog-). Real-time PCR and immunocytochemistry analysis confirmed the presence of the MEF-derived IMs. This study introduced a method for mESCs differentiation to RELCs followed by MEF conversion in an attempt to generate IM by circumventing pluripotency.
Collapse
Affiliation(s)
- Zohreh Mansoori-Moghadam
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Mehdi Totonchi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Mahdi Hesaraki
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Nasser Aghdami
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Reza Moghadasali
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
88
|
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
|
89
|
Phua YL, Chen KH, Hemker SL, Marrone AK, Bodnar AJ, Liu X, Clugston A, Kostka D, Butterworth MB, Ho J. Loss of miR-17~92 results in dysregulation of Cftr in nephron progenitors. Am J Physiol Renal Physiol 2019; 316:F993-F1005. [PMID: 30838872 DOI: 10.1152/ajprenal.00450.2018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
We have previously demonstrated that loss of miR-17~92 in nephron progenitors in a mouse model results in renal hypodysplasia and chronic kidney disease. Clinically, decreased congenital nephron endowment because of renal hypodysplasia is associated with an increased risk of hypertension and chronic kidney disease, and this is at least partly dependent on the self-renewal of nephron progenitors. Here, we present evidence for a novel molecular mechanism regulating the self-renewal of nephron progenitors and congenital nephron endowment by the highly conserved miR-17~92 cluster. Whole transcriptome sequencing revealed that nephron progenitors lacking this cluster demonstrated increased Cftr expression. We showed that one member of the cluster, miR-19b, is sufficient to repress Cftr expression in vitro and that perturbation of Cftr activity in nephron progenitors results in impaired proliferation. Together, these data suggest that miR-19b regulates Cftr expression in nephron progenitors, with this interaction playing a role in appropriate nephron progenitor self-renewal during kidney development to generate normal nephron endowment.
Collapse
Affiliation(s)
- Yu Leng Phua
- Rangos Research Center, UPMC Children's Hospital of Pittsburgh , Pittsburgh, Pennsylvania.,Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Kevin Hong Chen
- Rangos Research Center, UPMC Children's Hospital of Pittsburgh , Pittsburgh, Pennsylvania.,Department of Biological Sciences, Carnegie Mellon University , Pittsburgh, Pennsylvania
| | - Shelby L Hemker
- Rangos Research Center, UPMC Children's Hospital of Pittsburgh , Pittsburgh, Pennsylvania.,Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - April K Marrone
- Rangos Research Center, UPMC Children's Hospital of Pittsburgh , Pittsburgh, Pennsylvania.,Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Andrew J Bodnar
- Rangos Research Center, UPMC Children's Hospital of Pittsburgh , Pittsburgh, Pennsylvania.,Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Xiaoning Liu
- Department of Cell Biology, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Andrew Clugston
- Rangos Research Center, UPMC Children's Hospital of Pittsburgh , Pittsburgh, Pennsylvania.,Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania.,Department of Developmental Biology and Department of Computational and Systems Biology, Pittsburgh Center for Evolutionary Biology and Medicine, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Dennis Kostka
- Department of Developmental Biology and Department of Computational and Systems Biology, Pittsburgh Center for Evolutionary Biology and Medicine, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Michael B Butterworth
- Department of Cell Biology, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Jacqueline Ho
- Rangos Research Center, UPMC Children's Hospital of Pittsburgh , Pittsburgh, Pennsylvania.,Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| |
Collapse
|
90
|
Vanslambrouck JM, Woodard LE, Suhaimi N, Williams FM, Howden SE, Wilson SB, Lonsdale A, Er PX, Li J, Maksimovic J, Oshlack A, Wilson MH, Little MH. Direct reprogramming to human nephron progenitor-like cells using inducible piggyBac transposon expression of SNAI2-EYA1-SIX1. Kidney Int 2019; 95:1153-1166. [PMID: 30827514 DOI: 10.1016/j.kint.2018.11.041] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/15/2018] [Accepted: 11/21/2018] [Indexed: 01/01/2023]
Abstract
All nephrons in the mammalian kidney arise from a transient nephron progenitor population that is lost close to the time of birth. The generation of new nephron progenitors and their maintenance in culture are central to the success of kidney regenerative strategies. Using a lentiviral screening approach, we previously generated a human induced nephron progenitor-like state in vitro using a pool of six transcription factors. Here, we sought to develop a more efficient approach for direct reprogramming of human cells that could be applied in vivo. PiggyBac transposons are a non-viral integrating gene delivery system that is suitable for in vivo use and allows for simultaneous delivery of multiple genes. Using an inducible piggyBac transposon system, we optimized a protocol for the direct reprogramming of HK2 cells to induced nephron progenitor-like cells with expression of only 3 transcription factors (SNAI2, EYA1, and SIX1). Culture in conditions supportive of the nephron progenitor state further increased the expression of nephron progenitor genes. The refined protocol was then applied to primary human renal epithelial cells, which integrated into developing nephron structures in vitro and in vivo. Such inducible reprogramming to nephron progenitor-like cells could facilitate direct cellular reprogramming for kidney regeneration.
Collapse
Affiliation(s)
- Jessica M Vanslambrouck
- Murdoch Children's Research Institute, Parkville, Melbourne, Australia; Division of Genomics of Development and Disease, Institute for Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Lauren E Woodard
- Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, Tennessee, USA; Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Norseha Suhaimi
- Division of Genomics of Development and Disease, Institute for Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Felisha M Williams
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Sara E Howden
- Murdoch Children's Research Institute, Parkville, Melbourne, Australia; Department of Pediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Australia
| | - Sean B Wilson
- Murdoch Children's Research Institute, Parkville, Melbourne, Australia
| | - Andrew Lonsdale
- Murdoch Children's Research Institute, Parkville, Melbourne, Australia
| | - Pei X Er
- Murdoch Children's Research Institute, Parkville, Melbourne, Australia
| | - Joan Li
- Division of Genomics of Development and Disease, Institute for Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Jovana Maksimovic
- Murdoch Children's Research Institute, Parkville, Melbourne, Australia
| | - Alicia Oshlack
- Murdoch Children's Research Institute, Parkville, Melbourne, Australia
| | - Matthew H Wilson
- Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, Tennessee, USA; Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Melissa H Little
- Murdoch Children's Research Institute, Parkville, Melbourne, Australia; Division of Genomics of Development and Disease, Institute for Molecular Biosciences, The University of Queensland, Brisbane, Australia; Department of Pediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Australia.
| |
Collapse
|
91
|
Affiliation(s)
- Mo Li
- From the King Abdullah University of Science and Technology, Thuwal, Saudi Arabia (M.L.); and the Salk Institute for Biological Studies, La Jolla, CA (J.C.I.B.)
| | - Juan C Izpisua Belmonte
- From the King Abdullah University of Science and Technology, Thuwal, Saudi Arabia (M.L.); and the Salk Institute for Biological Studies, La Jolla, CA (J.C.I.B.)
| |
Collapse
|
92
|
Naganuma H, Nishinakamura R. From organoids to transplantable artificial kidneys. Transpl Int 2019; 32:563-570. [PMID: 30710378 DOI: 10.1111/tri.13404] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 12/13/2018] [Accepted: 01/28/2019] [Indexed: 12/16/2022]
Abstract
It is difficult to restore kidney function following chronic kidney damage. Although dialysis is currently used to treat patients with chronic kidney disease, it does not cure the disease, while severely restricting the patient's daily and social activities. Kidney transplantation is an alternative and curative therapy, but donor numbers remain limited. However, the generation of kidney organoids from human induced pluripotent stem cells represents an important recent advance in regenerative medicine. Kidney organoids are expected to be used for disease modeling and drug discovery, and may eventually be applicable for transplantation. In this review, we describe the current status of kidney organoids and discuss the hurdles that need to be overcome to generate transplantable artificial kidneys.
Collapse
Affiliation(s)
- Hidekazu Naganuma
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan.,Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ryuichi Nishinakamura
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
93
|
Lawlor KT, Zappia L, Lefevre J, Park JS, Hamilton NA, Oshlack A, Little MH, Combes AN. Nephron progenitor commitment is a stochastic process influenced by cell migration. eLife 2019; 8:41156. [PMID: 30676318 PMCID: PMC6363379 DOI: 10.7554/elife.41156] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 01/23/2019] [Indexed: 12/31/2022] Open
Abstract
Progenitor self-renewal and differentiation is often regulated by spatially restricted cues within a tissue microenvironment. Here, we examine how progenitor cell migration impacts regionally induced commitment within the nephrogenic niche in mice. We identify a subset of cells that express Wnt4, an early marker of nephron commitment, but migrate back into the progenitor population where they accumulate over time. Single cell RNA-seq and computational modelling of returning cells reveals that nephron progenitors can traverse the transcriptional hierarchy between self-renewal and commitment in either direction. This plasticity may enable robust regulation of nephrogenesis as niches remodel and grow during organogenesis.
Collapse
Affiliation(s)
- Kynan T Lawlor
- Murdoch Children's Research Institute, Parkville, Australia
| | - Luke Zappia
- Murdoch Children's Research Institute, Parkville, Australia.,School of Biosciences, University of Melbourne, Melbourne, Australia
| | - James Lefevre
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Joo-Seop Park
- Division of Pediatric Urology and Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, United States
| | - Nicholas A Hamilton
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Alicia Oshlack
- Murdoch Children's Research Institute, Parkville, Australia.,School of Biosciences, University of Melbourne, Melbourne, Australia
| | - Melissa H Little
- Murdoch Children's Research Institute, Parkville, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, Australia.,Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Australia
| | - Alexander N Combes
- Murdoch Children's Research Institute, Parkville, Australia.,Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Australia
| |
Collapse
|
94
|
Hariharan K, Stachelscheid H, Rossbach B, Oh SJ, Mah N, Schmidt-Ott K, Kurtz A, Reinke P. Parallel generation of easily selectable multiple nephronal cell types from human pluripotent stem cells. Cell Mol Life Sci 2019; 76:179-192. [PMID: 30310934 PMCID: PMC11105784 DOI: 10.1007/s00018-018-2929-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 09/21/2018] [Accepted: 09/26/2018] [Indexed: 02/05/2023]
Abstract
Human pluripotent stem cells (hPSCs) provide a source for the generation of defined kidney cells and renal organoids applicable in regenerative medicine, disease modeling, and drug screening. These applications require the provision of hPSC-derived renal cells by reproducible, scalable, and efficient methods. We established a chemically defined protocol by application of Activin A, BMP4, and Retinoic acid followed by GDNF, which steered hPSCs to the renal lineage and resulted in populations of SIX2+/CITED1+ metanephric mesenchyme- (MM) and of HOXB7+/GRHL2+ ureteric bud (UB)-like cells already by 6 days. Transcriptome analysis corroborated that the PSC-derived cell types at day 8 resemble their renal vesicle and ureteric epithelial counterpart in vivo, forming tubular and glomerular renal cells 6 days later. We demonstrate that starting from hPSCs, our in vitro protocol generates a pool of nephrogenic progenitors at the renal vesicle stage, which can be further directed into specialized nephronal cell types including mesangial-, proximal tubular-, distal tubular, collecting duct epithelial cells, and podocyte precursors after 14 days. This simple and rapid method to produce renal cells from a common precursor pool in 2D culture provides the basis for scaled-up production of tailored renal cell types, which are applicable for drug testing or cell-based regenerative therapies.
Collapse
Affiliation(s)
- Krithika Hariharan
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Harald Stachelscheid
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- Berlin Institute of Health (BIH), Stem Cell Core, Berlin, Germany
| | - Bella Rossbach
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Su-Jun Oh
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Nancy Mah
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Kai Schmidt-Ott
- Department of Nephrology and Intensive Care, Charité University Medicine Berlin, Berlin, Germany
- Max Delbrueck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Andreas Kurtz
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.
| | - Petra Reinke
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- Department of Nephrology and Intensive Care, Charité University Medicine Berlin, Berlin, Germany
| |
Collapse
|
95
|
Abstract
Nephrons differentiate from the cap mesenchyme of the fetal kidney. Nephron progenitor cells that populate the cap mesenchyme efficiently balance self-renewal and epithelial differentiation to enable repeated rounds of nephron formation during development. Here we describe a method to isolate and propagate these cells from the embryonic mouse kidney. Using this method, nephron progenitor cells from a single litter of mice can be propagated to hundreds of millions of cells that express appropriate markers of the undifferentiated state and retain epithelial differentiation capacity in vitro.
Collapse
Affiliation(s)
- Aaron C Brown
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME, USA
| | - Ashwani K Gupta
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME, USA
| | - Leif Oxburgh
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME, USA.
| |
Collapse
|
96
|
Abstract
During kidney development, Six2+ nephron progenitor cells (NPCs) generate nephrons, the functional units of the kidney. Isolating and expanding NPCs in vitro have enabled wide applications in both basic and translational research. Here we describe the methods to derive NPC lines from mouse embryonic kidneys in a 3D culture format as well as gene editing in the NPC lines.
Collapse
Affiliation(s)
- Zhongwei Li
- Division of Nephrology and Hypertension, Department of Medicine and USC/UKRO Kidney Research Center, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA. .,Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA.
| | - Toshikazu Araoka
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | | |
Collapse
|
97
|
Phua YL, Clugston A, Chen KH, Kostka D, Ho J. Small non-coding RNA expression in mouse nephrogenic mesenchymal progenitors. Sci Data 2018; 5:180218. [PMID: 30422124 PMCID: PMC6233257 DOI: 10.1038/sdata.2018.218] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 08/15/2018] [Indexed: 01/02/2023] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that are essential for the regulation of gene expression and play critical roles in human health and disease. Here we present comprehensive miRNA profiling data for mouse nephrogenic mesenchymal progenitors, a population of cells enriched for nephron progenitors that give rise to most cell-types of the nephron, the functional unit of the kidney. We describe a miRNA expression in nephrogenic mesenchymal progenitors, with 162 miRNAs differentially expressed in progenitors when compared to whole kidney. We also annotated 49 novel miRNAs in the developing kidney and experimentally validated 4 of them. Our data are available as a public resource, so that it can be integrated into future studies and analyzed in the context of other functional and epigenomic data in kidney development. Specifically, it will be useful in the effort to shed light on molecular mechanisms underlying processes essential for normal kidney development, like nephron progenitor specification, self-renewal and differentiation.
Collapse
Affiliation(s)
- Yu Leng Phua
- Rangos Research Center, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, USA.,Department of Pediatrics, Division of Nephrology, University of Pittsburgh School of Medicine, PA, USA
| | - Andrew Clugston
- Rangos Research Center, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, USA.,Department of Pediatrics, Division of Nephrology, University of Pittsburgh School of Medicine, PA, USA.,Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kevin Hong Chen
- Rangos Research Center, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, USA.,Department of Pediatrics, Division of Nephrology, University of Pittsburgh School of Medicine, PA, USA
| | - Dennis Kostka
- Rangos Research Center, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, USA.,Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Computational & Systems Biology and Pittsburgh Center for Evolutionary Biology and Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jacqueline Ho
- Rangos Research Center, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, USA.,Department of Pediatrics, Division of Nephrology, University of Pittsburgh School of Medicine, PA, USA
| |
Collapse
|
98
|
Kurtzeborn K, Cebrian C, Kuure S. Regulation of Renal Differentiation by Trophic Factors. Front Physiol 2018; 9:1588. [PMID: 30483151 PMCID: PMC6240607 DOI: 10.3389/fphys.2018.01588] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 10/23/2018] [Indexed: 12/11/2022] Open
Abstract
Classically, trophic factors are considered as proteins which support neurons in their growth, survival, and differentiation. However, most neurotrophic factors also have important functions outside of the nervous system. Especially essential renal growth and differentiation regulators are glial cell line-derived neurotrophic factor (GDNF), bone morphogenetic proteins (BMPs), and fibroblast growth factors (FGFs). Here we discuss how trophic factor-induced signaling contributes to the control of ureteric bud (UB) branching morphogenesis and to maintenance and differentiation of nephrogenic mesenchyme in embryonic kidney. The review includes recent advances in trophic factor functions during the guidance of branching morphogenesis and self-renewal versus differentiation decisions, both of which dictate the control of kidney size and nephron number. Creative utilization of current information may help better recapitulate renal differentiation in vitro, but it is obvious that significantly more basic knowledge is needed for development of regeneration-based renal therapies.
Collapse
Affiliation(s)
- Kristen Kurtzeborn
- Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Medicum, University of Helsinki, Helsinki, Finland
| | - Cristina Cebrian
- Developmental Biology Division, Cincinnati Children’s Hospital, Cincinnati, OH, United States
| | - Satu Kuure
- Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Medicum, University of Helsinki, Helsinki, Finland
- GM-Unit, Laboratory Animal Centre, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| |
Collapse
|
99
|
Recapitulating kidney development: Progress and challenges. Semin Cell Dev Biol 2018; 91:153-168. [PMID: 30184476 DOI: 10.1016/j.semcdb.2018.08.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 08/22/2018] [Accepted: 08/28/2018] [Indexed: 12/14/2022]
Abstract
Decades of research into the molecular and cellular regulation of kidney morphogenesis in rodent models, particularly the mouse, has provided both an atlas of the mammalian kidney and a roadmap for recreating kidney cell types with potential applications for the treatment of kidney disease. With advances in both our capacity to maintain nephron progenitors in culture, reprogram to kidney cell types and direct the differentiation of human pluripotent stem cells to kidney endpoints, renal regeneration via cellular therapy or tissue engineering may be possible. Human kidney models also have potential for disease modelling and drug screening. Such applications will rely upon the accuracy of the model at the cellular level and the capacity for stem-cell derived kidney tissue to recapitulate both normal and diseased kidney tissue. In this review, we will discuss the available cell sources, how well they model the human kidney and how far we are from application either as models or for tissue engineering.
Collapse
|
100
|
Nishikawa M, Yuri S, Kimura H, Yanagawa N, Hamon M, Hauser P, Zhao L, Jo OD, Yanagawa N. Comprehensive analysis of chromatin signature and transcriptome uncovers functional lncRNAs expressed in nephron progenitor cells. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2018; 1862:58-70. [PMID: 30416088 DOI: 10.1016/j.bbagrm.2018.09.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 08/25/2018] [Accepted: 09/12/2018] [Indexed: 01/21/2023]
Abstract
Emerging evidence from recent studies has unraveled the roles of long noncoding RNAs (lncRNAs) in the function of various tissues. However, little is known about the roles of lncRNAs in kidney development. In our present study, we aimed to identify functional lncRNAs in one of the three lineages of kidney progenitor cells, i.e., metanephric mesenchymal (MM) cells. We conducted comprehensive analyses of the chromatin signature and transcriptome by RNA-seq and ChIP-seq. We found seventeen lncRNAs that were expressed specifically in MM cells with an active chromatin signature, while remaining silenced in a bivalent chromatin state in non-MM cells. Out of these MM specific lncRNAs, we identified a lncRNA, Gm29418, in a distal enhancer region of Six2, a key regulatory gene of MM cells. We further identified three transcript variants of Gm29418 by Rapid Amplification of cDNA Ends (RACE), and confirmed that the transcription-start-sites (TSSs) of these variants were consistent with the result of Cap Analysis Gene Expression (CAGE). In support of the enhancer-like function of Gm29418 on Six2 expression, we found that knock-down of Gm29418 by two independent anti-sense locked nucleic acid (LNA) phosphorothioate gapmers suppressed Six2 mRNA expression levels in MM cells. We also found that over-expression of Gm29418 led to an increase in Six2 mRNA expression levels in a mouse MM cell line. In conclusion, we identified a lncRNA, Gm29418, in nephron progenitor cells that has an enhancer-like function on a key regulatory gene, Six2.
Collapse
Affiliation(s)
- Masaki Nishikawa
- Medical and Research Services, Greater Los Angeles Veterans Affairs Healthcare System at Sepulveda, North Hills, CA, USA; University of California at Los Angeles, David Geffen School of Medicine, Los Angeles, CA, USA.
| | - Shunsuke Yuri
- Nara Institute of Science & Technology, Nara 630-0192, Japan
| | | | - Naomi Yanagawa
- Medical and Research Services, Greater Los Angeles Veterans Affairs Healthcare System at Sepulveda, North Hills, CA, USA; University of California at Los Angeles, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Morgan Hamon
- Medical and Research Services, Greater Los Angeles Veterans Affairs Healthcare System at Sepulveda, North Hills, CA, USA; University of California at Los Angeles, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Peter Hauser
- Medical and Research Services, Greater Los Angeles Veterans Affairs Healthcare System at Sepulveda, North Hills, CA, USA; University of California at Los Angeles, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Lifu Zhao
- Medical and Research Services, Greater Los Angeles Veterans Affairs Healthcare System at Sepulveda, North Hills, CA, USA
| | - Oak D Jo
- Medical and Research Services, Greater Los Angeles Veterans Affairs Healthcare System at Sepulveda, North Hills, CA, USA; University of California at Los Angeles, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Norimoto Yanagawa
- Medical and Research Services, Greater Los Angeles Veterans Affairs Healthcare System at Sepulveda, North Hills, CA, USA; University of California at Los Angeles, David Geffen School of Medicine, Los Angeles, CA, USA.
| |
Collapse
|