1
|
Petreski T, Gradišnik L, Varda L, Kovačič P, Dolenšek J, Stožer A, Bevc S, Maver U. A Novel Protocol for Culturing Polarized Proximal Tubular Epithelial Cells from Kidney Biopsies: Enhancing Platforms for Drug Excretion and Nephrotoxicity Studies. J Xenobiot 2025; 15:52. [PMID: 40278157 PMCID: PMC12028765 DOI: 10.3390/jox15020052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 03/27/2025] [Accepted: 03/31/2025] [Indexed: 04/26/2025] Open
Abstract
The kidneys are integral to homeostasis but are susceptible to nephrotoxic compounds. Proximal tubular epithelial cells (PTECs) mediate drug metabolism and transport and are widely used in preclinical studies. However, commercial PTECs are limited in availability and physiological relevance. This study aimed to develop a novel, reliable protocol for isolating and culturing PTECs from human kidney biopsies. Primary PTECs were isolated from kidney biopsies of two patients (MFUM-RPTEC-1 and MFUM-RPTEC-2). Their morphology, population doubling time, transepithelial electrical resistance (TEER), and phenotypic markers were evaluated. Polarization and transporter expression were analyzed using cells cultured on Transwell inserts. Colonies formed within 24-48 h, with confluence reached by 8-10 days and dome (hemicyst) formation by day 13. TEER values peaked at 190 Ω/cm2 after 7-14 days, confirming tight junction formation. Immunostaining identified characteristic markers (e.g., SGLT2, OAT1/3, OCT2, P-gp, MRP4, MATE1, N-cadherin, ZO-1, CK-18). Cells cultured on Transwell plates exhibited native polarization, expressing transporters crucial for drug excretion on apical and basolateral surfaces. We present two robust protocols for isolating and characterizing PTECs, offering a scalable method to obtain functional, polarized cells from scarce biopsy material. The isolated PTECs, therefore, present a valuable platform for preclinical studies, especially for drug excretion testing through the expressed transporters. Drug competition for these transporters during tubular secretion is also a common cause of nephrotoxicity.
Collapse
Affiliation(s)
- Tadej Petreski
- Department of Nephrology, University Medical Centre Maribor, Ljubljanska ulica 5, 2000 Maribor, Slovenia;
| | - Lidija Gradišnik
- Institute of Biomedical Sciences, Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia;
| | - Luka Varda
- Department of Dialysis, University Medical Centre Maribor, Ljubljanska ulica 5, 2000 Maribor, Slovenia;
| | - Polona Kovačič
- Department of Physiology, Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia; (P.K.); (J.D.); (A.S.)
| | - Jurij Dolenšek
- Department of Physiology, Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia; (P.K.); (J.D.); (A.S.)
- Department of Biology, Faculty of Natural Sciences and Mathematics, University of Maribor, Koroška cesta 160, 2000 Maribor, Slovenia
| | - Andraž Stožer
- Department of Physiology, Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia; (P.K.); (J.D.); (A.S.)
| | - Sebastjan Bevc
- Department of Nephrology, University Medical Centre Maribor, Ljubljanska ulica 5, 2000 Maribor, Slovenia;
- Department of Pharmacology, Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia
- Department of Internal Medicine, Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia
| | - Uroš Maver
- Institute of Biomedical Sciences, Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia;
- Department of Pharmacology, Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia
| |
Collapse
|
2
|
Mao R, Zhang J, Qin H, Liu Y, Xing Y, Zeng W. Application progress of bio-manufacturing technology in kidney organoids. Biofabrication 2025; 17:022007. [PMID: 39933190 DOI: 10.1088/1758-5090/adb4a1] [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: 08/04/2024] [Accepted: 02/11/2025] [Indexed: 02/13/2025]
Abstract
Kidney transplantation remains a pivotal treatment modality for kidney disease, yet its progress is significantly hindered by the scarcity of donor kidneys and ethical dilemmas surrounding their procurement. As organoid technology evolves and matures, the creation of bionic human kidney organoids offers profound potential for advancing kidney disease research, drug nephrotoxicity screening, and regenerative medicine. Nevertheless, current kidney organoid models grapple with limitations such as constrained cellular differentiation, underdeveloped functional structures, and a crucial absence of vascularization. This deficiency in vascularization, in particular, stunts organoid development, restricts their size, diminishes filtration capabilities, and may trigger immune inflammatory reactions through the resulting ischemic microenvironment. Hence, the achievement of vascularization within kidney organoids and the successful establishment of functional microvascular networks constitutes a paramount goal for their future progression. In this review, we provide an overview of recent advancements in biotechnology domains, encompassing organ-on-a-chip technology, biomimetic matrices, and bioprinting, with the aim of catalyzing technological breakthroughs that can enhance the vascularization of kidney organoids and broaden their applicability. These technologies hold the key to unlocking the full potential of kidney organoids as a transformative therapeutic option for kidney disease.
Collapse
Affiliation(s)
- Runqi Mao
- Department of Cell Biology, Third Military Medical University, Chongqing, People's Republic of China
| | - Junming Zhang
- Department of Cell Biology, Third Military Medical University, Chongqing, People's Republic of China
| | - Haoxiang Qin
- Department of Cell Biology, Third Military Medical University, Chongqing, People's Republic of China
| | - Yuanyuan Liu
- Department of Cell Biology, Third Military Medical University, Chongqing, People's Republic of China
| | - Yuxin Xing
- Department of Cell Biology, Third Military Medical University, Chongqing, People's Republic of China
| | - Wen Zeng
- Department of Cell Biology, Third Military Medical University, Chongqing, People's Republic of China
- State Key Laboratory of Trauma, Burn and Combined Injury, Chongqing, People's Republic of China
- Jinfeng Laboratory, Chongqing 401329, People's Republic of China
| |
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
|
Ceccotti E, Semnani A, Bussolati B, Bruno S. Human kidney organoids for modeling the development of different diseases. Curr Top Dev Biol 2025; 163:364-393. [PMID: 40254349 DOI: 10.1016/bs.ctdb.2024.12.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
The increasing incidence of kidney diseases has highlighted the need for in vitro experimental models to mimic disease development and to test new therapeutic approaches. Traditional two-dimensional in vitro experimental models are not fully able to recapitulate renal diseases. Instead, kidney organoids represent three-dimensional models that better mimic the human organ from both structural and functional points of view. Human pluripotent stem cells (PSCs), both embryonic and induced, are ideal sources for generating renal organoids. These organoids contain all renal cell types and the protocols to differentiate PSCs into renal organoids consist of three different stages that recapitulate embryonic development: mesodermal induction, nephron progenitor formation, and nephron differentiation. Recently it has been establish a renal organoid model where collecting ducts are also present. In this case, the presence of ureteric bud progenitor cells is essential. Renal organoids are particularly useful for studying genetic diseases, by introducing the specific mutations in PSCs by genome editing or generating organoids from patient-derived PSCs. Moreover, renal organoids represent promising models in toxicology studies and testing new therapeutic approaches. Renal organoids can be established also from adult stem cells. This type of organoid, named tubuloid, is composed only of epithelial cells and recapitulates the tissue repair process. The tubuloids can be generated from adult stem or progenitor cells, obtained from renal biopsies or urine, and are promising in vitro models for studying tubular functions, diseases, and regeneration. Tubuloids can be derived from patients and permit the study of genetic diseases, performing personalized drug screening and modeling renal pathologies.
Collapse
Affiliation(s)
- Elena Ceccotti
- Department of Medical Sciences, University of Torino, Corso Dogliotti, Torino, Italy
| | - Armina Semnani
- Department of Medical Sciences, University of Torino, Corso Dogliotti, Torino, Italy
| | - Benedetta Bussolati
- Department of Medical Sciences, University of Torino, Corso Dogliotti, Torino, Italy; Molecular Biotechnology Center "Guido Tarone", Via Nizza, Torino, Italy
| | - Stefania Bruno
- Department of Medical Sciences, University of Torino, Corso Dogliotti, Torino, Italy.
| |
Collapse
|
5
|
Shao Y, Wang J, Jin A, Jiang S, Lei L, Liu L. Biomaterial-assisted organoid technology for disease modeling and drug screening. Mater Today Bio 2025; 30:101438. [PMID: 39866785 PMCID: PMC11757232 DOI: 10.1016/j.mtbio.2024.101438] [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: 10/17/2024] [Revised: 12/10/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025] Open
Abstract
Developing disease models and screening for effective drugs are key areas of modern medical research. Traditional methodologies frequently fall short in precisely replicating the intricate architecture of bodily tissues and organs. Nevertheless, recent advancements in biomaterial-assisted organoid technology have ushered in a paradigm shift in biomedical research. This innovative approach enables the cultivation of three-dimensional cellular structures in vitro that closely emulate the structural and functional attributes of organs, offering physiologically superior models compared to conventional techniques. The evolution of biomaterials plays a pivotal role in supporting the culture and development of organ tissues, thereby facilitating more accurate disease state modeling and the rigorous evaluation of drug efficacy and safety profiles. In this review, we will explore the roles that various biomaterials play in organoid development, examine the fundamental principles and advantages of utilizing these technologies in constructing disease models, and highlight recent advances and practical applications in drug screening using disease-specific organoid models. Additionally, the challenges and future directions of organoid technology are discussed. Through continued research and innovation, we aim to make remarkable strides in disease treatment and drug development, ultimately enhancing patient quality of life.
Collapse
Affiliation(s)
- Yunyuan Shao
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, 310015, China
| | - Juncheng Wang
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| | - Anqi Jin
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, 310015, China
| | - Shicui Jiang
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| | - Lanjie Lei
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, 310015, China
| | - Liangle Liu
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| |
Collapse
|
6
|
Oliveira M, Sarker PP, Skovorodkin I, Kalantarifard A, Haskavuk T, Mac Intyre J, Nallukunnel Raju E, Nooranian S, Shioda H, Nishikawa M, Sakai Y, Vainio SJ, Elbuken C, Raykhel I. From ex ovo to in vitro: xenotransplantation and vascularization of mouse embryonic kidneys in a microfluidic chip. LAB ON A CHIP 2024; 24:4816-4826. [PMID: 39290081 PMCID: PMC11408908 DOI: 10.1039/d4lc00547c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 09/01/2024] [Indexed: 09/19/2024]
Abstract
Organoids are emerging as a powerful tool to investigate complex biological structures in vitro. Vascularization of organoids is crucial to recapitulate the morphology and function of the represented human organ, especially in the case of the kidney, whose primary function of blood filtration is closely associated with blood circulation. Current in vitro microfluidic approaches have only provided initial vascularization of kidney organoids, whereas in vivo transplantation to animal models is problematic due to ethical problems, with the exception of xenotransplantation onto a chicken chorioallantoic membrane (CAM). Although CAM can serve as a good environment for vascularization, it can only be used for a fixed length of time, limited by development of the embryo. Here, we propose a novel lab on a chip design that allows organoids of different origin to be cultured and vascularized on a CAM, as well as to be transferred to in vitro conditions when required. Mouse embryonic kidneys cultured on the CAM showed enhanced vascularization by intrinsic endothelial cells, and made connections with the chicken vasculature, as evidenced by blood flowing through them. After the chips were transferred to in vitro conditions, the vasculature inside the organoids was successfully maintained. To our knowledge, this is the first demonstration of the combination of in vivo and in vitro approaches applied to microfluidic chip design.
Collapse
Affiliation(s)
- Micaela Oliveira
- Microfluidics and Biosensor Research Group, Disease Networks Research Unit, Department of Biochemistry and Molecular Medicine, University of Oulu, Finland.
| | - Partha Protim Sarker
- Microfluidics and Biosensor Research Group, Disease Networks Research Unit, Department of Biochemistry and Molecular Medicine, University of Oulu, Finland.
- Developmental Biology Laboratory, Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.
| | - Ilya Skovorodkin
- Microfluidics and Biosensor Research Group, Disease Networks Research Unit, Department of Biochemistry and Molecular Medicine, University of Oulu, Finland.
- Developmental Biology Laboratory, Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.
| | - Ali Kalantarifard
- Microfluidics and Biosensor Research Group, Disease Networks Research Unit, Department of Biochemistry and Molecular Medicine, University of Oulu, Finland.
| | - Tugce Haskavuk
- Microfluidics and Biosensor Research Group, Disease Networks Research Unit, Department of Biochemistry and Molecular Medicine, University of Oulu, Finland.
- Developmental Biology Laboratory, Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.
| | - Jonatan Mac Intyre
- Microfluidics and Biosensor Research Group, Disease Networks Research Unit, Department of Biochemistry and Molecular Medicine, University of Oulu, Finland.
| | - Elizabath Nallukunnel Raju
- Microfluidics and Biosensor Research Group, Disease Networks Research Unit, Department of Biochemistry and Molecular Medicine, University of Oulu, Finland.
| | - Samin Nooranian
- Microfluidics and Biosensor Research Group, Disease Networks Research Unit, Department of Biochemistry and Molecular Medicine, University of Oulu, Finland.
| | - Hiroki Shioda
- Laboratory of Organs and Biosystems Engineering, Department of Chemical System Engineering, University of Tokyo, Tokyo, Japan
| | - Masaki Nishikawa
- Laboratory of Organs and Biosystems Engineering, Department of Chemical System Engineering, University of Tokyo, Tokyo, Japan
| | - Yasuyuki Sakai
- Laboratory of Organs and Biosystems Engineering, Department of Chemical System Engineering, University of Tokyo, Tokyo, Japan
| | - Seppo J Vainio
- Developmental Biology Laboratory, Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.
- Infotech Oulu, University of Oulu, Oulu, Finland
- Kvantum Institute, University of Oulu, Oulu, Finland
| | - Caglar Elbuken
- Microfluidics and Biosensor Research Group, Disease Networks Research Unit, Department of Biochemistry and Molecular Medicine, University of Oulu, Finland.
- VTT Technical Research Centre of Finland Ltd., Finland
| | - Irina Raykhel
- Developmental Biology Laboratory, Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.
- Laboratory of Organs and Biosystems Engineering, Department of Chemical System Engineering, University of Tokyo, Tokyo, Japan
| |
Collapse
|
7
|
Gu S, Wu G, Lu D, Meng G, Wang Y, Tang L, Zhang W. Nephrotoxicity assessment of Esculentoside A using human-induced pluripotent stem cell-derived organoids. Phytother Res 2024; 38:4893-4903. [PMID: 36722705 DOI: 10.1002/ptr.7721] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 11/28/2022] [Accepted: 12/20/2022] [Indexed: 02/02/2023]
Abstract
Drug-induced nephrotoxicity is a leading cause of acute kidney injury (AKI). A major obstacle in predicting AKI is the lack of a comprehensive experimental model that mimics stable and physiologically relevant kidney functions and accurately reflects the changes a drug induces. Organoids derived from human-induced pluripotent stem cells (iPSCs) are promising models because of their reproducibility and similarity to the in vivo conditions. In this study, Esculentoside A, the triterpene saponin with the highest concentration isolated from the root of Phytolacca acinose Roxb., was used to induce kidney injury models in vivo and kidney organoids. Esculentoside A induced AKI in mice, together with pathological changes and enhanced apoptosis. Moreover, Esculentoside A damaged podocytes and proximal tubular endothelial cells in kidney organoids in a similar way as in vivo. We also found that treatment with 60 μM Esculentoside A induced the known biomarkers of kidney damage and inflammatory cytokines (such as kidney injury molecule (KIM-1), β2-microglobulin (β2-M), and cystatin C (CysC)) in the organoids, in which activation of Cleaved Caspase-3 was involved, possibly due to lowered mitochondrial membrane potential. In summary, this study strongly suggests using kidney organoids as a reliable platform to assess Chinese medicine-induced nephrotoxicity.
Collapse
Affiliation(s)
- Shuyi Gu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
- Shanghai Frontiers Science Center of TCM Chemical Biology, Shanghai, PR China
| | - Gaosong Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
- Shanghai Frontiers Science Center of TCM Chemical Biology, Shanghai, PR China
| | - Dong Lu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
- Shanghai Frontiers Science Center of TCM Chemical Biology, Shanghai, PR China
| | - Guofeng Meng
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
- Shanghai Frontiers Science Center of TCM Chemical Biology, Shanghai, PR China
| | - Yu Wang
- Pharmacology and Toxicology Department, Shanghai Institute for Food and Drug Control, Shanghai, PR China
| | - Liming Tang
- Pharmacology and Toxicology Department, Shanghai Institute for Food and Drug Control, Shanghai, PR China
| | - Weidong Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
- Shanghai Frontiers Science Center of TCM Chemical Biology, Shanghai, PR China
| |
Collapse
|
8
|
Sendino Garví E, van Slobbe GJJ, Zaal EA, de Baaij JHF, Hoenderop JG, Masereeuw R, Janssen MJ, van Genderen AM. KCNJ16-depleted kidney organoids recapitulate tubulopathy and lipid recovery upon statins treatment. Stem Cell Res Ther 2024; 15:268. [PMID: 39183338 PMCID: PMC11346019 DOI: 10.1186/s13287-024-03881-3] [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: 05/29/2024] [Accepted: 08/11/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND The KCNJ16 gene has been associated with a novel kidney tubulopathy phenotype, viz. disturbed acid-base homeostasis, hypokalemia and altered renal salt transport. KCNJ16 encodes for Kir5.1, which together with Kir4.1 constitutes a potassium channel located at kidney tubular cell basolateral membranes. Preclinical studies provided mechanistic links between Kir5.1 and tubulopathy, however, the disease pathology remains poorly understood. Here, we aimed at generating and characterizing a novel advanced in vitro human kidney model that recapitulates the disease phenotype to investigate further the pathophysiological mechanisms underlying the tubulopathy and potential therapeutic interventions. METHODS We used CRISPR/Cas9 to generate KCNJ16 mutant (KCNJ16+/- and KCNJ16-/-) cell lines from healthy human induced pluripotent stem cells (iPSC) KCNJ16 control (KCNJ16WT). The iPSCs were differentiated following an optimized protocol into kidney organoids in an air-liquid interface. RESULTS KCNJ16-depleted kidney organoids showed transcriptomic and potential functional impairment of key voltage-dependent electrolyte and water-balance transporters. We observed cysts formation, lipid droplet accumulation and fibrosis upon Kir5.1 function loss. Furthermore, a large scale, glutamine tracer flux metabolomics analysis demonstrated that KCNJ16-/- organoids display TCA cycle and lipid metabolism impairments. Drug screening revealed that treatment with statins, particularly the combination of simvastatin and C75, prevented lipid droplet accumulation and collagen-I deposition in KCNJ16-/- kidney organoids. CONCLUSIONS Mature kidney organoids represent a relevant in vitro model for investigating the function of Kir5.1. We discovered novel molecular targets for this genetic tubulopathy and identified statins as a potential therapeutic strategy for KCNJ16 defects in the kidney.
Collapse
Affiliation(s)
- E Sendino Garví
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands
| | - G J J van Slobbe
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands
| | - E A Zaal
- Division of Cell Biology, Metabolism and Cancer, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - J H F de Baaij
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - J G Hoenderop
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - R Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands
| | - M J Janssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands.
| | - A M van Genderen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands.
| |
Collapse
|
9
|
Kang HM, Kim DS, Kim YK, Shin K, Ahn SJ, Jung CR. Guidelines for Manufacturing and Application of Organoids: Kidney. Int J Stem Cells 2024; 17:141-146. [PMID: 38764433 PMCID: PMC11170122 DOI: 10.15283/ijsc24040] [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: 04/08/2024] [Revised: 04/25/2024] [Accepted: 05/02/2024] [Indexed: 05/21/2024] Open
Abstract
Recent advancements in organoid technology have led to a vigorous movement towards utilizing it as a substitute for animal experiments. Organoid technology offers versatile applications, particularly in toxicity testing of pharmaceuticals or chemical substances. However, for the practical use in toxicity testing, minimal guidance is required to ensure reliability and relevance. This paper aims to provide minimal guidelines for practical uses of kidney organoids derived from human pluripotent stem cells as a toxicity evaluation model in vitro.
Collapse
Affiliation(s)
- Hyun Mi Kang
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea
- Department of Functional Genomics, Korea University of Science and Technology, Daejeon, Korea
- Organoid Standards Initiative
| | - Dong Sung Kim
- Organoid Standards Initiative
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Korea
- Department of Chemical Engineering, POSTECH, Pohang, Korea
- School of Interdisciplinary Bioscience and Bioengineering, POSTECH, Pohang, Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul, Korea
| | - Yong Kyun Kim
- Organoid Standards Initiative
- Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, St. Vincent’s Hospital, Suwon, Korea
| | - Kunyoo Shin
- Organoid Standards Initiative
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Korea
- School of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, Korea
| | - Sun-Ju Ahn
- Organoid Standards Initiative
- Department of Biophysics, Sungkyunkwan University, Suwon, Korea
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Korea
| | - Cho-Rok Jung
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea
- Department of Functional Genomics, Korea University of Science and Technology, Daejeon, Korea
- Organoid Standards Initiative
| |
Collapse
|
10
|
Jin H, Xue Z, Liu J, Ma B, Yang J, Lei L. Advancing Organoid Engineering for Tissue Regeneration and Biofunctional Reconstruction. Biomater Res 2024; 28:0016. [PMID: 38628309 PMCID: PMC11018530 DOI: 10.34133/bmr.0016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 03/04/2024] [Indexed: 04/19/2024] Open
Abstract
Tissue damage and functional abnormalities in organs have become a considerable clinical challenge. Organoids are often applied as disease models and in drug discovery and screening. Indeed, several studies have shown that organoids are an important strategy for achieving tissue repair and biofunction reconstruction. In contrast to established stem cell therapies, organoids have high clinical relevance. However, conventional approaches have limited the application of organoids in clinical regenerative medicine. Engineered organoids might have the capacity to overcome these challenges. Bioengineering-a multidisciplinary field that applies engineering principles to biomedicine-has bridged the gap between engineering and medicine to promote human health. More specifically, bioengineering principles have been applied to organoids to accelerate their clinical translation. In this review, beginning with the basic concepts of organoids, we describe strategies for cultivating engineered organoids and discuss the multiple engineering modes to create conditions for breakthroughs in organoid research. Subsequently, studies on the application of engineered organoids in biofunction reconstruction and tissue repair are presented. Finally, we highlight the limitations and challenges hindering the utilization of engineered organoids in clinical applications. Future research will focus on cultivating engineered organoids using advanced bioengineering tools for personalized tissue repair and biofunction reconstruction.
Collapse
Affiliation(s)
- Hairong Jin
- Institute of Translational Medicine,
Zhejiang Shuren University, Hangzhou 310015, China
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
- Ningxia Medical University, Ningxia 750004, China
| | - Zengqi Xue
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Jinnv Liu
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Binbin Ma
- Department of Biology,
The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Jianfeng Yang
- Institute of Translational Medicine,
Zhejiang Shuren University, Hangzhou 310015, China
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Lanjie Lei
- Institute of Translational Medicine,
Zhejiang Shuren University, Hangzhou 310015, China
| |
Collapse
|
11
|
Chambers BE, Weaver NE, Lara CM, Nguyen TK, Wingert RA. (Zebra)fishing for nephrogenesis genes. Tissue Barriers 2024; 12:2219605. [PMID: 37254823 PMCID: PMC11042071 DOI: 10.1080/21688370.2023.2219605] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/14/2023] [Indexed: 06/01/2023] Open
Abstract
Kidney disease is a devastating condition affecting millions of people worldwide, where over 100,000 patients in the United States alone remain waiting for a lifesaving organ transplant. Concomitant with a surge in personalized medicine, single-gene mutations, and polygenic risk alleles have been brought to the forefront as core causes of a spectrum of renal disorders. With the increasing prevalence of kidney disease, it is imperative to make substantial strides in the field of kidney genetics. Nephrons, the core functional units of the kidney, are epithelial tubules that act as gatekeepers of body homeostasis by absorbing and secreting ions, water, and small molecules to filter the blood. Each nephron contains a series of proximal and distal segments with explicit metabolic functions. The embryonic zebrafish provides an ideal platform to systematically dissect the genetic cues governing kidney development. Here, we review the use of zebrafish to discover nephrogenesis genes.
Collapse
Affiliation(s)
- Brooke E. Chambers
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, Indiana (IN), USA
| | - Nicole E. Weaver
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, Indiana (IN), USA
| | - Caroline M. Lara
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, Indiana (IN), USA
| | - Thanh Khoa Nguyen
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, Indiana (IN), USA
| | - Rebecca A. Wingert
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, Indiana (IN), USA
| |
Collapse
|
12
|
Smandri A, Al-Masawa ME, Hwei NM, Fauzi MB. ECM-derived biomaterials for regulating tissue multicellularity and maturation. iScience 2024; 27:109141. [PMID: 38405613 PMCID: PMC10884934 DOI: 10.1016/j.isci.2024.109141] [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] [Indexed: 02/27/2024] Open
Abstract
Recent breakthroughs in developing human-relevant organotypic models led to the building of highly resemblant tissue constructs that hold immense potential for transplantation, drug screening, and disease modeling. Despite the progress in fine-tuning stem cell multilineage differentiation in highly controlled spatiotemporal conditions and hosting microenvironments, 3D models still experience naive and incomplete morphogenesis. In particular, existing systems and induction protocols fail to maintain stem cell long-term potency, induce high tissue-level multicellularity, or drive the maturity of stem cell-derived 3D models to levels seen in their in vivo counterparts. In this review, we highlight the use of extracellular matrix (ECM)-derived biomaterials in providing stem cell niche-mimicking microenvironment capable of preserving stem cell long-term potency and inducing spatial and region-specific differentiation. We also examine the maturation of different 3D models, including organoids, encapsulated in ECM biomaterials and provide looking-forward perspectives on employing ECM biomaterials in building more innovative, transplantable, and functional organs.
Collapse
Affiliation(s)
- Ali Smandri
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Maimonah Eissa Al-Masawa
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Ng Min Hwei
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Mh Busra Fauzi
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
13
|
Cui Z, Wei H, Goding C, Cui R. Stem cell heterogeneity, plasticity, and regulation. Life Sci 2023; 334:122240. [PMID: 37925141 DOI: 10.1016/j.lfs.2023.122240] [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: 09/08/2023] [Revised: 10/30/2023] [Accepted: 10/31/2023] [Indexed: 11/06/2023]
Abstract
As a population of homogeneous cells with both self-renewal and differentiation potential, stem cell pools are highly compartmentalized and contain distinct subsets that exhibit stable but limited heterogeneity during homeostasis. However, their striking plasticity is showcased under natural or artificial stress, such as injury, transplantation, cancer, and aging, leading to changes in their phenotype, constitution, metabolism, and function. The complex and diverse network of cell-extrinsic niches and signaling pathways, together with cell-intrinsic genetic and epigenetic regulators, tightly regulate both the heterogeneity during homeostasis and the plasticity under perturbation. Manipulating these factors offers better control of stem cell behavior and a potential revolution in the current state of regenerative medicine. However, disruptions of normal regulation by genetic mutation or excessive plasticity acquisition may contribute to the formation of tumors. By harnessing innovative techniques that enhance our understanding of stem cell heterogeneity and employing novel approaches to maximize the utilization of stem cell plasticity, stem cell therapy holds immense promise for revolutionizing the future of medicine.
Collapse
Affiliation(s)
- Ziyang Cui
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing 100034, China.
| | - Hope Wei
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, United States of America
| | - Colin Goding
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Headington, Oxford OX37DQ, UK
| | - Rutao Cui
- Skin Disease Research Institute, The 2nd Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| |
Collapse
|
14
|
Kim KH, Kim EJ, Kim HY, Li S, Jung HS. Fabrication of functional ameloblasts from hiPSCs for dental application. Front Cell Dev Biol 2023; 11:1164811. [PMID: 37457296 PMCID: PMC10339106 DOI: 10.3389/fcell.2023.1164811] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 06/15/2023] [Indexed: 07/18/2023] Open
Abstract
Tooth formation relies on two types of dental cell populations, namely, the dental epithelium and dental mesenchyme, and the interactions between these cell populations are important during tooth development. Although human-induced pluripotent stem cells (hiPSCs) can differentiate into dental epithelial and mesenchymal cells, organoid research on tooth development has not been established yet. This study focused on the hiPSC-derived human ameloblast organoid (hAO) using a three-dimensional (3D) culture system. hAOs had similar properties to ameloblasts, forming enamel in response to calcium and mineralization by interaction with the dental mesenchyme. hAOs simultaneously had osteogenic and odontogenic differentiation potential. Furthermore, hAOs demonstrated tooth regenerative potential upon interaction with the mouse dental mesenchyme. Our findings provide new insights into a suitable hiPSC-derived dental source and demonstrate that hAOs can be beneficial not only for tooth regeneration but also for the study of various dental diseases for which treatment has not been developed yet.
Collapse
Affiliation(s)
- Ka-Hwa Kim
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Taste Research Center, Oral Science Research Center, BK21 FOUR Project, Yonsei University College of Dentistry, Seoul, Republic of Korea
| | - Eun-Jung Kim
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Taste Research Center, Oral Science Research Center, BK21 FOUR Project, Yonsei University College of Dentistry, Seoul, Republic of Korea
| | | | - Shujin Li
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Taste Research Center, Oral Science Research Center, BK21 FOUR Project, Yonsei University College of Dentistry, Seoul, Republic of Korea
| | - Han-Sung Jung
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Taste Research Center, Oral Science Research Center, BK21 FOUR Project, Yonsei University College of Dentistry, Seoul, Republic of Korea
| |
Collapse
|
15
|
Zhu Y, Shi Z, Ding W, Li C. On-chip construction of a fully structured scaffold-free vascularized renal tubule. Biomed Microdevices 2023; 25:8. [PMID: 36826720 DOI: 10.1007/s10544-023-00648-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2023] [Indexed: 02/25/2023]
Abstract
Renal tubule chips have emerged as a promising platform for drug nephrotoxicity testing. However, the reported renal tubule chips hardly replicate the unique structure of renal tubules with thick proximal and distal tubules and a thin loop of Henle. In this study, we developed a fully structured scaffold-free vascularized renal tubule on a microfluidic chip. On the chip, the renal epithelial cell-laden hollow calcium-polymerized alginate tube with thick segments at both ends and a thin middle segment was U-shaped embedded in collagen hydrogel, parallel to the endothelial cell-laden hollow calcium-polymerized alginate tube with uniform tube diameter. After the alginate tubes were on-chip degraded, the renal epithelial cells and endothelial cells automatically attached to the collagen hydrogel and proliferated to form the renal tubule with proximal tubule, loop of Henle and distal tubule as well as peritubular blood vessel. We evaluated the viability of cells on the hollow alginate tubes, characterized the distribution and morphology of cells before and after the degradation of the alginate tube, and confirmed the proliferation of cells and the metabolic function of cells in terms of ATP synthesis, fibronectin secretion and VEGFR2 expression on the chip. The enhanced metabolic functions of renal epithelial cells and endothelial cells were preliminarily demonstrated. This study provides new insights into designing a more biomimetic renal tubule on a microfluidic chip.
Collapse
Affiliation(s)
- Yuntian Zhu
- , Hefei No.1 High School, 230041, Hefei, Anhui, China
| | - Zhengdi Shi
- School of Information Science and Technology, University of Science and Technology of China, 230027, Hefei, Anhui, China
| | - Weiping Ding
- School of Information Science and Technology, University of Science and Technology of China, 230027, Hefei, Anhui, China.
| | - Chengpan Li
- School of Information Science and Technology, University of Science and Technology of China, 230027, Hefei, Anhui, China. .,Center for Biomedical Imaging, University of Science and Technology of China, 230027, Hefei, Anhui, China.
| |
Collapse
|
16
|
Qian S, Mao J, Liu Z, Zhao B, Zhao Q, Lu B, Zhang L, Mao X, Cheng L, Cui W, Zhang Y, Sun X. Stem cells for organoids. SMART MEDICINE 2022; 1:e20220007. [PMID: 39188738 PMCID: PMC11235201 DOI: 10.1002/smmd.20220007] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 08/23/2022] [Indexed: 08/28/2024]
Abstract
Organoids are three-dimensional (3D) cell culture systems that simulate the structures and functions of organs, involving applications in disease modeling, drug screening, and cellular developmental biology. The material matrix in organoids can provide a 3D environment for stem cells to differentiate into different cell types and continuously self-renew, thereby realizing the in vitro culture of organs, which has received extensive attention in recent years. However, some challenges still exist in organoids, including low maturity, high heterogeneity, and lack of spatiotemporal regulation. Therefore, in this review, we summarized the culturing protocols and various applications of stem cell-derived organoids and proposed insightful thoughts for engineering stem cells into organoids in view of the current shortcomings, to achieve the further application and clinical translation of stem cells and engineered stem cells in organoid research.
Collapse
Affiliation(s)
- Shutong Qian
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jiayi Mao
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Zhimo Liu
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Binfan Zhao
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Qiuyu Zhao
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Bolun Lu
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Liucheng Zhang
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiyuan Mao
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Liying Cheng
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Wenguo Cui
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yuguang Zhang
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiaoming Sun
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
17
|
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
|
18
|
Kalejaiye TD, Barreto AD, Musah S. Translating Organoids into Artificial Kidneys. CURRENT TRANSPLANTATION REPORTS 2022; 9:276-286. [PMID: 36311696 PMCID: PMC9592871 DOI: 10.1007/s40472-022-00383-0] [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] [Accepted: 09/27/2022] [Indexed: 11/21/2022]
Abstract
Purpose of Review
Kidney disease affects more than 13% of the world population, and current treatment options are limited to dialysis and organ transplantation. The generation of kidney organoids from human-induced pluripotent stem (hiPS) cells could be harnessed to engineer artificial organs and help overcome the challenges associated with the limited supply of transplantable kidneys. The purpose of this article is to review the progress in kidney organoid generation and transplantation and highlight some existing challenges in the field. We also examined possible improvements that could help realize the potential of organoids as artificial organs or alternatives for kidney transplantation therapy. Recent Findings Organoids are useful for understanding the mechanisms of kidney development, and they provide robust platforms for drug screening, disease modeling, and generation of tissues for organ replacement therapies. Efforts to design organoids rely on the ability of cells to self-assemble and pattern themselves into recognizable tissues. While existing protocols for generating organoids result in multicellular structures reminiscent of the developing kidney, many do not yet fully recapitulate the complex cellular composition, structure, and functions of the intact kidney. Recent advances toward achieving these goals include identifying cell culture conditions that produce organoids with improved vasculature and cell maturation and functional states. Still, additional improvements are needed to enhance tissue patterning, specialization, and function, and avoid tumorigenicity after transplantation. Summary This report focuses on kidney organoid studies, advancements and limitations, and future directions for improvements towards transplantation.
Collapse
Affiliation(s)
- Titilola D. Kalejaiye
- grid.26009.3d0000 0004 1936 7961Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC USA
| | - Amanda D. Barreto
- grid.26009.3d0000 0004 1936 7961Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC USA
| | - Samira Musah
- grid.26009.3d0000 0004 1936 7961Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC USA ,grid.26009.3d0000 0004 1936 7961Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC USA ,grid.26009.3d0000 0004 1936 7961Department of Cell Biology, Duke University, Durham, NC USA ,Affiliate Faculty of the Developmental and Stem Cell Biology Program, Duke Regeneration Center, and Duke MEDx Initiative, Durham, NC USA
| |
Collapse
|
19
|
Harnessing conserved signaling and metabolic pathways to enhance the maturation of functional engineered tissues. NPJ Regen Med 2022; 7:44. [PMID: 36057642 PMCID: PMC9440900 DOI: 10.1038/s41536-022-00246-3] [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: 02/22/2022] [Accepted: 08/05/2022] [Indexed: 11/08/2022] Open
Abstract
The development of induced-pluripotent stem cell (iPSC)-derived cell types offers promise for basic science, drug testing, disease modeling, personalized medicine, and translatable cell therapies across many tissue types. However, in practice many iPSC-derived cells have presented as immature in physiological function, and despite efforts to recapitulate adult maturity, most have yet to meet the necessary benchmarks for the intended tissues. Here, we summarize the available state of knowledge surrounding the physiological mechanisms underlying cell maturation in several key tissues. Common signaling consolidators, as well as potential synergies between critical signaling pathways are explored. Finally, current practices in physiologically relevant tissue engineering and experimental design are critically examined, with the goal of integrating greater decision paradigms and frameworks towards achieving efficient maturation strategies, which in turn may produce higher-valued iPSC-derived tissues.
Collapse
|
20
|
Wang Q, Lu J, Fan K, Xu Y, Xiong Y, Sun Z, Zhai M, Zhang Z, Zhang S, Song Y, Luo J, You M, Guo M, Zhang X. High-throughput "read-on-ski" automated imaging and label-free detection system for toxicity screening of compounds using personalised human kidney organoids. J Zhejiang Univ Sci B 2022; 23:564-577. [PMID: 35794686 PMCID: PMC9264113 DOI: 10.1631/jzus.b2100701] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 02/04/2022] [Indexed: 11/11/2022]
Abstract
Organoid models are used to study kidney physiology, such as the assessment of nephrotoxicity and underlying disease processes. Personalized human pluripotent stem cell-derived kidney organoids are ideal models for compound toxicity studies, but there is a need to accelerate basic and translational research in the field. Here, we developed an automated continuous imaging setup with the "read-on-ski" law of control to maximize temporal resolution with minimum culture plate vibration. High-accuracy performance was achieved: organoid screening and imaging were performed at a spatial resolution of 1.1 μm for the entire multi-well plate under 3 min. We used the in-house developed multi-well spinning device and cisplatin-induced nephrotoxicity model to evaluate the toxicity in kidney organoids using this system. The acquired images were processed via machine learning-based classification and segmentation algorithms, and the toxicity in kidney organoids was determined with 95% accuracy. The results obtained by the automated "read-on-ski" imaging device, combined with label-free and non-invasive algorithms for detection, were verified using conventional biological procedures. Taking advantage of the close-to-in vivo-kidney organoid model, this new development opens the door for further application of scaled-up screening using organoids in basic research and drug discovery.
Collapse
Affiliation(s)
- Qizheng Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Jun Lu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510320, China
| | - Ke Fan
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Yiwei Xu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Yucui Xiong
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Zhiyong Sun
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510320, China
| | - Man Zhai
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Zhizhong Zhang
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510320, China
| | - Sheng Zhang
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Yan Song
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Jianzhong Luo
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Mingliang You
- Hangzhou Cancer Institute, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou 310002, China
| | - Meijin Guo
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China. ,
| | - Xiao Zhang
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510320, China.
| |
Collapse
|
21
|
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
|
22
|
Bonanini F, Kurek D, Previdi S, Nicolas A, Hendriks D, de Ruiter S, Meyer M, Clapés Cabrer M, Dinkelberg R, García SB, Kramer B, Olivier T, Hu H, López-Iglesias C, Schavemaker F, Walinga E, Dutta D, Queiroz K, Domansky K, Ronden B, Joore J, Lanz HL, Peters PJ, Trietsch SJ, Clevers H, Vulto P. In vitro grafting of hepatic spheroids and organoids on a microfluidic vascular bed. Angiogenesis 2022; 25:455-470. [PMID: 35704148 PMCID: PMC9519670 DOI: 10.1007/s10456-022-09842-9] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 05/14/2022] [Indexed: 12/12/2022]
Abstract
With recent progress in modeling liver organogenesis and regeneration, the lack of vasculature is becoming the bottleneck in progressing our ability to model human hepatic tissues in vitro. Here, we introduce a platform for routine grafting of liver and other tissues on an in vitro grown microvascular bed. The platform consists of 64 microfluidic chips patterned underneath a 384-well microtiter plate. Each chip allows the formation of a microvascular bed between two main lateral vessels by inducing angiogenesis. Chips consist of an open-top microfluidic chamber, which enables addition of a target tissue by manual or robotic pipetting. Upon grafting a liver microtissue, the microvascular bed undergoes anastomosis, resulting in a stable, perfusable vascular network. Interactions with vasculature were found in spheroids and organoids upon 7 days of co-culture with space of Disse-like architecture in between hepatocytes and endothelium. Veno-occlusive disease was induced by azathioprine exposure, leading to impeded perfusion of the vascularized spheroid. The platform holds the potential to replace animals with an in vitro alternative for routine grafting of spheroids, organoids, or (patient-derived) explants.
Collapse
Affiliation(s)
| | | | | | | | - Delilah Hendriks
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, 3584 CT, Utrecht, The Netherlands
| | | | | | | | | | | | | | | | - Huili Hu
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, 3584 CT, Utrecht, The Netherlands
| | - Carmen López-Iglesias
- The Maastricht Multimodal Molecular Imaging Institute, Maastricht University, Maastricht, The Netherlands
| | | | | | - Devanjali Dutta
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, 3584 CT, Utrecht, The Netherlands
| | | | | | | | | | | | - Peter J Peters
- The Maastricht Multimodal Molecular Imaging Institute, Maastricht University, Maastricht, The Netherlands
| | | | - Hans Clevers
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, 3584 CT, Utrecht, The Netherlands
| | | |
Collapse
|
23
|
Genderen AMV, G Valverde M, Capendale PE, Kersten V, Sendino Garví E, Schuurmans CCL, Ruelas M, Soeiro JT, Tang G, Janssen MJ, Jansen J, Mihăilă SM, Vermonden T, Zhang YS, Masereeuw R. Co-axial Printing of Convoluted Proximal Tubule for Kidney Disease Modeling. Biofabrication 2022; 14. [PMID: 35700695 DOI: 10.1088/1758-5090/ac7895] [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] [Received: 02/21/2022] [Accepted: 06/14/2022] [Indexed: 11/11/2022]
Abstract
Despite the increasing incidence of kidney-related diseases, we are still far from understanding the underlying mechanisms of these diseases and their progression. This lack of understanding is partly because of a poor replication of the diseases in vitro, limited to planar culture. Advancing towards three-dimensional models, hereby we propose coaxial printing to obtain microfibers containing a helical hollow microchannel. These recapitulate the architecture of the proximal tubule (PT), an important nephron segment often affected in kidney disorders. A stable gelatin/alginate-based ink was formulated to allow printability while maintaining structural properties. Fine tuning of the composition, printing temperature and extrusion rate allowed for optimal ink viscosity that led to coiling of the microfiber's inner channel. The printed microfibers exhibited prolonged structural stability (42 days) and cytocompatibility in culture. Healthy conditionally immortalized PT epithelial cells and a knockout cell model for cystinosis (CTNS-/-) were seeded to mimic two genotypes of PT. Upon culturing for 14 days, engineered PT showed homogenous cytoskeleton organization as indicated by staining for filamentous actin, barrier-formation and polarization with apical marker α-tubulin and basolateral marker Na+/K+-ATPase. Cell viability was slightly decreased upon prolonged culturing for 14 days, which was more pronounced inCTNS-/-microfibers. Finally, cystinosis cells showed reduced apical transport activity in the microfibers compared to healthy PT epithelial cells when looking at breast cancer resistance protein and multidrug resistance-associated protein 4. Engineered PT incorporated in a custom-designed microfluidic chip allowed to assess leak-tightness of the epithelium, which appeared less tight in cystinosis PT compared to healthy PT, in agreement with its in vivo phenotype. While we are still on the verge of patient-oriented medicine, this system holds great promise for further research in establishing advanced in vitro disease models.
Collapse
Affiliation(s)
- Anne Metje van Genderen
- Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, NETHERLANDS
| | - Marta G Valverde
- Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, NETHERLANDS
| | - Pamela E Capendale
- Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, NETHERLANDS
| | - Valerie Kersten
- Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, NETHERLANDS
| | - Elena Sendino Garví
- Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, NETHERLANDS
| | - Carl C L Schuurmans
- Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, NETHERLANDS
| | - Marina Ruelas
- Harvard Medical School, 65 Landsdowne Street, Cambridge, Massachusetts, 02139, UNITED STATES
| | - Joana T Soeiro
- Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, NETHERLANDS
| | - Guosheng Tang
- Harvard Medical School, 65 Landsdowne Street, Cambridge, Massachusetts, 02139, UNITED STATES
| | - Manoe J Janssen
- Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, NETHERLANDS
| | - Jitske Jansen
- Pathology and Pediatric Nephrology, Radboud University Medical Center, -, Nijmegen, 6525 GA, NETHERLANDS
| | - Silvia M Mihăilă
- Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, NETHERLANDS
| | - Tina Vermonden
- Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Universiteit Utrecht, Universiteitsweg 99, Utrecht, 3584 CG, NETHERLANDS
| | - Y Shrike Zhang
- Harvard Medical School, 65 Landsdowne Street, Cambridge, Massachusetts, 02139, UNITED STATES
| | - Rosalinde Masereeuw
- Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, NETHERLANDS
| |
Collapse
|
24
|
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
|
25
|
Hautefort I, Poletti M, Papp D, Korcsmaros T. Everything You Always Wanted to Know About Organoid-Based Models (and Never Dared to Ask). Cell Mol Gastroenterol Hepatol 2022; 14:311-331. [PMID: 35643188 PMCID: PMC9233279 DOI: 10.1016/j.jcmgh.2022.04.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 12/12/2022]
Abstract
Homeostatic functions of a living tissue, such as the gastrointestinal tract, rely on highly sophisticated and finely tuned cell-to-cell interactions. These crosstalks evolve and continuously are refined as the tissue develops and give rise to specialized cells performing general and tissue-specific functions. To study these systems, stem cell-based in vitro models, often called organoids, and non-stem cell-based primary cell aggregates (called spheroids) appeared just over a decade ago. These models still are evolving and gaining complexity, making them the state-of-the-art models for studying cellular crosstalk in the gastrointestinal tract, and to investigate digestive pathologies, such as inflammatory bowel disease, colorectal cancer, and liver diseases. However, the use of organoid- or spheroid-based models to recapitulate in vitro the highly complex structure of in vivo tissue remains challenging, and mainly restricted to expert developmental cell biologists. Here, we condense the founding knowledge and key literature information that scientists adopting the organoid technology for the first time need to consider when using these models for novel biological questions. We also include information that current organoid/spheroid users could use to add to increase the complexity to their existing models. We highlight the current and prospective evolution of these models through bridging stem cell biology with biomaterial and scaffold engineering research areas. Linking these complementary fields will increase the in vitro mimicry of in vivo tissue, and potentially lead to more successful translational biomedical applications. Deepening our understanding of the nature and dynamic fine-tuning of intercellular crosstalks will enable identifying novel signaling targets for new or repurposed therapeutics used in many multifactorial diseases.
Collapse
Affiliation(s)
- Isabelle Hautefort
- Earlham Institute, Organisms and Ecosystems Programme, Norwich, United Kingdom
| | - Martina Poletti
- Earlham Institute, Organisms and Ecosystems Programme, Norwich, United Kingdom; Quadram Institute Bioscience, Gut Microbes and Health Programme, Norwich, United Kingdom
| | - Diana Papp
- Quadram Institute Bioscience, Gut Microbes and Health Programme, Norwich, United Kingdom
| | - Tamas Korcsmaros
- Earlham Institute, Organisms and Ecosystems Programme, Norwich, United Kingdom; Quadram Institute Bioscience, Gut Microbes and Health Programme, Norwich, United Kingdom; Imperial College London, Department of Metabolism, Digestion and Reproduction, London, United Kingdom.
| |
Collapse
|
26
|
Nunez-Nescolarde AB, Nikolic-Paterson DJ, Combes AN. Human Kidney Organoids and Tubuloids as Models of Complex Kidney Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:738-749. [PMID: 35181335 DOI: 10.1016/j.ajpath.2022.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/18/2022] [Accepted: 01/24/2022] [Indexed: 10/19/2022]
Abstract
Kidney organoids derived from pluripotent stem cells and epithelial organoids derived from adult tissue (tubuloids) have been used to study various kidney disorders with a strong genetic component, such as polycystic kidney disease, Wilms tumor, and congenital nephrotic syndrome. However, complex disorders without clear genetic associations, such as acute kidney injury and many forms of chronic kidney disease, are only just beginning to be investigated using these in vitro approaches. Although organoids are a reductionist model, they contain clinically relevant cell populations that may help to elucidate human-specific pathogenic mechanisms. Thus, organoids may complement animal disease models to accelerate the translation of laboratory proof-of-concept research into clinical practice. This review discusses whether kidney organoids and tubuloids are suitable models for the study of complex human kidney disease and highlights their advantages and limitations compared with monolayer cell culture and animal models.
Collapse
Affiliation(s)
- Ana B Nunez-Nescolarde
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - David J Nikolic-Paterson
- Department of Nephrology, Monash Health and Monash University Centre for Inflammatory Diseases, Monash Medical Centre, Clayton, Victoria, Australia.
| | - Alexander N Combes
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
27
|
Li M, Gong J, Gao L, Zou T, Kang J, Xu H. Advanced human developmental toxicity and teratogenicity assessment using human organoid models. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 235:113429. [PMID: 35325609 DOI: 10.1016/j.ecoenv.2022.113429] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/12/2022] [Accepted: 03/16/2022] [Indexed: 06/14/2023]
Abstract
Tremendous progress has been made in the field of toxicology leading to the advance of developmental toxicity assessment. Conventional animal models and in vitro two-dimensional models cannot accurately describe toxic effects and predict actual in vivo responses due to obvious inter-species differences between humans and animals, as well as the lack of a physiologically relevant tissue microenvironment. Human embryonic stem cell (hESC)- and induced pluripotent stem cell (iPSC)-derived three-dimensional organoids are ideal complex and multicellular organotypic models, which are indispensable in recapitulating morphogenesis, cellular interactions, and molecular processes of early human organ development. Recently, human organoids have been used for drug discovery, chemical toxicity and safety in vitro assessment. This review discusses the recent advances in the use of human organoid models, (i.e., brain, retinal, cardiac, liver, kidney, lung, and intestinal organoid models) for developmental toxicity and teratogenicity assessment of distinct tissues/organs following exposure to pharmaceutical compounds, heavy metals, persistent organic pollutants, nanomaterials, and ambient air pollutants. Combining next-generation organoid models with innovative engineering technologies generates novel and powerful tools for developmental toxicity and teratogenicity assessment, and the rapid progress in this field is expected to continue.
Collapse
Affiliation(s)
- Minghui Li
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Jing Gong
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Lixiong Gao
- Department of Ophthalmology, Third Medical Center of PLA General Hospital, Beijing 100039, China
| | - Ting Zou
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Jiahui Kang
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Haiwei Xu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China.
| |
Collapse
|
28
|
Molema G, Zijlstra JG, van Meurs M, Kamps JAAM. Renal microvascular endothelial cell responses in sepsis-induced acute kidney injury. Nat Rev Nephrol 2022; 18:95-112. [PMID: 34667283 DOI: 10.1038/s41581-021-00489-1] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2021] [Indexed: 12/29/2022]
Abstract
Microvascular endothelial cells in the kidney have been a neglected cell type in sepsis-induced acute kidney injury (sepsis-AKI) research; yet, they offer tremendous potential as pharmacological targets. As endothelial cells in distinct cortical microvascular segments are highly heterogeneous, this Review focuses on endothelial cells in their anatomical niche. In animal models of sepsis-AKI, reduced glomerular blood flow has been attributed to inhibition of endothelial nitric oxide synthase activation in arterioles and glomeruli, whereas decreased cortex peritubular capillary perfusion is associated with epithelial redox stress. Elevated systemic levels of vascular endothelial growth factor, reduced levels of circulating sphingosine 1-phosphate and loss of components of the glycocalyx from glomerular endothelial cells lead to increased microvascular permeability. Although coagulation disbalance occurs in all microvascular segments, the molecules involved differ between segments. Induction of the expression of adhesion molecules and leukocyte recruitment also occurs in a heterogeneous manner. Evidence of similar endothelial cell responses has been found in kidney and blood samples from patients with sepsis. Comprehensive studies are needed to investigate the relationships between segment-specific changes in the microvasculature and kidney function loss in sepsis-AKI. The application of omics technologies to kidney tissues from animals and patients will be key in identifying these relationships and in developing novel therapeutics for sepsis.
Collapse
Affiliation(s)
- Grietje Molema
- Dept. Pathology and Medical Biology, Medical Biology section, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
| | - Jan G Zijlstra
- Dept. Critical Care, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Matijs van Meurs
- Dept. Pathology and Medical Biology, Medical Biology section, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.,Dept. Critical Care, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Jan A A M Kamps
- Dept. Pathology and Medical Biology, Medical Biology section, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
29
|
Abstract
The kidney is a highly complex organ in the human body. Although creating an in vitro model of the human kidney is challenging, tremendous advances have been made in recent years. Kidney organoids are in vitro kidney models that are generated from stem cells in three-dimensional (3D) cultures. They exhibit remarkable degree of similarities with the native tissue in terms of cell type, morphology, and function. The establishment of 3D kidney organoids facilitates a mechanistic study of cell communications, and these organoids can be used for drug screening, disease modeling, and regenerative medicine applications. This review discusses the cellular complexity during in vitro kidney generation. We intend to highlight recent progress in kidney organoids and the applications of these relatively new technologies.
Collapse
|
30
|
Pou Casellas C, Rookmaaker MB, Verhaar MC. Controlling cellular plasticity to improve in vitro models for kidney regeneration. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2021. [DOI: 10.1016/j.cobme.2021.100345] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
31
|
Schneider N, Sundaresan Y, Gopalakrishnan P, Beryozkin A, Hanany M, Levanon EY, Banin E, Ben-Aroya S, Sharon D. Inherited retinal diseases: Linking genes, disease-causing variants, and relevant therapeutic modalities. Prog Retin Eye Res 2021; 89:101029. [PMID: 34839010 DOI: 10.1016/j.preteyeres.2021.101029] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 11/11/2021] [Accepted: 11/16/2021] [Indexed: 12/11/2022]
Abstract
Inherited retinal diseases (IRDs) are a clinically complex and heterogenous group of visual impairment phenotypes caused by pathogenic variants in at least 277 nuclear and mitochondrial genes, affecting different retinal regions, and depleting the vision of affected individuals. Genes that cause IRDs when mutated are unique by possessing differing genotype-phenotype correlations, varying inheritance patterns, hypomorphic alleles, and modifier genes thus complicating genetic interpretation. Next-generation sequencing has greatly advanced the identification of novel IRD-related genes and pathogenic variants in the last decade. For this review, we performed an in-depth literature search which allowed for compilation of the Global Retinal Inherited Disease (GRID) dataset containing 4,798 discrete variants and 17,299 alleles published in 31 papers, showing a wide range of frequencies and complexities among the 194 genes reported in GRID, with 65% of pathogenic variants being unique to a single individual. A better understanding of IRD-related gene distribution, gene complexity, and variant types allow for improved genetic testing and therapies. Current genetic therapeutic methods are also quite diverse and rely on variant identification, and range from whole gene replacement to single nucleotide editing at the DNA or RNA levels. IRDs and their suitable therapies thus require a range of effective disease modelling in human cells, granting insight into disease mechanisms and testing of possible treatments. This review summarizes genetic and therapeutic modalities of IRDs, provides new analyses of IRD-related genes (GRID and complexity scores), and provides information to match genetic-based therapies such as gene-specific and variant-specific therapies to the appropriate individuals.
Collapse
Affiliation(s)
- Nina Schneider
- Department of Ophthalmology, Hadassah Medical Center, Faculty of Medicine, The Hebrew University of Jerusalem, 91120, Israel
| | - Yogapriya Sundaresan
- Department of Ophthalmology, Hadassah Medical Center, Faculty of Medicine, The Hebrew University of Jerusalem, 91120, Israel
| | - Prakadeeswari Gopalakrishnan
- Department of Ophthalmology, Hadassah Medical Center, Faculty of Medicine, The Hebrew University of Jerusalem, 91120, Israel
| | - Avigail Beryozkin
- Department of Ophthalmology, Hadassah Medical Center, Faculty of Medicine, The Hebrew University of Jerusalem, 91120, Israel
| | - Mor Hanany
- Department of Ophthalmology, Hadassah Medical Center, Faculty of Medicine, The Hebrew University of Jerusalem, 91120, Israel
| | - Erez Y Levanon
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, 5290002, Israel
| | - Eyal Banin
- Department of Ophthalmology, Hadassah Medical Center, Faculty of Medicine, The Hebrew University of Jerusalem, 91120, Israel
| | - Shay Ben-Aroya
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, 5290002, Israel
| | - Dror Sharon
- Department of Ophthalmology, Hadassah Medical Center, Faculty of Medicine, The Hebrew University of Jerusalem, 91120, Israel.
| |
Collapse
|
32
|
Yanofsky SM, Dugas CM, Katsurada A, Liu J, Saifudeen Z, El-Dahr SS, Satou R. Angiotensin II biphasically regulates cell differentiation in human iPSC-derived kidney organoids. Am J Physiol Renal Physiol 2021; 321:F559-F571. [PMID: 34448643 PMCID: PMC8616599 DOI: 10.1152/ajprenal.00134.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 08/09/2021] [Accepted: 08/25/2021] [Indexed: 12/28/2022] Open
Abstract
Human kidney organoid technology holds promise for novel kidney disease treatment strategies and utility in pharmacological and basic science. Given the crucial roles of the intrarenal renin-angiotensin system (RAS) and angiotensin II (ANG II) in the progression of kidney development and injury, we investigated the expression of RAS components and effects of ANG II on cell differentiation in human kidney organoids. Human induced pluripotent stem cell-derived kidney organoids were induced using a modified 18-day Takasato protocol. Gene expression analysis by digital PCR and immunostaining demonstrated the formation of renal compartments and expression of RAS components. The ANG II type 1 receptor (AT1R) was strongly expressed in the early phase of organoid development (around day 0), whereas ANG II type 2 receptor (AT2R) expression levels peaked on day 5. Thus, the organoids were treated with 100 nM ANG II in the early phase on days 0-5 (ANG II-E) or during the middle phase on days 5-10 (ANG II-M). ANG II-E was observed to decrease levels of marker genes for renal tubules and proximal tubules, and the downregulation of renal tubules was inhibited by an AT1R antagonist. In contrast, ANG II-M increased levels of markers for podocytes, the ureteric tip, and the nephrogenic mesenchyme, and an AT2R blocker attenuated the ANG II-M-induced augmentation of podocyte formation. These findings demonstrate RAS expression and ANG II exertion of biphasic effects on cell differentiation through distinct mediatory roles of AT1R and AT2R, providing a novel strategy to establish and further characterize the developmental potential of human induced pluripotent stem cell-derived kidney organoids.NEW & NOTEWORTHY This study demonstrates angiotensin II exertion of biphasic effects on cell differentiation through distinct mediatory roles of angiotensin II type 1 receptor and type 2 receptor in human induced pluripotent stem cell-derived kidney organoids, providing a novel strategy to establish and further characterize the developmental potential of the human kidney organoids.
Collapse
MESH Headings
- Angiotensin II/pharmacology
- Cell Differentiation/drug effects
- Cell Line
- Gene Expression Regulation, Developmental
- Humans
- Induced Pluripotent Stem Cells/drug effects
- Induced Pluripotent Stem Cells/metabolism
- Kidney/cytology
- Kidney/drug effects
- Kidney/metabolism
- Organoids/cytology
- Organoids/drug effects
- Organoids/metabolism
- Receptor, Angiotensin, Type 1/agonists
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/metabolism
- Receptor, Angiotensin, Type 2/agonists
- Receptor, Angiotensin, Type 2/genetics
- Receptor, Angiotensin, Type 2/metabolism
- Renin-Angiotensin System/drug effects
- Signal Transduction
- Time Factors
Collapse
Affiliation(s)
- Stacy M Yanofsky
- Department of Physiology and Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, Louisiana
| | - Courtney M Dugas
- Department of Physiology and Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, Louisiana
| | - Akemi Katsurada
- Department of Physiology and Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, Louisiana
| | - Jiao Liu
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, Louisiana
| | - Zubaida Saifudeen
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, Louisiana
| | - Samir S El-Dahr
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, Louisiana
| | - Ryousuke Satou
- Department of Physiology and Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, Louisiana
| |
Collapse
|
33
|
Lu S, Zhang J, Lin S, Zheng D, Shen Y, Qin J, Li Y, Wang S. Recent advances in the development of in vitro liver models for hepatotoxicity testing. Biodes Manuf 2021. [DOI: 10.1007/s42242-021-00142-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
34
|
Ebefors K, Lassén E, Anandakrishnan N, Azeloglu EU, Daehn IS. Modeling the Glomerular Filtration Barrier and Intercellular Crosstalk. Front Physiol 2021; 12:689083. [PMID: 34149462 PMCID: PMC8206562 DOI: 10.3389/fphys.2021.689083] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/05/2021] [Indexed: 12/11/2022] Open
Abstract
The glomerulus is a compact cluster of capillaries responsible for blood filtration and initiating urine production in the renal nephrons. A trilaminar structure in the capillary wall forms the glomerular filtration barrier (GFB), composed of glycocalyx-enriched and fenestrated endothelial cells adhering to the glomerular basement membrane and specialized visceral epithelial cells, podocytes, forming the outermost layer with a molecular slit diaphragm between their interdigitating foot processes. The unique dynamic and selective nature of blood filtration to produce urine requires the functionality of each of the GFB components, and hence, mimicking the glomerular filter in vitro has been challenging, though critical for various research applications and drug screening. Research efforts in the past few years have transformed our understanding of the structure and multifaceted roles of the cells and their intricate crosstalk in development and disease pathogenesis. In this review, we present a new wave of technologies that include glomerulus-on-a-chip, three-dimensional microfluidic models, and organoids all promising to improve our understanding of glomerular biology and to enable the development of GFB-targeted therapies. Here, we also outline the challenges and the opportunities of these emerging biomimetic systems that aim to recapitulate the complex glomerular filter, and the evolving perspectives on the sophisticated repertoire of cellular signaling that comprise the glomerular milieu.
Collapse
Affiliation(s)
- Kerstin Ebefors
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Emelie Lassén
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Nanditha Anandakrishnan
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Evren U Azeloglu
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ilse S Daehn
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
35
|
Abbas M, Moradi F, Hu W, Regudo KL, Osborne M, Pettipas J, Atallah DS, Hachem R, Ott-Peron N, Stuart JA. Vertebrate cell culture as an experimental approach – limitations and solutions. Comp Biochem Physiol B Biochem Mol Biol 2021; 254:110570. [DOI: 10.1016/j.cbpb.2021.110570] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 01/13/2021] [Accepted: 01/21/2021] [Indexed: 02/06/2023]
|
36
|
Zhou Z, Zhu J, Jiang M, Sang L, Hao K, He H. The Combination of Cell Cultured Technology and In Silico Model to Inform the Drug Development. Pharmaceutics 2021; 13:pharmaceutics13050704. [PMID: 34065907 PMCID: PMC8151315 DOI: 10.3390/pharmaceutics13050704] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/29/2021] [Accepted: 05/05/2021] [Indexed: 12/12/2022] Open
Abstract
Human-derived in vitro models can provide high-throughput efficacy and toxicity data without a species gap in drug development. Challenges are still encountered regarding the full utilisation of massive data in clinical settings. The lack of translated methods hinders the reliable prediction of clinical outcomes. Therefore, in this study, in silico models were proposed to tackle these obstacles from in vitro to in vivo translation, and the current major cell culture methods were introduced, such as human-induced pluripotent stem cells (hiPSCs), 3D cells, organoids, and microphysiological systems (MPS). Furthermore, the role and applications of several in silico models were summarised, including the physiologically based pharmacokinetic model (PBPK), pharmacokinetic/pharmacodynamic model (PK/PD), quantitative systems pharmacology model (QSP), and virtual clinical trials. These credible translation cases will provide templates for subsequent in vitro to in vivo translation. We believe that synergising high-quality in vitro data with existing models can better guide drug development and clinical use.
Collapse
Affiliation(s)
- Zhengying Zhou
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China; (Z.Z.); (M.J.)
| | - Jinwei Zhu
- State Key Laboratory of Natural Medicines, Jiangsu Province Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China; (J.Z.); (L.S.)
| | - Muhan Jiang
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China; (Z.Z.); (M.J.)
| | - Lan Sang
- State Key Laboratory of Natural Medicines, Jiangsu Province Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China; (J.Z.); (L.S.)
| | - Kun Hao
- State Key Laboratory of Natural Medicines, Jiangsu Province Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China; (J.Z.); (L.S.)
- Correspondence: (K.H.); (H.H.)
| | - Hua He
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China; (Z.Z.); (M.J.)
- Correspondence: (K.H.); (H.H.)
| |
Collapse
|
37
|
Namestnikov M, Pleniceanu O, Dekel B. Mixing Cells for Vascularized Kidney Regeneration. Cells 2021; 10:1119. [PMID: 34066487 PMCID: PMC8148539 DOI: 10.3390/cells10051119] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/26/2021] [Accepted: 05/01/2021] [Indexed: 02/06/2023] Open
Abstract
The worldwide rise in prevalence of chronic kidney disease (CKD) demands innovative bio-medical solutions for millions of kidney patients. Kidney regenerative medicine aims to replenish tissue which is lost due to a common pathological pathway of fibrosis/inflammation and rejuvenate remaining tissue to maintain sufficient kidney function. To this end, cellular therapy strategies devised so far utilize kidney tissue-forming cells (KTFCs) from various cell sources, fetal, adult, and pluripotent stem-cells (PSCs). However, to increase engraftment and potency of the transplanted cells in a harsh hypoxic diseased environment, it is of importance to co-transplant KTFCs with vessel forming cells (VFCs). VFCs, consisting of endothelial cells (ECs) and mesenchymal stem-cells (MSCs), synergize to generate stable blood vessels, facilitating the vascularization of self-organizing KTFCs into renovascular units. In this paper, we review the different sources of KTFCs and VFCs which can be mixed, and report recent advances made in the field of kidney regeneration with emphasis on generation of vascularized kidney tissue by cell transplantation.
Collapse
Affiliation(s)
- Michael Namestnikov
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel Hashomer, Ramat Gan 52621, Israel;
- Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel;
- ediatric Nephrology Division, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel Hashomer, Ramat Gan 52621, Israel;
| | - Oren Pleniceanu
- Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel;
- The Kidney Research Lab, Institute of Nephrology and Hypertension, Sheba Medical Center, Tel Hashomer, Ramat Gan 52621, Israel
| | - Benjamin Dekel
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel Hashomer, Ramat Gan 52621, Israel;
- Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel;
| |
Collapse
|
38
|
Zhao X, Xu Z, Xiao L, Shi T, Xiao H, Wang Y, Li Y, Xue F, Zeng W. Review on the Vascularization of Organoids and Organoids-on-a- Chip. Front Bioeng Biotechnol 2021; 9:637048. [PMID: 33912545 PMCID: PMC8072266 DOI: 10.3389/fbioe.2021.637048] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 03/04/2021] [Indexed: 11/16/2022] Open
Abstract
The use of human cells for the construction of 3D organ models in vitro based on cell self-assembly and engineering design has recently increased in popularity in the field of biological science. Although the organoids are able to simulate the structures and functions of organs in vitro, the 3D models have difficulty in forming a complex vascular network that can recreate the interaction between tissue and vascular systems. Therefore, organoids are unable to survive, due to the lack of oxygen and nutrients, as well as the accumulation of metabolic waste. Organoids-on-a-chip provides a more controllable and favorable design platform for co-culture of different cells and tissue types in organoid systems, overcoming some of the limitations present in organoid culture. However, the majority of them has vascular networks that are not adequately elaborate to simulate signal communications between bionic microenvironment (e.g., fluid shear force) and multiple organs. Here, we will review the technological progress of the vascularization in organoids and organoids-on-a-chip and the development of intravital 3D and 4D bioprinting as a new way for vascularization, which can aid in further study on tissue or organ development, disease research and regenerative medicine.
Collapse
Affiliation(s)
- Xingli Zhao
- Department of Cell Biology, Third Military Medical University, Chongqing, China
| | - Zilu Xu
- Department of Cell Biology, Third Military Medical University, Chongqing, China
| | - Lang Xiao
- Department of Cell Biology, Third Military Medical University, Chongqing, China
| | - Tuo Shi
- Department of Cell Biology, Third Military Medical University, Chongqing, China
| | - Haoran Xiao
- Department of Cell Biology, Third Military Medical University, Chongqing, China
| | - Yeqin Wang
- Department of Cell Biology, Third Military Medical University, Chongqing, China
| | - Yanzhao Li
- Department of Cell Biology, Third Military Medical University, Chongqing, China
| | - Fangchao Xue
- Department of Cell Biology, Third Military Medical University, Chongqing, China
| | - Wen Zeng
- Department of Cell Biology, Third Military Medical University, Chongqing, China.,State Key Laboratory of Trauma, Burn and Combined Injury, Chongqing, China.,Department of Neurology, Southwest Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
39
|
Shechtman LA, Piarowski CM, Scott JK, Golden EJ, Gaillard D, Barlow LA. Generation and Culture of Lingual Organoids Derived from Adult Mouse Taste Stem Cells. J Vis Exp 2021. [PMID: 33871462 DOI: 10.3791/62300] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The sense of taste is mediated by taste buds on the tongue, which are composed of rapidly renewing taste receptor cells (TRCs). This continual turnover is powered by local progenitor cells and renders taste function prone to disruption by a multitude of medical treatments, which in turn severely impacts the quality of life. Thus, studying this process in the context of drug treatment is vital to understanding if and how taste progenitor function and TRC production are affected. Given the ethical concerns and limited availability of human taste tissue, mouse models, which have a taste system similar to humans, are commonly used. Compared to in vivo methods, which are time-consuming, expensive, and not amenable to high throughput studies, murine lingual organoids can enable experiments to be run rapidly with many replicates and fewer mice. Here, previously published protocols have been adapted and a standardized method for generating taste organoids from taste progenitor cells isolated from the circumvallate papilla (CVP) of adult mice is presented. Taste progenitor cells in the CVP express LGR5 and can be isolated via EGFP fluorescence-activated cell sorting (FACS) from mice carrying an Lgr5EGFP-IRES-CreERT2 allele. Sorted cells are plated onto a matrix gel-based 3D culture system and cultured for 12 days. Organoids expand for the first 6 days of the culture period via proliferation and then enter a differentiation phase, during which they generate all three taste cell types along with non-taste epithelial cells. Organoids can be harvested upon maturation at day 12 or at any time during the growth process for RNA expression and immunohistochemical analysis. Standardizing culture methods for production of lingual organoids from adult stem cells will improve reproducibility and advance lingual organoids as a powerful drug screening tool in the fight to help patients experiencing taste dysfunction.
Collapse
Affiliation(s)
- Lauren A Shechtman
- Department of Cell and Developmental Biology and the Rocky Mountain Taste and Smell Center, University of Colorado Anschutz Medical Campus
| | - Christina M Piarowski
- Department of Cell and Developmental Biology and the Rocky Mountain Taste and Smell Center, University of Colorado Anschutz Medical Campus
| | - Jennifer K Scott
- Department of Cell and Developmental Biology and the Rocky Mountain Taste and Smell Center, University of Colorado Anschutz Medical Campus
| | - Erin J Golden
- Department of Cell and Developmental Biology and the Rocky Mountain Taste and Smell Center, University of Colorado Anschutz Medical Campus
| | - Dany Gaillard
- Department of Cell and Developmental Biology and the Rocky Mountain Taste and Smell Center, University of Colorado Anschutz Medical Campus;
| | - Linda A Barlow
- Department of Cell and Developmental Biology and the Rocky Mountain Taste and Smell Center, University of Colorado Anschutz Medical Campus;
| |
Collapse
|
40
|
Human reconstructed kidney models. In Vitro Cell Dev Biol Anim 2021; 57:133-147. [PMID: 33594607 DOI: 10.1007/s11626-021-00548-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 01/12/2021] [Indexed: 02/07/2023]
Abstract
The human kidney, which consists of up to 2 million nephrons, is critical for blood filtration, electrolyte balance, pH regulation, and fluid balance in the body. Animal experiments, particularly mice and rats, combined with advances in genetically modified technology have been the primary mechanism to study kidney injury in recent years. Mouse or rat kidneys, however, differ substantially from human kidneys at the anatomical, histological, and molecular levels. These differences combined with increased regulatory hurdles and shifting attitudes towards animal testing by non-specialists have led scientists to develop new and more relevant models of kidney injury. Although in vitro tissue culture studies are a valuable tool to study kidney injury and have yielded a great deal of insight, they are not a perfect model. Perhaps, the biggest limitation of tissue culture is that it cannot replicate the complex architecture, consisting of multiple cell types, of the kidney, and the interplay between these cells. Recent studies have found that pluripotent stem cells (PSCs), which are capable of differentiation into any cell type, can be used to generate kidney organoids. Organoids recapitulate the multicellular relationships and microenvironments of complex organs like kidney. Kidney organoids have been used to successfully model nephrotoxin-induced tubular and glomerular disease as well as complex diseases such as chronic kidney disease (CKD), which involves multiple cell types. In combination with genetic engineering techniques, such as CRISPR-Cas9, genetic diseases of the kidney can be reproduced in organoids. Thus, organoid models have the potential to predict drug toxicity and enhance drug discovery for human disease more accurately than animal models.
Collapse
|
41
|
Peelen DM, Hoogduijn MJ, Hesselink DA, Baan CC. Advanced in vitro Research Models to Study the Role of Endothelial Cells in Solid Organ Transplantation. Front Immunol 2021; 12:607953. [PMID: 33664744 PMCID: PMC7921837 DOI: 10.3389/fimmu.2021.607953] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 01/21/2021] [Indexed: 12/26/2022] Open
Abstract
The endothelium plays a key role in acute and chronic rejection of solid organ transplants. During both processes the endothelium is damaged often with major consequences for organ function. Also, endothelial cells (EC) have antigen-presenting properties and can in this manner initiate and enhance alloreactive immune responses. For decades, knowledge about these roles of EC have been obtained by studying both in vitro and in vivo models. These experimental models poorly imitate the immune response in patients and might explain why the discovery and development of agents that control EC responses is hampered. In recent years, various innovative human 3D in vitro models mimicking in vivo organ structure and function have been developed. These models will extend the knowledge about the diverse roles of EC in allograft rejection and will hopefully lead to discoveries of new targets that are involved in the interactions between the donor organ EC and the recipient's immune system. Moreover, these models can be used to gain a better insight in the mode of action of the currently prescribed immunosuppression and will enhance the development of novel therapeutics aiming to reduce allograft rejection and prolong graft survival.
Collapse
Affiliation(s)
- Daphne M Peelen
- Rotterdam Transplant Group, Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Martin J Hoogduijn
- Rotterdam Transplant Group, Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Dennis A Hesselink
- Rotterdam Transplant Group, Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Carla C Baan
- Rotterdam Transplant Group, Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC, Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
42
|
Olde Hanhof CJA, Yousef Yengej FA, Rookmaaker MB, Verhaar MC, van der Wijst J, Hoenderop JG. Modeling Distal Convoluted Tubule (Patho)Physiology: An Overview of Past Developments and an Outlook Toward the Future. Tissue Eng Part C Methods 2021; 27:200-212. [PMID: 33544049 DOI: 10.1089/ten.tec.2020.0345] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The kidneys are essential for maintaining electrolyte homeostasis. Blood electrolyte composition is controlled by active reabsorption and secretion processes in dedicated segments of the kidney tubule. Specifically, the distal convoluted tubule (DCT) and connecting tubule are important for regulating the final excretion of sodium, magnesium, and calcium. Studies unravelling the specific function of these segments have greatly improved our understanding of DCT (patho)physiology. Over the years, experimental models used to study the DCT have changed and the field has advanced from early dissection studies with rats and rabbits to the use of various transgenic mouse models. Developments in dissection techniques and cell culture methods have resulted in immortalized mouse DCT cell lines and made it possible to specifically obtain DCT fragments for ex vivo studies. However, we still do not fully understand the complex (patho)physiology of this segment and there is need for advanced human DCT models. Recently, kidney organoids and tubuloids have emerged as new complex cell models that provide excellent opportunities for physiological studies, disease modeling, drug discovery, and even personalized medicine in the future. This review presents an overview of cell models used to study the DCT and provides an outlook on kidney organoids and tubuloids as model for DCT (patho)physiology. Impact statement This study provides a detailed overview of past and future developments on cell models used to study kidney (patho)physiology and specifically the distal convoluted tubule (DCT) segment. Hereby, we highlight the need for an advanced human cell model of this segment and summarize recent advances in the field of kidney organoids and tubuloids with a focus on DCT properties. The findings reported in this review are significant for future developments toward an advanced human model of the DCT that will help to increase our understanding of DCT (patho)physiology.
Collapse
Affiliation(s)
- Charlotte J A Olde Hanhof
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Fjodor A Yousef Yengej
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands.,Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Maarten B Rookmaaker
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marianne C Verhaar
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jenny van der Wijst
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joost G Hoenderop
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
43
|
Sinenko SA, Ponomartsev SV, Tomilin AN. Pluripotent stem cell-based gene therapy approach: human de novo synthesized chromosomes. Cell Mol Life Sci 2021; 78:1207-1220. [PMID: 33011821 PMCID: PMC11072874 DOI: 10.1007/s00018-020-03653-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 09/14/2020] [Accepted: 09/22/2020] [Indexed: 02/06/2023]
Abstract
A novel approach in gene therapy was introduced 20 years ago since artificial non-integrative chromosome-based vectors containing gene loci size inserts were engineered. To date, different human artificial chromosomes (HAC) were generated with the use of de novo construction or "top-down" engineering approaches. The HAC-based therapeutic approach includes ex vivo gene transferring and correction of pluripotent stem cells (PSCs) or highly proliferative modified stem cells. The current progress in the technology of induced PSCs, integrating with the HAC technology, resulted in a novel platform of stem cell-based tissue replacement therapy for the treatment of genetic disease. Nowadays, the sophisticated and laborious HAC technology has significantly improved and is now closer to clinical studies. In here, we reviewed the achievements in the technology of de novo synthesized HACs for a chromosome transfer for developing gene therapy tissue replacement models of monogenic human diseases.
Collapse
Affiliation(s)
- Sergey A Sinenko
- Institute of Cytology, Russian Academy of Sciences, 4 Tikhoretsky Ave, St-Petersburg, 194064, Russia.
| | - Sergey V Ponomartsev
- Institute of Cytology, Russian Academy of Sciences, 4 Tikhoretsky Ave, St-Petersburg, 194064, Russia
| | - Alexey N Tomilin
- Institute of Cytology, Russian Academy of Sciences, 4 Tikhoretsky Ave, St-Petersburg, 194064, Russia.
- Institute of Translational Biomedicine, St-Petersburg State University, 7-9, Universitetskaya Emb, St-Petersburg, 199034, Russia.
| |
Collapse
|
44
|
Abstract
Complex multicellular life in mammals relies on functional cooperation of different organs for the survival of the whole organism. The kidneys play a critical part in this process through the maintenance of fluid volume and composition homeostasis, which enables other organs to fulfil their tasks. The renal endothelium exhibits phenotypic and molecular traits that distinguish it from endothelia of other organs. Moreover, the adult kidney vasculature comprises diverse populations of mostly quiescent, but not metabolically inactive, endothelial cells (ECs) that reside within the kidney glomeruli, cortex and medulla. Each of these populations supports specific functions, for example, in the filtration of blood plasma, the reabsorption and secretion of water and solutes, and the concentration of urine. Transcriptional profiling of these diverse EC populations suggests they have adapted to local microenvironmental conditions (hypoxia, shear stress, hyperosmolarity), enabling them to support kidney functions. Exposure of ECs to microenvironment-derived angiogenic factors affects their metabolism, and sustains kidney development and homeostasis, whereas EC-derived angiocrine factors preserve distinct microenvironment niches. In the context of kidney disease, renal ECs show alteration in their metabolism and phenotype in response to pathological changes in the local microenvironment, further promoting kidney dysfunction. Understanding the diversity and specialization of kidney ECs could provide new avenues for the treatment of kidney diseases and kidney regeneration.
Collapse
|
45
|
|
46
|
van den Berg CW, Koudijs A, Ritsma L, Rabelink TJ. In Vivo Assessment of Size-Selective Glomerular Sieving in Transplanted Human Induced Pluripotent Stem Cell-Derived Kidney Organoids. J Am Soc Nephrol 2020; 31:921-929. [PMID: 32354986 DOI: 10.1681/asn.2019060573] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 02/19/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND The utility of kidney organoids in regenerative medicine will rely on the functionality of the glomerular and tubular structures in these tissues. Recent studies have demonstrated the vascularization and subsequent maturation of human pluripotent stem cell-derived kidney organoids after renal subcapsular transplantation. This raises the question of whether the glomeruli also become functional upon transplantation. METHODS We transplanted kidney organoids under the renal capsule of the left kidney in immunodeficient mice followed by the implantation of a titanium imaging window on top of the kidney organoid. To assess glomerular function in the transplanted human pluripotent stem cell-derived kidney tissue 1, 2, and 3 weeks after transplantation, we applied high-resolution intravital multiphoton imaging through the imaging window during intravenous infusion of fluorescently labeled low and high molecular mass dextran molecules or albumin. RESULTS After vascularization, glomerular structures in the organoid displayed dextran and albumin size selectivity across their glomerular filtration barrier. We also observed evidence of proximal tubular dextran reuptake. CONCLUSIONS Our results demonstrate that human pluripotent stem cell-derived glomeruli can develop an appropriate barrier function and discriminate between molecules of varying size. These characteristics together with tubular presence of low molecular mass dextran provide clear evidence of functional filtration. This approach to visualizing glomerular filtration function will be instrumental for translation of organoid technology for clinical applications as well as for disease modeling.
Collapse
Affiliation(s)
- Cathelijne W van den Berg
- Department of Internal Medicine-Nephrology, Leiden University Medical Center, Leiden, The Netherlands .,Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Angela Koudijs
- Department of Internal Medicine-Nephrology, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Laila Ritsma
- Department of Cell and Chemical Biology, Cancer Genomics Centre Netherlands, Leiden University Medical Center, Leiden, The Netherlands
| | - Ton J Rabelink
- Department of Internal Medicine-Nephrology, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
47
|
Gupta✉ N, Dilmen E, Morizane R. 3D kidney organoids for bench-to-bedside translation. J Mol Med (Berl) 2020; 99:477-487. [PMID: 33034708 PMCID: PMC8026465 DOI: 10.1007/s00109-020-01983-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 06/30/2020] [Accepted: 09/22/2020] [Indexed: 12/23/2022]
Abstract
The kidneys are essential organs that filter the blood, removing urinary waste while maintaining fluid and electrolyte homeostasis. Current conventional research models such as static cell cultures and animal models are insufficient to grasp the complex human in vivo situation or lack translational value. To accelerate kidney research, novel research tools are required. Recent developments have allowed the directed differentiation of induced pluripotent stem cells to generate kidney organoids. Kidney organoids resemble the human kidney in vitro and can be applied in regenerative medicine and as developmental, toxicity, and disease models. Although current studies have shown great promise, challenges remain including the immaturity, limited reproducibility, and lack of perfusable vascular and collecting duct systems. This review gives an overview of our current understanding of nephrogenesis that enabled the generation of kidney organoids. Next, the potential applications of kidney organoids are discussed followed by future perspectives. This review proposes that advancement in kidney organoid research will be facilitated through our increasing knowledge on nephrogenesis and combining promising techniques such as organ-on-a-chip models.
Collapse
Affiliation(s)
- Navin Gupta✉
- Nephrology Division, Massachusetts General Hospital, Boston, MA USA
- Department of Medicine, Harvard Medical School, Boston, MA USA
- The Wyss Institute, Harvard University, Cambridge, MA USA
| | - Emre Dilmen
- Nephrology Division, Massachusetts General Hospital, Boston, MA USA
| | - Ryuji Morizane
- Nephrology Division, Massachusetts General Hospital, Boston, MA USA
- Department of Medicine, Harvard Medical School, Boston, MA USA
- The Wyss Institute, Harvard University, Cambridge, MA USA
- Harvard Stem Cell Institute, Cambridge, MA USA
| |
Collapse
|
48
|
Rajan A, Robertson MJ, Carter HE, Poole NM, Clark JR, Green SI, Criss ZK, Zhao B, Karandikar U, Xing Y, Margalef-Català M, Jain N, Wilson RL, Bai F, Hyser JM, Petrosino J, Shroyer NF, Blutt SE, Coarfa C, Song X, Prasad BVV, Amieva MR, Grande-Allen J, Estes MK, Okhuysen PC, Maresso AW. Enteroaggregative E. coli Adherence to Human Heparan Sulfate Proteoglycans Drives Segment and Host Specific Responses to Infection. PLoS Pathog 2020; 16:e1008851. [PMID: 32986782 PMCID: PMC7553275 DOI: 10.1371/journal.ppat.1008851] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 10/13/2020] [Accepted: 08/01/2020] [Indexed: 02/06/2023] Open
Abstract
Enteroaggregative Escherichia coli (EAEC) is a significant cause of acute and chronic diarrhea, foodborne outbreaks, infections of the immunocompromised, and growth stunting in children in developing nations. There is no vaccine and resistance to antibiotics is rising. Unlike related E. coli pathotypes that are often associated with acute bouts of infection, EAEC is associated with persistent diarrhea and subclinical long-term colonization. Several secreted virulence factors have been associated with EAEC pathogenesis and linked to disease in humans, less certain are the molecular drivers of adherence to the intestinal mucosa. We previously established human intestinal enteroids (HIEs) as a model system to study host-EAEC interactions and aggregative adherence fimbriae A (AafA) as a major driver of EAEC adherence to HIEs. Here, we report a large-scale assessment of the host response to EAEC adherence from all four segments of the intestine across at least three donor lines for five E. coli pathotypes. The data demonstrate that the host response in the duodenum is driven largely by the infecting pathotype, whereas the response in the colon diverges in a patient-specific manner. Major pathways altered in gene expression in each of the four enteroid segments differed dramatically, with responses observed for inflammation, apoptosis and an overwhelming response to different mucin genes. In particular, EAEC both associated with large mucus droplets and specific mucins at the epithelial surface, binding that was ameliorated when mucins were removed, a process dependent on AafA. Pan-screening for glycans for binding to purified AafA identified the human ligand as heparan sulfate proteoglycans (HSPGs). Removal of HSPG abrogated EAEC association with HIEs. These results may mean that the human intestine responds remarkably different to distinct pathobionts that is dependent on the both the individual and intestinal segment in question, and uncover a major role for surface heparan sulfate proteoglycans as tropism-driving factor in adherence and/or colonization.
Collapse
Affiliation(s)
- Anubama Rajan
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Matthew J. Robertson
- Molecular and Cell Biology-Mol. Regulation, Baylor College of Medicine, Houston, TX, United States of America
| | - Hannah E. Carter
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Nina M. Poole
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Justin R. Clark
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Sabrina I. Green
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Zachary K. Criss
- Department of Medicine Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, TX, United States of America
| | - Boyang Zhao
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, United States of America
| | - Umesh Karandikar
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Yikun Xing
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Mar Margalef-Català
- Department of Pediatrics, Division of Infectious Diseases, Stanford University, Stanford, CA, United States of America
| | - Nikhil Jain
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, United States of America
| | - Reid L. Wilson
- Department of Bioengineering, Rice University, Houston, TX, United States of America
| | - Fan Bai
- Department of Biochemistry, Emory Comprehensive Glycomics Core, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Joseph M. Hyser
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Joseph Petrosino
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Noah F. Shroyer
- Department of Medicine Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, TX, United States of America
| | - Sarah E. Blutt
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Cristian Coarfa
- Molecular and Cell Biology-Mol. Regulation, Baylor College of Medicine, Houston, TX, United States of America
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States of America
| | - Xuezheng Song
- Department of Biochemistry, Emory Comprehensive Glycomics Core, Emory University School of Medicine, Atlanta, GA, United States of America
| | - BV Venkataram Prasad
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, United States of America
| | - Manuel R. Amieva
- Department of Pediatrics, Division of Infectious Diseases, Stanford University, Stanford, CA, United States of America
| | - Jane Grande-Allen
- Department of Bioengineering, Rice University, Houston, TX, United States of America
| | - Mary K. Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Pablo C. Okhuysen
- Department of Infectious Diseases, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Anthony W. Maresso
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| |
Collapse
|
49
|
Yousef Yengej FA, Jansen J, Rookmaaker MB, Verhaar MC, Clevers H. Kidney Organoids and Tubuloids. Cells 2020; 9:E1326. [PMID: 32466429 PMCID: PMC7349753 DOI: 10.3390/cells9061326] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/18/2020] [Accepted: 05/23/2020] [Indexed: 02/07/2023] Open
Abstract
In the past five years, pluripotent stem cell (PSC)-derived kidney organoids and adult stem or progenitor cell (ASC)-based kidney tubuloids have emerged as advanced in vitro models of kidney development, physiology, and disease. PSC-derived organoids mimic nephrogenesis. After differentiation towards the kidney precursor tissues ureteric bud and metanephric mesenchyme, their reciprocal interaction causes self-organization and patterning in vitro to generate nephron structures that resemble the fetal kidney. ASC tubuloids on the other hand recapitulate renewal and repair in the adult kidney tubule and give rise to long-term expandable and genetically stable cultures that consist of adult proximal tubule, loop of Henle, distal tubule, and collecting duct epithelium. Both organoid types hold great potential for: (1) studies of kidney physiology, (2) disease modeling, (3) high-throughput screening for drug efficacy and toxicity, and (4) regenerative medicine. Currently, organoids and tubuloids are successfully used to model hereditary, infectious, toxic, metabolic, and malignant kidney diseases and to screen for effective therapies. Furthermore, a tumor tubuloid biobank was established, which allows studies of pathogenic mutations and novel drug targets in a large group of patients. In this review, we discuss the nature of kidney organoids and tubuloids and their current and future applications in science and medicine.
Collapse
Affiliation(s)
- Fjodor A. Yousef Yengej
- Hubrecht Institute—Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands;
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (M.B.R.); (M.C.V.)
| | - Jitske Jansen
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 24, 6500 HB Nijmegen, The Netherlands;
- Department of Pediatric Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Amalia Children’s Hospital, Geert Grooteplein 24, 6500 HB Nijmegen, The Netherlands
| | - Maarten B. Rookmaaker
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (M.B.R.); (M.C.V.)
| | - Marianne C. Verhaar
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (M.B.R.); (M.C.V.)
| | - Hans Clevers
- Hubrecht Institute—Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands;
| |
Collapse
|