1
|
Bahrami M, Abbaszadeh HA, Norouzian M, Abdollahifar MA, Roozbahany NA, Saber M, Azimi M, Ehsani E, Bakhtiyari M, Serra AL, Moghadasali R. Enriched human embryonic stem cells-derived CD133 +, CD24 + renal progenitors engraft and restore function in a gentamicin-induced kidney injury in mice. Regen Ther 2024; 27:506-518. [PMID: 38745839 PMCID: PMC11091464 DOI: 10.1016/j.reth.2024.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 03/30/2024] [Accepted: 04/25/2024] [Indexed: 05/16/2024] Open
Abstract
Introduction Acute kidney injury (AKI) is a common health problem that leads to high morbidity and potential mortality. The failure of conventional treatments to improve forms of this condition highlights the need for innovative and effective treatment approaches. Regenerative therapies with Renal Progenitor Cells (RPCs) have been proposed as a promising new strategy. A growing body of evidence suggests that progenitor cells differentiated from different sources, including human embryonic stem cells (hESCs), can effectively treat AKI. Methods Here, we describe a method for generating RPCs and directed human Embryoid Bodies (EBs) towards CD133+CD24+ renal progenitor cells and evaluate their functional activity in alleviating AKI. Results The obtained results show that hESCs-derived CD133+CD24+ RPCs can engraft into damaged renal tubules and restore renal function and structure in mice with gentamicin-induced kidney injury, and significantly decrease blood urea nitrogen levels, suppress oxidative stress and inflammation, and attenuate histopathological disturbances, including tubular necrosis, tubular dilation, urinary casts, and interstitial fibrosis. Conclusion The results suggest that RPCs have a promising regenerative potential in improving renal disease and can lay the foundation for future cell therapy and disease modeling.
Collapse
Affiliation(s)
- Maryam Bahrami
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Laser Applications in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hojjat Allah Abbaszadeh
- Laser Applications in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Norouzian
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad-Amin Abdollahifar
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Navid Ahmady Roozbahany
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Private Practice, Bradford ON, Canada
| | - Maryam Saber
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Masoumeh Azimi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Ehsan Ehsani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Biology, Roudehen Branch, Islamic Azad University, Roudehen, Iran
| | - Mohsen Bakhtiyari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Andreas L. Serra
- Department of Internal Medicine and Nephrology, Klinik Hirslanden, Zurich, Switzerland
| | - Reza Moghadasali
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
2
|
Long HY, Qian ZP, Lan Q, Xu YJ, Da JJ, Yu FX, Zha Y. Human pluripotent stem cell-derived kidney organoids: Current progress and challenges. World J Stem Cells 2024; 16:114-125. [PMID: 38455108 PMCID: PMC10915962 DOI: 10.4252/wjsc.v16.i2.114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/18/2023] [Accepted: 01/29/2024] [Indexed: 02/26/2024] Open
Abstract
Human pluripotent stem cell (hPSC)-derived kidney organoids share similarities with the fetal kidney. However, the current hPSC-derived kidney organoids have some limitations, including the inability to perform nephrogenesis and lack of a corticomedullary definition, uniform vascular system, and coordinated exit pathway for urinary filtrate. Therefore, further studies are required to produce hPSC-derived kidney organoids that accurately mimic human kidneys to facilitate research on kidney development, regeneration, disease modeling, and drug screening. In this review, we discussed recent advances in the generation of hPSC-derived kidney organoids, how these organoids contribute to the understanding of human kidney development and research in disease modeling. Additionally, the limitations, future research focus, and applications of hPSC-derived kidney organoids were highlighted.
Collapse
Affiliation(s)
- Hong-Yan Long
- Graduate School, Zunyi Medical University, Zunyi 563000, Guizhou Province, China
| | - Zu-Ping Qian
- Graduate School, Zunyi Medical University, Zunyi 563000, Guizhou Province, China
| | - Qin Lan
- Graduate School, Zunyi Medical University, Zunyi 563000, Guizhou Province, China
| | - Yong-Jie Xu
- Department of Laboratory Medicine, Guizhou Provincial People's Hospital, Guiyang 550002, Guizhou Province, China
| | - Jing-Jing Da
- Department of Nephrology, Guizhou Provincial People's Hospital, Guiyang 550002, Guizhou Province, China
| | - Fu-Xun Yu
- Key Laboratory of Diagnosis and Treatment of Pulmonary Immune Diseases, National Health Commission, Guizhou Provincial People's Hospital, Guiyang 550002, Guizhou Province, China
| | - Yan Zha
- Graduate School, Zunyi Medical University, Zunyi 563000, Guizhou Province, China
- Department of Nephrology, Guizhou Provincial People's Hospital, Guiyang 550002, Guizhou Province, China.
| |
Collapse
|
3
|
Lacueva-Aparicio A, Lindoso RS, Mihăilă SM, Giménez I. Role of extracellular matrix components and structure in new renal models in vitro. Front Physiol 2022; 13:1048738. [PMID: 36569770 PMCID: PMC9767975 DOI: 10.3389/fphys.2022.1048738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 10/31/2022] [Indexed: 12/12/2022] Open
Abstract
The extracellular matrix (ECM), a complex set of fibrillar proteins and proteoglycans, supports the renal parenchyma and provides biomechanical and biochemical cues critical for spatial-temporal patterning of cell development and acquisition of specialized functions. As in vitro models progress towards biomimicry, more attention is paid to reproducing ECM-mediated stimuli. ECM's role in in vitro models of renal function and disease used to investigate kidney injury and regeneration is discussed. Availability, affordability, and lot-to-lot consistency are the main factors determining the selection of materials to recreate ECM in vitro. While simpler components can be synthesized in vitro, others must be isolated from animal or human tissues, either as single isolated components or as complex mixtures, such as Matrigel or decellularized formulations. Synthetic polymeric materials with dynamic and instructive capacities are also being explored for cell mechanical support to overcome the issues with natural products. ECM components can be used as simple 2D coatings or complex 3D scaffolds combining natural and synthetic materials. The goal is to recreate the biochemical signals provided by glycosaminoglycans and other signaling molecules, together with the stiffness, elasticity, segmentation, and dimensionality of the original kidney tissue, to support the specialized functions of glomerular, tubular, and vascular compartments. ECM mimicking also plays a central role in recent developments aiming to reproduce renal tissue in vitro or even in therapeutical strategies to regenerate renal function. Bioprinting of renal tubules, recellularization of kidney ECM scaffolds, and development of kidney organoids are examples. Future solutions will probably combine these technologies.
Collapse
Affiliation(s)
- Alodia Lacueva-Aparicio
- Renal and Cardiovascular Physiopathology (FISIOPREN), Aragon’s Health Sciences Institute, Zaragoza, Spain,Tissue Microenvironment Lab (TME Lab), I3A, University of Zaragoza, Zaragoza, Spain
| | - Rafael Soares Lindoso
- Carlos Chagas Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Silvia M. Mihăilă
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Ignacio Giménez
- Renal and Cardiovascular Physiopathology (FISIOPREN), Aragon’s Health Sciences Institute, Zaragoza, Spain,Institute for Health Research Aragon (IIS Aragon), Zaragoza, Spain,School of Medicine, University of Zaragoza, Zaragoza, Spain,*Correspondence: Ignacio Giménez,
| |
Collapse
|
4
|
Khelifi G, Chow T, Whiteley J, Fort V, Humphreys BD, Hussein SM, Rogers IM. Determining epigenetic memory in kidney proximal tubule cell derived induced pluripotent stem cells using a quadruple transgenic reprogrammable mouse. Sci Rep 2022; 12:20340. [PMID: 36434072 PMCID: PMC9700797 DOI: 10.1038/s41598-022-24581-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 11/17/2022] [Indexed: 11/27/2022] Open
Abstract
The majority of nucleated somatic cells can be reprogrammed to induced pluripotent stem cells (iPSCs). The process of reprogramming involves epigenetic remodelling to turn on pluripotency-associated genes and turn off lineage-specific genes. Some evidence shows that iPSCs retain epigenetic marks of their cell of origin and this "epigenetic memory" influences their differentiation potential, with a preference towards their cell of origin. Here, we reprogrammed proximal tubule cells (PTC) and tail tip fibroblasts (TTF), from a reprogrammable mouse to iPSCs and differentiated the iPSCs to renal progenitors to understand if epigenetic memory plays a role in renal differentiation. This model allowed us to eliminate experimental variability due to donor genetic differences and transfection of the reprogramming factors such as copy number and integration site. In this study we demonstrated that early passage PTC iPSCs and TTF iPSCs expressed low levels of renal progenitor genes and high levels of pluripotency-associated genes, and the transcriptional levels of these genes were not significantly different between PTC iPSCs and TTF iPSCs. We used ChIP-seq of H3K4me3, H3K27me3, H3K36me3 and global DNA methylation profiles of PTC iPSCs and TTF iPSCs to demonstrate that global epigenetic marks were not different between the cells from the two different sets of tissue samples. There were also no epigenetic differences observed when kidney developmental genes and pluripotency-associated genes were closely examined. We did observe that during differentiation to renal progenitor cells the PTC iPSC-derived renal cells expressed higher levels of three renal progenitor genes compared to progenitors derived from TTF iPSCs but the underlying DNA methylation and histone methylation patterns did not suggest an epigenetic memory basis for this.
Collapse
Affiliation(s)
- Gabriel Khelifi
- grid.23856.3a0000 0004 1936 8390Cancer Research Center, Université Laval, Quebec City, QC Canada ,grid.411081.d0000 0000 9471 1794Oncology Division, CHU of Québec-Université Laval Research Center, Quebec City, QC Canada
| | - Theresa Chow
- grid.250674.20000 0004 0626 6184Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Physiology, University of Toronto, Toronto, ON Canada
| | - Jennifer Whiteley
- grid.250674.20000 0004 0626 6184Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON Canada
| | - Victoire Fort
- grid.23856.3a0000 0004 1936 8390Cancer Research Center, Université Laval, Quebec City, QC Canada ,grid.411081.d0000 0000 9471 1794Oncology Division, CHU of Québec-Université Laval Research Center, Quebec City, QC Canada
| | - Benjamin D. Humphreys
- grid.4367.60000 0001 2355 7002Division of Nephrology, Department of Medicine, Department of Developmental Biology, Washington University in St. Louis School of Medicine, St. Louis, MO USA
| | - Samer M.I. Hussein
- grid.23856.3a0000 0004 1936 8390Cancer Research Center, Université Laval, Quebec City, QC Canada ,grid.411081.d0000 0000 9471 1794Oncology Division, CHU of Québec-Université Laval Research Center, Quebec City, QC Canada
| | - Ian M. Rogers
- grid.250674.20000 0004 0626 6184Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Physiology, University of Toronto, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Obstetrics and Gynaecology, University of Toronto, Toronto, ON Canada ,grid.231844.80000 0004 0474 0428Ajmera Transplant Center, UHN, Toronto, Canada
| |
Collapse
|
5
|
Miranda CC, Gomes MR, Moço M, Cabral JMS, Ferreira FC, Sanjuan-Alberte P. A Concise Review on Electrospun Scaffolds for Kidney Tissue Engineering. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 9:bioengineering9100554. [PMID: 36290522 PMCID: PMC9598616 DOI: 10.3390/bioengineering9100554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/06/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022]
Abstract
Chronic kidney disease is one of the deadliest diseases globally and treatment methods are still insufficient, relying mostly on transplantation and dialysis. Engineering of kidney tissues in vitro from induced pluripotent stem cells (iPSCs) could provide a solution to this medical need by restoring the function of damaged kidneys. However, implementation of such approaches is still challenging to achieve due to the complexity of mature kidneys in vivo. Several strategies have been defined to obtain kidney progenitor endothelial and epithelial cells that could form nephrons and proximal tube cells, but these lack tissue maturity and vascularisation to be further implemented. Electrospinning is a technique that has shown promise in the development of physiological microenvironments of several tissues and could be applied in the engineering of kidney tissues. Synthetic polymers such as polycaprolactone, polylactic acid, and poly(vinyl alcohol) have been explored in the manufacturing of fibres that align and promote the proliferation and cell-to-cell interactions of kidney cells. Natural polymers including silk fibroin and decellularised extracellular matrix have also been explored alone and in combination with synthetic polymers promoting the differentiation of podocytes and tubular-specific cells. Despite these attempts, further work is still required to advance the applications of electrospun fibres in kidney tissue engineering and explore this technique in combination with other manufacturing methods such as bioprinting to develop more organised, mature and reproducible kidney organoids.
Collapse
Affiliation(s)
- Cláudia C. Miranda
- Department of Bioengineering, Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Correspondence: (C.C.M.); (P.S.-A.)
| | - Mariana Ramalho Gomes
- Department of Bioengineering, Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Mariana Moço
- Department of Bioengineering, Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Joaquim M. S. Cabral
- Department of Bioengineering, Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Frederico Castelo Ferreira
- Department of Bioengineering, Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Paola Sanjuan-Alberte
- Department of Bioengineering, Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Correspondence: (C.C.M.); (P.S.-A.)
| |
Collapse
|
6
|
Chen H, Zhang W, Maskey N, Yang F, Zheng Z, Li C, Wang R, Wu P, Mao S, Zhang J, Yan Y, Li W, Yao X. Urological cancer organoids, patients' avatars for precision medicine: past, present and future. Cell Biosci 2022; 12:132. [PMID: 35986387 PMCID: PMC9389738 DOI: 10.1186/s13578-022-00866-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 07/31/2022] [Indexed: 11/29/2022] Open
Abstract
Urological cancers are common malignant cancers worldwide, with annually increasing morbidity and mortality rates. For decades, two-dimensional cell cultures and animal models have been widely used to study the development and underlying molecular mechanisms of urological cancers. However, they either fail to reflect cancer heterogeneity or are time-consuming and labour-intensive. The recent emergence of a three-dimensional culture model called organoid has the potential to overcome the shortcomings of traditional models. For example, organoids can recapitulate the histopathological and molecular diversity of original cancer and reflect the interaction between cancer and surrounding cells or stroma by simulating tumour microenvironments. Emerging evidence suggests that urine-derived organoids can be generated, which could be a novel non-invasive liquid biopsy method that provides new ideas for clinical precision therapy. However, the current research on organoids has encountered some bottlenecks, such as the lack of a standard culture process, the need to optimize the culture medium and the inability to completely simulate the immune system in vivo. Nonetheless, cell co-culture and organoid-on-a-chip have significant potential to solve these problems. In this review, the latest applications of organoids in drug screening, cancer origin investigation and combined single-cell sequencing are illustrated. Furthermore, the development and application of organoids in urological cancers and their challenges are summarised.
Collapse
|
7
|
Bejoy J, Qian ES, Woodard LE. Tissue Culture Models of AKI: From Tubule Cells to Human Kidney Organoids. J Am Soc Nephrol 2022; 33:487-501. [PMID: 35031569 PMCID: PMC8975068 DOI: 10.1681/asn.2021050693] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
AKI affects approximately 13.3 million people around the world each year, causing CKD and/or mortality. The mammalian kidney cannot generate new nephrons after postnatal renal damage and regenerative therapies for AKI are not available. Human kidney tissue culture systems can complement animal models of AKI and/or address some of their limitations. Donor-derived somatic cells, such as renal tubule epithelial cells or cell lines (RPTEC/hTERT, ciPTEC, HK-2, Nki-2, and CIHP-1), have been used for decades to permit drug toxicity screening and studies into potential AKI mechanisms. However, tubule cell lines do not fully recapitulate tubular epithelial cell properties in situ when grown under classic tissue culture conditions. Improving tissue culture models of AKI would increase our understanding of the mechanisms, leading to new therapeutics. Human pluripotent stem cells (hPSCs) can be differentiated into kidney organoids and various renal cell types. Injury to human kidney organoids results in renal cell-type crosstalk and upregulation of kidney injury biomarkers that are difficult to induce in primary tubule cell cultures. However, current protocols produce kidney organoids that are not mature and contain off-target cell types. Promising bioengineering techniques, such as bioprinting and "kidney-on-a-chip" methods, as applied to kidney nephrotoxicity modeling advantages and limitations are discussed. This review explores the mechanisms and detection of AKI in tissue culture, with an emphasis on bioengineered approaches such as human kidney organoid models.
Collapse
Affiliation(s)
- Julie Bejoy
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Eddie S. Qian
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Lauren E. Woodard
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
8
|
HEK293-Conditioned Medium Altered the Expression of Renal Markers WT1, CD2AP, and CDH16 in the Human Adipose Mesenchymal Stem Cells. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2022. [DOI: 10.1007/s40883-021-00246-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
9
|
Kidney development to kidney organoids and back again. Semin Cell Dev Biol 2021; 127:68-76. [PMID: 34627669 DOI: 10.1016/j.semcdb.2021.09.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/01/2021] [Accepted: 09/28/2021] [Indexed: 12/14/2022]
Abstract
Kidney organoid technology has led to a renaissance in kidney developmental biology. The complex underpinnings of mammalian kidney development have provided a framework for the generation of kidney cells and tissues from human pluripotent stem cells. Termed kidney organoids, these 3-dimensional structures contain kidney-specific cell types distributed similarly to in vivo architecture. The adult human kidney forms from the reciprocal induction of two disparate tissues, the metanephric mesenchyme (MM) and ureteric bud (UB), to form nephrons and collecting ducts, respectively. Although nephrons and collecting ducts are derived from the intermediate mesoderm (IM), their development deviates in time and space to impart distinctive inductive signaling for which separate differentiation protocols are required. Here we summarize the directed differentiation protocols which generate nephron kidney organoids and collecting duct kidney organoids, making note of similarities as much as differences. We discuss limitations of these present approaches and discuss future directions to improve kidney organoid technology, including a greater understanding of anterior IM and its derivatives to enable an improved differentiation protocol to collecting duct organoids for which historic and future developmental biology studies will be instrumental.
Collapse
|
10
|
Liu D, Zheng W, Pan S, Liu Z. Concise review: current trends on applications of stem cells in diabetic nephropathy. Cell Death Dis 2020; 11:1000. [PMID: 33221823 PMCID: PMC7680458 DOI: 10.1038/s41419-020-03206-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/27/2020] [Accepted: 10/27/2020] [Indexed: 12/15/2022]
Abstract
Diabetic nephropathy, with high prevalence, is the main cause of renal failure in diabetic patients. The strategies for treating DN are limited with not only high cost but an unsatisfied effect. Therefore, the effective treatment of DN needs to be explored urgently. In recent years, due to their self-renewal ability and multi-directional differentiation potential, stem cells have exerted therapeutic effects in many diseases, such as graft-versus-host disease, autoimmune diseases, pancreatic diseases, and even acute kidney injury. With the development of stem cell technology, stem cell-based regenerative medicine has been tried to be applied to the treatment of DN. Related stem cells include embryonic stem cells, induced pluripotent stem cells, mesenchymal cells, and endothelial progenitor cells. Undoubtedly, stem cell transplantation has achieved certain results in the treatment of DN animal models. However, stem cell therapy still remains certain thorny issues during treatment. For instance, poor engraftment and limited differentiation of stem cells caused by the diabetic microenvironment, differentiation into unwanted cell lineages, and malignant transformation or genetic aberrations of stem cells. At present, various researches on the therapeutic effects of stem cells in DN with different opinions are reported and the specific mechanism of stem cells is still unclear. We review here the potential mechanism of stem cells as new therapeutic agents in the treatment of DN. Also, we review recent findings and updated information about not only the utilization of stem cells on DN in both preclinical and clinical trials but limitations and future expectations of stem cell-based therapy for DN.
Collapse
Affiliation(s)
- Dongwei Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P.R. China.,Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, P.R. China.,Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P.R. China.,Core Unit of National Clinical Medical Research Center of Kidney Disease, Zhengzhou, 450052, P.R. China
| | - Wen Zheng
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P.R. China.,Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, P.R. China.,Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P.R. China.,Core Unit of National Clinical Medical Research Center of Kidney Disease, Zhengzhou, 450052, P.R. China
| | - Shaokang Pan
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P.R. China.,Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, P.R. China.,Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P.R. China.,Core Unit of National Clinical Medical Research Center of Kidney Disease, Zhengzhou, 450052, P.R. China
| | - Zhangsuo Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P.R. China. .,Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, P.R. China. .,Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P.R. China. .,Core Unit of National Clinical Medical Research Center of Kidney Disease, Zhengzhou, 450052, P.R. China.
| |
Collapse
|
11
|
de Carvalho Ribeiro P, Oliveira LF, Filho MA, Caldas HC. Differentiating Induced Pluripotent Stem Cells into Renal Cells: A New Approach to Treat Kidney Diseases. Stem Cells Int 2020; 2020:8894590. [PMID: 32831854 PMCID: PMC7428838 DOI: 10.1155/2020/8894590] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/21/2020] [Accepted: 07/28/2020] [Indexed: 12/16/2022] Open
Abstract
Renal disease is a major issue for global public health. Despite some progress in supportive care, the mortality rates among patients with this condition remain alarmingly high. Studies in pursuit of innovative strategies to treat renal diseases, especially stimulating kidney regeneration, have been developed. In this field, stem cell-based therapy has been a promising area. Induced pluripotent stem cell-derived renal cells (iPSC-RCs) represent an interesting source of cells for treating kidney diseases. Advances in regenerative medicine using iPSC-RCs and their application to the kidney are discussed in this review. Furthermore, the way differentiation protocols of induced pluripotent stem cells into renal cells may also be applied for the generation of kidney organoids is also described, contributing to studies in renal development, kidney diseases, and drug toxicity tests. The translation of the differentiation methodologies into animal model studies and the safety and feasibility of renal differentiated cells as a treatment for kidney injury are also highlighted. Although only few studies were published in this field, the results seem promising and support the use of iPSC-RCs as a potential therapy in the future.
Collapse
Affiliation(s)
- Patrícia de Carvalho Ribeiro
- Laboratory of Immunology and Experimental Transplantation-LITEX, Medical School of Sao Jose do Rio Preto, Sao Jose do Rio Preto, Sao Paulo, Brazil
| | - Lucas Felipe Oliveira
- Physiology Division, Natural and Biological Sciences Institute, Triangulo Mineiro Federal University, Uberaba, Minas Gerais, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mario Abbud Filho
- Laboratory of Immunology and Experimental Transplantation-LITEX, Medical School of Sao Jose do Rio Preto, Sao Jose do Rio Preto, Sao Paulo, Brazil
- Kidney Transplant Unit, Hospital de Base, FAMERP/FUNFARME, Sao Jose do Rio Preto, Sao Paulo, Brazil
- Urology and Nephrology Institute, Sao Jose Rio Preto, Sao Paulo, Brazil
| | - Heloisa Cristina Caldas
- Laboratory of Immunology and Experimental Transplantation-LITEX, Medical School of Sao Jose do Rio Preto, Sao Jose do Rio Preto, Sao Paulo, Brazil
- Kidney Transplant Unit, Hospital de Base, FAMERP/FUNFARME, Sao Jose do Rio Preto, Sao Paulo, Brazil
| |
Collapse
|
12
|
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
|
13
|
Chow T, Wong FTM, Monetti C, Nagy A, Cox B, Rogers IM. Recapitulating kidney development in vitro by priming and differentiating mouse embryonic stem cells in monolayers. NPJ Regen Med 2020; 5:7. [PMID: 32351711 PMCID: PMC7171095 DOI: 10.1038/s41536-020-0092-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 03/13/2020] [Indexed: 12/26/2022] Open
Abstract
In order to harness the potential of pluripotent stem cells, we need to understand how to differentiate them to our target cell types. Here, we developed a protocol to differentiate mouse embryonic stem cells (ESCs) to renal progenitors in a step-wise manner. Microarrays were used to track the transcriptional changes at each stage of differentiation and we observed that genes associated with metanephros, ureteric bud, and blood vessel development were significantly upregulated as the cells differentiated towards renal progenitors. Priming the ESCs and optimizing seeding cell density and growth factor concentrations helped improve differentiation efficiency. Organoids were used to determine the developmental potential of the renal progenitor cells. Aggregated renal progenitors gave rise to organoids consisting of LTL+/E-cadherin+ proximal tubules, cytokeratin+ ureteric bud-derived tubules, and extracellular matrix proteins secreted by the cells themselves. Over-expression of key kidney developmental genes, Pax2, Six1, Eya1, and Hox11 paralogs, during differentiation did not improve differentiation efficiency. Altogether, we developed a protocol to differentiate mouse ESCs in a manner that recapitulates embryonic kidney development and showed that precise gene regulation is essential for proper differentiation to occur.
Collapse
Affiliation(s)
- Theresa Chow
- 1Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON Canada.,2Department of Physiology, University of Toronto, Toronto, ON Canada
| | - Frances T M Wong
- 2Department of Physiology, University of Toronto, Toronto, ON Canada
| | - Claudio Monetti
- 1Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON Canada
| | - Andras Nagy
- 1Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON Canada.,3Department of Obstetrics and Gynaecology, University of Toronto, Toronto, ON Canada.,4Institute of Medical Science, University of Toronto, Toronto, ON Canada
| | - Brian Cox
- 2Department of Physiology, University of Toronto, Toronto, ON Canada.,3Department of Obstetrics and Gynaecology, University of Toronto, Toronto, ON Canada
| | - Ian M Rogers
- 1Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON Canada.,2Department of Physiology, University of Toronto, Toronto, ON Canada.,3Department of Obstetrics and Gynaecology, University of Toronto, Toronto, ON Canada
| |
Collapse
|
14
|
Ahmadi A, Moghadasali R, Ezzatizadeh V, Taghizadeh Z, Nassiri SM, Asghari-Vostikolaee MH, Alikhani M, Hadi F, Rahbarghazi R, Yazdi RS, Baharvand H, Aghdami N. Transplantation of Mouse Induced Pluripotent Stem Cell-Derived Podocytes in a Mouse Model of Membranous Nephropathy Attenuates Proteinuria. Sci Rep 2019; 9:15467. [PMID: 31664077 PMCID: PMC6820764 DOI: 10.1038/s41598-019-51770-0] [Citation(s) in RCA: 8] [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: 03/21/2019] [Accepted: 09/26/2019] [Indexed: 12/31/2022] Open
Abstract
Injury to podocytes is a principle cause of initiation and progression of both immune and non-immune mediated glomerular diseases that result in proteinuria and decreased function of the kidney. Current advances in regenerative medicine shed light on the therapeutic potential of cell-based strategies for treatment of such disorders. Thus, there is hope that generation and transplantation of podocytes from induced pluripotent stem cells (iPSCs), could potentially be used as a curative treatment for glomerulonephritis caused by podocytes injury and loss. Despite several reports on the generation of iPSC-derived podocytes, there are rare reports about successful use of these cells in animal models. In this study, we first generated a model of anti-podocyte antibody-induced heavy proteinuria that resembled human membranous nephropathy and was characterized by the presence of sub-epithelial immune deposits and podocytes loss. Thereafter, we showed that transplantation of functional iPSC-derived podocytes following podocytes depletion results in recruitment of iPSC-derived podocytes within the damaged glomerulus, and leads to attenuation of proteinuria and histological alterations. These results provided evidence that application of iPSCs-derived renal cells could be a possible therapeutic strategy to favorably influence glomerular diseases outcomes.
Collapse
Affiliation(s)
- Amin Ahmadi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Reza Moghadasali
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Vahid Ezzatizadeh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Medical Genetics Department, Medical Laboratory Center, Royesh Medical Group, Tehran, Iran
| | - Zeinab Taghizadeh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Seyed Mahdi Nassiri
- Department of Clinical Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | | | - Mehdi Alikhani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Fatemeh Hadi
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Salman Yazdi
- Department of Andrology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
- Department of Developmental Biology, University of Science and Culture, Tehran, Iran.
| | - Nasser Aghdami
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
15
|
Abstract
The worldwide increase in the number of patients with end-stage renal disease leads to a growing waiting list for kidney transplantation resulting from the scarcity of kidney donors. Therefore, alternative treatment options for patients with end-stage renal disease are being sought. In vitro differentiation of stem cells into renal tissue is a promising approach to repair nonfunctional kidney tissue. Impressive headway has been made in the use of stem cells with the use of adult renal progenitor cells, embryonic stem cells, and induced pluripotent stem cells for the development toward primitive kidney structures. Currently, efforts are directed at improving long-term maintenance and stability of the cells. This review aims to provide a comprehensive overview of the cell sources used for the generation of kidney cells and strategies used for transplantation in in vivo models. Furthermore, it provides a perspective on stability and safety during future clinical application of in vitro generated kidney cells.
Collapse
|
16
|
Qian T, Hernday SE, Bao X, Olson WR, Panzer SE, Shusta EV, Palecek SP. Directed Differentiation of Human Pluripotent Stem Cells to Podocytes under Defined Conditions. Sci Rep 2019; 9:2765. [PMID: 30808965 PMCID: PMC6391455 DOI: 10.1038/s41598-019-39504-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 12/10/2018] [Indexed: 01/20/2023] Open
Abstract
A major cause of chronic kidney disease (CKD) is glomerular disease, which can be attributed to a spectrum of podocyte disorders. Podocytes are non-proliferative, terminally differentiated cells. Thus, the limited supply of primary podocytes impedes CKD research. Differentiation of human pluripotent stem cells (hPSCs) into podocytes has the potential to produce podocytes for disease modeling, drug screening, and cell therapies. In the podocyte differentiation process described here, hPSCs are first induced to primitive streak-like cells by activating canonical Wnt signaling. Next, these cells progress to mesoderm precursors, proliferative nephron progenitors, and eventually become mature podocytes by culturing in a serum-free medium. Podocytes generated via this protocol adopt podocyte morphology, express canonical podocyte markers, and exhibit podocyte phenotypes, including albumin uptake and TGF-β1 triggered cell death. This study provides a simple, defined strategy to generate podocytes for in vitro modeling of podocyte development and disease or for cell therapies.
Collapse
Affiliation(s)
- Tongcheng Qian
- Department of Chemical & Biological Engineering, University of Wisconsin, Madison, WI, 53706, USA
| | - Shaenah E Hernday
- Department of Chemical & Biological Engineering, University of Wisconsin, Madison, WI, 53706, USA
| | - Xiaoping Bao
- Department of Chemical & Biological Engineering, University of Wisconsin, Madison, WI, 53706, USA
| | - William R Olson
- Department of Chemical & Biological Engineering, University of Wisconsin, Madison, WI, 53706, USA
| | - Sarah E Panzer
- School of Medicine and Public Health, University of Wisconsin, Madison, WI, 53706, USA
| | - Eric V Shusta
- Department of Chemical & Biological Engineering, University of Wisconsin, Madison, WI, 53706, USA.
| | - Sean P Palecek
- Department of Chemical & Biological Engineering, University of Wisconsin, Madison, WI, 53706, USA.
| |
Collapse
|
17
|
Begum S. Engineering renal epithelial cells: programming and directed differentiation towards glomerular podocyte's progenitor and mature podocyte. Am J Transl Res 2019; 11:1102-1115. [PMID: 30899410 PMCID: PMC6413241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 12/26/2018] [Indexed: 06/09/2023]
Abstract
Current knowledge of normal developmental physiology and identification of specific cell types of the kidney at molecular levels enables us to generate various cells of the kidney. The generation of renal specialized cells in vitro with its correct molecular and functional implications is the urgent need for cellular therapy in chronic kidney diseases and for organ formation. Glomerular podocytes are one of the major renal cells lose its functionality to maintain glomerular blood filtration function. In vitro, many inductions or reprogramming methods have been established for podocytes development. In these methods transcription factors, small molecules, and growth factors play the major role to remodel stem cells into podocyte progenitors and towards mature podocytes. Micro ribonucleic acids (miRNAs) have been utilizing as another strategy to generate podocyte. In this review, current protocols for in vitro glomerular podocyte differentiation have summarized emphasizing programming methods, signaling modulation, and cytoskeletal changes. Novel ideas are also pointed out, which are required for efficient optimal glomerular podocyte generation and their functional characterization in vitro with nanoarchitecture impression of the glomerular basement membrane.
Collapse
Affiliation(s)
- Sumreen Begum
- Stem Cells Research Laboratory (SCRL), Sindh Institute of Urology and Transplantation (SIUT) Karachi, Sindh, Pakistan
| |
Collapse
|
18
|
Lau RW, Wang B, Ricardo SD. Gene editing of stem cells for kidney disease modelling and therapeutic intervention. Nephrology (Carlton) 2019; 23:981-990. [PMID: 29851168 DOI: 10.1111/nep.13410] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/26/2018] [Indexed: 12/13/2022]
Abstract
Recent developments in targeted gene editing have paved the way for the wide adoption of clustered regularly interspaced short palindromic repeats (CRISPR)-associated protein-9 nucleases (Cas9) as an RNA-guided molecular tool to modify the genome of eukaryotic cells of animals. Theoretically, the translation of CRISPR-Cas9 can be applied to the treatment of inherited or acquired kidney disease, kidney transplantation and genetic corrections of somatic cells from kidneys with inherited mutations, such as polycystic kidney disease. Human pluripotent stem cells have been used to generate an unlimited source of kidney progenitor cells or, when spontaneously differentiated into three-dimensional kidney organoids, to model kidney organogenesis or the pathogenesis of disease. Gene editing now allows for the tagging and selection of specific kidney cell types or disease-specific gene knock in/out, which enables more precise understanding of kidney organogenesis and genetic diseases. This review discusses the mechanisms of action, in addition to the advantages and disadvantages, of the three major gene editing technologies, namely, CRISPR-Cas9, zinc finger nucleases and transcription activator-like effector nucleases. The implications of using gene editing to better understand kidney disease is reviewed in detail. In addition, the ethical issues of gene editing, which could be easily neglected in the modern, fast-paced research environment, are highlighted.
Collapse
Affiliation(s)
- Ricky Wk Lau
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Bo Wang
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Sharon D Ricardo
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
19
|
Minocha E, Chaturvedi CP, Nityanand S. Renogenic characterization and in vitro differentiation of rat amniotic fluid stem cells into renal proximal tubular- and juxtaglomerular-like cells. In Vitro Cell Dev Biol Anim 2019; 55:138-147. [PMID: 30645697 DOI: 10.1007/s11626-018-00315-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 12/16/2018] [Indexed: 12/31/2022]
Abstract
The aim of the present study was to investigate the renogenic characteristics of amniotic fluid stem cells (AFSCs) and to evaluate their in vitro differentiation potential into renal proximal tubular-like cells and juxtaglomerular-like cells. We culture expanded AFSCs derived from rat amniotic fluid. The AFSCs grew as adherent spindle-shaped cells and expressed mesenchymal markers CD73, CD90, and CD105 as well as renal progenitor markers WT1, PAX2, SIX2, SALL1, and CITED1. AFSCs exhibited an in vitro differentiation potential into renal proximal tubular epithelial-like cells, as shown by the upregulation of expression of proximal tubular cell-specific genes like AQP1, CD13, PEPT1, GLUT5, OAT1, and OCT1. AFSCs could also be differentiated into juxtaglomerular-like cells as demonstrated by the expression of renin and α-SMA. The AFSCs also expressed pluripotency markers OCT4, NANOG, and SOX2 and could be induced into embryoid bodies with differentiation into all the three germ layers, highlighting the pluripotent nature of these cells. Our results show that amniotic fluid contains a population of primitive stem cells that express renal-progenitor markers and also possess the propensity to differentiate into two renal lineage cell types and, thus, may have a therapeutic potential in renal regenerative medicine.
Collapse
Affiliation(s)
- Ekta Minocha
- Stem Cell Research Facility, Department of Hematology, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Raebareli Road, Lucknow, UP, 226014, India
| | - Chandra Prakash Chaturvedi
- Stem Cell Research Facility, Department of Hematology, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Raebareli Road, Lucknow, UP, 226014, India
| | - Soniya Nityanand
- Stem Cell Research Facility, Department of Hematology, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Raebareli Road, Lucknow, UP, 226014, India.
| |
Collapse
|
20
|
Recapitulating kidney development: Progress and challenges. Semin Cell Dev Biol 2018; 91:153-168. [PMID: 30184476 DOI: 10.1016/j.semcdb.2018.08.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 08/22/2018] [Accepted: 08/28/2018] [Indexed: 12/14/2022]
Abstract
Decades of research into the molecular and cellular regulation of kidney morphogenesis in rodent models, particularly the mouse, has provided both an atlas of the mammalian kidney and a roadmap for recreating kidney cell types with potential applications for the treatment of kidney disease. With advances in both our capacity to maintain nephron progenitors in culture, reprogram to kidney cell types and direct the differentiation of human pluripotent stem cells to kidney endpoints, renal regeneration via cellular therapy or tissue engineering may be possible. Human kidney models also have potential for disease modelling and drug screening. Such applications will rely upon the accuracy of the model at the cellular level and the capacity for stem-cell derived kidney tissue to recapitulate both normal and diseased kidney tissue. In this review, we will discuss the available cell sources, how well they model the human kidney and how far we are from application either as models or for tissue engineering.
Collapse
|
21
|
O'Brien LL. Nephron progenitor cell commitment: Striking the right balance. Semin Cell Dev Biol 2018; 91:94-103. [PMID: 30030141 DOI: 10.1016/j.semcdb.2018.07.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 06/29/2018] [Accepted: 07/16/2018] [Indexed: 10/28/2022]
Abstract
The filtering component of the kidney, the nephron, arises from a single progenitor population. These nephron progenitor cells (NPCs) both self-renew and differentiate throughout the course of kidney development ensuring sufficient nephron endowment. An appropriate balance of these processes must be struck as deficiencies in nephron numbers are associated with hypertension and kidney disease. This review will discuss the mechanisms and molecules supporting NPC maintenance and differentiation. A focus on recent work will highlight new molecular insights into NPC regulation and their dynamic behavior in both space and time.
Collapse
Affiliation(s)
- Lori L O'Brien
- Department of Cell Biology and Physiology, UNC Kidney Center, University of North Carolina at Chapel Hill, 111 Mason Farm Road, Chapel Hill, NC, 27599, United States.
| |
Collapse
|
22
|
Generation of Functioning Nephrons by Implanting Human Pluripotent Stem Cell-Derived Kidney Progenitors. Stem Cell Reports 2018; 10:766-779. [PMID: 29429961 PMCID: PMC5918196 DOI: 10.1016/j.stemcr.2018.01.008] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 01/11/2018] [Accepted: 01/12/2018] [Indexed: 12/14/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) hold great promise for understanding kidney development and disease. We reproducibly differentiated three genetically distinct wild-type hPSC lines to kidney precursors that underwent rudimentary morphogenesis in vitro. They expressed nephron and collecting duct lineage marker genes, several of which are mutated in human kidney disease. Lentiviral-transduced hPSCs expressing reporter genes differentiated similarly to controls in vitro. Kidney progenitors were subcutaneously implanted into immunodeficient mice. By 12 weeks, they formed organ-like masses detectable by bioluminescence imaging. Implants included perfused glomeruli containing human capillaries, podocytes with regions of mature basement membrane, and mesangial cells. After intravenous injection of fluorescent low-molecular-weight dextran, signal was detected in tubules, demonstrating uptake from glomerular filtrate. Thus, we have developed methods to trace hPSC-derived kidney precursors that formed functioning nephrons in vivo. These advances beyond in vitro culture are critical steps toward using hPSCs to model and treat kidney diseases. Reproducible differentiation to kidney progenitors in 3 hESC lines After subcutaneous implantation, kidney-like tissues detectable by bioluminescence Implant nephrons contain glomeruli, proximal and distal tubules, and collecting ducts Vascularized glomeruli filter intravenously injected low-molecular-weight dextran
Collapse
|
23
|
Turunen S, Kaisto S, Skovorodkin I, Mironov V, Kalpio T, Vainio S, Rak-Raszewska A. 3D bioprinting of the kidney—hype or hope? ACTA ACUST UNITED AC 2018. [DOI: 10.3934/celltissue.2018.3.119] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
24
|
Gupta N, Susa K, Morizane R. Regenerative Medicine, Disease Modeling, and Drug Discovery in Human Pluripotent Stem Cell-derived Kidney Tissue. EUROPEAN MEDICAL JOURNAL. REPRODUCTIVE HEALTH 2017; 3:57-67. [PMID: 31157117 PMCID: PMC6544146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The multitude of research clarifying critical factors in embryonic organ development has been instrumental in human stem cell research. Mammalian organogenesis serves as the archetype for directed differentiation protocols, subdividing the process into a series of distinct intermediate stages that can be chemically induced and monitored for the expression of stage-specific markers. Significant advances over the past few years include established directed differentiation protocols of human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs) into human kidney organoids in vitro. Human kidney tissue in vitro simulate the in vivo response when subject to nephrotoxins, providing a novel screening platform during drug discovery to facilitate identification of lead candidates, reduce developmental expenditures, and reduce future rates of drug-induced acute kidney injury. Patient-derived hiPSCs, which bear naturally occurring DNA mutations, may allow for modeling of human genetic diseases to determine pathologic mechanisms and screen for novel therapeutics. In addition, recent advances in genome editing with CRISPR/Cas9 enable to generate specific mutations to study genetic disease with non-mutated lines serving as an ideal isogenic control. The growing population of patients with end-stage kidney disease (ESKD) is a world-wide healthcare problem with higher morbidity and mortality that warrants the discovery of novel forms of renal replacement therapy. Coupling the outlined advances in hiPSC research with innovative bioengineering techniques, such as decellularized kidney and 3D printed scaffolds, may contribute to the development of bioengineered transplantable human kidney tissue as a means of renal replacement therapy.
Collapse
Affiliation(s)
- Navin Gupta
- Department of Medicine, Renal Division, Brigham and Women’s Hospital, Boston, Massachusetts, 02115, USA
- Harvard Medical School, Boston, Massachusetts, 02115, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, 02138, USA
| | - Koichiro Susa
- Department of Medicine, Renal Division, Brigham and Women’s Hospital, Boston, Massachusetts, 02115, USA
- Harvard Medical School, Boston, Massachusetts, 02115, USA
| | - Ryuji Morizane
- Department of Medicine, Renal Division, Brigham and Women’s Hospital, Boston, Massachusetts, 02115, USA
- Harvard Medical School, Boston, Massachusetts, 02115, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, 02138, USA
| |
Collapse
|
25
|
Morizane R, Bonventre JV. Kidney Organoids: A Translational Journey. Trends Mol Med 2017; 23:246-263. [PMID: 28188103 DOI: 10.1016/j.molmed.2017.01.001] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 01/06/2017] [Accepted: 01/08/2017] [Indexed: 12/12/2022]
Abstract
Human pluripotent stem cells (hPSCs) are attractive sources for regenerative medicine and disease modeling in vitro. Directed hPSC differentiation approaches have derived from knowledge of cell development in vivo rather than from stochastic cell differentiation. Moreover, there has been great success in the generation of 3D organ-buds termed 'organoids' from hPSCs; these consist of a variety of cell types in vitro that mimic organs in vivo. The organoid bears great potential in the study of human diseases in vitro, especially when combined with CRISPR/Cas9-based genome-editing. We summarize the current literature describing organoid studies with a special focus on kidney organoids, and discuss goals and future opportunities for organoid-based studies.
Collapse
Affiliation(s)
- Ryuji Morizane
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA; Harvard Stem Cell Institute, Cambridge, MA, USA.
| | - Joseph V Bonventre
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA; Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
26
|
Yamaguchi S, Morizane R, Homma K, Monkawa T, Suzuki S, Fujii S, Koda M, Hiratsuka K, Yamashita M, Yoshida T, Wakino S, Hayashi K, Sasaki J, Hori S, Itoh H. Generation of kidney tubular organoids from human pluripotent stem cells. Sci Rep 2016; 6:38353. [PMID: 27982115 PMCID: PMC5159864 DOI: 10.1038/srep38353] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 11/08/2016] [Indexed: 12/12/2022] Open
Abstract
Recent advances in stem cell research have resulted in methods to generate kidney organoids from human pluripotent stem cells (hPSCs), which contain cells of multiple lineages including nephron epithelial cells. Methods to purify specific types of cells from differentiated hPSCs, however, have not been established well. For bioengineering, cell transplantation, and disease modeling, it would be useful to establish those methods to obtain pure populations of specific types of kidney cells. Here, we report a simple two-step differentiation protocol to generate kidney tubular organoids from hPSCs with direct purification of KSP (kidney specific protein)-positive cells using anti-KSP antibody. We first differentiated hPSCs into mesoderm cells using a glycogen synthase kinase-3β inhibitor for 3 days, then cultured cells in renal epithelial growth medium to induce KSP+ cells. We purified KSP+ cells using flow cytometry with anti-KSP antibody, which exhibited characteristics of all segments of kidney tubular cells and cultured KSP+ cells in 3D Matrigel, which formed tubular organoids in vitro. The formation of tubular organoids by KSP+ cells induced the acquisition of functional kidney tubules. KSP+ cells also allowed for the generation of chimeric kidney cultures in which human cells self-assembled into 3D tubular structures in combination with mouse embryonic kidney cells.
Collapse
Affiliation(s)
- Shintaro Yamaguchi
- Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Ryuji Morizane
- Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.,Renal Division, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA.,Department of Medicine, Harvard Medical School, 25 Shattuck St, Boston, MA 02115, USA.,Harvard Stem Cell Institute, 7 Divinity Ave, Cambridge, MA 02138, USA
| | - Koichiro Homma
- Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.,Emergency and Critical Care Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Toshiaki Monkawa
- Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.,Medical Education Center, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Sayuri Suzuki
- Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Shizuka Fujii
- Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Muneaki Koda
- Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Ken Hiratsuka
- Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Maho Yamashita
- Apheresis and Dialysis Center, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Tadashi Yoshida
- Apheresis and Dialysis Center, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Shu Wakino
- Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Koichi Hayashi
- Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Junichi Sasaki
- Emergency and Critical Care Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Shingo Hori
- Emergency and Critical Care Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hiroshi Itoh
- Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
27
|
Abstract
The human kidney develops from four progenitor populations-nephron progenitors, ureteric epithelial progenitors, renal interstitial progenitors and endothelial progenitors-resulting in the formation of maximally 2 million nephrons. Until recently, the reported methods differentiated human pluripotent stem cells (hPSCs) into either nephron progenitor or ureteric epithelial progenitor cells, consequently forming only nephrons or collecting ducts, respectively. Here we detail a protocol that simultaneously induces all four progenitors to generate kidney organoids within which segmented nephrons are connected to collecting ducts and surrounded by renal interstitial cells and an endothelial network. As evidence of functional maturity, proximal tubules within organoids display megalin-mediated and cubilin-mediated endocytosis, and they respond to a nephrotoxicant to undergo apoptosis. This protocol consists of 7 d of monolayer culture for intermediate mesoderm induction, followed by 18 d of 3D culture to facilitate self-organizing renogenic events leading to organoid formation. Personnel experienced in culturing hPSCs are required to conduct this protocol.
Collapse
|
28
|
Du C, Narayanan K, Leong MF, Ibrahim MS, Chua YP, Khoo VMH, Wan ACA. Functional Kidney Bioengineering with Pluripotent Stem-Cell-Derived Renal Progenitor Cells and Decellularized Kidney Scaffolds. Adv Healthc Mater 2016; 5:2080-91. [PMID: 27294565 DOI: 10.1002/adhm.201600120] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 04/20/2016] [Indexed: 11/11/2022]
Abstract
Recent advances in developmental biology and stem cell technology have led to the engineering of functional organs in a dish. However, the limited size of these organoids and absence of a large circulatory system poses limits to its clinical translation. To overcome these issues, decellularized whole kidney scaffolds with native microstructure and extracellular matrix (ECM) are employed for kidney bioengineering, using human-induced pluripotent-stem-cell-derived renal progenitor cells and endothelial cells. To demonstrate ECM-guided cellular assembly, the present work is focused on generating the functional unit of the kidney, the glomerulus. In the repopulated organ, the presence of endothelial cells broadly upregulates the expression level of genes related to renal development. When the cellularized native scaffolds are implanted in SCID mice, glomeruli assembly can be achieved by co-culture of the renal progenitors and endothelial cells. These individual glomerular units are shown to be functional in the context of the whole organ using a simulated bio-reactor set-up with urea and creatinine excretion and albumin reabsorption. Our results indicate that the repopulation of decellularized native kidney using clinically relevant, expandable patient-specific renal progenitors and endothelial cells may be a viable approach for the generation of a functional whole kidney.
Collapse
Affiliation(s)
- Chan Du
- Institute of Bioengineering and Nanotechnology; 31 Biopolis Way Singapore 138669 Singapore
| | - Karthikeyan Narayanan
- Institute of Bioengineering and Nanotechnology; 31 Biopolis Way Singapore 138669 Singapore
| | - Meng Fatt Leong
- Institute of Bioengineering and Nanotechnology; 31 Biopolis Way Singapore 138669 Singapore
| | | | - Ying Ping Chua
- Institute of Bioengineering and Nanotechnology; 31 Biopolis Way Singapore 138669 Singapore
| | - Vanessa Mei Hui Khoo
- Institute of Bioengineering and Nanotechnology; 31 Biopolis Way Singapore 138669 Singapore
| | - Andrew C. A. Wan
- Institute of Bioengineering and Nanotechnology; 31 Biopolis Way Singapore 138669 Singapore
| |
Collapse
|
29
|
Schutgens F, Verhaar MC, Rookmaaker MB. Pluripotent stem cell-derived kidney organoids: An in vivo-like in vitro technology. Eur J Pharmacol 2016; 790:12-20. [PMID: 27375081 DOI: 10.1016/j.ejphar.2016.06.059] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 06/14/2016] [Accepted: 06/30/2016] [Indexed: 12/12/2022]
Abstract
Organoids are self-organizing, multicellular structures that contain multiple cell types, represent organ structure and function, and can be used to model organ development, maintenance and repair ex vivo. Organoids, derived from embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs) or adult stem cells, are cultured in extracellular matrix (ECM). Organoid cultures have been developed for multiple organs and for the kidney, pluripotent stem cell (PSCs) derived organoid technology has rapidly developed in the last three years. Here, we review available PSC differentiation protocols, focusing on the pluripotent stem cells to initiate the organoid culture, as well as on growth factors and ECM used to regulate differentiation and expansion. In addition, we will discuss the read out strategies to evaluate organoid phenotype and function. Finally, we will indicate how the choice of both culture parameters and read out strategy should be tailored to specific applications of the organoid culture.
Collapse
Affiliation(s)
- Frans Schutgens
- UMC Utrecht, Department of Nephrology and Hypertension, Postbus 85500, 3508 GA Utrecht, The Netherlands; Hubrecht Institute, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands.
| | - Marianne C Verhaar
- UMC Utrecht, Department of Nephrology and Hypertension, Postbus 85500, 3508 GA Utrecht, The Netherlands.
| | - Maarten B Rookmaaker
- UMC Utrecht, Department of Nephrology and Hypertension, Postbus 85500, 3508 GA Utrecht, The Netherlands; Hubrecht Institute, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands.
| |
Collapse
|
30
|
Generation of functional podocytes from human induced pluripotent stem cells. Stem Cell Res 2016; 17:130-9. [PMID: 27299470 PMCID: PMC5009184 DOI: 10.1016/j.scr.2016.06.001] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 05/05/2016] [Accepted: 06/02/2016] [Indexed: 02/08/2023] Open
Abstract
Generating human podocytes in vitro could offer a unique opportunity to study human diseases. Here, we describe a simple and efficient protocol for obtaining functional podocytes in vitro from human induced pluripotent stem cells. Cells were exposed to a three-step protocol, which induced their differentiation into intermediate mesoderm, then into nephron progenitors and, finally, into mature podocytes. After differentiation, cells expressed the main podocyte markers, such as synaptopodin, WT1, α-Actinin-4, P-cadherin and nephrin at the protein and mRNA level, and showed the low proliferation rate typical of mature podocytes. Exposure to Angiotensin II significantly decreased the expression of podocyte genes and cells underwent cytoskeleton rearrangement. Cells were able to internalize albumin and self-assembled into chimeric 3D structures in combination with dissociated embryonic mouse kidney cells. Overall, these findings demonstrate the establishment of a robust protocol that, mimicking developmental stages, makes it possible to derive functional podocytes in vitro. Human iPSC differentiation into podocytes recapitulates kidney developmental stages. The differentiation protocol is reproducible and highly efficient. The generated podocytes reflect primary cell behaviour and are functional.
Collapse
|
31
|
Morizane R, Lam AQ, Freedman BS, Kishi S, Valerius MT, Bonventre JV. Nephron organoids derived from human pluripotent stem cells model kidney development and injury. Nat Biotechnol 2016; 33:1193-200. [PMID: 26458176 PMCID: PMC4747858 DOI: 10.1038/nbt.3392] [Citation(s) in RCA: 608] [Impact Index Per Article: 67.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 10/06/2015] [Indexed: 12/23/2022]
Abstract
Kidney cells and tissues derived from human pluripotent stem cells (hPSCs) would enable organ regeneration, disease modeling, and drug screening in vitro. We established an efficient, chemically defined protocol for differentiating hPSCs into multipotent nephron progenitor cells (NPCs) that can form nephron-like structures. By recapitulating metanephric kidney development in vitro, we generate SIX2+SALL1+WT1+PAX2+ NPCs with 90% efficiency within 9 days of differentiation. The NPCs possess the developmental potential of their in vivo counterparts and form PAX8+LHX1+ renal vesicles that self-pattern into nephron structures. In both 2D and 3D culture, NPCs form kidney organoids containing epithelial nephron-like structures expressing markers of podocytes, proximal tubules, loops of Henle, and distal tubules in an organized, continuous arrangement that resembles the nephron in vivo. We also show that this organoid culture system can be used to study mechanisms of human kidney development and toxicity.
Collapse
|
32
|
Poornejad N, Schaumann LB, Buckmiller EM, Roeder BL, Cook AD. Current Cell-Based Strategies for Whole Kidney Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2016; 22:358-370. [PMID: 26905375 DOI: 10.1089/ten.teb.2015.0520] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Chronic kidney diseases affect thousands of people worldwide. Although hemodialysis alleviates the situation by filtering the patient's blood, it does not replace other kidney functions such as hormone release or homeostasis regulation. Consequently, orthotopic transplantation of donor organs is the ultimate treatment for patients suffering from end-stage renal failure. Unfortunately, the number of patients on the waiting list far exceeds the number of donors. In addition, recipients must remain on immunosuppressive medications for the remainder of their lives, which increases the risk of morbidity due to their weakened immune system. Despite recent advancements in whole organ transplantation, 40% of recipients will face rejection of implanted organs with a life expectancy of only 10 years. Bioengineered patient-specific kidneys could be an inexhaustible source of healthy kidneys without the risk of immune rejection. The purpose of this article is to review the pros and cons of several bioengineering strategies used in recent years and their unresolved issues. These strategies include repopulation of natural scaffolds with a patient's cells, de-novo generation of kidneys using patient-induced pluripotent stem cells combined with stepwise differentiation, and the creation of a patient's kidney in the embryos of other mammalian species.
Collapse
Affiliation(s)
- Nafiseh Poornejad
- 1 Department of Chemical Engineering, Brigham Young University , Provo, Utah
| | - Lara B Schaumann
- 1 Department of Chemical Engineering, Brigham Young University , Provo, Utah
| | - Evan M Buckmiller
- 2 Department of Genetics and Biotechnology, Brigham Young University , Provo, Utah
| | | | - Alonzo D Cook
- 1 Department of Chemical Engineering, Brigham Young University , Provo, Utah
| |
Collapse
|
33
|
Poornejad N, Momtahan N, Salehi ASM, Scott DR, Fronk CA, Roeder BL, Reynolds PR, Bundy BC, Cook AD. Efficient decellularization of whole porcine kidneys improves reseeded cell behavior. Biomed Mater 2016; 11:025003. [DOI: 10.1088/1748-6041/11/2/025003] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
34
|
Kidney organoids from human iPS cells contain multiple lineages and model human nephrogenesis. Nature 2015; 526:564-8. [PMID: 26444236 DOI: 10.1038/nature15695] [Citation(s) in RCA: 974] [Impact Index Per Article: 97.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 09/08/2015] [Indexed: 01/09/2023]
Abstract
The human kidney contains up to 2 million epithelial nephrons responsible for blood filtration. Regenerating the kidney requires the induction of the more than 20 distinct cell types required for excretion and the regulation of pH, and electrolyte and fluid balance. We have previously described the simultaneous induction of progenitors for both collecting duct and nephrons via the directed differentiation of human pluripotent stem cells. Paradoxically, although both are of intermediate mesoderm in origin, collecting duct and nephrons have distinct temporospatial origins. Here we identify the developmental mechanism regulating the preferential induction of collecting duct versus kidney mesenchyme progenitors. Using this knowledge, we have generated kidney organoids that contain nephrons associated with a collecting duct network surrounded by renal interstitium and endothelial cells. Within these organoids, individual nephrons segment into distal and proximal tubules, early loops of Henle, and glomeruli containing podocytes elaborating foot processes and undergoing vascularization. When transcription profiles of kidney organoids were compared to human fetal tissues, they showed highest congruence with first trimester human kidney. Furthermore, the proximal tubules endocytose dextran and differentially apoptose in response to cisplatin, a nephrotoxicant. Such kidney organoids represent powerful models of the human organ for future applications, including nephrotoxicity screening, disease modelling and as a source of cells for therapy.
Collapse
|
35
|
Hyperglycemic Stress Impairs the Stemness Capacity of Kidney Stem Cells in Rats. PLoS One 2015; 10:e0139607. [PMID: 26431335 PMCID: PMC4592017 DOI: 10.1371/journal.pone.0139607] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 09/14/2015] [Indexed: 12/31/2022] Open
Abstract
The incidence of acute kidney injury in patients with diabetes is significantly higher than that of patients without diabetes, and may be associated with the poor stemness capacity of kidney stem cells (KSCs) and limited recovery of injured renal tubules. To investigate the effects of hyperglycemic stress on KSC stemness, KSCs were isolated from the rat renal papilla and analyzed for their self-renewal and differentiation abilities. Our results showed that isolated KSCs expressed the mesenchymal stem cell markers N-cadherin, Nestin, CD133, CD29, CD90, and CD73. Moreover, KSCs co-cultured with hypoxia-injured renal tubular epithelial cell (RTECs) induced the expression of the mature epithelial cell marker CK18, suggesting that the KSCs could differentiate into RTECs in vitro. However, KSC proliferation, differentiation ability and tolerance to hypoxia were decreased in high-glucose cultures. Taken together, these results suggest the high-glucose microenvironment can damage the reparative ability of KSCs. It may result in a decreased of recovery capability of renal tubules from injury.
Collapse
|
36
|
Morizane R, Lam AQ. Directed Differentiation of Pluripotent Stem Cells into Kidney. Biomark Insights 2015; 10:147-52. [PMID: 26417199 PMCID: PMC4571990 DOI: 10.4137/bmi.s20055] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 06/08/2015] [Accepted: 06/10/2015] [Indexed: 01/10/2023] Open
Abstract
Pluripotent stem cells (PSCs), including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), represent an ideal substrate for regenerating kidney cells and tissue lost through injury and disease. Recent studies have demonstrated the ability to differentiate PSCs into populations of nephron progenitor cells that can organize into kidney epithelial structures in three-dimensional contexts. While these findings are highly encouraging, further studies need to be performed to improve the efficiency and specificity of kidney differentiation. The identification of specific markers of the differentiation process is critical to the development of protocols that effectively recapitulate nephrogenesis in vitro. In this review, we summarize the current studies describing the differentiation of ESCs and iPSCs into cells of the kidney lineage. We also present an analysis of the markers relevant to the stages of kidney development and differentiation and propose a new roadmap for the directed differentiation of PSCs into nephron progenitor cells of the metanephric mesenchyme.
Collapse
Affiliation(s)
- Ryuji Morizane
- Division of Kidney Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Albert Q Lam
- Division of Kidney Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA. ; Harvard Stem Cell Institute, Cambridge, MA, USA
| |
Collapse
|
37
|
Abstract
PURPOSE OF REVIEW Nephrogenesis in humans is limited to the period of embryonic kidney development in utero, with no new nephrons formed after birth. Although the kidneys possess the capacity to self-repair segments of the nephron, nephron loss from acute or chronic kidney injury is irreversible and results in impaired function. Human pluripotent stem cells (PSCs), including embryonic stem cells and induced pluripotent stem cells, are an attractive source of cells to regenerate nephron progenitor cells (NPCs) and ultimately functional kidney tissue. NPCs are found exclusively during the period of embryonic development, but their nephron-forming capacity makes them an ideal cell population to regenerate with PSCs. RECENT FINDINGS Significant progress has been made in the effort to direct the differentiation of human PSCs into NPCs. Differentiation protocols designed to recapitulate the complex process of kidney organogenesis in vitro can generate cells that express characteristic NPC markers and these cells can assemble into three-dimensional nephron-like structures. Additional studies are required to evaluate the functionality of these putative kidney cells and to test their ability to integrate into three-dimensional organized kidney tissue structures, either spontaneously or facilitated by bioengineered structures or scaffolds with appropriate matrix materials. SUMMARY The successful recreation of human nephrons from PSCs would offer a novel therapeutic approach to treating patients with kidney disease.
Collapse
|
38
|
Abstract
PURPOSE OF REVIEW Human induced pluripotent stem cells (iPSCs) and embryonic stem cells (ESCs) are potential unlimited cell sources for renal cells in regenerative medicine. This review highlights recent advance in the directed differentiation of human iPSCs into kidney lineages and discusses the remaining challenges to generate functional or mature renal cells from human iPSCs. RECENT FINDINGS Recently, directed differentiation methods from human iPSCs/ESCs into embryonic renal progenitor cells, such as those included in metanephric mesenchyme and ureteric bud, that mimic embryonic development have been reported. These studies show the developmental potential of progenitor cells by forming renal tubule-like or glomerulus-like structures in vitro. However, it has not been verified whether the physiological functions of the induced progenitors are equivalent to their in-vivo counterparts. The establishment of definitive marker genes for kidney lineages and functional assay systems is essential for the verification. Such achievement is needed before kidney regeneration can provide cell replacement therapy, reliable disease models and elucidation of the mechanisms of kidney development. SUMMARY In conclusion, this review outlines milestones in directed differentiation methods for functional renal cell types from human iPSCs toward clinical application and practical use.
Collapse
|
39
|
Prediction of drug-induced nephrotoxicity and injury mechanisms with human induced pluripotent stem cell-derived cells and machine learning methods. Sci Rep 2015. [PMID: 26212763 PMCID: PMC4515747 DOI: 10.1038/srep12337] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The renal proximal tubule is a main target for drug-induced toxicity. The prediction of proximal tubular toxicity during drug development remains difficult. Any in vitro methods based on induced pluripotent stem cell-derived renal cells had not been developed, so far. Here, we developed a rapid 1-step protocol for the differentiation of human induced pluripotent stem cells (hiPSC) into proximal tubular-like cells. These proximal tubular-like cells had a purity of >90% after 8 days of differentiation and could be directly applied for compound screening. The nephrotoxicity prediction performance of the cells was determined by evaluating their responses to 30 compounds. The results were automatically determined using a machine learning algorithm called random forest. In this way, proximal tubular toxicity in humans could be predicted with 99.8% training accuracy and 87.0% test accuracy. Further, we studied the underlying mechanisms of injury and drug-induced cellular pathways in these hiPSC-derived renal cells, and the results were in agreement with human and animal data. Our methods will enable the development of personalized or disease-specific hiPSC-based renal in vitro models for compound screening and nephrotoxicity prediction.
Collapse
|
40
|
Toyohara T, Mae SI, Sueta SI, Inoue T, Yamagishi Y, Kawamoto T, Kasahara T, Hoshina A, Toyoda T, Tanaka H, Araoka T, Sato-Otsubo A, Takahashi K, Sato Y, Yamaji N, Ogawa S, Yamanaka S, Osafune K. Cell Therapy Using Human Induced Pluripotent Stem Cell-Derived Renal Progenitors Ameliorates Acute Kidney Injury in Mice. Stem Cells Transl Med 2015. [PMID: 26198166 DOI: 10.5966/sctm.2014-0219] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
UNLABELLED Acute kidney injury (AKI) is defined as a rapid loss of renal function resulting from various etiologies, with a mortality rate exceeding 60% among intensive care patients. Because conventional treatments have failed to alleviate this condition, the development of regenerative therapies using human induced pluripotent stem cells (hiPSCs) presents a promising new therapeutic option for AKI. We describe our methodology for generating renal progenitors from hiPSCs that show potential in ameliorating AKI. We established a multistep differentiation protocol for inducing hiPSCs into OSR1+SIX2+ renal progenitors capable of reconstituting three-dimensional proximal renal tubule-like structures in vitro and in vivo. Moreover, we found that renal subcapsular transplantation of hiPSC-derived renal progenitors ameliorated the AKI in mice induced by ischemia/reperfusion injury, significantly suppressing the elevation of blood urea nitrogen and serum creatinine levels and attenuating histopathological changes, such as tubular necrosis, tubule dilatation with casts, and interstitial fibrosis. To our knowledge, few reports demonstrating the therapeutic efficacy of cell therapy with renal lineage cells generated from hiPSCs have been published. Our results suggest that regenerative medicine strategies for kidney diseases could be developed using hiPSC-derived renal cells. SIGNIFICANCE This report is the first to demonstrate that the transplantation of renal progenitor cells differentiated from human induced pluripotent stem (iPS) cells has therapeutic effectiveness in mouse models of acute kidney injury induced by ischemia/reperfusion injury. In addition, this report clearly demonstrates that the therapeutic benefits come from trophic effects by the renal progenitor cells, and it identifies the renoprotective factors secreted by the progenitors. The results of this study indicate the feasibility of developing regenerative medicine strategy using iPS cells against renal diseases.
Collapse
Affiliation(s)
- Takafumi Toyohara
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan; Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan; Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Clinical Research Center, Chiba University of Medicine, Chiba, Japan; Gladstone Institute of Cardiovascular Disease, San Francisco, California, USA
| | - Shin-Ichi Mae
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan; Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan; Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Clinical Research Center, Chiba University of Medicine, Chiba, Japan; Gladstone Institute of Cardiovascular Disease, San Francisco, California, USA
| | - Shin-Ichi Sueta
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan; Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan; Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Clinical Research Center, Chiba University of Medicine, Chiba, Japan; Gladstone Institute of Cardiovascular Disease, San Francisco, California, USA
| | - Tatsuyuki Inoue
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan; Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan; Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Clinical Research Center, Chiba University of Medicine, Chiba, Japan; Gladstone Institute of Cardiovascular Disease, San Francisco, California, USA
| | - Yukiko Yamagishi
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan; Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan; Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Clinical Research Center, Chiba University of Medicine, Chiba, Japan; Gladstone Institute of Cardiovascular Disease, San Francisco, California, USA
| | - Tatsuya Kawamoto
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan; Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan; Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Clinical Research Center, Chiba University of Medicine, Chiba, Japan; Gladstone Institute of Cardiovascular Disease, San Francisco, California, USA
| | - Tomoko Kasahara
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan; Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan; Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Clinical Research Center, Chiba University of Medicine, Chiba, Japan; Gladstone Institute of Cardiovascular Disease, San Francisco, California, USA
| | - Azusa Hoshina
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan; Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan; Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Clinical Research Center, Chiba University of Medicine, Chiba, Japan; Gladstone Institute of Cardiovascular Disease, San Francisco, California, USA
| | - Taro Toyoda
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan; Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan; Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Clinical Research Center, Chiba University of Medicine, Chiba, Japan; Gladstone Institute of Cardiovascular Disease, San Francisco, California, USA
| | - Hiromi Tanaka
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan; Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan; Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Clinical Research Center, Chiba University of Medicine, Chiba, Japan; Gladstone Institute of Cardiovascular Disease, San Francisco, California, USA
| | - Toshikazu Araoka
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan; Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan; Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Clinical Research Center, Chiba University of Medicine, Chiba, Japan; Gladstone Institute of Cardiovascular Disease, San Francisco, California, USA
| | - Aiko Sato-Otsubo
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan; Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan; Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Clinical Research Center, Chiba University of Medicine, Chiba, Japan; Gladstone Institute of Cardiovascular Disease, San Francisco, California, USA
| | - Kazutoshi Takahashi
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan; Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan; Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Clinical Research Center, Chiba University of Medicine, Chiba, Japan; Gladstone Institute of Cardiovascular Disease, San Francisco, California, USA
| | - Yasunori Sato
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan; Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan; Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Clinical Research Center, Chiba University of Medicine, Chiba, Japan; Gladstone Institute of Cardiovascular Disease, San Francisco, California, USA
| | - Noboru Yamaji
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan; Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan; Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Clinical Research Center, Chiba University of Medicine, Chiba, Japan; Gladstone Institute of Cardiovascular Disease, San Francisco, California, USA
| | - Seishi Ogawa
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan; Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan; Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Clinical Research Center, Chiba University of Medicine, Chiba, Japan; Gladstone Institute of Cardiovascular Disease, San Francisco, California, USA
| | - Shinya Yamanaka
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan; Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan; Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Clinical Research Center, Chiba University of Medicine, Chiba, Japan; Gladstone Institute of Cardiovascular Disease, San Francisco, California, USA
| | - Kenji Osafune
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan; Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan; Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Clinical Research Center, Chiba University of Medicine, Chiba, Japan; Gladstone Institute of Cardiovascular Disease, San Francisco, California, USA
| |
Collapse
|
41
|
Gupta AK, Jadhav SH, Tripathy NK, Nityanand S. Fetal kidney stem cells ameliorate cisplatin induced acute renal failure and promote renal angiogenesis. World J Stem Cells 2015; 7:776-788. [PMID: 26029348 PMCID: PMC4444617 DOI: 10.4252/wjsc.v7.i4.776] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 02/05/2015] [Accepted: 03/09/2015] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate whether fetal kidney stem cells (fKSC) ameliorate cisplatin induced acute renal failure (ARF) in rats and promote renal angiogenesis.
METHODS: The fKSC were isolated from rat fetuses of gestation day 16 and expanded in vitro up to 3rd passage. They were characterized for the expression of mesenchymal and renal progenitor markers by flow cytometry and immunocytochemistry, respectively. The in vitro differentiation of fKSC towards epithelial lineage was evaluated by the treatment with specific induction medium and their angiogenic potential by matrigel induced tube formation assay. To study the effect of fKSC in ARF, fKSC labeled with PKH26 were infused in rats with cisplatin induced ARF and, the blood and renal tissues of the rats were collected at different time points. Blood biochemical parameters were studied to evaluate renal function. Renal tissues were evaluated for renal architecture, renal cell proliferation and angiogenesis by immunohistochemistry, renal cell apoptosis by terminal deoxynucleotidyl transferase nick-end labeling assay and early expression of angiogenic molecules viz. vascular endothelial growth factor (VEGF), hypoxia-inducible factor (HIF)-1α and endothelial nitric oxide synthase (eNOS) by western blot.
RESULTS: The fKSC expressed mesenchymal markers viz. CD29, CD44, CD73, CD90 and CD105 as well as renal progenitor markers viz. Wt1, Pax2 and Six2. They exhibited a potential to form CD31 and Von Willebrand factor expressing capillary-like structures and could be differentiated into cytokeratin (CK)18 and CK19 positive epithelial cells. Administration of fKSC in rats with ARF as compared to administration of saline alone, resulted in a significant improvement in renal function and histology on day 3 (2.33 ± 0.33 vs 3.50 ± 0.34, P < 0.05) and on day 7 (0.83 ± 0.16 vs 2.00 ± 0.25, P < 0.05). The infused PKH26 labeled fKSC were observed to engraft in damaged renal tubules and showed increased proliferation and reduced apoptosis (P < 0.05) of renal cells. The kidneys of fKSC as compared to saline treated rats had a higher capillary density on day 3 [13.30 ± 1.54 vs 7.10 ± 1.29, capillaries/high-power fields (HPF), P < 0.05], and on day 7 (21.10 ± 1.46 vs 15.00 ± 1.30, capillaries/HPF, P < 0.05). In addition, kidneys of fKSC treated rats had an up-regulation of angiogenic proteins hypoxia-inducible factor-1α, VEGF and eNOS on day 3 (P < 0.05).
CONCLUSION: Our study shows that fKSC ameliorate cisplatin induced ARF in rats and promote renal angiogenesis, which may be an important therapeutic mechanism of these stem cells in the disease.
Collapse
|
42
|
Song D, Yue L, Wu G, Ma S, Guo L, Yang H, Liu Q, Zhang D, Xia Z, Wang L, Zhang J, Zhao W, Guo F, Wang J. Assessment of promoter methylation and expression of SIX2 as a diagnostic and prognostic biomarker in Wilms' tumor. Tumour Biol 2015; 36:7591-8. [PMID: 25921281 DOI: 10.1007/s13277-015-3456-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 04/10/2015] [Indexed: 11/29/2022] Open
Abstract
This study was designed to evaluate the utility of expression and DNA methylation patterns of the sine oculis homeobox homolog 2 (SIX2) gene in early diagnosis and prognosis of Wilms' tumor (WT). Methylation-specific polymerase chain reaction (MSP), real-time quantitative polymerase chain reaction (qRT-PCR), receiver operating characteristic (ROC), and survival curve analyses were utilized to measure the expression and DNA methylation patterns of SIX2 in a cohort of WT tissues, with a view to assessing their diagnostic and prognostic value. Relative expression of SIX2 mRNA was higher, while the promoter methylation level was lower in the WT than control group (P < 0.05) and closely associated with poor survival prognosis of WT children (P < 0.05). Increased expression and decreased methylation of SIX2 were correlated with increasing tumor size, clinical stage, vascular invasion, and unfavorable histological differentiation (P < 0.05). ROC curve analysis showed areas under the curve (AUCs) of 0.579 for methylation and 0.917 for expression in WT venous blood, indicating higher diagnostic yield of preoperative SIX2 expression. The preoperative venous blood SIX2 expression level serves as an underlying biomarker for early diagnosis of WT. SIX2 overexpression and concomitantly decreased promoter methylation are significantly associated with poor survival of WT children.
Collapse
Affiliation(s)
- Dongjian Song
- Department of Pediatric Surgery, First Affiliated Hospital of Zhengzhou University, Construction East Road 1, Erqi District, Zhengzhou, Henan, 450000, People's Republic of China
| | - Lifang Yue
- Department of Ultrasonography, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, People's Republic of China
| | - Gang Wu
- Department of Interventional Radiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, People's Republic of China
| | - Shanshan Ma
- School of Life Science, Zhengzhou University, Zhengzhou, Henan, 450000, People's Republic of China
| | - Lihua Guo
- Department of Pediatric Surgery, First Affiliated Hospital of Zhengzhou University, Construction East Road 1, Erqi District, Zhengzhou, Henan, 450000, People's Republic of China
| | - Heying Yang
- Department of Pediatric Surgery, First Affiliated Hospital of Zhengzhou University, Construction East Road 1, Erqi District, Zhengzhou, Henan, 450000, People's Republic of China
| | - Qiuliang Liu
- Department of Pediatric Surgery, First Affiliated Hospital of Zhengzhou University, Construction East Road 1, Erqi District, Zhengzhou, Henan, 450000, People's Republic of China
| | - Da Zhang
- Department of Pediatric Surgery, First Affiliated Hospital of Zhengzhou University, Construction East Road 1, Erqi District, Zhengzhou, Henan, 450000, People's Republic of China
| | - Ziqiang Xia
- Department of Pediatric Surgery, First Affiliated Hospital of Zhengzhou University, Construction East Road 1, Erqi District, Zhengzhou, Henan, 450000, People's Republic of China
| | - Lei Wang
- Department of Pediatric Surgery, First Affiliated Hospital of Zhengzhou University, Construction East Road 1, Erqi District, Zhengzhou, Henan, 450000, People's Republic of China
| | - Junjie Zhang
- Department of Pediatric Surgery, First Affiliated Hospital of Zhengzhou University, Construction East Road 1, Erqi District, Zhengzhou, Henan, 450000, People's Republic of China
| | - Wei Zhao
- Department of Pediatric Surgery, First Affiliated Hospital of Zhengzhou University, Construction East Road 1, Erqi District, Zhengzhou, Henan, 450000, People's Republic of China
| | - Fei Guo
- Department of Pediatric Surgery, First Affiliated Hospital of Zhengzhou University, Construction East Road 1, Erqi District, Zhengzhou, Henan, 450000, People's Republic of China
| | - Jiaxiang Wang
- Department of Pediatric Surgery, First Affiliated Hospital of Zhengzhou University, Construction East Road 1, Erqi District, Zhengzhou, Henan, 450000, People's Republic of China.
| |
Collapse
|
43
|
Suter-Dick L, Alves PM, Blaauboer BJ, Bremm KD, Brito C, Coecke S, Flick B, Fowler P, Hescheler J, Ingelman-Sundberg M, Jennings P, Kelm JM, Manou I, Mistry P, Moretto A, Roth A, Stedman D, van de Water B, Beilmann M. Stem cell-derived systems in toxicology assessment. Stem Cells Dev 2015; 24:1284-96. [PMID: 25675366 DOI: 10.1089/scd.2014.0540] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Industrial sectors perform toxicological assessments of their potential products to ensure human safety and to fulfill regulatory requirements. These assessments often involve animal testing, but ethical, cost, and time concerns, together with a ban on it in specific sectors, make appropriate in vitro systems indispensable in toxicology. In this study, we summarize the outcome of an EPAA (European Partnership of Alternatives to Animal Testing)-organized workshop on the use of stem cell-derived (SCD) systems in toxicology, with a focus on industrial applications. SCD systems, in particular, induced pluripotent stem cell-derived, provide physiological cell culture systems of easy access and amenable to a variety of assays. They also present the opportunity to apply the vast repository of existing nonclinical data for the understanding of in vitro to in vivo translation. SCD systems from several toxicologically relevant tissues exist; they generally recapitulate many aspects of physiology and respond to toxicological and pharmacological interventions. However, focused research is necessary to accelerate implementation of SCD systems in an industrial setting and subsequent use of such systems by regulatory authorities. Research is required into the phenotypic characterization of the systems, since methods and protocols for generating terminally differentiated SCD cells are still lacking. Organotypical 3D culture systems in bioreactors and microscale tissue engineering technologies should be fostered, as they promote and maintain differentiation and support coculture systems. They need further development and validation for their successful implementation in toxicity testing in industry. Analytical measures also need to be implemented to enable compound exposure and metabolism measurements for in vitro to in vivo extrapolation. The future of SCD toxicological tests will combine advanced cell culture technologies and biokinetic measurements to support regulatory and research applications. However, scientific and technical hurdles must be overcome before SCD in vitro methods undergo appropriate validation and become accepted in the regulatory arena.
Collapse
Affiliation(s)
- Laura Suter-Dick
- 1University of Applied Sciences Northwestern Switzerland, School of Life Sciences, Muttenz, Switzerland
| | - Paula M Alves
- 2iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,3Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Bas J Blaauboer
- 4Division of Toxicology, Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, the Netherlands
| | - Klaus-Dieter Bremm
- 5Bayer Pharma AG, Global Drug Discovery-Global Early Development, Wuppertal, Germany
| | - Catarina Brito
- 2iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,3Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Sandra Coecke
- 6European Commission Joint Research Centre, Institute for Health and Consumer Protection, EURL ECVAM, Ispra, Italy
| | - Burkhard Flick
- 7BASF SE, Experimental Toxicology and Ecology, Ludwigshafen, Germany
| | | | - Jürgen Hescheler
- 9Institut for Neurophysiology, University of Cologne, Cologne, Germany
| | | | - Paul Jennings
- 11Division of Physiology, Department of Physiology and Medical Physics, Innsbruck Medical University, Innsbruck, Austria
| | | | - Irene Manou
- 13European Partnership for Alternative Approaches to Animal Testing (EPAA), B-Brussels, Belgium
| | - Pratibha Mistry
- 14Syngenta Ltd., Product Safety, Jealott's Hill International Research Station, Berkshire, United Kingdom
| | - Angelo Moretto
- 15Dipartimento di Scienze Biochimiche e Cliniche, Università degli Studi di Milano, Milano, Italy.,16Centro Internazionale per gli Antiparassitari e la Prevenzione Sanitaria, Luigi Sacco Hospital, Milano, Italy
| | - Adrian Roth
- 17F. Hoffmann-La Roche Ltd., Innovation Center Basel, Pharmaceutical Sciences, Basel, Switzerland
| | - Donald Stedman
- 18Pfizer Worldwide Research and Development, Cambridge, Massachusetts
| | - Bob van de Water
- 19Division of Toxicology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | | |
Collapse
|
44
|
van de Stolpe A, Kauffmann RH. Innovative human-specific investigational approaches to autoimmune disease. RSC Adv 2015. [DOI: 10.1039/c4ra15794j] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
An organ-on-chip disease model approach, including “pre-clinical trial-on-chip” is introduced for understanding of human autoimmune disease pathophysiology and drug development.
Collapse
Affiliation(s)
- Anja van de Stolpe
- Precision & Decentralized Diagnostics
- Philips Research
- Eindhoven
- The Netherlands
| | | |
Collapse
|
45
|
Lash LH, Putt DA, Benipal B. Multigenerational study of chemically induced cytotoxicity and proliferation in cultures of human proximal tubular cells. Int J Mol Sci 2014; 15:21348-65. [PMID: 25411799 PMCID: PMC4264229 DOI: 10.3390/ijms151121348] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 10/24/2014] [Accepted: 11/07/2014] [Indexed: 11/16/2022] Open
Abstract
Primary cultures of human proximal tubular (hPT) cells are a useful experimental model to study transport, metabolism, cytotoxicity, and effects on gene expression of a diverse array of drugs and environmental chemicals because they are derived directly from the in vivo human kidney. To extend the model to investigate longer-term processes, primary cultures (P0) were passaged for up to four generations (P1-P4). hPT cells retained epithelial morphology and stained positively for cytokeratins through P4, although cell growth and proliferation successively slowed with each passage. Necrotic cell death due to the model oxidants tert-butyl hydroperoxide (tBH) and methyl vinyl ketone (MVK) increased with increasing passage number, whereas that due to the selective nephrotoxicant S-(1,2-dichlorovinyl)-l-cysteine (DCVC) was modest and did not change with passage number. Mitochondrial activity was lower in P2-P4 cells than in either P0 or P1 cells. P1 and P2 cells were most sensitive to DCVC-induced apoptosis. DCVC also increased cell proliferation most prominently in P1 and P2 cells. Modest differences with respect to passage number and response to DCVC exposure were observed in expression of three key proteins (Hsp27, GADD153, p53) involved in stress response. Hence, although there are some modest differences in function with passage, these results support the use of multiple generations of hPT cells as an experimental model.
Collapse
Affiliation(s)
- Lawrence H Lash
- Department of Pharmacology, Wayne State University School of Medicine, 540 East Canfield Avenue, Detroit, MI 48201, USA.
| | - David A Putt
- Department of Pharmacology, Wayne State University School of Medicine, 540 East Canfield Avenue, Detroit, MI 48201, USA.
| | - Bavneet Benipal
- Department of Pharmacology, Wayne State University School of Medicine, 540 East Canfield Avenue, Detroit, MI 48201, USA.
| |
Collapse
|
46
|
Biotechnological challenges of bioartificial kidney engineering. Biotechnol Adv 2014; 32:1317-1327. [PMID: 25135479 DOI: 10.1016/j.biotechadv.2014.08.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 08/05/2014] [Accepted: 08/09/2014] [Indexed: 12/14/2022]
Abstract
With the world-wide increase of patients with renal failure, the development of functional renal replacement therapies have gained significant interest and novel technologies are rapidly evolving. Currently used renal replacement therapies insufficiently remove accumulating waste products, resulting in the uremic syndrome. A more preferred treatment option is kidney transplantation, but the shortage of donor organs and the increasing number of patients waiting for a transplant warrant the development of novel technologies. The bioartificial kidney (BAK) is such promising biotechnological approach to replace essential renal functions together with the active secretion of waste products. The development of the BAK requires a multidisciplinary approach and evolves at the intersection of regenerative medicine and renal replacement therapy. Here we provide a concise review embracing a compact historical overview of bioartificial kidney development and highlighting the current state-of-the-art, including implementation of living-membranes and the relevance of extracellular matrices. We focus further on the choice of relevant renal epithelial cell lines versus the use of stem cells and co-cultures that need to be implemented in a suitable device. Moreover, the future of the BAK in regenerative nephrology is discussed.
Collapse
|
47
|
Schaap-Oziemlak AM, Kühn PT, van Kooten TG, van Rijn P. Biomaterial–stem cell interactions and their impact on stem cell response. RSC Adv 2014. [DOI: 10.1039/c4ra07915a] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
In this review, current research in the field of biomaterial properties for directing stem cells are discussed and placed in a critical perspective.
Collapse
Affiliation(s)
- Aneta M. Schaap-Oziemlak
- Department of Biomedical Engineering-FB40
- W.J. Kolff Institute for Biomedical Engineering and Materials Science
- University of Groningen
- University Medical Center Groningen
- 9713 AVGroningen, The Netherlands
| | - Philipp T. Kühn
- Department of Biomedical Engineering-FB40
- W.J. Kolff Institute for Biomedical Engineering and Materials Science
- University of Groningen
- University Medical Center Groningen
- 9713 AVGroningen, The Netherlands
| | - Theo G. van Kooten
- Department of Biomedical Engineering-FB40
- W.J. Kolff Institute for Biomedical Engineering and Materials Science
- University of Groningen
- University Medical Center Groningen
- 9713 AVGroningen, The Netherlands
| | - Patrick van Rijn
- Department of Biomedical Engineering-FB40
- W.J. Kolff Institute for Biomedical Engineering and Materials Science
- University of Groningen
- University Medical Center Groningen
- 9713 AVGroningen, The Netherlands
| |
Collapse
|