151
|
Tran T, Lindström NO, Ransick A, De Sena Brandine G, Guo Q, Kim AD, Der B, Peti-Peterdi J, Smith AD, Thornton M, Grubbs B, McMahon JA, McMahon AP. In Vivo Developmental Trajectories of Human Podocyte Inform In Vitro Differentiation of Pluripotent Stem Cell-Derived Podocytes. Dev Cell 2020; 50:102-116.e6. [PMID: 31265809 DOI: 10.1016/j.devcel.2019.06.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 03/27/2019] [Accepted: 05/31/2019] [Indexed: 12/21/2022]
Abstract
The renal corpuscle of the kidney comprises a glomerular vasculature embraced by podocytes and supported by mesangial myofibroblasts, which ensure plasma filtration at the podocyte-generated slit diaphragm. With a spectrum of podocyte-expressed gene mutations causing chronic disease, an enhanced understanding of podocyte development and function to create relevant in vitro podocyte models is a clinical imperative. To characterize podocyte development, scRNA-seq was performed on human fetal kidneys, identifying distinct transcriptional signatures accompanying the differentiation of functional podocytes from progenitors. Interestingly, organoid-generated podocytes exhibited highly similar, progressive transcriptional profiles despite an absence of the vasculature, although abnormal gene expression was pinpointed in late podocytes. On transplantation into mice, organoid-derived podocytes recruited the host vasculature and partially corrected transcriptional profiles. Thus, human podocyte development is mostly intrinsically regulated and vascular interactions refine maturation. These studies support the application of organoid-derived podocytes to model disease and to restore or replace normal kidney functions.
Collapse
Affiliation(s)
- Tracy Tran
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Nils O Lindström
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Andrew Ransick
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Guilherme De Sena Brandine
- Molecular and Computational Biology, Division of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Qiuyu Guo
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Albert D Kim
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Balint Der
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Janos Peti-Peterdi
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Andrew D Smith
- Molecular and Computational Biology, Division of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Matthew Thornton
- Maternal Fetal Medicine Division, University of Southern California, Los Angeles, CA 90089, USA
| | - Brendan Grubbs
- Maternal Fetal Medicine Division, University of Southern California, Los Angeles, CA 90089, USA
| | - Jill A McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
152
|
Genova E, Cavion F, Lucafò M, Leo LD, Pelin M, Stocco G, Decorti G. Induced pluripotent stem cells for therapy personalization in pediatric patients: Focus on drug-induced adverse events. World J Stem Cells 2019; 11:1020-1044. [PMID: 31875867 PMCID: PMC6904863 DOI: 10.4252/wjsc.v11.i12.1020] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 09/05/2019] [Accepted: 10/14/2019] [Indexed: 02/06/2023] Open
Abstract
Adverse drug reactions (ADRs) are major clinical problems, particularly in special populations such as pediatric patients. Indeed, ADRs may be caused by a plethora of different drugs leading, in some cases, to hospitalization, disability or even death. In addition, pediatric patients may respond differently to drugs with respect to adults and may be prone to developing different kinds of ADRs, leading, in some cases, to more severe consequences. To improve the comprehension, and thus the prevention, of ADRs, the set-up of sensitive and personalized assays is urgently needed. Important progress is represented by the possibility of setting up groundbreaking patient-specific assays. This goal has been powerfully achieved using induced pluripotent stem cells (iPSCs). Due to their genetic and physiological species-specific differences and their ability to be differentiated ideally into all tissues of the human body, this model may be accurate in predicting drug toxicity, especially when this toxicity is related to individual genetic differences. This review is an up-to-date summary of the employment of iPSCs as a model to study ADRs, with particular attention to drugs used in the pediatric field. We especially focused on the intestinal, hepatic, pancreatic, renal, cardiac, and neuronal levels, also discussing progress in organoids creation. The latter are three-dimensional in vitro culture systems derived from pluripotent or adult stem cells simulating the architecture and functionality of native organs such as the intestine, liver, pancreas, kidney, heart, and brain. Based on the existing knowledge, these models are powerful and promising tools in multiple clinical applications including toxicity screening, disease modeling, personalized and regenerative medicine.
Collapse
Affiliation(s)
- Elena Genova
- PhD School in Reproduction and Development Sciences, University of Trieste, Trieste 34127, Italy
| | - Federica Cavion
- Department of Life Sciences, University of Trieste, Trieste 34127, Italy
| | - Marianna Lucafò
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste 34137, Italy
| | - Luigina De Leo
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste 34137, Italy
| | - Marco Pelin
- Department of Life Sciences, University of Trieste, Trieste 34127, Italy
| | - Gabriele Stocco
- Department of Life Sciences, University of Trieste, Trieste 34127, Italy.
| | - Giuliana Decorti
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste 34137, Italy
| |
Collapse
|
153
|
Misra PS, Silva E Silva V, Collister D. Roadblocks and Opportunities to the Implementation of Novel Therapies for Acute Kidney Injury: A Narrative Review. Can J Kidney Health Dis 2019; 6:2054358119880519. [PMID: 31636913 PMCID: PMC6787878 DOI: 10.1177/2054358119880519] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 08/12/2019] [Indexed: 11/29/2022] Open
Abstract
Background: Acute kidney injury (AKI) is a complex and heterogeneous clinical syndrome
with limited effective treatment options. Therefore, a coherent research
structure considering AKI pathophysiology, treatment, translation, and
implementation is critical to advancing patient care in this area. Purpose of review: In this narrative review, we discuss novel therapies for AKI from their
journey from bench to bedside to population and focus on roadblocks and
opportunities to their successful implementation. Sources of information: Peer-reviewed articles, opinion pieces from research leaders and research
funding agencies, and clinical and research expertise. Methods: This narrative review details the challenges of translation of preclinical
studies in AKI and highlights trending research areas and innovative designs
in the field. Key developments in preclinical research, clinical trials, and
knowledge translation are discussed. Furthermore, this article discusses the
current need to involve patients in clinical research and the barriers and
opportunities for effective knowledge translation. Key findings: Preclinical studies have largely been unsuccessful in generating novel
therapies for AKI, due both to the complexity and heterogeneity of the
disease, as well as the limitations of commonly available preclinical models
of AKI. The emergence of kidney organoid technology may be an opportunity to
reverse this trend. However, the roadblocks encountered at the bench have
not precluded researchers from running well-designed and impactful clinical
trials, and the field of renal replacement therapy in AKI is highlighted as
an area that has been particularly active. Meanwhile, knowledge translation
initiatives are bolstered by the presence of large administrative databases
to permit ongoing monitoring of clinical practices and outcomes, with
research output from such evaluations having the potential to directly
impact patient care and inform the generation of meaningful clinical
practice guidelines. Limitations: There are limited objective data examining the process of knowledge creation
and translation in AKI, and as such the opinions and research areas of the
authors are significantly drawn upon in the discussion. Implications: The use of an organized knowledge-to-action framework involving multiple
stakeholders, especially patient partners, is critical to translating basic
research findings to improvements in patient care in AKI, an area where
effective treatment options are lacking.
Collapse
Affiliation(s)
- Paraish S Misra
- Kidney Research Scientist Core Education and National Training Program, Canada.,McEwen Stem Cell Institute, Department of Medicine, University of Toronto, ON, Canada
| | - Vanessa Silva E Silva
- Kidney Research Scientist Core Education and National Training Program, Canada.,The Canadian Donation and Transplantation Research Program, Canada.,School of Nursing, Queen's University, Kingston, ON, Canada.,School of Nursing, Federal University of Sao Paulo, Brazil
| | - David Collister
- Kidney Research Scientist Core Education and National Training Program, Canada.,Department of Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
154
|
Shen JX, Youhanna S, Zandi Shafagh R, Kele J, Lauschke VM. Organotypic and Microphysiological Models of Liver, Gut, and Kidney for Studies of Drug Metabolism, Pharmacokinetics, and Toxicity. Chem Res Toxicol 2019; 33:38-60. [DOI: 10.1021/acs.chemrestox.9b00245] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Joanne X. Shen
- Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Sonia Youhanna
- Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Reza Zandi Shafagh
- Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
- Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Julianna Kele
- Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Volker M. Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| |
Collapse
|
155
|
Abstract
There are now many reports of human kidney organoids generated via the directed differentiation of human pluripotent stem cells (PSCs) based on an existing understanding of mammalian kidney organogenesis. Such kidney organoids potentially represent tractable tools for the study of normal human development and disease with improvements in scale, structure, and functional maturation potentially providing future options for renal regeneration. The utility of such organotypic models, however, will ultimately be determined by their developmental accuracy. While initially inferred from mouse models, recent transcriptional analyses of human fetal kidney have provided greater insight into nephrogenesis. In this review, we discuss how well human kidney organoids model the human fetal kidney and how the remaining differences challenge their utility.
Collapse
Affiliation(s)
- Melissa H Little
- Murdoch Children's Research Institute, Parkville, Victoria 3052, Australia
- Department of Anatomy and Neuroscience, The University of Melbourne, Victoria 3052, Australia
- Department of Paediatrics, The University of Melbourne, Victoria 3052, Australia
| | - Alexander N Combes
- Murdoch Children's Research Institute, Parkville, Victoria 3052, Australia
- Department of Anatomy and Neuroscience, The University of Melbourne, Victoria 3052, Australia
| |
Collapse
|
156
|
van der Wijst J, Belge H, Bindels RJM, Devuyst O. Learning Physiology From Inherited Kidney Disorders. Physiol Rev 2019; 99:1575-1653. [PMID: 31215303 DOI: 10.1152/physrev.00008.2018] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The identification of genes causing inherited kidney diseases yielded crucial insights in the molecular basis of disease and improved our understanding of physiological processes that operate in the kidney. Monogenic kidney disorders are caused by mutations in genes coding for a large variety of proteins including receptors, channels and transporters, enzymes, transcription factors, and structural components, operating in specialized cell types that perform highly regulated homeostatic functions. Common variants in some of these genes are also associated with complex traits, as evidenced by genome-wide association studies in the general population. In this review, we discuss how the molecular genetics of inherited disorders affecting different tubular segments of the nephron improved our understanding of various transport processes and of their involvement in homeostasis, while providing novel therapeutic targets. These include inherited disorders causing a dysfunction of the proximal tubule (renal Fanconi syndrome), with emphasis on epithelial differentiation and receptor-mediated endocytosis, or affecting the reabsorption of glucose, the handling of uric acid, and the reabsorption of sodium, calcium, and magnesium along the kidney tubule.
Collapse
Affiliation(s)
- Jenny van der Wijst
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; Institute of Physiology, University of Zurich , Zurich , Switzerland ; and Division of Nephrology, Institute of Experimental and Clinical Research (IREC), Medical School, Université catholique de Louvain, Brussels, Belgium
| | - Hendrica Belge
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; Institute of Physiology, University of Zurich , Zurich , Switzerland ; and Division of Nephrology, Institute of Experimental and Clinical Research (IREC), Medical School, Université catholique de Louvain, Brussels, Belgium
| | - René J M Bindels
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; Institute of Physiology, University of Zurich , Zurich , Switzerland ; and Division of Nephrology, Institute of Experimental and Clinical Research (IREC), Medical School, Université catholique de Louvain, Brussels, Belgium
| | - Olivier Devuyst
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; Institute of Physiology, University of Zurich , Zurich , Switzerland ; and Division of Nephrology, Institute of Experimental and Clinical Research (IREC), Medical School, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
157
|
Abdallah M, Martin M, El Tahchi MR, Balme S, Faour WH, Varga B, Cloitre T, Páll O, Cuisinier FJG, Gergely C, Bassil MJ, Bechelany M. Influence of Hydrolyzed Polyacrylamide Hydrogel Stiffness on Podocyte Morphology, Phenotype, and Mechanical Properties. ACS APPLIED MATERIALS & INTERFACES 2019; 11:32623-32632. [PMID: 31424195 DOI: 10.1021/acsami.9b09337] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Chronic kidney disease is characterized by a gradual decline in renal function that progresses toward end-stage renal disease. Podocytes are highly specialized glomerular epithelial cells which form with the glomerular basement membrane (GBM) and capillary endothelium the glomerular filtration barrier. GBM is an extracellular matrix (ECM) that acts as a mechanical support and provides biophysical signals that control normal podocytes behavior in the process of glomerular filtration. Thus, the ECM stiffness represents an essential characteristic that controls podocyte function. Hydrolyzed Polyacrylamide (PAAm) hydrogels are smart polyelectrolyte materials. Their biophysical properties can be tuned as desired to mimic the natural ECM. Therefore, these hydrogels are investigated as new ECM-like constructs to engineer a podocyte-like basement membrane that forms with cultured human podocytes a functional glomerular-like filtration barrier. Such ECM-like PAAm hydrogel construct will provide unique opportunity to reveal podocyte cell biological responses in an in vivo-like setting by controlling the physical properties of the PAAm membranes. In this work, Hydrolyzed PAAm scaffolds having different stiffness ranging between 0.6-44 kPa are prepared. The correlation between the hydrogel structural and mechanical properties and Podocyte morphology, elasticity, cytoskeleton reorganization, and podocin expression is evaluated. Results show that hydrolyzed PAAm hydrogels promote good cell adhesion and growth and are suitable materials for the development of future 3D smart scaffolds. In addition, the hydrogel properties can be easily modulated over a wide physiological range by controlling the cross-linker concentration. Finally, tuning the hydrogel properties is an effective strategy to control the cells function. This work addressed the complexity of podocytes behavior which will further enhance our knowledge to develop a kidney-on-chip model much needed in kidney function studies in both healthy and diseased states.
Collapse
Affiliation(s)
- Maya Abdallah
- Institut Européen des Membranes, ENSCM, CNRS , Université de Montpellier , Montpellier 34090 , France
- Biomaterials and Intelligent Materials Research Laboratory (LBMI) , Lebanese University , Faculty of Sciences 2, Physic Department , Jdeidet 90656 , Lebanon
| | - Marta Martin
- Laboratoire Charles Coulomb , Université de Montpellier , CNRS , Montpellier 34095 , France
| | - Mario R El Tahchi
- Biomaterials and Intelligent Materials Research Laboratory (LBMI) , Lebanese University , Faculty of Sciences 2, Physic Department , Jdeidet 90656 , Lebanon
| | - Sebastien Balme
- Institut Européen des Membranes, ENSCM, CNRS , Université de Montpellier , Montpellier 34090 , France
| | - Wissam H Faour
- Gilbert and Rose-Marie Chagoury School of Medicine , Lebanese American University , P.O. Box 36 , Byblos , Lebanon
| | - Béla Varga
- Laboratoire Charles Coulomb , Université de Montpellier , CNRS , Montpellier 34095 , France
| | - Thierry Cloitre
- Laboratoire Charles Coulomb , Université de Montpellier , CNRS , Montpellier 34095 , France
| | - Orsolya Páll
- Laboratoire de Bioingénierie et Nanosciences , Université de Montpellier , Montpellier 34090 , France
| | - Frédéric J G Cuisinier
- Laboratoire de Bioingénierie et Nanosciences , Université de Montpellier , Montpellier 34090 , France
| | - Csilla Gergely
- Laboratoire Charles Coulomb , Université de Montpellier , CNRS , Montpellier 34095 , France
| | - Maria J Bassil
- Biomaterials and Intelligent Materials Research Laboratory (LBMI) , Lebanese University , Faculty of Sciences 2, Physic Department , Jdeidet 90656 , Lebanon
| | - Mikhael Bechelany
- Institut Européen des Membranes, ENSCM, CNRS , Université de Montpellier , Montpellier 34090 , France
| |
Collapse
|
158
|
Vanslambrouck JM, Wilson SB, Tan KS, Soo JYC, Scurr M, Spijker HS, Starks LT, Neilson A, Cui X, Jain S, Little MH, Howden SE. A Toolbox to Characterize Human Induced Pluripotent Stem Cell-Derived Kidney Cell Types and Organoids. J Am Soc Nephrol 2019; 30:1811-1823. [PMID: 31492807 DOI: 10.1681/asn.2019030303] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 07/25/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The generation of reporter lines for cell identity, lineage, and physiologic state has provided a powerful tool in advancing the dissection of mouse kidney morphogenesis at a molecular level. Although use of this approach is not an option for studying human development in vivo, its application in human induced pluripotent stem cells (iPSCs) is now feasible. METHODS We used CRISPR/Cas9 gene editing to generate ten fluorescence reporter iPSC lines designed to identify nephron progenitors, podocytes, proximal and distal nephron, and ureteric epithelium. Directed differentiation to kidney organoids was performed according to published protocols. Using immunofluorescence and live confocal microscopy, flow cytometry, and cell sorting techniques, we investigated organoid patterning and reporter expression characteristics. RESULTS Each iPSC reporter line formed well patterned kidney organoids. All reporter lines showed congruence of endogenous gene and protein expression, enabling isolation and characterization of kidney cell types of interest. We also demonstrated successful application of reporter lines for time-lapse imaging and mouse transplantation experiments. CONCLUSIONS We generated, validated, and applied a suite of fluorescence iPSC reporter lines for the study of morphogenesis within human kidney organoids. This fluorescent iPSC reporter toolbox enables the visualization and isolation of key populations in forming kidney organoids, facilitating a range of applications, including cellular isolation, time-lapse imaging, protocol optimization, and lineage-tracing approaches. These tools offer promise for enhancing our understanding of this model system and its correspondence with human kidney morphogenesis.
Collapse
Affiliation(s)
| | - Sean B Wilson
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Ker Sin Tan
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Joanne Y-C Soo
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Michelle Scurr
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - H Siebe Spijker
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | - Lakshi T Starks
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Amber Neilson
- Department of Genetics, Genome Engineering and iPSC Center and
| | - Xiaoxia Cui
- Department of Genetics, Genome Engineering and iPSC Center and
| | - Sanjay Jain
- Department of Medicine, Kidney Translational Research Center, Washington University School of Medicine, St. Louis, Missouri; and
| | - Melissa Helen Little
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia; .,Faculty of Medicine, Dentistry and Health Sciences, Department of Paediatrics and.,Department of Anatomy and Neuroscience, University of Melbourne, Victoria, Australia
| | - Sara E Howden
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Faculty of Medicine, Dentistry and Health Sciences, Department of Paediatrics and
| |
Collapse
|
159
|
Lindoso RS, Kasai-Brunswick TH, Monnerat Cahli G, Collino F, Bastos Carvalho A, Campos de Carvalho AC, Vieyra A. Proteomics in the World of Induced Pluripotent Stem Cells. Cells 2019; 8:703. [PMID: 31336746 PMCID: PMC6678893 DOI: 10.3390/cells8070703] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 06/24/2019] [Accepted: 06/25/2019] [Indexed: 02/05/2023] Open
Abstract
Omics approaches have significantly impacted knowledge about molecular signaling pathways driving cell function. Induced pluripotent stem cells (iPSC) have revolutionized the field of biological sciences and proteomics and, in particular, has been instrumental in identifying key elements operating during the maintenance of the pluripotent state and the differentiation process to the diverse cell types that form organisms. This review covers the evolution of conceptual and methodological strategies in proteomics; briefly describes the generation of iPSC from a historical perspective, the state-of-the-art of iPSC-based proteomics; and compares data on the proteome and transcriptome of iPSC to that of embryonic stem cells (ESC). Finally, proteomics of healthy and diseased cells and organoids differentiated from iPSC are analyzed.
Collapse
Affiliation(s)
- Rafael Soares Lindoso
- Carlos Chagas Filho Institute of Biophysics and National Center for Structural Biology and Bioimaging/CENABIO, Federal University of Rio de Janeiro, Rio de Janeiro 21941-102, Brazil
| | - Tais H Kasai-Brunswick
- Carlos Chagas Filho Institute of Biophysics and National Center for Structural Biology and Bioimaging/CENABIO, Federal University of Rio de Janeiro, Rio de Janeiro 21941-102, Brazil
| | - Gustavo Monnerat Cahli
- Carlos Chagas Filho Institute of Biophysics and National Center for Structural Biology and Bioimaging/CENABIO, Federal University of Rio de Janeiro, Rio de Janeiro 21941-102, Brazil
- Laboratory of Proteomics, LADETEC, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro 21941-598, Brazil
| | - Federica Collino
- Carlos Chagas Filho Institute of Biophysics and National Center for Structural Biology and Bioimaging/CENABIO, Federal University of Rio de Janeiro, Rio de Janeiro 21941-102, Brazil
- Department of Biomedical Sciences, University of Padova, 35131 Padua, Italy
| | - Adriana Bastos Carvalho
- Carlos Chagas Filho Institute of Biophysics and National Center for Structural Biology and Bioimaging/CENABIO, Federal University of Rio de Janeiro, Rio de Janeiro 21941-102, Brazil
| | - Antonio Carlos Campos de Carvalho
- Carlos Chagas Filho Institute of Biophysics and National Center for Structural Biology and Bioimaging/CENABIO, Federal University of Rio de Janeiro, Rio de Janeiro 21941-102, Brazil.
| | - Adalberto Vieyra
- Carlos Chagas Filho Institute of Biophysics and National Center for Structural Biology and Bioimaging/CENABIO, Federal University of Rio de Janeiro, Rio de Janeiro 21941-102, Brazil.
- Graduate Program in Translational Biomedicine, Grande Rio University, Duque de Caxias 25071-202, Brazil.
| |
Collapse
|
160
|
Yoshimura Y, Nishinakamura R. Podocyte development, disease, and stem cell research. Kidney Int 2019; 96:1077-1082. [PMID: 31420196 DOI: 10.1016/j.kint.2019.04.044] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/04/2019] [Accepted: 04/23/2019] [Indexed: 12/18/2022]
Abstract
The glomerular podocyte is one of the major targets of kidney research. Recent establishment of kidney organoids from pluripotent stem cells has enabled the detailed analysis of human podocytes in both development and disease. The podocytes in organoids express slit diaphragm-related genes and proteins and exhibit characteristic morphology, especially upon experimental transplantation. Organoid technology is now used to reproduce hereditary podocyte diseases, and selective podocyte induction methods have also been reported. Moreover, single-cell RNA-sequencing of human fetal and adult kidneys has revealed the detailed molecular features of this cell lineage, as well as serving as references for kidney organoids in which podocytes are still immature. Here, we discuss the recent progress and limitations of podocyte research from the viewpoint of developmental biology and kidney organoids.
Collapse
Affiliation(s)
- Yasuhiro Yoshimura
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan; Department of Nephrology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Ryuichi Nishinakamura
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
161
|
Woolf AS. Growing a new human kidney. Kidney Int 2019; 96:871-882. [PMID: 31399199 PMCID: PMC6856720 DOI: 10.1016/j.kint.2019.04.040] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 03/01/2019] [Accepted: 04/01/2019] [Indexed: 12/17/2022]
Abstract
There are 3 reasons to generate a new human kidney. The first is to learn more about the biology of the developing and mature organ. The second is to generate tissues with which to model congenital and acquired kidney diseases. In particular, growing human kidneys in this manner ultimately should help us understand the mechanisms of common chronic kidney diseases such as diabetic nephropathy and others featuring fibrosis, as well as nephrotoxicity. The third reason is to provide functional kidney tissues that can be used directly in regenerative medicine therapies. The second and third reasons to grow new human kidneys are especially compelling given the millions of persons worldwide whose lives depend on a functioning kidney transplant or long-term dialysis, as well as those with end-stage renal disease who die prematurely because they are unable to access these treatments. As shown in this review, the aim to create healthy human kidney tissues has been partially realized. Moreover, the technology shows promise in terms of modeling genetic disease. In contrast, barely the first steps have been taken toward modeling nongenetic chronic kidney diseases or using newly grown human kidney tissue for regenerative medicine therapies.
Collapse
Affiliation(s)
- Adrian S Woolf
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, United Kingdom; Royal Manchester Children's Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom.
| |
Collapse
|
162
|
Miyoshi T, Hiratsuka K, Saiz EG, Morizane R. Kidney organoids in translational medicine: Disease modeling and regenerative medicine. Dev Dyn 2019; 249:34-45. [PMID: 30843293 DOI: 10.1002/dvdy.22] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 03/04/2019] [Accepted: 03/04/2019] [Indexed: 12/15/2022] Open
Abstract
The kidney is one of the most complex organs composed of multiple cell types, functioning to maintain homeostasis by means of the filtering of metabolic wastes, balancing of blood electrolytes, and adjustment of blood pressure. Recent advances in 3D culture technologies in vitro enabled the generation of "organoids" which mimic the structure and function of in vivo organs. Organoid technology has allowed for new insights into human organ development and human pathophysiology, with great potential for translational research. Increasing evidence shows that kidney organoids are a useful platform for disease modeling of genetic kidney diseases when derived from genetic patient iPSCs and/or CRISPR-mutated stem cells. Although single cell RNA-seq studies highlight the technical difficulties underlying kidney organoid generation reproducibility and variation in differentiation protocols, kidney organoids still hold great potential to understand kidney pathophysiology as applied to kidney injury and fibrosis. In this review, we summarize various studies of kidney organoids, disease modeling, genome-editing, and bioengineering, and additionally discuss the potential of and current challenges to kidney organoid research.
Collapse
Affiliation(s)
- Tomoya Miyoshi
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Ken Hiratsuka
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Edgar Garcia Saiz
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Ryuji Morizane
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts.,Harvard Stem Cell Institute, Cambridge, Massachusetts.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, Massachusetts
| |
Collapse
|
163
|
Abstract
Regeneration of a functional kidney from pluripotent stem cells (PSCs) is challenging because of its complex structure. Kidneys are derived from embryonic metanephros, which are composed of three progenitor cells: nephron progenitors, ureteric bud, and stromal progenitors. Nephron progenitors and ureteric bud have been induced successfully from PSCs as a result of the understanding of their detailed developmental process through cell-lineage tracing analysis. Moreover, these induced progenitors can be used to reconstruct the three-dimensional (3D) structure of kidneys in vitro, including glomeruli with podocytes, renal tubules, and the branching ureters. Induction of the remaining renal progenitors (that is, stromal progenitors from PSCs and the further maturation of reconstructed kidneys) needs to be studied extensively to regenerate functional and sophisticated kidneys from PSCs. In addition to the proper induction of renal progenitors, new bioengineering methods such as decellularization and 3D bioprinting and the recent advancements in the regeneration of kidneys in other species are promising leads for regenerating the complex spatial arrangement of kidneys, including the vascular network and urinary excretion pathway in humans.
Collapse
Affiliation(s)
- Sho Hasegawa
- Division of Nephrology and Endocrinology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Tetsuhiro Tanaka
- Division of Nephrology and Endocrinology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Masaomi Nangaku
- Division of Nephrology and Endocrinology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|