1
|
Shi M, Crouse B, Sundaram N, Pode Shakked N, Thorner K, King NM, Dutta P, Ester L, Zhang W, Govindarajah V, Kopan R, Cebrian C, Mayhew CN, Helmrath MA, Bonventre JV, McCracken KW. Integrating collecting systems in human kidney organoids through fusion of distal nephron to ureteric bud. Cell Stem Cell 2025:S1934-5909(25)00143-2. [PMID: 40345193 DOI: 10.1016/j.stem.2025.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 03/07/2025] [Accepted: 04/17/2025] [Indexed: 05/11/2025]
Abstract
Kidneys maintain homeostasis through an array of parallel nephrons, which fuse during development to a system of collecting ducts (CDs), establishing the essential luminal pathway for excretion of metabolic waste. Human kidney organoids derived from pluripotent stem cells (human pluripotent stem cells [hPSCs]) generate nephrons that lack CDs and terminate as blind-ended tubules, limiting their functional potential. Here, we describe a developmentally inspired hPSC differentiation system that addresses this deficiency through assembly of induced nephrogenic mesenchyme with ureteric bud (UB) progenitors, leading to a CD network functionally integrated in kidney organoids through fusion with the distal tubule. The nephron fusion occurs stereotypically and is regulated by proximal-distal nephron patterning, which can be modulated through temporal manipulation of developmental pathways. This work provides a platform for interrogating the principles and mechanisms underlying nephron-UB fusion and a framework for engineering unobstructed nephrons with collecting systems, an important step toward de novo generation of functional kidney tissue.
Collapse
Affiliation(s)
- Min Shi
- Division of Nephrology and Hypertension, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Brittney Crouse
- Division of Nephrology and Hypertension, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Nambirajan Sundaram
- Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Naomi Pode Shakked
- Division of Nephrology and Hypertension, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Pediatric Nephrology Unit, Dana Dwek Children's Hospital, Tel Aviv Medical Center, Tel Aviv 6423906, Israel; Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Konrad Thorner
- Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Nathaniel M King
- Division of Nephrology and Hypertension, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Parna Dutta
- Division of Nephrology and Hypertension, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Lioba Ester
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne and University Hospital Cologne, Cologne 50937, Germany
| | - Weitao Zhang
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Vinothini Govindarajah
- Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Raphael Kopan
- Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Cristina Cebrian
- Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Christopher N Mayhew
- Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Michael A Helmrath
- Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Joseph V Bonventre
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02138, USA
| | - Kyle W McCracken
- Division of Nephrology and Hypertension, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
| |
Collapse
|
2
|
Goux Corredera I, Amato G, Moya-Rull D, Garreta E, Montserrat N. Unlocking the full potential of human pluripotent stem cell-derived kidney organoids through bioengineering. Kidney Int 2025:S0085-2538(25)00327-8. [PMID: 40280411 DOI: 10.1016/j.kint.2025.01.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 01/17/2025] [Accepted: 01/28/2025] [Indexed: 04/29/2025]
Abstract
Human pluripotent stem cells hold inherent properties, allowing researchers to recapitulate key morphogenetic processes. These characteristics, coupled with bioengineering techniques, have led to the definition of early procedures to derive organ-like cell cultures, the so-called organoids. With regard to kidney organoids, challenges stand ahead, such as the need to enhance cellular composition, maturation, and function to that found in the native organ. To this end, the kidney organoid field has begun to nourish from innovative engineering approaches aiming to gain control on the externally imposed biochemical and biophysical cues. In this review, we first introduce how previous research in kidney development and human pluripotent stem cells has informed the establishment of current kidney organoid procedures. We then discuss recent engineering approaches to guide kidney organoid self-organization, differentiation, and maturation. In addition, we present current strategies to engineer vascularization and promote in vivo-like physiological microenvironments as potential solutions to increase kidney organoid lifespan and functionality. We finally emphasize how working at the cusp of cell mechanics and computational biology will set the ground for successful translational applications of kidney organoids.
Collapse
Affiliation(s)
- Iphigénie Goux Corredera
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Gaia Amato
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Daniel Moya-Rull
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Elena Garreta
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain; University of Barcelona, Barcelona, Spain.
| | - Nuria Montserrat
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain; University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina, Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
| |
Collapse
|
3
|
Gu J, Liu F, Li L, Mao J. Advances and Challenges in Modeling Autosomal Dominant Polycystic Kidney Disease: A Focus on Kidney Organoids. Biomedicines 2025; 13:523. [PMID: 40002937 PMCID: PMC11852630 DOI: 10.3390/biomedicines13020523] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/04/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a prevalent hereditary disorder characterized by distinct phenotypic variability that has posed challenges for advancing in-depth research. Recent advancements in kidney organoid construction technologies have enabled researchers to simulate kidney development and create simplified in vitro experimental environments, allowing for more direct observation of how genetic mutations drive pathological phenotypes and disrupt physiological functions. Emerging technologies, such as microfluidic bioreactor culture systems and single-cell transcriptomics, have further supported the development of complex ADPKD organoids, offering robust models for exploring disease mechanisms and facilitating drug discovery. Nevertheless, significant challenges remain in constructing more accurate ADPKD disease models. This review will summarize recent advances in ADPKD organoid construction, focusing on the limitations of the current techniques and the critical issues that need to be addressed for future breakthroughs. New and Noteworthy: This review presents recent advancements in ADPKD organoid construction, particularly iPSC-derived models, offering new insights into disease mechanisms and drug discovery. It focuses on challenges such as limited vascularization and maturity, proposing potential solutions through emerging technologies. The ongoing optimization of ADPKD organoid models is expected to enhance understanding of the disease and drive breakthroughs in disease mechanisms and targeted therapy development.
Collapse
Affiliation(s)
| | | | | | - Jianhua Mao
- Department of Nephrology, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310058, China; (J.G.); (F.L.); (L.L.)
| |
Collapse
|
4
|
Michalopulos MG, Liu Y, Raju DR, Lafin JT, Ma Y, Gaur D, Khadka S, Xing C, McMahon AP, Carroll TJ, Drake KA. Defects in nephrogenesis result in an expansion of the Foxd1+ stromal progenitor population. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.10.637031. [PMID: 39990396 PMCID: PMC11844377 DOI: 10.1101/2025.02.10.637031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Reciprocal signaling interactions coordinate multiple aspects of kidney development. While signals from the stroma have been shown to regulate nephron progenitor cell (NPC) differentiation, much less is known about regulation of the stromal progenitor population. Here, we demonstrate that disruption of the NPC lineage via loss of Wt1 (i.e., Six2cre;Wt1 c/c ) results in an expansion of Foxd1+ stromal progenitor cells. Analyses of the developing stroma in two additional models, including Wnt4-null mutants (which fail to form nephron structures similar to Six2cre;Wt1 c/c kidneys) and NPC ablation via diphtheria toxin (i.e., Six2cre;RosaDTA c/+ ), both phenocopy Six2cre;Wt1 c/c mutants, thus further confirming that defects in the NPC lineage result in abnormal development of the stromal progenitor population. Furthermore, we identify a subcluster of the Foxd1+ stroma that appears expanded in the three mutant mouse models and conserved in human fetal kidneys. Overall, the findings from this study suggest that loss of differentiating nephron structures may result in possible over proliferation of the stromal progenitor population and/or a block in stromal differentiation and further highlight how crosstalk amongst the progenitor cell lineages coordinates multiple aspects of kidney development.
Collapse
Affiliation(s)
- Michael G Michalopulos
- Department of Pediatrics, Division of Pediatric Nephrology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Yan Liu
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Dinesh Ravindra Raju
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - John T Lafin
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yanru Ma
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Dhruv Gaur
- Midwestern University, Chicago College of Osteopathic Medicine, Chicago, IL, USA
| | - Sadiksha Khadka
- Division of Pediatric Nephrology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Chao Xing
- Department of Bioinformatics and O'Donnell School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Thomas J Carroll
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Keri A Drake
- Division of Pediatric Nephrology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
5
|
van den Berg CW, Dumas SJ, Little MH, Rabelink TJ. Challenges in maturation and integration of kidney organoids for stem cell-based renal replacement therapy. Kidney Int 2025; 107:262-270. [PMID: 39571903 DOI: 10.1016/j.kint.2024.10.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 09/20/2024] [Accepted: 10/03/2024] [Indexed: 12/10/2024]
Abstract
Human pluripotent stem cell-derived kidney organoids hold promise for future applications in regenerative medicine. However, significant biological hurdles need to be overcome to enable their use as a transplantable stem cell-derived therapeutic graft. Current kidney organoid protocols do not recapitulate a complete integrated developing kidney, but embryonic kidney transplantations have provided clues for advancing maturation and functionality of kidney organoids. Transplantation, subsequent vascularization, and blood perfusion of kidney organoids improve nephron patterning and maturation, suggesting a role for angiocrine factors as well as metabolic wiring in these processes. Transplanted organoids exhibit filtration, but the resulting filtrate has no apparent exit path for excretion. Improved in vitro patterning of kidney organoids may be required such that a more structurally correct tissue is formed before transplant. Here we review current progress with transplantation of kidney organoids, as well as their engraftment and integration, and identify the key obstacles to therapeutic success and how these might be achieved.
Collapse
Affiliation(s)
- Cathelijne W van den Berg
- Department of Internal Medicine-Nephrology, Leiden University Medical Center, Leiden, the Netherlands; Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, the Netherlands.
| | - Sébastien J Dumas
- Department of Internal Medicine-Nephrology, Leiden University Medical Center, Leiden, the Netherlands; Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, the Netherlands
| | - Melissa H Little
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, Victoria, Australia; Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), University of Copenhagen, Copenhagen, Denmark
| | - Ton J Rabelink
- Department of Internal Medicine-Nephrology, Leiden University Medical Center, Leiden, the Netherlands; Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
6
|
Kimura H, Nishikawa M, Kutsuzawa N, Tokito F, Kobayashi T, Kurniawan DA, Shioda H, Cao W, Shinha K, Nakamura H, Doi K, Sakai Y. Advancements in Microphysiological systems: Exploring organoids and organ-on-a-chip technologies in drug development -focus on pharmacokinetics related organs. Drug Metab Pharmacokinet 2025; 60:101046. [PMID: 39847980 DOI: 10.1016/j.dmpk.2024.101046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 12/05/2024] [Accepted: 12/14/2024] [Indexed: 01/25/2025]
Abstract
This study explored the evolving landscape of Microphysiological Systems (MPS), with a focus on organoids and organ-on-a-chip (OoC) technologies, which are promising alternatives to animal testing in drug discovery. MPS technology offers in vitro models with high physiological relevance, simulating organ function for pharmacokinetic studies. Organoids composed of 3D cell aggregates and OoCs mimicking in vivo environments based on microfluidic platforms represent the forefront of MPS. This paper provides a comprehensive overview of their application in studying the gut, liver, and kidney and their challenges in becoming reliable alternatives to in vivo models. Although MPS technology is not yet fully comparable to in vivo systems, its continued development, aided by in silico, automation, and AI approaches, is anticipated to bring about further advancements. Collaboration across multiple disciplines and ongoing regulatory discussions will be crucial in driving MPS toward practical and ethical applications in biomedical research and drug development.
Collapse
Affiliation(s)
- Hiroshi Kimura
- Micro/Nano Technology Center, Tokai University, 4-1-1 Kitakaname, Hiratsuka, Kanagawa, 259-1292, Japan.
| | - Masaki Nishikawa
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo, 113-8654, Japan
| | - Naokata Kutsuzawa
- Micro/Nano Technology Center, Tokai University, 4-1-1 Kitakaname, Hiratsuka, Kanagawa, 259-1292, Japan; Division of Pulmonary Medicine, Department of Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1143, Japan
| | - Fumiya Tokito
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo, 113-8654, Japan
| | - Takuma Kobayashi
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo, 113-8654, Japan
| | - Dhimas Agung Kurniawan
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo, 113-8654, Japan
| | - Hiroki Shioda
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo, 113-8654, Japan
| | - Wenxin Cao
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo, 113-8654, Japan
| | - Kenta Shinha
- Micro/Nano Technology Center, Tokai University, 4-1-1 Kitakaname, Hiratsuka, Kanagawa, 259-1292, Japan
| | - Hiroko Nakamura
- Micro/Nano Technology Center, Tokai University, 4-1-1 Kitakaname, Hiratsuka, Kanagawa, 259-1292, Japan
| | - Kotaro Doi
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo, Tokyo, 113-8655, Japan
| | - Yasuyuki Sakai
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo, 113-8654, Japan
| |
Collapse
|
7
|
Elhendawi M, Lawrence ML, Davies JA. Toxicity Testing Using Organoids Made from Human-Induced Pluripotent Stem Cells Engineered to Report Oxidative Stress. Methods Mol Biol 2025; 2924:131-143. [PMID: 40307640 DOI: 10.1007/978-1-0716-4530-7_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
This article describes the engineering of human induced pluripotent stem cells to report a cellular response to oxidative stress, which is a common (but not universal) result of cells being exposed to a toxicant. It then describes the use of these cells to generate renal organoids that will report oxidative stress, and the use of these organoids to screen a panel of compounds for nephrotoxicity. This application is intended as an illustration; in principle, other organoids could be made from these cells, and other cell lines could be made to report different common markers of cellular stress (for example, the release of pro-inflammatory signals).
Collapse
Affiliation(s)
- Mona Elhendawi
- Deanery of Biomedical Sciences and Centre for Engineering Biology, University of Edinburgh, Edinburgh, UK
| | - Melanie L Lawrence
- Deanery of Biomedical Sciences and Centre for Engineering Biology, University of Edinburgh, Edinburgh, UK
| | - Jamie A Davies
- Deanery of Biomedical Sciences and Centre for Engineering Biology, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
8
|
Ide H, Miike K, Ohmori T, Maruyama K, Izumi Y, Tanigawa S, Nishinakamura R. Mouse embryonic kidney transplantation identifies maturation defects in the medulla. Sci Rep 2024; 14:30293. [PMID: 39639083 PMCID: PMC11621804 DOI: 10.1038/s41598-024-81984-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024] Open
Abstract
Kidney organoids are connected to the host circulation and mature after transplantation. However, they are still immature compared to the adult kidneys, and their precise maturation stages remain unclear. By transplanting the mouse embryonic kidney as a model system for organoid transplantation, we report here the maturation defects of the graft, especially in the medulla. Single cell profiling of the developing kidneys in vivo identified gene sets associated with the maturation of the collecting duct epithelium and medullary stroma. These data revealed an upregulation of genes associated with channel/transporter functions and immune defense, as well as a downregulation of neuronal genes. Using these marker genes, we found that the maturation of the collecting duct and medullary stroma in the grafts barely corresponds to the perinatal stage, which was confirmed histologically by using representative genes. Thus, the gene sets obtained serve as maturation coordinates for the renal medulla and will be helpful in analyzing its maturation defects after transplantation. They will also provide a useful basis for further maturation of transplanted kidney organoids.
Collapse
Affiliation(s)
- Hiroshi Ide
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
| | - Koichiro Miike
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
| | - Tomoko Ohmori
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
| | - Kosuke Maruyama
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Yuichiro Izumi
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Shunsuke Tanigawa
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
| | - Ryuichi Nishinakamura
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan.
| |
Collapse
|
9
|
Shi M, Crouse B, Sundaram N, Pode Shakked N, Ester L, Zhang W, Janakiram V, Kopan R, Helmrath MA, Bonventre JV, McCracken KW. Integrating collecting systems in kidney organoids through fusion of distal nephron to ureteric bud. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.19.613645. [PMID: 39345524 PMCID: PMC11429897 DOI: 10.1101/2024.09.19.613645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
The kidney maintains homeostasis through an array of parallel nephrons, which all originate in development as isolated epithelial structures that later fuse through their distal poles to a system of collecting ducts (CD). This connection is required to generate functional nephrons by providing a pathway for excretion of metabolic waste and byproducts. Currently, methods for differentiating human pluripotent stem cells into kidney organoids generate nephrons that lack CDs and instead terminate as blind-ended tubules. Here we describe a developmentally inspired system that addresses this deficiency through assembly of induced nephrogenic mesenchyme with ureteric bud (UB) tissues, the embryonic building blocks of the kidney's collecting system. The UB progenitors grow and develop into a network of CDs within the organoid, and importantly, they functionally integrate with the nephrons through recapitulating fusion between the distal tubule and CD to create a continuous epithelial lumen. We further showed that proximal-distal nephron specification, fusion frequency, and maturation of the CD can be augmented through temporal manipulation of developmental signaling pathways. This work provides a platform for interrogating the principles and mechanisms underlying nephron-UB fusion and a framework for engineering unobstructed nephrons with patterned collecting systems, an important step toward the de novo generation of functional kidney tissue.
Collapse
Affiliation(s)
- Min Shi
- Division of Nephrology and Hypertension, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Center for Stem Cell and Organoid Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Brittney Crouse
- Division of Nephrology and Hypertension, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Center for Stem Cell and Organoid Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Nambirajan Sundaram
- Center for Stem Cell and Organoid Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Surgery, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Naomi Pode Shakked
- Division of Nephrology and Hypertension, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Developmental Biology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Current address: Faculty of Medicine and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Lioba Ester
- Division of Renal Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Weitao Zhang
- Division of Renal Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Vinothini Janakiram
- Center for Stem Cell and Organoid Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Surgery, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Raphael Kopan
- Division of Developmental Biology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Michael A. Helmrath
- Center for Stem Cell and Organoid Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Surgery, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Joseph V. Bonventre
- Division of Renal Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| | - Kyle W. McCracken
- Division of Nephrology and Hypertension, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Center for Stem Cell and Organoid Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Developmental Biology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Lead contact
| |
Collapse
|
10
|
Liu Q, Yue L, Deng J, Tan Y, Wu C. Progress and breakthroughs in human kidney organoid research. Biochem Biophys Rep 2024; 39:101736. [PMID: 38910872 PMCID: PMC11190488 DOI: 10.1016/j.bbrep.2024.101736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 04/03/2024] [Accepted: 05/17/2024] [Indexed: 06/25/2024] Open
Abstract
The three-dimensional (3D) kidney organoid is a breakthrough model for recapitulating renal morphology and function in vitro, which is grown from stem cells and resembles mammalian kidney organogenesis. Currently, protocols for cultivating this model from induced pluripotent stem cells (iPSCs) and patient-derived adult stem cells (ASCs) have been widely reported. In recent years, scientists have focused on combining cutting-edge bioengineering and bioinformatics technologies to improve the developmental accuracy of kidney organoids and achieve high-throughput experimentation. As a remarkable tool for mechanistic research of the renal system, kidney organoid has both potential and challenges. In this review, we have described the evolution of kidney organoid establishment methods and highlighted the latest progress leading to a more sophisticated kidney transformation research model. Finally, we have summarized the main applications of renal organoids in exploring kidney disease.
Collapse
Affiliation(s)
- Qi Liu
- School of Biomedical Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Liang Yue
- Department of Stem Cell and Regenerative Medicine, Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Jiu Deng
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, 266071, China
| | - Yingxia Tan
- Department of Stem Cell and Regenerative Medicine, Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Chengjun Wu
- School of Biomedical Engineering, Dalian University of Technology, Dalian, 116024, China
| |
Collapse
|
11
|
Davies JA, Holland I, Gül H. Kidney organoids: steps towards better organization and function. Biochem Soc Trans 2024; 52:1861-1871. [PMID: 38934505 PMCID: PMC11668298 DOI: 10.1042/bst20231554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/20/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024]
Abstract
Kidney organoids - 3D representations of kidneys made either from pluripotent or tissue stem cells - have been available for well over a decade. Their application could confer notable benefits over longstanding in vivo approaches with the potential for clinically aligned human cells and reduced ethical burdens. They been used, at a proof-of-concept level, in development in disease modeling (including with patient-derived stem cells), and in screening drugs for efficacy/toxicity. They differ from real kidneys: they represent only foetal-stage tissue, in their simplest forms they lack organ-scale anatomical organization, they lack a properly arranged vascular system, and include non-renal cells. Cell specificity may be improved by better techniques for differentiation and/or sorting. Sequential assembly techniques that mimic the sequence of natural development, and localized sources of differentiation-inducing signals, improve organ-scale anatomy. Organotypic vascularization remains a challenge: capillaries are easy, but the large vessels that should serve them are absent from organoids and, even in cultured real kidneys, these large vessels do not survive without blood flow. Transplantation of organoids into hosts results in their being vascularized (though probably not organotypically) and in some renal function. It will be important to transplant more advanced organoids, with a urine exit, in the near future to assess function more stringently. Transplantation of human foetal kidneys, followed by nephrectomy of host kidneys, keeps rats alive for many weeks, raising hope that, if organoids can be produced even to the limited size and complexity of foetal kidneys, they may one day be useful in renal replacement.
Collapse
Affiliation(s)
- Jamie A. Davies
- Deanery of Biomedical Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, U.K
| | - Ian Holland
- Deanery of Biomedical Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, U.K
| | - Huseyin Gül
- Deanery of Biomedical Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, U.K
| |
Collapse
|
12
|
Ibi Y, Nishinakamura R. Generating kidney organoids based on developmental nephrology. Eur J Cell Biol 2024; 103:151450. [PMID: 39137450 DOI: 10.1016/j.ejcb.2024.151450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 08/05/2024] [Accepted: 08/08/2024] [Indexed: 08/15/2024] Open
Abstract
Over the past decade, the induction protocols for the two types of kidney organoids (nephron organoids and ureteric bud organoids) from pluripotent stem cells (PSCs) have been established based on the knowledge gained in developmental nephrology. Kidney organoids are now used for disease modeling and drug screening, but they also have potential as tools for clinical transplantation therapy. One of the options to achieve this goal would be to assemble multiple renal progenitor cells (nephron progenitor, ureteric bud, stromal progenitor) to reproduce the organotypic kidney structure from PSCs. At least from mouse PSCs, all the three progenitors have been induced and assembled into such "higher order" kidney organoids. We will provide an overview of the developmental nephrology required for the induction of renal progenitors and discuss recent advances and remaining challenges of kidney organoids for clinical transplantation therapy.
Collapse
Affiliation(s)
- Yutaro Ibi
- 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
|
13
|
Morino-Koga S, Tsuruda M, Zhao X, Oshiro S, Yokomizo T, Yamane M, Tanigawa S, Miike K, Usuki S, Yasunaga KI, Nishinakamura R, Suda T, Ogawa M. Transition of signal requirement in hematopoietic stem cell development from hemogenic endothelial cells. Proc Natl Acad Sci U S A 2024; 121:e2404193121. [PMID: 39042698 PMCID: PMC11294991 DOI: 10.1073/pnas.2404193121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 06/19/2024] [Indexed: 07/25/2024] Open
Abstract
Hematopoietic stem cells (HSCs) develop from hemogenic endothelial cells (HECs) in vivo during mouse embryogenesis. When cultured in vitro, cells from the embryo phenotypically defined as pre-HSC-I and pre-HSC-II have the potential to differentiate into HSCs. However, minimal factors required for HSC induction from HECs have not yet been determined. In this study, we demonstrated that stem cell factor (SCF) and thrombopoietin (TPO) induced engrafting HSCs from embryonic day (E) 11.5 pre-HSC-I in a serum-free and feeder-free culture condition. In contrast, E10.5 pre-HSC-I and HECs required an endothelial cell layer in addition to SCF and TPO to differentiate into HSCs. A single-cell RNA sequencing analysis of E10.5 to 11.5 dorsal aortae with surrounding tissues and fetal livers detected TPO expression confined in hepatoblasts, while SCF was expressed in various tissues, including endothelial cells and hepatoblasts. Our results suggest a transition of signal requirement during HSC development from HECs. The differentiation of E10.5 HECs to E11.5 pre-HSC-I in the aorta-gonad-mesonephros region depends on SCF and endothelial cell-derived factors. Subsequently, SCF and TPO drive the differentiation of E11.5 pre-HSC-I to pre-HSC-II/HSCs in the fetal liver. The culture system established in this study provides a beneficial tool for exploring the molecular mechanisms underlying the development of HSCs from HECs.
Collapse
Affiliation(s)
- Saori Morino-Koga
- Department of Cell Differentiation, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto860-0811, Japan
| | - Mariko Tsuruda
- Department of Cell Differentiation, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto860-0811, Japan
| | - Xueyu Zhao
- Department of Cell Differentiation, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto860-0811, Japan
| | - Shogo Oshiro
- Department of Cell Differentiation, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto860-0811, Japan
| | - Tomomasa Yokomizo
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto860-0811, Japan
- Department of Microscopic and Developmental Anatomy, Tokyo Women’s Medical University, Tokyo162-8666, Japan
| | - Mariko Yamane
- Department of Pluripotent Stem Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto860-0811, Japan
- Department of Functional Genome Informatics, Division of Medical Genomics, Medical Research Institute, Tokyo Medical and Dental University, Tokyo113-8510, Japan
- Laboratory for Bioinformatics Research, RIKEN Center for Biosystems Dynamics Research, Kobe650-0047, Japan
| | - Shunsuke Tanigawa
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto860-0811, Japan
| | - Koichiro Miike
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto860-0811, Japan
| | - Shingo Usuki
- Liaison Laboratory Research Promotion Center, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto860-0811, Japan
| | - Kei-ichiro Yasunaga
- Liaison Laboratory Research Promotion Center, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto860-0811, Japan
| | - Ryuichi Nishinakamura
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto860-0811, Japan
| | - Toshio Suda
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto860-0811, Japan
| | - Minetaro Ogawa
- Department of Cell Differentiation, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto860-0811, Japan
| |
Collapse
|
14
|
Tadokoro T, Murata S, Kato M, Ueno Y, Tsuchida T, Okumura A, Kuse Y, Konno T, Uchida Y, Yamakawa Y, Zushi M, Yajima M, Kobayashi T, Hasegawa S, Kawakatsu-Hatada Y, Hayashi Y, Osakabe S, Maeda T, Kimura K, Mori A, Tanaka M, Kamishibahara Y, Matsuo M, Nie YZ, Okamoto S, Oba T, Tanimizu N, Taniguchi H. Human iPSC-liver organoid transplantation reduces fibrosis through immunomodulation. Sci Transl Med 2024; 16:eadg0338. [PMID: 39047116 DOI: 10.1126/scitranslmed.adg0338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/31/2024] [Accepted: 07/01/2024] [Indexed: 07/27/2024]
Abstract
Donor organ shortages for transplantation remain a serious global concern, and alternative treatment is in high demand. Fetal cells and tissues have considerable therapeutic potential as, for example, organoid technology that uses human induced pluripotent stem cells (hiPSCs) to generate unlimited human fetal-like cells and tissues. We previously reported the in vivo vascularization of early fetal liver-like hiPSC-derived liver buds (LBs) and subsquent improved survival of recipient mice with subacute liver failure. Here, we show hiPSC-liver organoids (LOs) that recapitulate midgestational fetal liver promote de novo liver generation when grafted onto the surface of host livers in chemical fibrosis models, thereby recovering liver function. We found that fetal liver, a hematopoietic tissue, highly expressed macrophage-recruiting factors and antifibrotic M2 macrophage polarization factors compared with the adult liver, resulting in fibrosis reduction because of CD163+ M2-macrophage polarization. Next, we created midgestational fetal liver-like hiPSC-LOs by fusion of hiPSC-LBs to induce static cell-cell interactions and found that these contained complex structures such as hepatocytes, vasculature, and bile ducts after transplantation. This fusion allowed the generation of a large human tissue suitable for transplantation into immunodeficient rodent models of liver fibrosis. hiPSC-LOs showed superior liver function compared with hiPSC-LBs and improved survival and liver function upon transplantation. In addition, hiPSC-LO transplantation ameliorated chemically induced liver fibrosis, a symptom of liver cirrhosis that leads to organ dysfunction, through immunomodulatory effects, particularly on CD163+ phagocytic M2-macrophage polarization. Together, our results suggest hiPSC-LO transplantation as a promising therapeutic option for liver fibrosis.
Collapse
Affiliation(s)
- Tomomi Tadokoro
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regeneration Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Soichiro Murata
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regeneration Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Mimoko Kato
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
| | - Yasuharu Ueno
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regeneration Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Tomonori Tsuchida
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regeneration Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Ayumu Okumura
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regeneration Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Yoshiki Kuse
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regeneration Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Takahiro Konno
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
| | - Yutaro Uchida
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
| | - Yuriko Yamakawa
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regeneration Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Marina Zushi
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regeneration Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Megumi Yajima
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regeneration Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Tatsuya Kobayashi
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regeneration Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Shunsuke Hasegawa
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
| | - Yumi Kawakatsu-Hatada
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
| | - Yoshihito Hayashi
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regeneration Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Shun Osakabe
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
| | - Takuji Maeda
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
| | - Kodai Kimura
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
| | - Akihiro Mori
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
| | - Maiko Tanaka
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
| | - Yu Kamishibahara
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regeneration Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Megumi Matsuo
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regeneration Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Yun-Zhong Nie
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regeneration Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Satoshi Okamoto
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regeneration Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Takayoshi Oba
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regeneration Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Naoki Tanimizu
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regeneration Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Hideki Taniguchi
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regeneration Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| |
Collapse
|
15
|
Porter CM, Qian GC, Grindel SH, Hughes AJ. Highly parallel production of designer organoids by mosaic patterning of progenitors. Cell Syst 2024; 15:649-661.e9. [PMID: 38981488 PMCID: PMC11257788 DOI: 10.1016/j.cels.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 04/09/2024] [Accepted: 06/17/2024] [Indexed: 07/11/2024]
Abstract
Organoids derived from human stem cells are a promising approach for disease modeling, regenerative medicine, and fundamental research. However, organoid variability and limited control over morphological outcomes remain as challenges. One open question is the extent to which engineering control over culture conditions can guide organoids to specific compositions. Here, we extend a DNA "velcro" cell patterning approach, precisely controlling the number and ratio of human induced pluripotent stem cell-derived progenitors contributing to nephron progenitor (NP) organoids and mosaic NP/ureteric bud (UB) tip cell organoids within arrays of microwells. We demonstrate long-term control over organoid size and morphology, decoupled from geometric constraints. We then show emergent trends in organoid tissue proportions that depend on initial progenitor cell composition. These include higher nephron and stromal cell representation in mosaic NP/UB organoids vs. NP-only organoids and a "goldilocks" initial cell ratio in mosaic organoids that optimizes the formation of proximal tubule structures.
Collapse
Affiliation(s)
- Catherine M Porter
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA; Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Soft and Living Matter, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Precision Engineering for Health (CPE4H), University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Grace C Qian
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA; Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Samuel H Grindel
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA; Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Soft and Living Matter, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Precision Engineering for Health (CPE4H), University of Pennsylvania, Philadelphia, PA 19104, USA; Materials Research Science and Engineering Center (MRSEC), University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alex J Hughes
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA; Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Soft and Living Matter, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Precision Engineering for Health (CPE4H), University of Pennsylvania, Philadelphia, PA 19104, USA; Materials Research Science and Engineering Center (MRSEC), University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
16
|
Song L, Li Q, Xia L, Sahay AE, Qiu Q, Li Y, Li H, Sasaki K, Susztak K, Wu H, Wan L. Single-cell multiomics reveals ENL mutation perturbs kidney developmental trajectory by rewiring gene regulatory landscape. Nat Commun 2024; 15:5937. [PMID: 39009564 PMCID: PMC11250843 DOI: 10.1038/s41467-024-50171-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 07/02/2024] [Indexed: 07/17/2024] Open
Abstract
How disruptions to normal cell differentiation link to tumorigenesis remains incompletely understood. Wilms tumor, an embryonal tumor associated with disrupted organogenesis, often harbors mutations in epigenetic regulators, but their role in kidney development remains unexplored. Here, we show at single-cell resolution that a Wilms tumor-associated mutation in the histone acetylation reader ENL disrupts kidney differentiation in mice by rewiring the gene regulatory landscape. Mutant ENL promotes nephron progenitor commitment while restricting their differentiation by dysregulating transcription factors such as Hox clusters. It also induces abnormal progenitors that lose kidney-associated chromatin identity. Furthermore, mutant ENL alters the transcriptome and chromatin accessibility of stromal progenitors, resulting in hyperactivation of Wnt signaling. The impacts of mutant ENL on both nephron and stroma lineages lead to profound kidney developmental defects and postnatal mortality in mice. Notably, a small molecule inhibiting mutant ENL's histone acetylation binding activity largely reverses these defects. This study provides insights into how mutations in epigenetic regulators disrupt kidney development and suggests a potential therapeutic approach.
Collapse
Affiliation(s)
- Lele Song
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Qinglan Li
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Lingbo Xia
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of the School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Arushi Eesha Sahay
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Qi Qiu
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Penn Epigenetics Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Yuanyuan Li
- MOE Key Laboratory of Protein Sciences, Beijing Frontier Research Center for Biological Structure, School of Medicine, Tsinghua University, Beijing, 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China
| | - Haitao Li
- MOE Key Laboratory of Protein Sciences, Beijing Frontier Research Center for Biological Structure, School of Medicine, Tsinghua University, Beijing, 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China
| | - Kotaro Sasaki
- Department of Biomedical Sciences, University of Pennsylvania, School of Veterinary Medicine, Philadelphia, PA, 19104, USA
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Katalin Susztak
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Hao Wu
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Penn Epigenetics Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Liling Wan
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Penn Epigenetics Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
17
|
Song L, Li Q, Xia L, Sahay A, Qiu Q, Li Y, Li H, Sasaki K, Susztak K, Wu H, Wan L. Single-Cell multiomics reveals ENL mutation perturbs kidney developmental trajectory by rewiring gene regulatory landscape. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.09.591709. [PMID: 38766219 PMCID: PMC11100752 DOI: 10.1101/2024.05.09.591709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Cell differentiation during organogenesis relies on precise epigenetic and transcriptional control. Disruptions to this regulation can result in developmental abnormalities and malignancies, yet the underlying mechanisms are not well understood. Wilms tumors, a type of embryonal tumor closely linked to disrupted organogenesis, harbor mutations in epigenetic regulators in 30-50% of cases. However, the role of these regulators in kidney development and pathogenesis remains unexplored. By integrating mouse modeling, histological characterizations, and single-cell transcriptomics and chromatin accessibility profiling, we show that a Wilms tumor-associated mutation in the chromatin reader protein ENL disrupts kidney development trajectory by rewiring the gene regulatory landscape. Specifically, the mutant ENL promotes the commitment of nephron progenitors while simultaneously restricting their differentiation by dysregulating key transcription factor regulons, particularly the HOX clusters. It also induces the emergence of abnormal progenitor cells that lose their chromatin identity associated with kidney specification. Furthermore, the mutant ENL might modulate stroma-nephron interactions via paracrine Wnt signaling. These multifaceted effects caused by the mutation result in severe developmental defects in the kidney and early postnatal mortality in mice. Notably, transient inhibition of the histone acetylation binding activity of mutant ENL with a small molecule displaces transcriptional condensates formed by mutant ENL from target genes, abolishes its gene activation function, and restores developmental defects in mice. This work provides new insights into how mutations in epigenetic regulators can alter the gene regulatory landscape to disrupt kidney developmental programs at single-cell resolution in vivo . It also offers a proof-of-concept for the use of epigenetics-targeted agents to rectify developmental defects.
Collapse
|
18
|
Tsujimoto H, Hoshina A, Mae SI, Araoka T, Changting W, Ijiri Y, Nakajima-Koyama M, Sakurai S, Okita K, Mizuta K, Niwa A, Saito MK, Saitou M, Yamamoto T, Graneli C, Woollard KJ, Osafune K. Selective induction of human renal interstitial progenitor-like cell lineages from iPSCs reveals development of mesangial and EPO-producing cells. Cell Rep 2024; 43:113602. [PMID: 38237600 DOI: 10.1016/j.celrep.2023.113602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/13/2023] [Accepted: 12/05/2023] [Indexed: 03/02/2024] Open
Abstract
Recent regenerative studies using human pluripotent stem cells (hPSCs) have developed multiple kidney-lineage cells and organoids. However, to further form functional segments of the kidney, interactions of epithelial and interstitial cells are required. Here we describe a selective differentiation of renal interstitial progenitor-like cells (IPLCs) from human induced pluripotent stem cells (hiPSCs) by modifying our previous induction method for nephron progenitor cells (NPCs) and analyzing mouse embryonic interstitial progenitor cell (IPC) development. Our IPLCs combined with hiPSC-derived NPCs and nephric duct cells form nephrogenic niche- and mesangium-like structures in vitro. Furthermore, we successfully induce hiPSC-derived IPLCs to differentiate into mesangial and erythropoietin-producing cell lineages in vitro by screening differentiation-inducing factors and confirm that p38 MAPK, hypoxia, and VEGF signaling pathways are involved in the differentiation of mesangial-lineage cells. These findings indicate that our IPC-lineage induction method contributes to kidney regeneration and developmental research.
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; Rege Nephro Co., Ltd., Med-Pharm Collaboration Building, Kyoto University, 46-29 Yoshidashimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Azusa Hoshina
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Shin-Ichi Mae
- 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
| | - Wang Changting
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yoshihiro Ijiri
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - May Nakajima-Koyama
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Satoko Sakurai
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Kazusa Okita
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Ken Mizuta
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Akira Niwa
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Megumu K Saito
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Mitinori Saitou
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan; Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan; Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto 606-8501, Japan
| | - Takuya Yamamoto
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan; Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto 606-8501, Japan; Medical-risk Avoidance based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto 606-8507, Japan
| | - Cecilia Graneli
- BioPharmaceuticals R&D Cell Therapy, Research and Early Development, Cardiovascular, Renal and Metabolic (CVRM), BioPharmaceuticals R&D, AstraZeneca, 431 83 Gothenburg, Sweden
| | - Kevin J Woollard
- Bioscience Renal, Research and Early Development, Cardiovascular, Renal and Metabolic, BioPharmaceuticals R&D, AstraZeneca, Cambridge CB2 0AA, UK
| | - Kenji Osafune
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan.
| |
Collapse
|
19
|
Tabibzadeh N, Satlin LM, Jain S, Morizane R. Navigating the kidney organoid: insights into assessment and enhancement of nephron function. Am J Physiol Renal Physiol 2023; 325:F695-F706. [PMID: 37767571 PMCID: PMC10878724 DOI: 10.1152/ajprenal.00166.2023] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/18/2023] [Accepted: 09/20/2023] [Indexed: 09/29/2023] Open
Abstract
Kidney organoids are three-dimensional structures generated from pluripotent stem cells (PSCs) that are capable of recapitulating the major structures of mammalian kidneys. As this technology is expected to be a promising tool for studying renal biology, drug discovery, and regenerative medicine, the functional capacity of kidney organoids has emerged as a critical question in the field. Kidney organoids produced using several protocols harbor key structures of native kidneys. Here, we review the current state, recent advances, and future challenges in the functional characterization of kidney organoids, strategies to accelerate and enhance kidney organoid functions, and access to PSC resources to advance organoid research. The strategies to construct physiologically relevant kidney organoids include the use of organ-on-a-chip technologies that integrate fluid circulation and improve organoid maturation. These approaches result in increased expression of the major tubular transporters and elements of mechanosensory signaling pathways suggestive of improved functionality. Nevertheless, continuous efforts remain crucial to create kidney tissue that more faithfully replicates physiological conditions for future applications in kidney regeneration medicine and their ethical use in patient care.NEW & NOTEWORTHY Kidney organoids are three-dimensional structures derived from stem cells, mimicking the major components of mammalian kidneys. Although they show great promise, their functional capacity has become a critical question. This review explores the advancements and challenges in evaluating and enhancing kidney organoid function, including the use of organ-on-chip technologies, multiomics data, and in vivo transplantation. Integrating these approaches to further enhance their physiological relevance will continue to advance disease modeling and regenerative medicine applications.
Collapse
Affiliation(s)
- Nahid Tabibzadeh
- Nephrology Division, Massachusetts General Hospital, Boston, Massachusetts, United States
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States
| | - Lisa M Satlin
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Sanjay Jain
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Pathology, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Ryuji Morizane
- Nephrology Division, Massachusetts General Hospital, Boston, Massachusetts, United States
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
20
|
Nauryzgaliyeva Z, Goux Corredera I, Garreta E, Montserrat N. Harnessing mechanobiology for kidney organoid research. Front Cell Dev Biol 2023; 11:1273923. [PMID: 38077999 PMCID: PMC10704179 DOI: 10.3389/fcell.2023.1273923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 10/16/2023] [Indexed: 10/16/2024] Open
Abstract
Recently, organoids have emerged as revolutionizing tools with the unprecedented potential to recreate organ-specific microanatomy in vitro. Upon their derivation from human pluripotent stem cells (hPSCs), organoids reveal the blueprints of human organogenesis, further allowing the faithful recapitulation of their physiology. Nevertheless, along with the evolution of this field, advanced research exposed the organoids' shortcomings, particularly regarding poor reproducibility rates and overall immatureness. To resolve these challenges, many studies have started to underscore the relevance of mechanical cues as a relevant source to induce and externally control hPSCs differentiation. Indeed, established organoid generation protocols from hPSCs have mainly relyed on the biochemical induction of fundamental signalling pathways present during kidney formation in mammals, whereas mechanical cues have largely been unexplored. This review aims to discuss the pertinence of (bio) physical cues within hPSCs-derived organoid cultures, while deciphering their effect on morphogenesis. Moreover, we will explore state-of-the-art mechanobiology techniques as revolutionizing means for understanding the underlying role of mechanical forces in biological processes in organoid model systems.
Collapse
Affiliation(s)
- Zarina Nauryzgaliyeva
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Iphigénie Goux Corredera
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), University of Barcelona, Barcelona, Spain
| | - Elena Garreta
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), University of Barcelona, Barcelona, Spain
| | - Nuria Montserrat
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina, Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|
21
|
Ibi Y, Nishinakamura R. Kidney Bioengineering for Transplantation. Transplantation 2023; 107:1883-1894. [PMID: 36717963 DOI: 10.1097/tp.0000000000004526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The kidney is an important organ for maintenance of homeostasis in the human body. As renal failure progresses, renal replacement therapy becomes necessary. However, there is a chronic shortage of kidney donors, creating a major problem for transplantation. To solve this problem, many strategies for the generation of transplantable kidneys are under investigation. Since the first reports describing that nephron progenitors could be induced from human induced pluripotent stem cells, kidney organoids have been attracting attention as tools for studying human kidney development and diseases. Because the kidney is formed through the interactions of multiple renal progenitors, current studies are investigating ways to combine these progenitors derived from human induced pluripotent stem cells for the generation of transplantable kidney organoids. Other bioengineering strategies, such as decellularization and recellularization of scaffolds, 3-dimensional bioprinting, interspecies blastocyst complementation and progenitor replacement, and xenotransplantation, also have the potential to generate whole kidneys, although each of these strategies has its own challenges. Combinations of these approaches will lead to the generation of bioengineered kidneys that are transplantable into humans.
Collapse
Affiliation(s)
- Yutaro Ibi
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | | |
Collapse
|
22
|
Nishinakamura R. Advances and challenges toward developing kidney organoids for clinical applications. Cell Stem Cell 2023; 30:1017-1027. [PMID: 37541208 DOI: 10.1016/j.stem.2023.07.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 07/12/2023] [Accepted: 07/12/2023] [Indexed: 08/06/2023]
Abstract
Kidney organoids have enabled modeling of human development and disease. While methods of generating the nephron lineage are well established, new protocols to induce another lineage, the ureteric bud/collecting duct, have been reported in the past 5 years. Many reports have described modeling of various hereditary kidney diseases, with polycystic kidney disease serving as the archetypal disease, by using patient-derived or genome-edited kidney organoids. The generation of more organotypic kidneys is also becoming feasible. In this review, I also discuss the significant challenges for more sophisticated disease modeling and for realizing the ambitious goal of generating transplantable synthetic kidneys.
Collapse
Affiliation(s)
- Ryuichi Nishinakamura
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan.
| |
Collapse
|
23
|
Yang S, Hu H, Kung H, Zou R, Dai Y, Hu Y, Wang T, Lv T, Yu J, Li F. Organoids: The current status and biomedical applications. MedComm (Beijing) 2023; 4:e274. [PMID: 37215622 PMCID: PMC10192887 DOI: 10.1002/mco2.274] [Citation(s) in RCA: 82] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 04/22/2023] [Accepted: 04/27/2023] [Indexed: 05/24/2023] Open
Abstract
Organoids are three-dimensional (3D) miniaturized versions of organs or tissues that are derived from cells with stem potential and can self-organize and differentiate into 3D cell masses, recapitulating the morphology and functions of their in vivo counterparts. Organoid culture is an emerging 3D culture technology, and organoids derived from various organs and tissues, such as the brain, lung, heart, liver, and kidney, have been generated. Compared with traditional bidimensional culture, organoid culture systems have the unique advantage of conserving parental gene expression and mutation characteristics, as well as long-term maintenance of the function and biological characteristics of the parental cells in vitro. All these features of organoids open up new opportunities for drug discovery, large-scale drug screening, and precision medicine. Another major application of organoids is disease modeling, and especially various hereditary diseases that are difficult to model in vitro have been modeled with organoids by combining genome editing technologies. Herein, we introduce the development and current advances in the organoid technology field. We focus on the applications of organoids in basic biology and clinical research, and also highlight their limitations and future perspectives. We hope that this review can provide a valuable reference for the developments and applications of organoids.
Collapse
Affiliation(s)
- Siqi Yang
- Division of Biliary Tract SurgeryDepartment of General SurgeryWest China HospitalSichuan UniversityChengduSichuan ProvinceChina
| | - Haijie Hu
- Division of Biliary Tract SurgeryDepartment of General SurgeryWest China HospitalSichuan UniversityChengduSichuan ProvinceChina
| | - Hengchung Kung
- Krieger School of Arts and SciencesJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Ruiqi Zou
- Division of Biliary Tract SurgeryDepartment of General SurgeryWest China HospitalSichuan UniversityChengduSichuan ProvinceChina
| | - Yushi Dai
- Division of Biliary Tract SurgeryDepartment of General SurgeryWest China HospitalSichuan UniversityChengduSichuan ProvinceChina
| | - Yafei Hu
- Division of Biliary Tract SurgeryDepartment of General SurgeryWest China HospitalSichuan UniversityChengduSichuan ProvinceChina
| | - Tiantian Wang
- Key Laboratory of Rehabilitation Medicine in Sichuan ProvinceWest China HospitalSichuan UniversityChengduSichuanChina
| | - Tianrun Lv
- Division of Biliary Tract SurgeryDepartment of General SurgeryWest China HospitalSichuan UniversityChengduSichuan ProvinceChina
| | - Jun Yu
- Departments of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Departments of OncologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Fuyu Li
- Division of Biliary Tract SurgeryDepartment of General SurgeryWest China HospitalSichuan UniversityChengduSichuan ProvinceChina
| |
Collapse
|
24
|
Singh J, Singh S. Review on kidney diseases: types, treatment and potential of stem cell therapy. RENAL REPLACEMENT THERAPY 2023; 9:21. [PMID: 37131920 PMCID: PMC10134709 DOI: 10.1186/s41100-023-00475-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 04/11/2023] [Indexed: 05/04/2023] Open
Abstract
Renal disorders are an emerging global public health issue with a higher growth rate despite progress in supportive therapies. In order to find more promising treatments to stimulate renal repair, stem cell-based technology has been proposed as a potentially therapeutic option. The self-renewal and proliferative nature of stem cells raised the hope to fight against various diseases. Similarly, it opens a new path for the treatment and repair of damaged renal cells. This review focuses on the types of renal diseases; acute and chronic kidney disease-their statistical data, and the conventional drugs used for treatment. It includes the possible stem cell therapy mechanisms involved and outcomes recorded so far, the limitations of using these regenerative medicines, and the progressive improvement in stem cell therapy by adopting approaches like PiggyBac, Sleeping Beauty, and the Sendai virus. Specifically, about the paracrine activities of amniotic fluid stem cells, renal stem cells, embryonic stem cells, mesenchymal stem cell, induced pluripotent stem cells as well as other stem cells.
Collapse
Affiliation(s)
- Jaspreet Singh
- School of Bioengineering & Biosciences, Lovely Professional University, 15935, Block 56, Room No 202, Phagwara, Punjab 144411 India
| | - Sanjeev Singh
- School of Bioengineering & Biosciences, Lovely Professional University, 15935, Block 56, Room No 202, Phagwara, Punjab 144411 India
| |
Collapse
|
25
|
Freedman BS, Dekel B. Engraftment of Kidney Organoids In Vivo. CURRENT TRANSPLANTATION REPORTS 2023; 10:29-39. [PMID: 37128257 PMCID: PMC10126570 DOI: 10.1007/s40472-023-00397-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/05/2023] [Indexed: 05/03/2023]
Abstract
Purpose of Review Kidney organoids are heterocellular structures grown in vitro that resemble nephrons. Organoids contain diverse cell types, including podocytes, proximal tubules, and distal tubules in contiguous segments, patterned along a proximal-to-distal axis. Human organoids are being explored for their potential as regenerative grafts, as an alternative to allograft transplants and hemodialysis. Earlier work, analyzing grafts of developing human kidney tissue and whole human embryonic kidney rudiments, serves as a baseline for organoid implantation experiments. Recent Findings When transplanted into immunodeficient mice beneath the kidney capsule, kidney organoid xenografts can form vascularized, glomerulus-like structures, which exhibit a degree of filtration function. However, the absence of an appropriate collecting duct outlet and the presence of abundant stromal-like cells limits the functionality of such grafts and raises safety concerns. Recently, ureteric-like organoids have also been generated, which extend projections that resemble collecting ducts. Summary Combining nephron-like and ureteric-like organoids, along with renal stromal cells, may provide a path towards more functional grafts.
Collapse
Affiliation(s)
- Benjamin S. Freedman
- Division of Nephrology, Kidney Research Institute, and Institute for Stem Cell and Regenerative Medicine, Departments of Medicine, Pathology (Adjunct), and Bioengineering (Adjunct), University of Washington School of Medicine, Seattle, WA USA
- Plurexa LLC, Seattle, WA USA
| | - Benjamin Dekel
- Division of Pediatric Nephrology and the Pediatric Stem Cell Research Institute, Sagol Center for Regenerative Medicine, Sheba Medical Center, School of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
| |
Collapse
|
26
|
The "3Ds" of Growing Kidney Organoids: Advances in Nephron Development, Disease Modeling, and Drug Screening. Cells 2023; 12:cells12040549. [PMID: 36831216 PMCID: PMC9954122 DOI: 10.3390/cells12040549] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/03/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023] Open
Abstract
A kidney organoid is a three-dimensional (3D) cellular aggregate grown from stem cells in vitro that undergoes self-organization, recapitulating aspects of normal renal development to produce nephron structures that resemble the native kidney organ. These miniature kidney-like structures can also be derived from primary patient cells and thus provide simplified context to observe how mutations in kidney-disease-associated genes affect organogenesis and physiological function. In the past several years, advances in kidney organoid technologies have achieved the formation of renal organoids with enhanced numbers of specialized cell types, less heterogeneity, and more architectural complexity. Microfluidic bioreactor culture devices, single-cell transcriptomics, and bioinformatic analyses have accelerated the development of more sophisticated renal organoids and tailored them to become increasingly amenable to high-throughput experimentation. However, many significant challenges remain in realizing the use of kidney organoids for renal replacement therapies. This review presents an overview of the renal organoid field and selected highlights of recent cutting-edge kidney organoid research with a focus on embryonic development, modeling renal disease, and personalized drug screening.
Collapse
|
27
|
Production of kidney organoids arranged around single ureteric bud trees, and containing endogenous blood vessels, solely from embryonic stem cells. Sci Rep 2022; 12:12573. [PMID: 35869233 PMCID: PMC9307805 DOI: 10.1038/s41598-022-16768-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 07/15/2022] [Indexed: 11/09/2022] Open
Abstract
There is intense worldwide effort in generating kidney organoids from pluripotent stem cells, for research, for disease modelling and, perhaps, for making transplantable organs. Organoids generated from pluripotent stem cells (PSC) possess accurate micro-anatomy, but they lack higher-organization. This is a problem, especially for transplantation, as such organoids will not be able to perform their physiological functions. In this study, we develop a method for generating murine kidney organoids with improved higher-order structure, through stages using chimaeras of ex-fetu and PSC-derived cells to a system that works entirely from embryonic stem cells. These organoids have nephrons organised around a single ureteric bud tree and also make vessels, with the endothelial network approaching podocytes.
Collapse
|
28
|
Tekguc M, Gaal RCVAN, Uzel SGM, Gupta N, Riella LV, Lewis JA, Morizane R. Kidney organoids: a pioneering model for kidney diseases. Transl Res 2022; 250:1-17. [PMID: 35750295 PMCID: PMC9691572 DOI: 10.1016/j.trsl.2022.06.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 06/14/2022] [Accepted: 06/16/2022] [Indexed: 11/18/2022]
Abstract
The kidney is a vital organ that regulates the bodily fluid and electrolyte homeostasis via tailored urinary excretion. Kidney injuries that cause severe or progressive chronic kidney disease have driven the growing population of patients with end-stage kidney disease, leading to substantial patient morbidity and mortality. This irreversible kidney damage has also created a huge socioeconomical burden on the healthcare system, highlighting the need for novel translational research models for progressive kidney diseases. Conventional research methods such as in vitro 2D cell culture or animal models do not fully recapitulate complex human kidney diseases. By contrast, directed differentiation of human induced pluripotent stem cells enables in vitro generation of patient-specific 3D kidney organoids, which can be used to model acute or chronic forms of hereditary, developmental, and metabolic kidney diseases. Furthermore, when combined with biofabrication techniques, organoids can be used as building blocks to construct vascularized kidney tissues mimicking their in vivo counterpart. By applying gene editing technology, organoid building blocks may be modified to minimize the process of immune rejection in kidney transplant recipients. In the foreseeable future, the universal kidney organoids derived from HLA-edited/deleted induced pluripotent stem cell (iPSC) lines may enable the supply of bioengineered organotypic kidney structures that are immune-compatible for the majority of the world population. Here, we summarize recent advances in kidney organoid research coupled with novel technologies such as organoids-on-chip and biofabrication of 3D kidney tissues providing convenient platforms for high-throughput drug screening, disease modelling, and therapeutic applications.
Collapse
Affiliation(s)
- Murat Tekguc
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts; Harvard Stem Cell Institute (HSCI), Cambridge, Massachusetts
| | - Ronald C VAN Gaal
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts; School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts
| | - Sebastien G M Uzel
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts; School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts
| | - Navin Gupta
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts; Harvard Stem Cell Institute (HSCI), Cambridge, Massachusetts
| | - Leonardo V Riella
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts; Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts
| | - Jennifer A Lewis
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts; School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts
| | - Ryuji Morizane
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts; Harvard Stem Cell Institute (HSCI), Cambridge, Massachusetts; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts.
| |
Collapse
|
29
|
Tanigawa S, Nishinakamura R. Functional renal collecting ducts from human PSCs. Cell Stem Cell 2022; 29:1510-1512. [PMID: 36332567 DOI: 10.1016/j.stem.2022.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Collecting ducts in the kidney control the balance of water and electrolytes, as well as pH of the urine to maintain body homeostasis. Shi et al.1 recently reported in Nature Biotechnology a protocol to generate functional collecting duct cells from human pluripotent stem cells.
Collapse
Affiliation(s)
- Shunsuke Tanigawa
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Ryuichi Nishinakamura
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan.
| |
Collapse
|
30
|
Dorison A, Forbes TA, Little MH. What can we learn from kidney organoids? Kidney Int 2022; 102:1013-1029. [PMID: 35970244 DOI: 10.1016/j.kint.2022.06.032] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/15/2022] [Accepted: 06/24/2022] [Indexed: 12/14/2022]
Abstract
The ability to generate 3-dimensional models of the developing human kidney via the directed differentiation of pluripotent stem cells-termed kidney organoids-has been hailed as a major advance in experimental nephrology. Although these provide an opportunity to interrogate human development, model-specific kidney diseases facilitate drug screening and even deliver bioengineered tissue; most of these prophetic end points remain to be realized. Indeed, at present we are still finding out what we can learn and what we cannot learn from this approach. In this review, we will summarize the approaches available to generate models of the human kidney from stem cells, the existing successful applications of kidney organoids, their limitations, and remaining challenges.
Collapse
Affiliation(s)
- Aude Dorison
- Murdoch Children's Research Institute, Parkville, Melbourne, Australia; Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Melbourne, Australia; Novo Nordisk Foundation Centre for Stem Cell Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Thomas A Forbes
- Murdoch Children's Research Institute, Parkville, Melbourne, Australia; Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Melbourne, Australia; Department of Nephrology, Royal Children's Hospital, Parkville, Melbourne, Australia
| | - Melissa H Little
- Murdoch Children's Research Institute, Parkville, Melbourne, Australia; Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Melbourne, Australia; Novo Nordisk Foundation Centre for Stem Cell Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| |
Collapse
|
31
|
Vanslambrouck JM, Wilson SB, Tan KS, Groenewegen E, Rudraraju R, Neil J, Lawlor KT, Mah S, Scurr M, Howden SE, Subbarao K, Little MH. Enhanced metanephric specification to functional proximal tubule enables toxicity screening and infectious disease modelling in kidney organoids. Nat Commun 2022; 13:5943. [PMID: 36209212 PMCID: PMC9547573 DOI: 10.1038/s41467-022-33623-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 09/27/2022] [Indexed: 01/08/2023] Open
Abstract
While pluripotent stem cell-derived kidney organoids are now being used to model renal disease, the proximal nephron remains immature with limited evidence for key functional solute channels. This may reflect early mispatterning of the nephrogenic mesenchyme and/or insufficient maturation. Here we show that enhanced specification to metanephric nephron progenitors results in elongated and radially aligned proximalised nephrons with distinct S1 - S3 proximal tubule cell types. Such PT-enhanced organoids possess improved albumin and organic cation uptake, appropriate KIM-1 upregulation in response to cisplatin, and improved expression of SARS-CoV-2 entry factors resulting in increased viral replication. The striking proximo-distal orientation of nephrons resulted from localized WNT antagonism originating from the organoid stromal core. PT-enhanced organoids represent an improved model to study inherited and acquired proximal tubular disease as well as drug and viral responses.
Collapse
Affiliation(s)
- Jessica M Vanslambrouck
- Murdoch Children's Research Institute, Flemington Rd, Parkville, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
| | - Sean B Wilson
- Murdoch Children's Research Institute, Flemington Rd, Parkville, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
| | - Ker Sin Tan
- Murdoch Children's Research Institute, Flemington Rd, Parkville, VIC, Australia
| | - Ella Groenewegen
- Murdoch Children's Research Institute, Flemington Rd, Parkville, VIC, Australia
| | - Rajeev Rudraraju
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, Australia
| | - Jessica Neil
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, Australia
| | - Kynan T Lawlor
- Murdoch Children's Research Institute, Flemington Rd, Parkville, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
| | - Sophia Mah
- Murdoch Children's Research Institute, Flemington Rd, Parkville, VIC, Australia
| | - Michelle Scurr
- Murdoch Children's Research Institute, Flemington Rd, Parkville, VIC, Australia
| | - Sara E Howden
- Murdoch Children's Research Institute, Flemington Rd, Parkville, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
| | - Kanta Subbarao
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, Australia
| | - Melissa H Little
- Murdoch Children's Research Institute, Flemington Rd, 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.
| |
Collapse
|
32
|
Huang B, Zeng Z, Zhang CC, Schreiber ME, Li Z. Approaches to kidney replacement therapies—opportunities and challenges. Front Cell Dev Biol 2022; 10:953408. [PMID: 35982852 PMCID: PMC9380013 DOI: 10.3389/fcell.2022.953408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/01/2022] [Indexed: 11/29/2022] Open
Abstract
One out of seven people develop chronic kidney disease (CKD). When kidney function continues to decline, CKD patients may develop end-stage renal disease (ESRD, or kidney failure). More than 2 out of 1,000 adults develop ESRD and these patients must live on dialysis or get a kidney transplant to survive. Each year, more than $51 billion is spent to treat patients with ESRD in the United States. In addition, ESRD greatly reduces longevity and quality of life for patients. Compared to dialysis, kidney transplant offers the best chance of survival, but few donor organs are available. Thus, there is an urgent need for innovative solutions that address the shortage of kidneys available for transplantation. Here we summarize the status of current approaches that are being developed to solve the shortage of donor kidneys. These include the bioartificial kidney approach which aims to make a portable dialysis device, the recellularization approach which utilizes native kidney scaffold to make an engineered kidney, the stem cell-based approach which aims to generate a kidney de novo by recapitulating normal kidney organogenesis, the xenotransplantation approach which has the goal to make immunocompatible pig kidneys for transplantation, and the interspecies chimera approach which has potential to generate a human kidney in a host animal. We also discuss the interconnections among the different approaches, and the remaining challenges of translating these approaches into novel therapies.
Collapse
Affiliation(s)
- Biao Huang
- USC/UKRO Kidney Research Center, Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Deptartment of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Zipeng Zeng
- USC/UKRO Kidney Research Center, Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Deptartment of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Chennan C. Zhang
- USC/UKRO Kidney Research Center, Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Deptartment of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Megan E. Schreiber
- USC/UKRO Kidney Research Center, Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Deptartment of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Zhongwei Li
- USC/UKRO Kidney Research Center, Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Deptartment of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- *Correspondence: Zhongwei Li,
| |
Collapse
|
33
|
Trush O, Takasato M. Kidney organoid research: current status and applications. Curr Opin Genet Dev 2022; 75:101944. [PMID: 35785592 DOI: 10.1016/j.gde.2022.101944] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/19/2022] [Accepted: 05/25/2022] [Indexed: 11/03/2022]
Abstract
Organoids are being widely introduced as novel research models in multiple research fields. Human-induced pluripotent stem cells-derived kidney organoids became an indispensable tool to study human kidney development, model various diseases and infections leading to kidney damage, and offer a new route towards better drug development and validation, personalized drug screening, and regenerative medicine. In this review, we provide an update of the most recent developments in kidney organoid induction: their main goals, advantages, and shortcomings. We further discuss their current applications in providing modeling and treatment avenues to various kidney injuries, their use in genome-wide screening of kidney diseases, and the cell interactions occurring in these kidney structures.
Collapse
Affiliation(s)
- Olena Trush
- Laboratory for Human Organogenesis, RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan
| | - Minoru Takasato
- Laboratory for Human Organogenesis, RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan; Laboratory of Molecular Cell Biology and Development, Department of Animal Development and Physiology, Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan; Department of Development and Regeneration, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan.
| |
Collapse
|
34
|
Kobayashi A, Nishinakamura R. Building kidney organoids from pluripotent stem cells. Curr Opin Nephrol Hypertens 2022; 31:367-373. [PMID: 35727170 DOI: 10.1097/mnh.0000000000000807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW During embryogenesis, the kidney is mainly generated from three progenitor cells; nephron progenitors, ureteric bud progenitors and stromal progenitors. Mutual interactions of the all three progenitor populations are essential to form a functional kidney with the higher-order structure. Pluripotent stem cells have potential to differentiate into all cell types of the animal body, including the kidney. In this review, we will summarize recent advances in reconstructing kidney organoids from pluripotent stem cells. RECENT FINDINGS In the past years, major advances were reported to induce nephron and ureteric bud progenitors from pluripotent stem cells in mice and humans, and to create kidney organoids of nephron and/or ureteric bud-derived collecting duct tissues in vitro. These kidney organoid technologies were applied to high-throughput genetic screenings and small chemical screenings to identify key factors for kidney development and disease. Furthermore, a novel method was established to induce stromal progenitors from pluripotent stem cells, leading to creation of kidney organoids with the higher-order structures completely derived from pluripotent stem cells. SUMMARY These advances in kidney organoids from pluripotent stem cells should lay a foundation to establish a novel therapy for kidney disease, which ultimately eliminate the need of dialysis and kidney transplantation for patients with kidney disease in the future.
Collapse
Affiliation(s)
- Akio Kobayashi
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | | |
Collapse
|
35
|
Saito Y, Yamanaka S, Matsumoto N, Takamura T, Fujimoto T, Matsui K, Tajiri S, Matsumoto K, Kobayashi E, Yokoo T. Generation of functional chimeric kidney containing exogenous progenitor-derived stroma and nephron via a conditional empty niche. Cell Rep 2022; 39:110933. [PMID: 35705028 DOI: 10.1016/j.celrep.2022.110933] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 03/31/2022] [Accepted: 05/18/2022] [Indexed: 11/17/2022] Open
Abstract
Generation of new kidneys can be useful in various research fields, including organ transplantation. However, generating renal stroma, an important component tissue for structural support, endocrine function, and kidney development, remains difficult. Organ generation using an animal developmental niche can provide an appropriate in vivo environment for renal stroma differentiation. Here, we generate rat renal stroma with endocrine capacity by removing mouse stromal progenitor cells (SPCs) from the host developmental niche and transplanting rat SPCs. Furthermore, we develop a method to replace both nephron progenitor cells (NPCs) and SPCs, called the interspecies dual replacement of the progenitor (i-DROP) system, and successfully generate functional chimeric kidneys containing rat nephrons and stroma. This method can generate renal tissue from progenitors and reduce xenotransplant rejection. Moreover, it is a safe method, as donor cells do not stray into nontarget organs, thus accelerating research on stem cells, chimeras, and xenotransplantation.
Collapse
Affiliation(s)
- Yatsumu Saito
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Shuichiro Yamanaka
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Naoto Matsumoto
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Tsuyoshi Takamura
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Toshinari Fujimoto
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Kenji Matsui
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Susumu Tajiri
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Kei Matsumoto
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Eiji Kobayashi
- Department of Kidney Regenerative Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Takashi Yokoo
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan.
| |
Collapse
|
36
|
Freedman BS. Physiology assays in human kidney organoids. Am J Physiol Renal Physiol 2022; 322:F625-F638. [PMID: 35379001 PMCID: PMC9076410 DOI: 10.1152/ajprenal.00400.2021] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 01/15/2023] Open
Abstract
Kidney organoids derived from human pluripotent stem cells constitute a novel model of disease, development, and regenerative therapy. Organoids are human, experimentally accessible, high throughput, and enable reconstitution of tissue-scale biology in a petri dish. Although gene expression patterns in organoid cells have been analyzed extensively, less is known about the functionality of these structures. Here, we review assays of physiological function in human kidney organoids, including best practices for quality control, and future applications. Tubular structures in organoids accumulate specific molecules through active transport, including dextran and organic anions, and swell with fluid in response to cAMP stimulation. When engrafted into animal models in vivo, organoids form vascularized glomerulus-like structures capable of size-selective filtration. Organoids exhibit metabolic, endocrine, injury, and infection phenotypes, although their specificity is not yet fully clear. To properly interpret organoid physiology assays, it is important to incorporate appropriate negative and positive controls, statistical methods, data presentation, molecular mechanisms, and clinical data sets. Improvements in organoid perfusion, patterning, and maturation are needed to enable branching morphogenesis, urine production, and renal replacement. Reconstituting renal physiology with kidney organoids is a new field with potential to provide fresh insights into classical phenomena.
Collapse
Affiliation(s)
- Benjamin S Freedman
- Division of Nephrology, Kidney Research Institute, and Institute for Stem Cell and Regenerative Medicine, Department of Medicine, Department of Laboratory Medicine and Physiology (adjunct), and Department of Bioengineering (adjunct), University of Washington School of Medicine, Seattle, Washington
| |
Collapse
|
37
|
Vanslambrouck JM, Wilson SB, Tan KS, Groenewegen E, Rudraraju R, Neil J, Lawlor KT, Mah S, Scurr M, Howden SE, Subbarao K, Little MH. Enhanced metanephric specification to functional proximal tubule enables toxicity screening and infectious disease modelling in kidney organoids. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2021.10.14.464320. [PMID: 35665006 PMCID: PMC9164445 DOI: 10.1101/2021.10.14.464320] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
While pluripotent stem cell-derived kidney organoids are now being used to model renal disease, the proximal nephron remains immature with limited evidence for key functional solute channels. This may reflect early mispatterning of the nephrogenic mesenchyme and/or insufficient maturation. Here we show that enhanced specification to metanephric nephron progenitors results in elongated and radially aligned proximalised nephrons with distinct S1 - S3 proximal tubule cell types. Such PT-enhanced organoids possess improved albumin and organic cation uptake, appropriate KIM-1 upregulation in response to cisplatin, and improved expression of SARS-CoV-2 entry factors resulting in increased viral replication. The striking proximo-distal orientation of nephrons resulted from localized WNT antagonism originating from the organoid stromal core. PT-enhanced organoids represent an improved model to study inherited and acquired proximal tubular disease as well as drug and viral responses.
Collapse
Affiliation(s)
- Jessica M. Vanslambrouck
- Murdoch Children’s Research Institute, Flemington Rd, Parkville, VIC, Australia
- Department of Paediatrics, The University of Melbourne, VIC, Australia
| | - Sean B. Wilson
- Murdoch Children’s Research Institute, Flemington Rd, Parkville, VIC, Australia
- Department of Paediatrics, The University of Melbourne, VIC, Australia
| | - Ker Sin Tan
- Murdoch Children’s Research Institute, Flemington Rd, Parkville, VIC, Australia
| | - Ella Groenewegen
- Murdoch Children’s Research Institute, Flemington Rd, Parkville, VIC, Australia
| | - Rajeev Rudraraju
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, VIC, Australia
| | - Jessica Neil
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, VIC, Australia
| | - Kynan T. Lawlor
- Murdoch Children’s Research Institute, Flemington Rd, Parkville, VIC, Australia
- Department of Paediatrics, The University of Melbourne, VIC, Australia
| | - Sophia Mah
- Murdoch Children’s Research Institute, Flemington Rd, Parkville, VIC, Australia
| | - Michelle Scurr
- Murdoch Children’s Research Institute, Flemington Rd, Parkville, VIC, Australia
| | - Sara E. Howden
- Murdoch Children’s Research Institute, Flemington Rd, Parkville, VIC, Australia
- Department of Paediatrics, The University of Melbourne, VIC, Australia
| | - Kanta Subbarao
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, VIC, Australia
| | - Melissa H. Little
- Murdoch Children’s Research Institute, Flemington Rd, Parkville, VIC, Australia
- Department of Paediatrics, The University of Melbourne, VIC, Australia
- Department of Anatomy and Neuroscience, The University of Melbourne, VIC, Australia
- Author for correspondence: M.H.L.: +61 3 9936 6206;
| |
Collapse
|
38
|
Gabbin B, Meraviglia V, Mummery CL, Rabelink TJ, van Meer BJ, van den Berg CW, Bellin M. Toward Human Models of Cardiorenal Syndrome in vitro. Front Cardiovasc Med 2022; 9:889553. [PMID: 35694669 PMCID: PMC9177996 DOI: 10.3389/fcvm.2022.889553] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 05/09/2022] [Indexed: 11/13/2022] Open
Abstract
Heart and kidney diseases cause high morbidity and mortality. Heart and kidneys have vital functions in the human body and, interestingly, reciprocally influence each other’s behavior: pathological changes in one organ can damage the other. Cardiorenal syndrome (CRS) is a group of disorders in which there is combined dysfunction of both heart and kidney, but its underlying biological mechanisms are not fully understood. This is because complex, multifactorial, and dynamic mechanisms are likely involved. Effective treatments are currently unavailable, but this may be resolved if more was known about how the disease develops and progresses. To date, CRS has actually only been modeled in mice and rats in vivo. Even though these models can capture cardiorenal interaction, they are difficult to manipulate and control. Moreover, interspecies differences may limit extrapolation to patients. The questions we address here are what would it take to model CRS in vitro and how far are we? There are already multiple independent in vitro (human) models of heart and kidney, but none have so far captured their dynamic organ-organ crosstalk. Advanced in vitro human models can provide an insight in disease mechanisms and offer a platform for therapy development. CRS represents an exemplary disease illustrating the need to develop more complex models to study organ-organ interaction in-a-dish. Human induced pluripotent stem cells in combination with microfluidic chips are one powerful tool with potential to recapitulate the characteristics of CRS in vitro. In this review, we provide an overview of the existing in vivo and in vitro models to study CRS, their limitations and new perspectives on how heart-kidney physiological and pathological interaction could be investigated in vitro for future applications.
Collapse
Affiliation(s)
- Beatrice Gabbin
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
| | - Viviana Meraviglia
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
| | - Christine L. Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
- Department of Applied Stem Cell Technologies, University of Twente, Enschede, Netherlands
| | - Ton J. Rabelink
- Department of Internal Medicine-Nephrology, Leiden University Medical Center, Leiden, Netherlands
- Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Berend J. van Meer
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
| | - Cathelijne W. van den Berg
- Department of Internal Medicine-Nephrology, Leiden University Medical Center, Leiden, Netherlands
- Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Milena Bellin
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
- Department of Biology, University of Padua, Padua, Italy
- Veneto Institute of Molecular Medicine, Padua, Italy
- *Correspondence: Milena Bellin, ,
| |
Collapse
|
39
|
Ilic D, Liovic M. Industry updates from the field of stem cell research and regenerative medicine in February 2022. Regen Med 2022. [PMID: 35354328 DOI: 10.2217/rme-2022-0046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Latest developments in the field of stem cell research and regenerative medicine compiled from publicly available information and press releases from non academic institutions in February 2022.
Collapse
Affiliation(s)
- Dusko Ilic
- Department of Women & Children's Health, Stem Cell Laboratories, Guy's Assisted Conception Unit, Faculty of Life Sciences & Medicine, King's College London, London, SE1 9RT, UK
| | - Mirjana Liovic
- Medical Center for Molecular Biology, Faculty of Medicine, University of Ljubljana, Ljubljana, 1000, Slovenia
| |
Collapse
|
40
|
Liu M, Cardilla A, Ngeow J, Gong X, Xia Y. Studying Kidney Diseases Using Organoid Models. Front Cell Dev Biol 2022; 10:845401. [PMID: 35309912 PMCID: PMC8927804 DOI: 10.3389/fcell.2022.845401] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 02/14/2022] [Indexed: 12/24/2022] Open
Abstract
The prevalence of chronic kidney disease (CKD) is rapidly increasing over the last few decades, owing to the global increase in diabetes, and cardiovascular diseases. Dialysis greatly compromises the life quality of patients, while demand for transplantable kidney cannot be met, underscoring the need to develop novel therapeutic approaches to stop or reverse CKD progression. Our understanding of kidney disease is primarily derived from studies using animal models and cell culture. While cross-species differences made it challenging to fully translate findings from animal models into clinical practice, primary patient cells quickly lose the original phenotypes during in vitro culture. Over the last decade, remarkable achievements have been made for generating 3-dimensional (3D) miniature organs (organoids) by exposing stem cells to culture conditions that mimic the signaling cues required for the development of a particular organ or tissue. 3D kidney organoids have been successfully generated from different types of source cells, including human pluripotent stem cells (hPSCs), adult/fetal renal tissues, and kidney cancer biopsy. Alongside gene editing tools, hPSC-derived kidney organoids are being harnessed to model genetic kidney diseases. In comparison, adult kidney-derived tubuloids and kidney cancer-derived tumoroids are still in their infancy. Herein, we first summarize the currently available kidney organoid models. Next, we discuss recent advances in kidney disease modelling using organoid models. Finally, we consider the major challenges that have hindered the application of kidney organoids in disease modelling and drug evaluation and propose prospective solutions.
Collapse
Affiliation(s)
- Meng Liu
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
| | - Angelysia Cardilla
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
| | - Joanne Ngeow
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
- Cancer Genetics Service, National Cancer Centre Singapore, Singapore, Singapore
| | - Ximing Gong
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
- *Correspondence: Ximing Gong, ; Yun Xia,
| | - Yun Xia
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
- *Correspondence: Ximing Gong, ; Yun Xia,
| |
Collapse
|