1
|
Xue Z, Xuan H, Lau K, Su Y, Wegener M, Li K, Turner L, Adams M, Shi X, Wen H. Expression of ENL YEATS domain tumor mutations in nephrogenic or stromal lineage impairs kidney development. Nat Commun 2025; 16:2531. [PMID: 40087269 PMCID: PMC11909213 DOI: 10.1038/s41467-025-57926-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 03/05/2025] [Indexed: 03/17/2025] Open
Abstract
Recurrent gain-of-function mutations in the histone reader protein ENL have been identified in Wilms tumor, the most prevalent pediatric kidney cancer. However, their pathological significance in kidney development and tumorigenesis in vivo remains elusive. Here, we generate mouse models mimicking ENL tumor (ENLT) mutations and show that heterozygous mutant expression in Six2+ nephrogenic or Foxd1+ stromal lineages leads to severe, lineage-specific kidney defects, both resulting in neonatal lethality. Six2-ENLT mutant kidneys display compromised cap mesenchyme, scant nephron tubules, and cystic glomeruli, indicative of premature progenitor commitment and blocked differentiation. Bulk and spatial transcriptomic analyses reveal aberrant activation of Hox and Wnt signaling genes in mutant nephrogenic cells. In contrast, Foxd1-ENLT mutant kidneys exhibit expansion in renal capsule and cap mesenchyme, with dysregulated stromal gene expression affecting stroma-epithelium crosstalk. Our findings uncover distinct pathways through which ENL mutations disrupt nephrogenesis, providing a foundation for further investigations into their role in tumorigenesis.
Collapse
Affiliation(s)
- Zhaoyu Xue
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, 49503, USA
| | - Hongwen Xuan
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, 49503, USA
| | - Kin Lau
- Bioinformatics and Biostatistics Core, Van Andel Institute, Grand Rapids, MI, 49503, USA
| | - Yangzhou Su
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, 49503, USA
| | - Marc Wegener
- Genomics Core, Van Andel Institute, Grand Rapids, MI, 49503, USA
| | - Kuai Li
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, 49503, USA
| | - Lisa Turner
- Pathology Core, Van Andel Institute, Grand Rapids, MI, 49503, USA
| | - Marie Adams
- Genomics Core, Van Andel Institute, Grand Rapids, MI, 49503, USA
| | - Xiaobing Shi
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, 49503, USA
| | - Hong Wen
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, 49503, USA.
| |
Collapse
|
2
|
Kazeminia S, Eirin A. Role of mitochondria in endogenous renal repair. Clin Sci (Lond) 2024; 138:963-973. [PMID: 39076039 PMCID: PMC11410300 DOI: 10.1042/cs20231331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/03/2024] [Accepted: 07/15/2024] [Indexed: 07/31/2024]
Abstract
Renal tubules have potential to regenerate and repair after mild-to-moderate injury. Proliferation of tubular epithelial cells represents the initial step of this reparative process. Although for many years, it was believed that proliferating cells originated from a pre-existing intra-tubular stem cell population, there is now consensus that surviving tubular epithelial cells acquire progenitor properties to regenerate the damaged kidney. Scattered tubular-like cells (STCs) are dedifferentiated adult renal tubular epithelial cells that arise upon injury and contribute to renal self-healing and recovery by replacing lost neighboring tubular epithelial cells. These cells are characterized by the co-expression of the stem cell surface markers CD133 and CD24, as well as mesenchymal and kidney injury markers. Previous studies have shown that exogenous delivery of STCs ameliorates renal injury and dysfunction in murine models of acute kidney injury, underscoring the regenerative potential of this endogenous repair system. Although STCs contain fewer mitochondria than their surrounding terminally differentiated tubular epithelial cells, these organelles modulate several important cellular functions, and their integrity and function are critical to preserve the reparative capacity of STCs. Recent data suggest that the microenviroment induced by cardiovascular risk factors, such as obesity, hypertension, and renal ischemia may compromise STC mitochondrial integrity and function, limiting the capacity of these cells to repair injured renal tubules. This review summarizes current knowledge of the contribution of STCs to kidney repair and discusses recent insight into the key role of mitochondria in modulating STC function and their vulnerability in the setting of cardiovascular disease.
Collapse
Affiliation(s)
- Sara Kazeminia
- Department of Internal Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, U.S.A
| | - Alfonso Eirin
- Department of Internal Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, U.S.A
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, U.S.A
| |
Collapse
|
3
|
Bahrami M, Darabi S, Roozbahany NA, Abbaszadeh HA, Moghadasali R. Great potential of renal progenitor cells in kidney: From the development to clinic. Exp Cell Res 2024; 434:113875. [PMID: 38092345 DOI: 10.1016/j.yexcr.2023.113875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/02/2023] [Accepted: 12/03/2023] [Indexed: 12/23/2023]
Abstract
The mammalian renal organ represents a pinnacle of complexity, housing functional filtering units known as nephrons. During embryogenesis, the depletion of niches containing renal progenitor cells (RPCs) and the subsequent incapacity of adult kidneys to generate new nephrons have prompted the formulation of protocols aimed at isolating residual RPCs from mature kidneys and inducing their generation from diverse cell sources, notably pluripotent stem cells. Recent strides in the realm of regenerative medicine and the repair of tissues using stem cells have unveiled critical signaling pathways essential for the maintenance and generation of human RPCs in vitro. These findings have ushered in a new era for exploring novel strategies for renal protection. The present investigation delves into potential transcription factors and signaling cascades implicated in the realm of renal progenitor cells, focusing on their protection and differentiation. The discourse herein elucidates contemporary research endeavors dedicated to the acquisition of progenitor cells, offering crucial insights into the developmental mechanisms of these cells within the renal milieu and paving the way for the formulation of innovative treatment modalities.
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
| | - Shahram Darabi
- Cellular and Molecular Research Center, Research Institute for Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | | | - Hojjat Allah Abbaszadeh
- Laser Applications in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Reza Moghadasali
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
4
|
Lindoso RS, Collino F, Kasai-Brunswick TH, Costa MR, Verdoorn KS, Einicker-Lamas M, Vieira-Beiral HJ, Wessely O, Vieyra A. Resident Stem Cells in Kidney Tissue. RESIDENT STEM CELLS AND REGENERATIVE THERAPY 2024:159-203. [DOI: 10.1016/b978-0-443-15289-4.00009-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
5
|
Omer D, Zontag OC, Gnatek Y, Harari-Steinberg O, Pleniceanu O, Namestnikov M, Cohen AH, Nissim-Rafinia M, Tam G, Kalisky T, Meshorer E, Dekel B. OCT4 induces long-lived dedifferentiated kidney progenitors poised to redifferentiate in 3D kidney spheroids. Mol Ther Methods Clin Dev 2023; 29:329-346. [PMID: 37214315 PMCID: PMC10193171 DOI: 10.1016/j.omtm.2023.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 04/18/2023] [Indexed: 05/24/2023]
Abstract
Upscaling of kidney epithelial cells is crucial for renal regenerative medicine. Nonetheless, the adult kidney lacks a distinct stem cell hierarchy, limiting the ability to long-term propagate clonal populations of primary cells that retain renal identity. Toward this goal, we tested the paradigm of shifting the balance between differentiation and stemness in the kidney by introducing a single pluripotency factor, OCT4. Here we show that ectopic expression of OCT4 in human adult kidney epithelial cells (hKEpC) induces the cells to dedifferentiate, stably proliferate, and clonally emerge over many generations. Control hKEpC dedifferentiate, assume fibroblastic morphology, and completely lose clonogenic capacity. Analysis of gene expression and histone methylation patterns revealed that OCT4 represses the HNF1B gene module, which is critical for kidney epithelial differentiation, and concomitantly activates stemness-related pathways. OCT4-hKEpC can be long-term expanded in the dedifferentiated state that is primed for renal differentiation. Thus, when expanded OCT4-hKEpC are grown as kidney spheroids (OCT4-kSPH), they reactivate the HNF1B gene signature, redifferentiate, and efficiently generate renal structures in vivo. Hence, changes occurring in the cellular state of hKEpC following OCT4 induction, long-term propagation, and 3D aggregation afford rapid scale-up technology of primary renal tissue-forming cells.
Collapse
Affiliation(s)
- Dorit Omer
- Pediatric Stem Cell Research Institute, Edmond & Lily Safra Children’s Hospital, Sheba Medical Center, Tel Hashomer 5262000, Israel
- Sagol Center for Regenerative Medicine, School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Osnat Cohen Zontag
- Pediatric Stem Cell Research Institute, Edmond & Lily Safra Children’s Hospital, Sheba Medical Center, Tel Hashomer 5262000, Israel
- Sagol Center for Regenerative Medicine, School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yehudit Gnatek
- Pediatric Stem Cell Research Institute, Edmond & Lily Safra Children’s Hospital, Sheba Medical Center, Tel Hashomer 5262000, Israel
- Sagol Center for Regenerative Medicine, School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Orit Harari-Steinberg
- Pediatric Stem Cell Research Institute, Edmond & Lily Safra Children’s Hospital, Sheba Medical Center, Tel Hashomer 5262000, Israel
- Sagol Center for Regenerative Medicine, School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Oren Pleniceanu
- Pediatric Stem Cell Research Institute, Edmond & Lily Safra Children’s Hospital, Sheba Medical Center, Tel Hashomer 5262000, Israel
- Sagol Center for Regenerative Medicine, School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Michael Namestnikov
- Pediatric Stem Cell Research Institute, Edmond & Lily Safra Children’s Hospital, Sheba Medical Center, Tel Hashomer 5262000, Israel
- Sagol Center for Regenerative Medicine, School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ayelet-Hashahar Cohen
- Department of Genetics, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Jerusalem 9190401, Israel
| | - Malka Nissim-Rafinia
- Department of Genetics, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Jerusalem 9190401, Israel
| | - Gal Tam
- Faculty of Engineering and Nanotechnology Institute, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Tomer Kalisky
- Faculty of Engineering and Nanotechnology Institute, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Eran Meshorer
- Department of Genetics, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Jerusalem 9190401, Israel
- Edmond & Lily Safra Center for Brain Sciences (ELSC), The Hebrew University of Jerusalem, Edmond J. Safra Campus, Jerusalem 9190401, Israel
| | - Benjamin Dekel
- Pediatric Stem Cell Research Institute, Edmond & Lily Safra Children’s Hospital, Sheba Medical Center, Tel Hashomer 5262000, Israel
- Sagol Center for Regenerative Medicine, School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Division of Pediatric Nephrology, Edmond & Lily Safra Children’s Hospital, Sheba Medical Center, Tel Hashomer 5262000, Israel
| |
Collapse
|
6
|
Wang M, Zhou X, Zhou S, Wang M, Jiang J, Wu W, Liu T, Xu W, Zhang J, Liu D, Zou Y, Qiu W, Zhang M, Liu W, Li Z, Wang D, Li T, Li J, Liu W, Yang L, Lei M. Mechanical force drives the initial mesenchymal-epithelial interaction during skin organoid development. Theranostics 2023; 13:2930-2945. [PMID: 37284452 PMCID: PMC10240816 DOI: 10.7150/thno.83217] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 04/25/2023] [Indexed: 06/08/2023] Open
Abstract
Rationale: Stem cells self-organize to form organoids that generate mini-organs that resemble the physiologically-developed ones. The mechanism by which the stem cells acquire the initial potential for generating mini-organs remains elusive. Here we used skin organoids as an example to study how mechanical force drives initial epidermal-dermal interaction which potentiates skin organoids to regenerate hair follicles. Methods: Live imaging analysis, single-cell RNA-sequencing analysis, and immunofluorescence were used to analyze the contractile force of dermal cells in skin organoids. Bulk RNA-sequencing analysis, calcium probe detection, and functional perturbations were used to verify that calcium signaling pathways respond to the contractile force of dermal cells. In vitro mechanical loading experiment was used to prove that the stretching force triggers the epidermal Piezo1 expression which negatively regulates dermal cell attachment. Transplantation assay was used to test the regenerative ability of skin organoids. Results: We found that dermal cell-derived contraction force drives the movement of dermal cells surrounding the epidermal aggregates to trigger initial mesenchymal-epithelial interaction (MEI). In response to dermal cell contraction force, the arrangement of the dermal cytoskeleton was negatively regulated by the calcium signaling pathway which further influences dermal-epidermal attachment. The native contraction force generated from the dermal cell movement exerts a stretching force on the adjacent epidermal cells, activating the stretching force sensor Piezo1 in the epidermal basal cells during organoid culture. Epidermal Piezo1 in turn drives strong MEI to negatively regulate dermal cell attachment. Proper initial MEI by mechanical-chemical coupling during organoid culture is required for hair regeneration upon transplantation of the skin organoids into the back of the nude mice. Conclusion: Our study demonstrated that mechanical-chemical cascade drives the initial event of MEI during skin organoid development, which is fundamental to the organoid, developmental, and regenerative biology fields.
Collapse
Affiliation(s)
- Mengyue Wang
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Xun Zhou
- Department of Dermatology and Cosmetology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing 400021, China
| | - Siyi Zhou
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Miaomiao Wang
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Jingwei Jiang
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Wang Wu
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
- Three Gorges Hospital, Chongqing University, Chongqing 404000, China
| | - Tiantian Liu
- Three Gorges Hospital, Chongqing University, Chongqing 404000, China
| | - Wei Xu
- Department of Dermatology and Cosmetology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing 400021, China
| | - Jinwei Zhang
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
- Department of Dermatology and Cosmetology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing 400021, China
| | - Deming Liu
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
- Department of Dermatology and Cosmetology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing 400021, China
| | - Yi Zou
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
- Department of Burns and Plastic Surgery, Wuhan General Hospital of Chinese People's Liberation Army, Wuhan 430000, China
| | - Weiming Qiu
- Department of Burns and Plastic Surgery, Wuhan General Hospital of Chinese People's Liberation Army, Wuhan 430000, China
| | - Man Zhang
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Weiwei Liu
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Zeming Li
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Dehuan Wang
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Tingting Li
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Ji Li
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Wanqian Liu
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Li Yang
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Mingxing Lei
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| |
Collapse
|
7
|
Veljačić Visković D, Lozić M, Vukoja M, Šoljić V, Vukojević K, Glavina Durdov M, Filipović N, Lozić B. Spatio-Temporal Expression Pattern of CAKUT Candidate Genes DLG1 and KIF12 during Human Kidney Development. Biomolecules 2023; 13:biom13020340. [PMID: 36830709 PMCID: PMC9953652 DOI: 10.3390/biom13020340] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/05/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023] Open
Abstract
We aimed to investigate expression of the novel susceptibility genes for CAKUT, DLG1 and KIF12, proposed by a systematic in silico approach, in developing and postnatal healthy human kidneys to provide information about their spatiotemporal expression pattern. We analyzed expression of their protein products by immunohistochemistry and immunofluorescence and quantified relative mRNA levels by RT-qPCR. Statistically significant differences in expression patterns were observed between certain developmental stages. Strong expression of DLG1 was observed in the developing kidney, with a gradual decrease from the first phase of kidney development (Ph1) until the third phase (Ph3), when most nephrons are formed; at later stages, the highest expression was observed in the tubules. KIF12 was highly expressed in the developing structures, especially in Ph1, with a gradual decrease until the postnatal phase, which would indicate a significant role in nephrogenesis. Co-localization of DLG1 and KIF12 was pronounced in Ph1, especially on the apical side of the tubular epithelial cells. Thereafter, their expression gradually became weaker and was only visible as punctate staining in Ph4. The direct association of DLG1 with KIF12 as control genes of normal kidney development may reveal their new functional aspect in renal tubular epithelial cells.
Collapse
Affiliation(s)
| | - Mirela Lozić
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2, 21 000 Split, Croatia
- Correspondence: ; Tel.: +385-21-557-800
| | - Martina Vukoja
- Laboratory of Morphology, Department of Histology and Embryology, School of Medicine, University of Mostar, 88 000 Mostar, Bosnia and Herzegovina
| | - Violeta Šoljić
- Laboratory of Morphology, Department of Histology and Embryology, School of Medicine, University of Mostar, 88 000 Mostar, Bosnia and Herzegovina
- Faculty of Health Studies, University of Mostar, 88 000 Mostar, Bosnia and Herzegovina
| | - Katarina Vukojević
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2, 21 000 Split, Croatia
- Laboratory of Morphology, Department of Histology and Embryology, School of Medicine, University of Mostar, 88 000 Mostar, Bosnia and Herzegovina
- Faculty of Health Studies, University of Mostar, 88 000 Mostar, Bosnia and Herzegovina
- Department of Anatomy, University of Mostar, 88 000 Mostar, Bosnia and Herzegovina
- Center for Translational Research in Biomedicine, University of Split School of Medicine, 21 000 Split, Croatia
| | - Merica Glavina Durdov
- Department of Pathology, University Hospital Split, 21 000 Split, Croatia
- School of Medicine, University of Split, Šoltanska 2, 21 000 Split, Croatia
| | - Natalija Filipović
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2, 21 000 Split, Croatia
- Department of Anatomy, University of Mostar, 88 000 Mostar, Bosnia and Herzegovina
- Center for Translational Research in Biomedicine, University of Split School of Medicine, 21 000 Split, Croatia
| | - Bernarda Lozić
- Paediatric Diseases Department, University Hospital of Split, Spinčićeva 1, 21 000 Split, Croatia
- School of Medicine, University of Split, Šoltanska 2, 21 000 Split, Croatia
| |
Collapse
|
8
|
Zhang Y, Xie Y, Lu W, Xu S, Wang X, Zhou W, Zhang Y, Ding X, Zhao S. Identification of resident progenitors labeled with Top2a responsible for proximal tubular regeneration in ischemia reperfusion-induced acute kidney injury. Cell Signal 2023; 101:110506. [PMID: 36309330 DOI: 10.1016/j.cellsig.2022.110506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 10/19/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND Acute kidney injury is a common fatal disease with complex etiology and limited treatment methods. Proximal tubules (PTs) are the most vulnerable segment. Not only in injured kidneys but also in normal kidneys, shedding of PTs often happens. However, the source cells and mechanism of their regeneration remain unclear. METHODS ScRNA and snRNA sequencing data of acute injured or normal kidney were downloaded from GEO database to identify the candidate biomarker of progenitor of proximal tubules. SLICE algorithm and CytoTRACE analyses were employed to evaluate the stemness of progenitors. Then the repairing trajectory was constructed through pseudotime analyses. SCENIC algorithm was used to detect cell-type-specific regulon. With spatial transcriptome data, the location of progenitors was simulated. Neonatal/ adult/ aged mice and preconditioning AKI mice model and deconvolution of 2 RNA-seq data were employed for validation. RESULTS Through cluster identification, PT cluster expressed Top2a specifically was identified to increase significantly during AKI. With relatively strong stemness, the Top2a-labeled PT cluster tended to be the origin of the repairing trajectory. Moreover, the cluster was regulated by Pbx3-based regulon and possessed great segmental heterogeneity. Changes of Top2a between neonatal and aged mice and among AKI models validated the progenitor role of Top2a-labeled cluster. CONCLUSIONS Our study provided transcriptomic evidence that resident proximal tubular progenitors labeled with Top2a participated in regeneration. Considering the segmental heterogeneity, we find that there is a group of reserve progenitor cells in each tubular segment. When AKI occurs, the reserve progenitors of each tubular segment proliferate and replenish first, and PT-progenitors, a cluster with no obvious PT markers replenish each subpopulation of the reserve cells.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Nephrology, Zhongshan Hospital, Fudan University
| | - Yeqing Xie
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney Disease; Kidney and Dialysis Institute of Shanghai; Kidney and Blood Purification Key Laboratory of Shanghai
| | - Wei Lu
- Department of Nephrology, Zhongshan Hospital, Fudan University
| | - Sujuan Xu
- Department of Nephrology, Zhongshan Hospital, Fudan University
| | - Xiaoyan Wang
- Department of Nephrology, Zhongshan Hospital, Fudan University
| | - Weiran Zhou
- Department of Nephrology, Zhongshan Hospital, Fudan University
| | - Yingjia Zhang
- Department of Nephrology, Zhongshan Hospital, Fudan University
| | - Xiaoqiang Ding
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney Disease; Kidney and Dialysis Institute of Shanghai; Kidney and Blood Purification Key Laboratory of Shanghai.
| | - Shuan Zhao
- Department of Nephrology, Zhongshan Hospital, Fudan University; Shanghai Medical Center of Kidney Disease; Kidney and Dialysis Institute of Shanghai; Kidney and Blood Purification Key Laboratory of Shanghai.
| |
Collapse
|
9
|
Kageyama T, Shimizu A, Anakama R, Nakajima R, Suzuki K, Okubo Y, Fukuda J. Reprogramming of three-dimensional microenvironments for in vitro hair follicle induction. SCIENCE ADVANCES 2022; 8:eadd4603. [PMID: 36269827 PMCID: PMC9586475 DOI: 10.1126/sciadv.add4603] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 09/02/2022] [Indexed: 06/08/2023]
Abstract
During embryonic development, reciprocal interactions between epidermal and mesenchymal layers trigger hair follicle morphogenesis. This study revealed that microenvironmental reprogramming via control over these interactions enabled hair follicle induction in vitro. A key approach is to modulate spatial distributions of epithelial and mesenchymal cells in their spontaneous organization. The de novo hair follicles with typical morphological features emerged in aggregates of the two cell types, termed hair follicloids, and hair shafts sprouted with near 100% efficiency in vitro. The hair shaft length reached ~3 mm in culture. Typical trichogenic signaling pathways were up-regulated in hair follicloids. Owing to replication of hair follicle morphogenesis in vitro, melanosome production and transportation were also monitored in the hair bulb region. This in vitro hair follicle model might be valuable for better understanding hair follicle induction, evaluating hair growth and inhibition of hair growth by drugs, and modeling gray hairs in a well-defined environment.
Collapse
Affiliation(s)
- Tatsuto Kageyama
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama, Kanagawa 240-8501, Japan
- Kanagawa Institute of Industrial Science and Technology, 3-2-1 Sakado Takatsu-ku, Kawasaki, Kanagawa 213-0012, Japan
- Japan Science and Technology Agency (JST)-PRESTO, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
| | - Akihiro Shimizu
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama, Kanagawa 240-8501, Japan
| | - Riki Anakama
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama, Kanagawa 240-8501, Japan
| | - Rikuma Nakajima
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama, Kanagawa 240-8501, Japan
| | - Kohei Suzuki
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama, Kanagawa 240-8501, Japan
- Nissan Chemical Corporation, 2-5-1 Nihonbashi, Chuo-ku, Tokyo 103-6119, Japan
| | - Yusuke Okubo
- Division of Cellular and Molecular Toxicology, Center for Biological Safety and Research, National Institute of Health Sciences, 3-25-26 Tono-machi, Kawasaki-ku, Kawasaki, Kanagawa 210-9501, Japan
| | - Junji Fukuda
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama, Kanagawa 240-8501, Japan
- Kanagawa Institute of Industrial Science and Technology, 3-2-1 Sakado Takatsu-ku, Kawasaki, Kanagawa 213-0012, Japan
| |
Collapse
|
10
|
Raghubar AM, Roberts MJ, Wood S, Healy HG, Kassianos AJ, Mallett AJ. Cellular milieu in clear cell renal cell carcinoma. Front Oncol 2022; 12:943583. [PMID: 36313721 PMCID: PMC9614096 DOI: 10.3389/fonc.2022.943583] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 09/21/2022] [Indexed: 11/13/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is globally the most prevalent renal cancer. The cells of origin in ccRCC have been identified as proximal tubular epithelial cells (PTEC); however, the transcriptomic pathways resulting in the transition from normal to malignant PTEC state have remained unclear. Immunotherapy targeting checkpoints have revolutionized the management of ccRCC, but a sustained clinical response is achieved in only a minority of ccRCC patients. This indicates that our understanding of the mechanisms involved in the malignant transition and resistance to immune checkpoint therapy in ccRCC is unclear. This review examines recent single-cell transcriptomics studies of ccRCC to clarify the transition of PTEC in ccRCC development, and the immune cell types, states, and interactions that may limit the response to targeted immune therapy, and finally suggests stromal cells as key drivers in recurrent and locally invasive ccRCC. These and future single-cell transcriptomics studies will continue to clarify the cellular milieu in the ccRCC microenvironment, thus defining actional clinical, therapeutic, and prognostic characteristics of ccRCC.
Collapse
Affiliation(s)
- Arti M. Raghubar
- Kidney Health Service, Royal Brisbane and Women’s Hospital, Herston, QLD, Australia
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Health Support Queensland, Herston, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
- Anatomical Pathology, Pathology Queensland, Health Support Queensland, Herston, QLD, Australia
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Matthew J. Roberts
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
- Department of Urology, Royal Brisbane and Women’s Hospital, Brisbane, QLD, Australia
- Department of Urology, Redcliffe Hospital, Redcliffe, QLD, Australia
- Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia
| | - Simon Wood
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
- Department of Urology, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Helen G. Healy
- Kidney Health Service, Royal Brisbane and Women’s Hospital, Herston, QLD, Australia
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Health Support Queensland, Herston, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Andrew J. Kassianos
- Kidney Health Service, Royal Brisbane and Women’s Hospital, Herston, QLD, Australia
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Health Support Queensland, Herston, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Andrew J. Mallett
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
- College of Medicine & Dentistry, James Cook University, Townsville, QLD, Australia
- Department of Renal Medicine, Townsville University Hospital, Townsville, QLD, Australia
- *Correspondence: Andrew J. Mallett,
| |
Collapse
|
11
|
Kagan M, Pleniceanu O, Vivante A. The genetic basis of congenital anomalies of the kidney and urinary tract. Pediatr Nephrol 2022; 37:2231-2243. [PMID: 35122119 DOI: 10.1007/s00467-021-05420-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 10/19/2022]
Abstract
During the past decades, remarkable progress has been made in our understanding of the molecular basis of kidney diseases, as well as in the ability to pinpoint disease-causing genetic changes. Congenital anomalies of the kidney and urinary tract (CAKUT) are remarkably diverse, and may be either isolated to the kidney or involve other systems, and are notorious in their variable genotype-phenotype correlations. Genetic conditions underlying CAKUT are individually rare, but collectively contribute to disease etiology in ~ 16% of children with CAKUT. In this review, we will discuss basic concepts of kidney development and genetics, common causes of monogenic CAKUT, and the approach to diagnosing and managing a patient with suspected monogenic CAKUT. Altogether, the concepts presented herein represent an introduction to the emergence of nephrogenetics, a fast-growing multi-disciplinary field that is focused on deciphering the causes and manifestations of genetic kidney diseases as well as providing the framework for managing patients with genetic forms of CAKUT.
Collapse
Affiliation(s)
- Maayan Kagan
- Pediatric Department B and Pediatric Nephrology Unit, Edmond and Lily Safra Children's Hospital, Sackler Faculty of Medicine, Sheba Medical Center, Tel Hashomer, 5265601, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Oren Pleniceanu
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Kidney Research Lab, The Institute of Nephrology and Hypertension, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Asaf Vivante
- Pediatric Department B and Pediatric Nephrology Unit, Edmond and Lily Safra Children's Hospital, Sackler Faculty of Medicine, Sheba Medical Center, Tel Hashomer, 5265601, Ramat Gan, Israel. .,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel. .,Talpiot Medical Leadership Program, Tel HaShomer, Ramat Gan, Israel.
| |
Collapse
|
12
|
Gao C, Zhang L, Chen E, Zhang W. Aqp2 + Progenitor Cells Maintain and Repair Distal Renal Segments. J Am Soc Nephrol 2022; 33:1357-1376. [PMID: 35318267 PMCID: PMC9257824 DOI: 10.1681/asn.2021081105] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 03/08/2022] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Adult progenitor cells presumably demonstrate clonogenicity, self-renewal, and multipotentiality, and can regenerate cells under various conditions. Definitive evidence demonstrating the existence of such progenitor cells in adult mammalian kidneys is lacking. METHOD We performed in vivo lineage tracing and thymidine analogue labeling using adult tamoxifen-inducible (Aqp2ECE/+ RFP/+, Aqp2ECE/+ Brainbow/+, and Aqp2ECE/+ Brainbow/Brainbow) and WT mice. The tamoxifen-inducible mice were analyzed between 1 and 300 days postinduction. Alternatively, WT and tamoxifen-induced mice were subjected to unilateral ureteral obstruction and thymidine analogue labeling and analyzed 2-14 days post-surgery. Multiple cell-specific markers were used for high-resolution immunofluorescence confocal microscopy to identify the cell types derived from Aqp2+ cells. RESULTS Like their embryonic counterparts, adult cells expressing Aqp2 and V-ATPase subunits B1 and B2 (Aqp2+ B1B2+) are the potential Aqp2+ progenitor cells (APs). Adult APs rarely divide to generate daughter cells, either maintaining the property of the AP (self-renewal) or differentiating into DCT2/CNT/CD cells (multipotentiality), forming single cell-derived, multiple-cell clones (clonogenicity) during tissue maintenance. APs selectively and continuously regenerate DCT2/CNT/CD cells in response to injury resulting from ureteral ligation. AP proliferation demonstrated direct correlation with Notch activation and was inversely correlated with development of kidney fibrosis. Derivation of both intercalated and DCT2 cells was found to be cell division-dependent and -independent, most likely through AP differentiation which requires cell division and through direct conversion of APs and/or regular principal cells without cell division, respectively. CONCLUSION Our study demonstrates that Aqp2+ B1B2+ cells behave as adult APs to maintain and repair DCT2/CNT1/CNT2/CD segments.
Collapse
Affiliation(s)
- Chao Gao
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York
| | - Long Zhang
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York
| | - Enuo Chen
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York
| | - Wenzheng Zhang
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York
| |
Collapse
|
13
|
Zheng CM, Chiu IJ, Chen YW, Hsu YH, Hung LY, Wu MY, Lin YF, Liao CT, Hung YP, Tsai CC, Cherng YG, Wu MS. Allogeneic adipose tissue-derived stem cells ELIXCYTE ® in chronic kidney disease: A phase I study assessing safety and clinical feasibility. J Cell Mol Med 2022; 26:2972-2980. [PMID: 35415928 PMCID: PMC9097837 DOI: 10.1111/jcmm.17310] [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: 07/03/2021] [Revised: 03/07/2022] [Accepted: 03/14/2022] [Indexed: 11/27/2022] Open
Abstract
The purpose of this phase I clinical trial is to assess the safety and tolerability of allogeneic adipose tissue‐derived stem cells (ADSCs) among chronic kidney disease (CKD) patients. 12 eligible CKD patients with an estimated glomerular filtration rate (eGFR) of 15–44 ml/min/1.73 m2 received one dose of intravenous allogeneic ADSCs (ELIXCYTE®), as 3 groups: 3 low dose (6.4 × 107 cells in total of 8 ml), 3 middle dose (19.2 × 107 cells in total of 24 ml) and 6 high dose (32.0 × 107 cells in total of 40 ml) of ELIXCYTE® and evaluated after 48 weeks. Primary endpoint was the safety profiles in terms of incidence of adverse events (AEs) and serious adverse event (SAE). Two subjects in high dose group experienced a total of 2 treatment‐related AEs which are Grade 1 slow speech and Grade 1 bradyphrenia after the infusion. One subject in middle dose group experienced an SAE unlikely related to treatment, grade 2 proteinuria. No fatal AE was reported in this study. An increase in eGFR was observed in 7 out of 12 subjects (58%) at Week 24 and in 6 of 12 subjects (50%) by Week 48. By Week 24, an increase in eGFR by more than 20% among all CKD patients with baseline eGFR ≧ 30 ml/min/1.73 m2 as compared to only 2 subjects in baseline eGFR < 30 ml/min/1.73 m2 group. No significant reduction in proteinuria was noted among all subjects. This phase I trial demonstrated single‐dose intravenous ELIXCYTE was well tolerated in moderate‐to‐severe CKD patients and its preliminary efficacy warrants future studies.
Collapse
Affiliation(s)
- Cai-Mei Zheng
- Division of Nephrology, Department of Internal Medicine, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan.,Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Taipei Medical University-Research Center of Urology and Kidney (TMU-RCUK), School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - I-Jen Chiu
- Division of Nephrology, Department of Internal Medicine, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan.,Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Taipei Medical University-Research Center of Urology and Kidney (TMU-RCUK), School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Wei Chen
- Division of Nephrology, Department of Internal Medicine, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan.,Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Taipei Medical University-Research Center of Urology and Kidney (TMU-RCUK), School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,College of Medicine, Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yung-Ho Hsu
- Division of Nephrology, Department of Internal Medicine, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan.,Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Taipei Medical University-Research Center of Urology and Kidney (TMU-RCUK), School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Nephrology, Department of Internal Medicine, Hsin Kuo Min Hospital, Taipei Medical University, Taoyuan City, Taiwan
| | - Lie-Yee Hung
- Division of Nephrology, Department of Internal Medicine, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan.,Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Taipei Medical University-Research Center of Urology and Kidney (TMU-RCUK), School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Mei-Yi Wu
- Division of Nephrology, Department of Internal Medicine, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan.,Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Taipei Medical University-Research Center of Urology and Kidney (TMU-RCUK), School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,College of Public Health, Institute of Epidemiology and Preventive Medicine, National Taiwan University, Taipei, Taiwan
| | - Yuh-Feng Lin
- Division of Nephrology, Department of Internal Medicine, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan.,Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Taipei Medical University-Research Center of Urology and Kidney (TMU-RCUK), School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,College of Medicine, Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chia-Te Liao
- Division of Nephrology, Department of Internal Medicine, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan.,Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Taipei Medical University-Research Center of Urology and Kidney (TMU-RCUK), School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | | | | | - Yih-Giun Cherng
- Department of Anesthesiology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Mai-Szu Wu
- Division of Nephrology, Department of Internal Medicine, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan.,Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Taipei Medical University-Research Center of Urology and Kidney (TMU-RCUK), School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
14
|
Weng X, Li J, Guan Q, Zhao H, Wang Z, Gleave ME, Nguan CY, Du C. The functions of clusterin in renal mesenchymal stromal cells: Promotion of cell growth and regulation of macrophage activation. Exp Cell Res 2022; 413:113081. [PMID: 35218723 DOI: 10.1016/j.yexcr.2022.113081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 01/31/2022] [Accepted: 02/15/2022] [Indexed: 12/24/2022]
Abstract
Clusterin (CLU) increases resistance to renal ischemia-reperfusion injury and promotes renal tissue repair. However, the mechanisms underlying of the renal protection of CLU remain unknown. Mesenchymal stromal cells (MSCs) may contribute to kidney cell turnover and injury repair. This study investigated the in vitro functions of CLU in kidney mesenchymal stromal cells (KMSCs). KMSCs were grown in plastic culture plates. Cell surface markers, apoptosis and phagocytosis were determined by flow cytometry, and CLU protein by Western blot. There were no differences in the expression of MSC markers (positive: CD133, Sca-1, CD44, CD117 and NG2, and negative: CD34, CD45, CD163, CD41, CD276, CD138, CD79a, CD146 and CD140b) and in the trilineage differentiation to chondrocytes, adipocytes and osteocytes between wild type (WT) and CLU knockout (KO) KMSCs. CLU was expressed intracellularly and secreted by WT KMSCs, and it was up-regulated by hypoxia. CLU did not prevent hypoxia-induced cell apoptosis but promoted cell growth in KMSC cultures. Furthermore, incubation with CLU-containing culture medium from WT KMSCs increased CD206 expression and phagocytic capacity of macrophages. In conclusion, our data for the first time demonstrate the function of CLU in the promotion of KMSCs proliferation, and it may be required for KMSCs-regulated macrophage M2 polarization and phagocytic activity.
Collapse
Affiliation(s)
- Xiaodong Weng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China; Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Jing Li
- Department of Ophthamology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430060, China
| | - Qiunong Guan
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Haimei Zhao
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada; College of Traditional Chinese Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, Jiangxi Province, China
| | - Zihuan Wang
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada; First Clinical Medical School, Southern Medical University, Guangzhou, 510000, China
| | - Martin E Gleave
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Christopher Yc Nguan
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Caigan Du
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
15
|
Zhang W, Gao C, Tsilosani A, Samarakoon R, Plews R, Higgins P. Potential renal stem/progenitor cells identified by in vivo lineage tracing. Am J Physiol Renal Physiol 2022; 322:F379-F391. [PMID: 35100814 PMCID: PMC8934668 DOI: 10.1152/ajprenal.00326.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Mammalian kidneys consist of more than 30 different types of cells. A challenging task is to identify and characterize the stem/progenitor subpopulations that establish the lineage relationships among these cellular elements during nephrogenesis in the embryonic and neonate kidneys and during tissue homeostasis and/or injury repair in the mature kidney. Moreover, the potential clinical utility of stem/progenitor cells holds promise for development of new regenerative medicine approaches for the treatment of renal diseases. Stem cells are defined by unlimited self-renewal capacity and pluripotentiality. Progenitor cells have pluripotentiality, but no or limited self-renewal potential. Cre-LoxP-based in vivo genetic lineage tracing is a powerful tool to identify the stem/progenitor cells in their native environment. Hypothetically, this technique enables investigators to accurately track the progeny of a single cell, or a group of cells. The Cre/loxP system has been widely employed to uncover the function of genes in various mammalian tissues and to identify stem/progenitor cells through in vivo lineage tracing analyses. In this review, we summarize the recent advances in the development and characterization of various Cre drivers, and their use in identifying potential renal stem/progenitor cells in both developing and mature mouse kidneys.
Collapse
Affiliation(s)
- Wenzheng Zhang
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Chao Gao
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Akaki Tsilosani
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Rohan Samarakoon
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Robert Plews
- Department of General Surgery, Albany Medical College, Albany, NY, United States
| | - Paul Higgins
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| |
Collapse
|
16
|
Harari-Steinberg O, Omer D, Gnatek Y, Pleniceanu O, Goldberg S, Cohen-Zontag O, Pri-Chen S, Kanter I, Ben Haim N, Becker E, Ankawa R, Fuchs Y, Kalisky T, Dotan Z, Dekel B. Ex Vivo Expanded 3D Human Kidney Spheres Engraft Long Term and Repair Chronic Renal Injury in Mice. Cell Rep 2021; 30:852-869.e4. [PMID: 31968258 DOI: 10.1016/j.celrep.2019.12.047] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 10/04/2019] [Accepted: 12/12/2019] [Indexed: 12/26/2022] Open
Abstract
End-stage renal disease is a worldwide epidemic requiring renal replacement therapy. Harvesting tissue from failing kidneys and autotransplantation of tissue progenitors could theoretically delay the need for dialysis. Here we use healthy and end-stage human adult kidneys to robustly expand proliferative kidney epithelial cells and establish 3D kidney epithelial cultures termed "nephrospheres." Formation of nephrospheres reestablishes renal identity and function in primary cultures. Transplantation into NOD/SCID mice shows that nephrospheres restore self-organogenetic properties lost in monolayer cultures, allowing long-term engraftment as tubular structures, potentially adding nephron segments and demonstrating self-organization as critical to survival. Furthermore, long-term tubular engraftment of nephrospheres is functionally beneficial in murine models of chronic kidney disease. Remarkably, nephrospheres inhibit pro-fibrotic collagen production in cultured fibroblasts via paracrine modulation, while transplanted nephrospheres induce transcriptional signatures of proliferation and release from quiescence, suggesting re-activation of endogenous repair. These data support the use of human nephrospheres for renal cell therapy.
Collapse
Affiliation(s)
- Orit Harari-Steinberg
- Pediatric Stem Cell Research Institute, Edmond and Lily Sara Children's Hospital, Sheba Medical Center, Tel Hashomer, Ramat-Gan, Israel; Pediatric Research Center for Genetics, Development and Environment, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dorit Omer
- Pediatric Stem Cell Research Institute, Edmond and Lily Sara Children's Hospital, Sheba Medical Center, Tel Hashomer, Ramat-Gan, Israel; Pediatric Research Center for Genetics, Development and Environment, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yehudit Gnatek
- Pediatric Stem Cell Research Institute, Edmond and Lily Sara Children's Hospital, Sheba Medical Center, Tel Hashomer, Ramat-Gan, Israel; Pediatric Research Center for Genetics, Development and Environment, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Oren Pleniceanu
- Pediatric Stem Cell Research Institute, Edmond and Lily Sara Children's Hospital, Sheba Medical Center, Tel Hashomer, Ramat-Gan, Israel; Pediatric Research Center for Genetics, Development and Environment, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sanja Goldberg
- Pediatric Stem Cell Research Institute, Edmond and Lily Sara Children's Hospital, Sheba Medical Center, Tel Hashomer, Ramat-Gan, Israel; Pediatric Research Center for Genetics, Development and Environment, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Osnat Cohen-Zontag
- Pediatric Stem Cell Research Institute, Edmond and Lily Sara Children's Hospital, Sheba Medical Center, Tel Hashomer, Ramat-Gan, Israel; Pediatric Research Center for Genetics, Development and Environment, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sara Pri-Chen
- The Maurice and Gabriela Goldschleger Eye Research Institute, Sheba Medical Center, Ramat-Gan, Israel
| | - Itamar Kanter
- Faculty of Engineering and Bar-Ilan Institute of Nanotechnology and Advanced Materials (BINA), Bar-Ilan University, Ramat-Gan, Israel
| | - Nissim Ben Haim
- Faculty of Engineering and Bar-Ilan Institute of Nanotechnology and Advanced Materials (BINA), Bar-Ilan University, Ramat-Gan, Israel
| | - Eli Becker
- Faculty of Engineering and Bar-Ilan Institute of Nanotechnology and Advanced Materials (BINA), Bar-Ilan University, Ramat-Gan, Israel
| | - Roi Ankawa
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion - Israel Institute of Technology, Haifa, Israel
| | - Yaron Fuchs
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion - Israel Institute of Technology, Haifa, Israel
| | - Tomer Kalisky
- Faculty of Engineering and Bar-Ilan Institute of Nanotechnology and Advanced Materials (BINA), Bar-Ilan University, Ramat-Gan, Israel
| | - Zohar Dotan
- Department of Urology, Sheba Medical Center, Tel Hashomer, Ramat-Gan, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Benjamin Dekel
- Pediatric Stem Cell Research Institute, Edmond and Lily Sara Children's Hospital, Sheba Medical Center, Tel Hashomer, Ramat-Gan, Israel; Pediatric Research Center for Genetics, Development and Environment, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Division of Pediatric Nephrology, Safra Children's Hospital, Sheba Medical Center, Ramat-Gan, Israel.
| |
Collapse
|
17
|
Calderon-Margalit R, Efron G, Pleniceanu O, Tzur D, Stern-Zimmer M, Afek A, Erlich T, Derazne E, Kark JD, Keinan-Boker L, Twig G, Vivante A. Congenital Anomalies of the Kidney and Urinary Tract and Adulthood risk of Urinary Tract Cancer. Kidney Int Rep 2021; 6:946-952. [PMID: 33912744 PMCID: PMC8071628 DOI: 10.1016/j.ekir.2021.01.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 12/21/2020] [Accepted: 01/04/2021] [Indexed: 11/13/2022] Open
Abstract
Introduction Congenital anomalies of the kidney and urinary tract (CAKUT) are the most common kidney diseases in childhood. Alterations in genes governing nephrogenesis may cause CAKUT, and in some cases may contribute to development of urinary tract (UT) tumors later in life. We aimed to assess the association between CAKUT and UT cancer in adulthood. Methods We conducted a population-based historical cohort study encompassing 1,510,042 recruits to the Israeli army between 1967 and 1997. CAKUT exposure was determined by army medical coding of CAKUT in childhood. Incidence of UT cancer (kidney, ureter, or bladder) was available through record linkage with the Israeli Cancer Registry. Recruits were followed from the prerecruitment assessment until cancer diagnosis, death, or study termination, in 2012. Cox proportional hazards models were constructed to estimate the hazard ratios (HRs) for UT cancer in participants with vs. without CAKUT. Results During a mean follow-up of 30.4 years, 2959 participants (2573 men and 386 women) developed UT cancer. Men with CAKUT exhibited an increased risk of UT cancer compared with men without CAKUT, yielding an adjusted HR of 1.98 (95% confidence interval [CI] 1.03-3.82). Among women CAKUT was associated with a HR of 5.88 (95% CI 2.19-15.76). Notably, upon stratification according to age of cancer diagnosis, the association between CAKUT and UT cancer was statistically significant only before 45 years of age in women and only after 45 years of age in men. Conclusion CAKUT is associated with a significantly increased risk of UT cancer, although the incidence and absolute risk remained quite low.
Collapse
Affiliation(s)
| | - Gil Efron
- Department of Military Medicine, Hebrew University of Jerusalem, Jerusalem and the Israel Defense Forces Medical Corps, Ramat Gan, Israel
| | - Oren Pleniceanu
- Department of Military Medicine, Hebrew University of Jerusalem, Jerusalem and the Israel Defense Forces Medical Corps, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,The Nephrology Research Lab, Institute of Nephrology and Hypertension, Sheba Medical Center, Tel Hashomer, Israel
| | - Dorit Tzur
- Department of Military Medicine, Hebrew University of Jerusalem, Jerusalem and the Israel Defense Forces Medical Corps, Ramat Gan, Israel
| | - Michal Stern-Zimmer
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Pediatrics B and Pediatric Nephrology Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
| | - Arnon Afek
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Central Management, Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Tomer Erlich
- Department of Military Medicine, Hebrew University of Jerusalem, Jerusalem and the Israel Defense Forces Medical Corps, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Urology Department, Sheba Medical Center, Ramat Gan, Israel
| | - Estela Derazne
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Jeremy D Kark
- Department of Military Medicine, Hebrew University of Jerusalem, Jerusalem and the Israel Defense Forces Medical Corps, Ramat Gan, Israel
| | - Lital Keinan-Boker
- School of Public Health, University of Haifa, Haifa, Israel.,Israel Center for Disease Control, Ministry of Health, Ramat Gan, Israel
| | - Gilad Twig
- Department of Military Medicine, Hebrew University of Jerusalem, Jerusalem and the Israel Defense Forces Medical Corps, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Talpiot Medical Leadership Program, Sheba Medical Center, Tel Hashomer, Israel
| | - Asaf Vivante
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Pediatrics B and Pediatric Nephrology Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
| |
Collapse
|
18
|
Maggio S, Polidori E, Ceccaroli P, Cioccoloni A, Stocchi V, Guescini M. Current Methods for the Isolation of Urinary Extracellular Vesicles. Methods Mol Biol 2021; 2292:153-172. [PMID: 33651360 DOI: 10.1007/978-1-0716-1354-2_14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Extracellular vesicles (EVs) are small membrane-bound particles released into extracellular space by almost all cell types, and found in body fluids like blood, urine, and saliva. Mounting evidence has demonstrated the clinical potential of EVs as diagnostic and therapeutic tools to analyse physiological/pathological processes due to their ability to transport biomolecules secreted from diverse tissues of an individual.For example, the urinary EVs (uEVs), released from all regions of the kidney's nephron and from other cells that line the urinary tract, retain proteomic and transcriptomic markers specific to their cell of origin representing a valuable tool for kidney disease diagnosis.Despite the numerous efforts in developing suitable methods to separate EVs from biofluids, providing material of high purity and low variability poses a limit to clinical translation.This chapter focuses on advantages and disadvantages of several EV isolation methodologies, and provides examples of uEV isolation protocols based on time, cost, and equipment considerations, as well as the sample requirements for any downstream analyses.
Collapse
Affiliation(s)
- Serena Maggio
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Emanuela Polidori
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Paola Ceccaroli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Andrea Cioccoloni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Vilberto Stocchi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Michele Guescini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy.
| |
Collapse
|
19
|
Zhou C, Zhou L, Liu J, Xu L, Xu Z, Chen Z, Ge Y, Zhao F, Wu R, Wang X, Jiang N, Mao L, Jia R. Kidney extracellular matrix hydrogel enhances therapeutic potential of adipose-derived mesenchymal stem cells for renal ischemia reperfusion injury. Acta Biomater 2020; 115:250-263. [PMID: 32771597 DOI: 10.1016/j.actbio.2020.07.056] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 07/28/2020] [Accepted: 07/30/2020] [Indexed: 12/11/2022]
Abstract
Stem cell-based therapy has been suggested as a promising option for the treatment of renal ischemia-reperfusion injury (IRI). However, how to efficiently deliver stem cells remains a challenge. In the present study, we firstly proposed the utilization of kidney extracellular matrix hydrogel (ECMH) as an injectable scaffold for delivering adipose-derived mesenchymal stem cells (ad-MSCs) into ischemic kidneys. A modified strategy of decellularization and gelation was introduced to prepare the ECMH, by which the bioactive ingredients were retained as much as possible. Bioluminescence living imaging and immunofluorescence revealed that ECMH could significantly elevate the retention and survival rate of transplanted ad-MSCs in damaged kidneys and reduce their escape rate to other organs, which consequently resulted to the enhanced therapeutic effect of ad-MSCs on renal IRI. Further, in vitro evidence demonstrated that ECMH could remarkably reduce the oxidative stress and apoptosis, promote the proliferation, secretion, and epithelial differentiation of ad-MSCs, as well as facilitate cell migration while acting as a sustained-release scaffold. This study establishes an effective approach to enhance the therapeutic potential of ad-MSCs for renal IRI. Our findings suggest that ECMH derived from organs or tissues would be a promising injectable scaffold for stem cell-based therapy. STATEMENT OF SIGNIFICANCE: It remains a challenge to efficiently deliver stem cells to target tissues, which may limit the clinical application of stem cell-based therapy. In this study, we developed a modified strategy of decellularization and gelation to prepare the kidney extracellular matrix hydrogel (ECMH). In vivo and in vitro evidence indicated that the kidney ECMH could improve the retention and survival rate, as well as multiple biological functions of adipose-derived mesenchymal stem cells, thereby contributing to the histological and functional recovery of injured kidneys induced by ischemia-reperfusion. Our findings highlight the use of organs or tissues derived ECMH as a promising stem cell delivery scaffold for tissue repair.
Collapse
Affiliation(s)
- Changcheng Zhou
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China; Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Liuhua Zhou
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China; Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Jingyu Liu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Luwei Xu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China; Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Zheng Xu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China; Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Zaozao Chen
- Institute of Biomaterials and Medical Devices, School of Biological Science and Medical Engineering, Southeast University, Dingjiaqiao 87, Nanjing 210009, China
| | - Yuzheng Ge
- Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Feng Zhao
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China; Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Ran Wu
- Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Xinning Wang
- Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Nan Jiang
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Liang Mao
- Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Ruipeng Jia
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China; Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China.
| |
Collapse
|
20
|
Pleniceanu O, Harari-Steinberg O, Omer D, Gnatek Y, Lachmi BE, Cohen-Zontag O, Manevitz-Mendelson E, Barzilai A, Yampolsky M, Fuchs Y, Rosenzweig B, Eisner A, Dotan Z, Fine LG, Dekel B, Greenberger S. Successful Introduction of Human Renovascular Units into the Mammalian Kidney. J Am Soc Nephrol 2020; 31:2757-2772. [PMID: 32753400 DOI: 10.1681/asn.2019050508] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Accepted: 06/22/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Cell-based therapies aimed at replenishing renal parenchyma have been proposed as an approach for treating CKD. However, pathogenic mechanisms involved in CKD such as renal hypoxia result in loss of kidney function and limit engraftment and therapeutic effects of renal epithelial progenitors. Jointly administering vessel-forming cells (human mesenchymal stromal cells [MSCs] and endothelial colony-forming cells [ECFCs]) may potentially result in in vivo formation of vascular networks. METHODS We administered renal tubule-forming cells derived from human adult and fetal kidneys (previously shown to exert a functional effect in CKD mice) into mice, alongside MSCs and ECFCs. We then assessed whether this would result in generation of "renovascular units" comprising both vessels and tubules with potential interaction. RESULTS Directly injecting vessel-forming cells and renal tubule-forming cells into the subcutaneous and subrenal capsular space resulted in self-organization of donor-derived vascular networks that connected to host vasculature, alongside renal tubules comprising tubular epithelia of different nephron segments. Vessels derived from MSCs and ECFCs augmented in vivo tubulogenesis by the renal tubule-forming cells. In vitro coculture experiments showed that MSCs and ECFCs induced self-renewal and genes associated with mesenchymal-epithelial transition in renal tubule-forming cells, indicating paracrine effects. Notably, after renal injury, renal tubule-forming cells and vessel-forming cells infused into the renal artery did not penetrate the renal vascular network to generate vessels; only administering them into the kidney parenchyma resulted in similar generation of human renovascular units in vivo. CONCLUSIONS Combined cell therapy of vessel-forming cells and renal tubule-forming cells aimed at alleviating renal hypoxia and enhancing tubulogenesis holds promise as the basis for new renal regenerative therapies.
Collapse
Affiliation(s)
- Oren Pleniceanu
- The Pediatric Stem Cell Research Institute and Pediatric Nephrology Division, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Orit Harari-Steinberg
- The Pediatric Stem Cell Research Institute and Pediatric Nephrology Division, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
| | - Dorit Omer
- The Pediatric Stem Cell Research Institute and Pediatric Nephrology Division, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
| | - Yehudit Gnatek
- The Pediatric Stem Cell Research Institute and Pediatric Nephrology Division, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
| | - Bat-El Lachmi
- The Pediatric Stem Cell Research Institute and Pediatric Nephrology Division, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
| | - Osnat Cohen-Zontag
- The Pediatric Stem Cell Research Institute and Pediatric Nephrology Division, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
| | | | - Aviv Barzilai
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Dermatology, Sheba Medical Center, Tel Hashomer, Israel
| | - Matan Yampolsky
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Yaron Fuchs
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Barak Rosenzweig
- Department of Urology, Sheba Medical Center, Tel Hashomer, Israel
| | - Alon Eisner
- Department of Urology, Sheba Medical Center, Tel Hashomer, Israel
| | - Zohar Dotan
- Department of Urology, Sheba Medical Center, Tel Hashomer, Israel
| | - Leon G Fine
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Benjamin Dekel
- The Pediatric Stem Cell Research Institute and Pediatric Nephrology Division, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel .,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shoshana Greenberger
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Dermatology, Sheba Medical Center, Tel Hashomer, Israel
| |
Collapse
|
21
|
Lau RWK, Al‐Rubaie A, Saini S, Wise AF, Ricardo SD. Percutaneous intrarenal transplantation of differentiated induced pluripotent stem cells into newborn mice. Anat Rec (Hoboken) 2020; 303:2603-2612. [DOI: 10.1002/ar.24371] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 11/22/2019] [Accepted: 12/07/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Ricky W. K. Lau
- Department of Anatomy and Developmental BiologyBiomedical Discovery Institute, Monash University Clayton Victoria Australia
| | - Ali Al‐Rubaie
- Department of Anatomy and Developmental BiologyBiomedical Discovery Institute, Monash University Clayton Victoria Australia
| | - Sheetal Saini
- Department of Anatomy and Developmental BiologyBiomedical Discovery Institute, Monash University Clayton Victoria Australia
| | - Andrea F. Wise
- Department of Anatomy and Developmental BiologyBiomedical Discovery Institute, Monash University Clayton Victoria Australia
| | - Sharon D. Ricardo
- Department of Anatomy and Developmental BiologyBiomedical Discovery Institute, Monash University Clayton Victoria Australia
| |
Collapse
|
22
|
Steichen C, Giraud S, Hauet T. Combining Kidney Organoids and Genome Editing Technologies for a Better Understanding of Physiopathological Mechanisms of Renal Diseases: State of the Art. Front Med (Lausanne) 2020; 7:10. [PMID: 32118002 PMCID: PMC7010937 DOI: 10.3389/fmed.2020.00010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 01/13/2020] [Indexed: 12/15/2022] Open
Abstract
Kidney organoids derived from pluripotent stem cells became a real alternative to the use of in vitro cellular models or in vivo animal models. Indeed, the comprehension of the key steps involved during kidney embryonic development led to the establishment of protocols enabling the differentiation of pluripotent stem cells into highly complex and organized structures, composed of various renal cell types. These organoids are linked with one major application based on iPSC technology advantage: the possibility to control iPSC genome, by selecting patients with specific disease or by genome editing tools such as CRISPR/Cas9 system. This allows the generation of kidney organoïds which recapitulate important physiopathological mechanisms such as cyst formation in renal polycystic disease for example. This review will focus on studies combining these both cutting edge technologies i.e., kidney organoid differentiation and genome editing and will describe what are the main advances performed in the comprehension of physiopathological mechanisms of renal diseases, as well as discuss remaining technical barriers and perspectives in the field.
Collapse
Affiliation(s)
- Clara Steichen
- INSERM U1082-IRTOMIT, Poitiers, France.,Université de Poitiers, Faculté de Médecine et de Pharmacie, Poitiers, France
| | - Sébastien Giraud
- INSERM U1082-IRTOMIT, Poitiers, France.,CHU Poitiers, Service de Biochimie, Poitiers, France
| | - Thierry Hauet
- INSERM U1082-IRTOMIT, Poitiers, France.,Université de Poitiers, Faculté de Médecine et de Pharmacie, Poitiers, France.,CHU Poitiers, Service de Biochimie, Poitiers, France
| |
Collapse
|
23
|
Rangel ÉB, Gomes SA, Kanashiro-Takeuchi R, Hare JM. Progenitor/Stem Cell Delivery by Suprarenal Aorta Route in Acute Kidney Injury. Cell Transplant 2019; 28:1390-1403. [PMID: 31409111 PMCID: PMC6802150 DOI: 10.1177/0963689719860826] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 05/14/2019] [Accepted: 06/03/2019] [Indexed: 02/06/2023] Open
Abstract
Progenitor/stem cell-based kidney regenerative strategies are a key step towards the development of novel therapeutic regimens for kidney disease treatment. However, the route of cell delivery, e.g., intravenous, intra-arterial, or intra-parenchymal, may affect the efficiency for kidney repair in different models of acute and chronic injury. Here, we describe a protocol of intra-aorta progenitor/stem cell injection in rats following either acute ischemia-reperfusion injury or acute proteinuria induced by puromycin aminonucleoside (PAN) - the experimental prototype of human minimal change disease and early stages of focal and segmental glomerulosclerosis. Vascular clips were applied across both renal pedicles for 35 min, or a single dose of PAN was injected via intra-peritoneal route, respectively. Subsequently, 2 x 106 stem cells [green fluorescent protein (GFP)-labeled c-Kit+ progenitor/stem cells or GFP-mesenchymal stem cells] or saline were injected into the suprarenal aorta, above the renal arteries, after application of a vascular clip to the abdominal aorta below the renal arteries. This approach contributed to engraftment rates of ∼10% at day 8 post ischemia-reperfusion injury, when c-Kit+ progenitor/stem cells were injected, which accelerated kidney recovery. Similar rates of engraftment were found after PAN-induced podocyte damage at day 21. With practice and gentle surgical technique, 100% of the rats could be injected successfully, and, in the week following injection, ∼ 85% of the injected rats will recover completely. Given the similarities in mammals, much of the data obtained from intra-arterial delivery of progenitor/stem cells in rodents can be tested in translational research and clinical trials with endovascular catheters in humans.
Collapse
Affiliation(s)
- Érika B. Rangel
- Interdisciplinary Stem Cell Institute, Leonard M Miller School of
Medicine, University of Miami, USA
- Hospital Israelita Albert Einstein, São Paulo, Brazil
- Federal University of São Paulo, Brazil
| | - Samirah A. Gomes
- Interdisciplinary Stem Cell Institute, Leonard M Miller School of
Medicine, University of Miami, USA
- Laboratory of Cellular, Genetic, and Molecular Nephrology, Renal
Division, University of São Paulo, Brazil
| | - Rosemeire Kanashiro-Takeuchi
- Interdisciplinary Stem Cell Institute, Leonard M Miller School of
Medicine, University of Miami, USA
- Department of Molecular and Cellular Pharmacology, Leonard M Miller
School of Medicine, University of Miami, USA
| | - Joshua M. Hare
- Interdisciplinary Stem Cell Institute, Leonard M Miller School of
Medicine, University of Miami, USA
- Department of Molecular and Cellular Pharmacology, Leonard M Miller
School of Medicine, University of Miami, USA
- Division of Cardiology, Leonard M Miller School of Medicine,
University of Miami, USA
| |
Collapse
|
24
|
Abstract
This review focus on kidney organoids derived from pluripotent stem cells, which become a real alternative to the use of in vitro cellular models or in vivo animals models. The comprehension of the key steps involved during kidney embryonic development led to the establishment of protocols enabling the differentiation of pluripotent stem cells into kidney organoids that are highly complex and organized structures, composed of various renal cell types. These mini-organs are endowed with major applications: the possibility to control iPSC genome (by selecting patients with specific disease or by genome editing) allows the generation of kidney organoïds which recapitulate important physiopathological mechanisms such as cyste formation in renal polycystic disease. Kidney organoids can also be used in high-throughput screening to fasten the screening of nephrotoxic/therapeutic compounds. Finally, kidney organoids have a huge interest in the context of tissue repair, which remains for now a challenging goal linked with technological barriers that need still to be overcome.
Collapse
Affiliation(s)
- Clara Steichen
- Inserm U1082 - IRTOMIT (Ischémie reperfusion en transplantation d'organes mécanismes et innovations thérapeutiques), Poitiers, F-86000, France - Université de Poitiers, Faculté de médecine et de pharmacie, Poitiers, F-86000, France
| | - Sébastien Giraud
- Inserm U1082 - IRTOMIT (Ischémie reperfusion en transplantation d'organes mécanismes et innovations thérapeutiques), Poitiers, F-86000, France - CHU de Poitiers, service de biochimie, Poitiers, F-86000, France
| | - Thierry Hauet
- Inserm U1082 - IRTOMIT (Ischémie reperfusion en transplantation d'organes mécanismes et innovations thérapeutiques), Poitiers, F-86000, France - Université de Poitiers, Faculté de médecine et de pharmacie, Poitiers, F-86000, France - CHU de Poitiers, service de biochimie, Poitiers, F-86000, France
| |
Collapse
|
25
|
Lin28 and let-7 regulate the timing of cessation of murine nephrogenesis. Nat Commun 2019; 10:168. [PMID: 30635573 PMCID: PMC6329821 DOI: 10.1038/s41467-018-08127-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 12/12/2018] [Indexed: 01/10/2023] Open
Abstract
In humans and in mice the formation of nephrons during embryonic development reaches completion near the end of gestation, after which no new nephrons are formed. The final nephron complement can vary 10-fold, with reduced nephron number predisposing individuals to hypertension, renal, and cardiovascular diseases in later life. While the heterochronic genes lin28 and let-7 are well-established regulators of developmental timing in invertebrates, their role in mammalian organogenesis is not fully understood. Here we report that the Lin28b/let-7 axis controls the duration of kidney development in mice. Suppression of let-7 miRNAs, directly or via the transient overexpression of LIN28B, can prolong nephrogenesis and enhance kidney function potentially via upregulation of the Igf2/H19 locus. In contrast, kidney-specific loss of Lin28b impairs renal development. Our study reveals mechanisms regulating persistence of nephrogenic mesenchyme and provides a rationale for therapies aimed at increasing nephron mass. Nephrogenesis ceases after postnatal day 2 in the mouse or after the 36th week of gestation in humans, but how this is regulated is unclear. Here, the authors identify a role for the RNA-binding protein Lin28 and suppression of let-7 microRNA in regulating the duration of nephrogenesis.
Collapse
|
26
|
Zhao L, Hu C, Zhang P, Jiang H, Chen J. Preconditioning strategies for improving the survival rate and paracrine ability of mesenchymal stem cells in acute kidney injury. J Cell Mol Med 2018; 23:720-730. [PMID: 30484934 PMCID: PMC6349184 DOI: 10.1111/jcmm.14035] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 10/20/2018] [Accepted: 10/27/2018] [Indexed: 02/06/2023] Open
Abstract
Acute kidney injury (AKI) is a common, severe emergency case in clinics, with high incidence, significant mortality and increased costs. Despite development in the understanding of its pathophysiology, the therapeutic choices are still confined to dialysis and renal transplantation. Considering their antiapoptotic, immunomodulatory, antioxidative and pro‐angiogenic effects, mesenchymal stem cells (MSCs) may be a promising candidate for AKI management. Based on these findings, some clinical trials have been performed, but the results are contradictory (NCT00733876, NCT01602328). The low engraftment, poor survival rate, impaired paracrine ability and delayed administration of MSCs are the four main reasons for the limited clinical efficacy. Investigators have developed a series of preconditioning strategies to improve MSC survival rates and paracrine ability. In this review, by summarizing these encouraging studies, we intend to provide a comprehensive understanding of various preconditioning strategies on AKI therapy and improve the prognosis of AKI patients by regenerative medicine.
Collapse
Affiliation(s)
- Lingfei Zhao
- Key Laboratory of Kidney Disease Prevention and Control Technology, Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Chenxia Hu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Ping Zhang
- Key Laboratory of Kidney Disease Prevention and Control Technology, Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Hua Jiang
- Key Laboratory of Kidney Disease Prevention and Control Technology, Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Jianghua Chen
- Key Laboratory of Kidney Disease Prevention and Control Technology, Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, PR China
| |
Collapse
|
27
|
Legallais C, Kim D, Mihaila SM, Mihajlovic M, Figliuzzi M, Bonandrini B, Salerno S, Yousef Yengej FA, Rookmaaker MB, Sanchez Romero N, Sainz-Arnal P, Pereira U, Pasqua M, Gerritsen KGF, Verhaar MC, Remuzzi A, Baptista PM, De Bartolo L, Masereeuw R, Stamatialis D. Bioengineering Organs for Blood Detoxification. Adv Healthc Mater 2018; 7:e1800430. [PMID: 30230709 DOI: 10.1002/adhm.201800430] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 08/23/2018] [Indexed: 12/11/2022]
Abstract
For patients with severe kidney or liver failure the best solution is currently organ transplantation. However, not all patients are eligible for transplantation and due to limited organ availability, most patients are currently treated with therapies using artificial kidney and artificial liver devices. These therapies, despite their relative success in preserving the patients' life, have important limitations since they can only replace part of the natural kidney or liver functions. As blood detoxification (and other functions) in these highly perfused organs is achieved by specialized cells, it seems relevant to review the approaches leading to bioengineered organs fulfilling most of the native organ functions. There, the culture of cells of specific phenotypes on adapted scaffolds that can be perfused takes place. In this review paper, first the functions of kidney and liver organs are briefly described. Then artificial kidney/liver devices, bioartificial kidney devices, and bioartificial liver devices are focused on, as well as biohybrid constructs obtained by decellularization and recellularization of animal organs. For all organs, a thorough overview of the literature is given and the perspectives for their application in the clinic are discussed.
Collapse
Affiliation(s)
- Cécile Legallais
- UMR CNRS 7338 Biomechanics & Bioengineering; Université de technologie de Compiègne; Sorbonne Universités; 60203 Compiègne France
| | - Dooli Kim
- (Bio)artificial organs; Department of Biomaterials Science and Technology; Faculty of Science and Technology; TechMed Institute; University of Twente; P.O. Box 217 7500 AE Enschede The Netherlands
| | - Sylvia M. Mihaila
- Division of Pharmacology; Utrecht Institute for Pharmaceutical Sciences; Utrecht University; Universiteitsweg 99 3584 CG Utrecht The Netherlands
- Department of Nephrology and Hypertension; University Medical Center Utrecht and Regenerative Medicine Utrecht; Utrecht University; Heidelberglaan 100 3584 CX Utrecht The Netherlands
| | - Milos Mihajlovic
- Division of Pharmacology; Utrecht Institute for Pharmaceutical Sciences; Utrecht University; Universiteitsweg 99 3584 CG Utrecht The Netherlands
| | - Marina Figliuzzi
- IRCCS-Istituto di Ricerche Farmacologiche Mario Negri; via Stezzano 87 24126 Bergamo Italy
| | - Barbara Bonandrini
- Department of Chemistry; Materials and Chemical Engineering “Giulio Natta”; Politecnico di Milano; Piazza Leonardo da Vinci 32 20133 Milan Italy
| | - Simona Salerno
- Institute on Membrane Technology; National Research Council of Italy; ITM-CNR; Via Pietro BUCCI, Cubo 17C - 87036 Rende Italy
| | - Fjodor A. Yousef Yengej
- Department of Nephrology and Hypertension; University Medical Center Utrecht and Regenerative Medicine Utrecht; Utrecht University; Heidelberglaan 100 3584 CX Utrecht The Netherlands
| | - Maarten B. Rookmaaker
- Department of Nephrology and Hypertension; University Medical Center Utrecht and Regenerative Medicine Utrecht; Utrecht University; Heidelberglaan 100 3584 CX Utrecht The Netherlands
| | | | - Pilar Sainz-Arnal
- Instituto de Investigación Sanitaria de Aragón (IIS Aragon); 50009 Zaragoza Spain
- Instituto Aragonés de Ciencias de la Salud (IACS); 50009 Zaragoza Spain
| | - Ulysse Pereira
- UMR CNRS 7338 Biomechanics & Bioengineering; Université de technologie de Compiègne; Sorbonne Universités; 60203 Compiègne France
| | - Mattia Pasqua
- UMR CNRS 7338 Biomechanics & Bioengineering; Université de technologie de Compiègne; Sorbonne Universités; 60203 Compiègne France
| | - Karin G. F. Gerritsen
- Department of Nephrology and Hypertension; University Medical Center Utrecht and Regenerative Medicine Utrecht; Utrecht University; Heidelberglaan 100 3584 CX Utrecht The Netherlands
| | - Marianne C. Verhaar
- Department of Nephrology and Hypertension; University Medical Center Utrecht and Regenerative Medicine Utrecht; Utrecht University; Heidelberglaan 100 3584 CX Utrecht The Netherlands
| | - Andrea Remuzzi
- IRCCS-Istituto di Ricerche Farmacologiche Mario Negri; via Stezzano 87 24126 Bergamo Italy
- Department of Management; Information and Production Engineering; University of Bergamo; viale Marconi 5 24044 Dalmine Italy
| | - Pedro M. Baptista
- Instituto de Investigación Sanitaria de Aragón (IIS Aragon); 50009 Zaragoza Spain
- Department of Management; Information and Production Engineering; University of Bergamo; viale Marconi 5 24044 Dalmine Italy
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas (CIBERehd); 28029 Barcelona Spain
- Fundación ARAID; 50009 Zaragoza Spain
- Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz; 28040 Madrid Spain. Department of Biomedical and Aerospace Engineering; Universidad Carlos III de Madrid; 28911 Madrid Spain
| | - Loredana De Bartolo
- Institute on Membrane Technology; National Research Council of Italy; ITM-CNR; Via Pietro BUCCI, Cubo 17C - 87036 Rende Italy
| | - Rosalinde Masereeuw
- Division of Pharmacology; Utrecht Institute for Pharmaceutical Sciences; Utrecht University; Universiteitsweg 99 3584 CG Utrecht The Netherlands
| | - Dimitrios Stamatialis
- (Bio)artificial organs; Department of Biomaterials Science and Technology; Faculty of Science and Technology; TechMed Institute; University of Twente; P.O. Box 217 7500 AE Enschede The Netherlands
| |
Collapse
|
28
|
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
|
29
|
Corrò C, Moch H. Biomarker discovery for renal cancer stem cells. J Pathol Clin Res 2018; 4:3-18. [PMID: 29416873 PMCID: PMC5783955 DOI: 10.1002/cjp2.91] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 11/27/2017] [Accepted: 12/13/2017] [Indexed: 12/17/2022]
Abstract
Characterised by high intra- and inter-tumor heterogeneity, metastatic renal cell carcinoma (RCC) is resistant to chemo- and radiotherapy. Therefore, the development of new prognostic and diagnostic markers for RCC patients is needed. Cancer stem cells (CSCs) are a small population of neoplastic cells within a tumor which present characteristics reminiscent of normal stem cells. CSCs are characterised by unlimited cell division, maintenance of the stem cell pool (self-renewal), and capability to give rise to all cell types within a tumor; and contribute to metastasis in vivo (tumourigenicity), treatment resistance and recurrence. So far, many studies have tried to establish unique biomarkers to identify CSC populations in RCC. At the same time, different approaches have been developed with the aim to isolate CSCs. Consequently, several markers were found to be specifically expressed in CSCs and cancer stem-like cells derived from RCC such as CD105, ALDH1, OCT4, CD133, and CXCR4. However, the contribution of genetic and epigenetic mechanisms, and tumor microenvironment, to cellular plasticity have made the discovery of unique biomarkers a very difficult task. In fact, contrasting results regarding the applicability of such markers to the isolation of renal CSCs have been reported in the literature. Therefore, a better understanding of the mechanism underlying CSC may help dissecting tumor heterogeneity and drug treatment efficiency.
Collapse
Affiliation(s)
- Claudia Corrò
- Department of Pathology and Molecular PathologyUniversity Hospital ZurichSwitzerland
| | - Holger Moch
- Department of Pathology and Molecular PathologyUniversity Hospital ZurichSwitzerland
| |
Collapse
|
30
|
Renal lineage cells as a source for renal regeneration. Pediatr Res 2018; 83:267-274. [PMID: 28985199 DOI: 10.1038/pr.2017.255] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 09/29/2017] [Indexed: 12/24/2022]
Abstract
The mammalian kidney is a highly complex organ, composed of various cell types within a unique structural framework. Nonetheless, in recent years, giant leaps in our understanding of nephrogenesis and the origin of new cells in the adult kidney have resulted in novel routes to regenerate damaged nephrons. While several strategies can be envisioned to achieve this aim, one common theme is the reliance on renal lineage cells, as extrarenal cells, such as bone marrow-derived cells, have been shown to be devoid of renal differentiation capacity. Herein, we will present the main motivation for the pursuit for cell-based therapies, which is the ever growing problem of chronic kidney disease (CKD), and discuss different strategies toward replenishing the damaged renal parenchyma. These include transplantation of fetal kidney grafts or fetal kidney stem cells, directed differentiation of pluripotent stem cells into kidney epithelia, establishment of renal progenitors from the adult kidney, and genetic reprogramming of mature kidney cells into a progenitor state. Taken together with novel techniques recapitulating the three-dimensional developmental environment, these advances are expected to take the field into a new era, bringing us closer than ever to the day when kidney stem cell-based therapy becomes a viable therapeutic option.
Collapse
|
31
|
Pleniceanu O, Shukrun R, Omer D, Vax E, Kanter I, Dziedzic K, Pode-Shakked N, Mark-Daniei M, Pri-Chen S, Gnatek Y, Alfandary H, Varda-Bloom N, Bar-Lev DD, Bollag N, Shtainfeld R, Armon L, Urbach A, Kalisky T, Nagler A, Harari-Steinberg O, Arbiser JL, Dekel B. Peroxisome proliferator-activated receptor gamma (PPARγ) is central to the initiation and propagation of human angiomyolipoma, suggesting its potential as a therapeutic target. EMBO Mol Med 2017; 9:508-530. [PMID: 28275008 PMCID: PMC5376758 DOI: 10.15252/emmm.201506111] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Angiomyolipoma (AML), the most common benign renal tumor, can result in severe morbidity from hemorrhage and renal failure. While mTORC1 activation is involved in its growth, mTORC1 inhibitors fail to eradicate AML, highlighting the need for new therapies. Moreover, the identity of the AML cell of origin is obscure. AML research, however, is hampered by the lack of in vivo models. Here, we establish a human AML‐xenograft (Xn) model in mice, recapitulating AML at the histological and molecular levels. Microarray analysis demonstrated tumor growth in vivo to involve robust PPARG‐pathway activation. Similarly, immunostaining revealed strong PPARG expression in human AML specimens. Accordingly, we demonstrate that while PPARG agonism accelerates AML growth, PPARG antagonism is inhibitory, strongly suppressing AML proliferation and tumor‐initiating capacity, via a TGFB‐mediated inhibition of PDGFB and CTGF. Finally, we show striking similarity between AML cell lines and mesenchymal stem cells (MSCs) in terms of antigen and gene expression and differentiation potential. Altogether, we establish the first in vivo human AML model, which provides evidence that AML may originate in a PPARG‐activated renal MSC lineage that is skewed toward adipocytes and smooth muscle and away from osteoblasts, and uncover PPARG as a regulator of AML growth, which could serve as an attractive therapeutic target.
Collapse
Affiliation(s)
- Oren Pleniceanu
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Division of Hematology and Cord Blood Bank, Sheba Medical Center, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Racheli Shukrun
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dorit Omer
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Einav Vax
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Itamar Kanter
- Faculty of Engineering, Institute of Nanotechnology, Bar-Ilan University, Ramat Gan, Israel
| | - Klaudyna Dziedzic
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Naomi Pode-Shakked
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Michal Mark-Daniei
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Sara Pri-Chen
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Yehudit Gnatek
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Hadas Alfandary
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Institute of Nephrology, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
| | - Nira Varda-Bloom
- Division of Hematology and Cord Blood Bank, Sheba Medical Center, Ramat Gan, Israel
| | - Dekel D Bar-Lev
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Naomi Bollag
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Rachel Shtainfeld
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Leah Armon
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Achia Urbach
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Tomer Kalisky
- Faculty of Engineering, Institute of Nanotechnology, Bar-Ilan University, Ramat Gan, Israel
| | - Arnon Nagler
- Division of Hematology and Cord Blood Bank, Sheba Medical Center, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Orit Harari-Steinberg
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Jack L Arbiser
- Department of Dermatology, Emory University School of Medicine, Atlanta, GA, USA.,Winship Cancer Institute, Atlanta Veterans Administration Hospital, Atlanta, GA, USA
| | - Benjamin Dekel
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel .,Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
32
|
Towards a Bioengineered Kidney: Recellularization Strategies for Decellularized Native Kidney Scaffolds. Int J Artif Organs 2017; 40:150-158. [DOI: 10.5301/ijao.5000564] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/02/2017] [Indexed: 12/12/2022]
Abstract
Patients with end-stage renal disease often undergo dialysis as a partial substitute for kidney function while waiting for their only treatment option: a kidney transplant. Several research directions emerged for alternatives in support of the ever-growing numbers of patients. Recent years brought big steps forward in the field, with researchers questioning and improving the current dialysis devices as well as moving towards the design of a bioengineered kidney. Whole-organ engineering is also being explored as a possibility, making use of animal or human kidney scaffolds for engineering a transplantable organ. While this is not a new strategy, having been applied so far for thin tissues, it is a novel approach for complex organs such as the kidneys. Kidneys can be decellularized and the remaining scaffold consisting of an extracellular matrix can be repopulated with (autologous) cells, aiming at growing ex vivo a fully transplantable organ. In a broader view, such organs might also be used for a better understanding of fundamental biological concepts and disease mechanisms, drug screening and toxicological investigations, opening new pathways in the treatment of kidney disease. Decellularization of whole organs has been widely explored and described; therefore, this manuscript only briefly reviews some important considerations with an emphasis on scaffold decontamination, but focuses further on recellularization strategies. Critical aspects, including cell types and sources that can be used for recellularization, seeding strategies and possible applications beyond renal replacement are discussed.
Collapse
|
33
|
MacGregor-Ramiasa M, Hopp I, Bachhuka A, Murray P, Vasilev K. Surface nanotopography guides kidney-derived stem cell differentiation into podocytes. Acta Biomater 2017; 56:171-180. [PMID: 28232254 DOI: 10.1016/j.actbio.2017.02.036] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 02/09/2017] [Accepted: 02/17/2017] [Indexed: 02/06/2023]
Abstract
Stem cells have enormous potential for developing novel therapies for kidney disease but our current inability to direct their differentiation to specialised renal cells presents a barrier to their use in renal bioengineering and drug development programmes. Here, a plasma-based technology was used to produce a range of biocompatible substrates comprising controlled surface nanotopography and tailored outermost chemical functionalities. These novel substrata were used to investigate the response of mouse kidney-derived stem cells to changes in both substrate nanotopography and surface chemistry. The stem cells proliferated to a similar extent on all substrates, but specific combinations of nanotopography and surface chemistry promoted differentiation into either podocyte or proximal tubule-like cells. The data reveal that high density of surface nanodefects in association with amine rich chemistry primarily lead to differentiation into podocytes while surfaces with low amine content constituted better substrates for differentiation into proximal tubule cells regardless of the surface nanotopographic profile. Thus plasma coated nanorough substrate may provide useful platform for guiding the fate kidney stem cell in vitro. STATEMENT OF SIGNIFICANCE Adult kidney-derived stem cells have been identified as a promising way to regenerate damaged nephrons. Artificial growth platforms capable to guide the stem cells differentiation into useful cell lineages are needed to expand regenerative cell therapies for chronic kidney diseases. Chemically homogeneous growth substrates endowed with nanotopography gradients were generated via plasma assisted methods in order to investigate the effect of physical cues on the proliferation and differentiation of kidney-derived stem cells. For the first time it is shown that the surface density of the nano-structures had a greater impact on fate of the stem cells than their size. Careful design of the growth substrate nanotopography may help directing the differentiation into either podocytes or proximal tubule cells.
Collapse
|
34
|
Pode-Shakked N, Gershon R, Tam G, Omer D, Gnatek Y, Kanter I, Oriel S, Katz G, Harari-Steinberg O, Kalisky T, Dekel B. Evidence of In Vitro Preservation of Human Nephrogenesis at the Single-Cell Level. Stem Cell Reports 2017; 9:279-291. [PMID: 28552604 PMCID: PMC5511042 DOI: 10.1016/j.stemcr.2017.04.026] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 04/24/2017] [Accepted: 04/25/2017] [Indexed: 01/09/2023] Open
Abstract
During nephrogenesis, stem/progenitor cells differentiate and give rise to early nephron structures that segment to proximal and distal nephron cell types. Previously, we prospectively isolated progenitors from human fetal kidney (hFK) utilizing a combination of surface markers. However, upon culture nephron progenitors differentiated and could not be robustly maintained in vitro. Here, by culturing hFK in a modified medium used for in vitro growth of mouse nephron progenitors, and by dissection of NCAM+/CD133− progenitor cells according to EpCAM expression (NCAM+/CD133−/EpCAM−, NCAM+/CD133−/EpCAMdim, NCAM+/CD133−/EpCAMbright), we show at single-cell resolution a preservation of uninduced and induced cap mesenchyme as well as a transitioning mesenchymal-epithelial state. Concomitantly, differentiating and differentiated epithelial lineages are also maintained. In vitro expansion of discrete stages of early human nephrogenesis in nephron stem cell cultures may be used for drug screening on a full repertoire of developing kidney cells and for prospective isolation of mesenchymal or epithelial renal lineages for regenerative medicine. mNPEM enables in vitro preservation of human renal embryonic CM and epithelia EpCAM allows further dissection of expanded NCAM+CD133− early nephric population Single-cell analysis unveils a continuous lineage hierarchy in nephrogenesis and WT Splice isoform switching confirms a unified MET hierarchy in nephrogenesis and WT
Collapse
Affiliation(s)
- Naomi Pode-Shakked
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer 52621, Israel; Sheba Centers for Regenerative Medicine and Cancer Research, Sheba Medical Center, Tel-Hashomer 52621, Israel; The Dr. Pinchas Borenstein Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer 52621, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel
| | - Rotem Gershon
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer 52621, Israel; Sheba Centers for Regenerative Medicine and Cancer Research, Sheba Medical Center, Tel-Hashomer 52621, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel
| | - Gal Tam
- Faculty of Engineering and Bar-Ilan Institute of Nanotechnology and Advanced Materials (BINA), Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Dorit Omer
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer 52621, Israel; Sheba Centers for Regenerative Medicine and Cancer Research, Sheba Medical Center, Tel-Hashomer 52621, Israel
| | - Yehudit Gnatek
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer 52621, Israel; Sheba Centers for Regenerative Medicine and Cancer Research, Sheba Medical Center, Tel-Hashomer 52621, Israel
| | - Itamar Kanter
- Faculty of Engineering and Bar-Ilan Institute of Nanotechnology and Advanced Materials (BINA), Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Sarit Oriel
- Faculty of Engineering and Bar-Ilan Institute of Nanotechnology and Advanced Materials (BINA), Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Guy Katz
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer 52621, Israel; Sheba Centers for Regenerative Medicine and Cancer Research, Sheba Medical Center, Tel-Hashomer 52621, Israel; The Dr. Pinchas Borenstein Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer 52621, Israel; The Joseph Buchman Gynecology and Maternity Center, Sheba Medical Center, Tel-Hashomer 52621, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel
| | - Orit Harari-Steinberg
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer 52621, Israel; Sheba Centers for Regenerative Medicine and Cancer Research, Sheba Medical Center, Tel-Hashomer 52621, Israel
| | - Tomer Kalisky
- Faculty of Engineering and Bar-Ilan Institute of Nanotechnology and Advanced Materials (BINA), Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Benjamin Dekel
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer 52621, Israel; Sheba Centers for Regenerative Medicine and Cancer Research, Sheba Medical Center, Tel-Hashomer 52621, Israel; Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer 52621, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel.
| |
Collapse
|
35
|
Zhou L, Song Q, Shen J, Xu L, Xu Z, Wu R, Ge Y, Zhu J, Wu J, Dou Q, Jia R. Comparison of human adipose stromal vascular fraction and adipose-derived mesenchymal stem cells for the attenuation of acute renal ischemia/reperfusion injury. Sci Rep 2017; 7:44058. [PMID: 28276451 PMCID: PMC5343423 DOI: 10.1038/srep44058] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 02/03/2017] [Indexed: 02/06/2023] Open
Abstract
Stem cells therapy has been suggested as a promising option for the treatment of acute kidney injury (AKI). This study was performed to compare the abilities of xenogenic transplantation of human adipose stromal vascular fraction (SVF) and adipose-derived mesenchymal stem cells (AdMSCs) to facilitate the recovery of renal function and structure in a rat model of ischemia/reperfusion (IR) induced AKI. SVF or AdMSCs were transplanted to the injured kidney through intra-parenchymal injection. Significantly improved renal function and reduced tubular injury were observed in SVF and AdMSCs groups. Administration of SVF or AdMSCs contributed to significantly improved cell proliferation and markedly reduced cell apoptosis in parallel with reduced microvascular rarefaction in injured kidney. IR injury resulted in higher levels of inflammatory cytokines, whereas xenogenic transplantation of SVF or AdMSCs reduced but not induced inflammatory cytokines expression. Additionally, in vitro study showed that administration of SVF or AdMSCs could also significantly promote the proliferation and survival of renal tubular epithelial cells underwent hypoxia/reoxygenation injury through secreting various growth factors. However, cell proliferation was significantly promoted in SVF group than in AdMSCs group. In conclusion, our study demonstrated that administration of SVF or AdMSCs was equally effective in attenuating acute renal IR injury.
Collapse
Affiliation(s)
- Liuhua Zhou
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, Jiangsu 210006, China.,Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, Jiangsu 210006, China
| | - Qun Song
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, Jiangsu 210006, China
| | - Jiangwei Shen
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, Jiangsu 210006, China
| | - Luwei Xu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, Jiangsu 210006, China.,Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, Jiangsu 210006, China
| | - Zheng Xu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, Jiangsu 210006, China.,Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, Jiangsu 210006, China
| | - Ran Wu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, Jiangsu 210006, China.,Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, Jiangsu 210006, China
| | - Yuzheng Ge
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, Jiangsu 210006, China.,Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, Jiangsu 210006, China
| | - Jiageng Zhu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, Jiangsu 210006, China.,Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, Jiangsu 210006, China
| | - Jianping Wu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, Jiangsu 210006, China.,Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, Jiangsu 210006, China
| | - Quanliang Dou
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, Jiangsu 210006, China.,Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, Jiangsu 210006, China
| | - Ruipeng Jia
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, Jiangsu 210006, China.,Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, Jiangsu 210006, China
| |
Collapse
|
36
|
Chambers BE, Wingert RA. Renal progenitors: Roles in kidney disease and regeneration. World J Stem Cells 2016; 8:367-375. [PMID: 27928463 PMCID: PMC5120241 DOI: 10.4252/wjsc.v8.i11.367] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 08/17/2016] [Accepted: 09/08/2016] [Indexed: 02/06/2023] Open
Abstract
Kidney disease is a devastating condition that affects millions of people worldwide, and its prevalence is predicted to significantly increase. The kidney is a complex organ encompassing many diverse cell types organized in a elaborate tissue architecture, making regeneration a challenging feat. In recent years, there has been a surge in the field of stem cell research to develop regenerative therapies for various organ systems. Here, we review some recent progressions in characterizing the role of renal progenitors in development, regeneration, and kidney disease in mammals. We also discuss how the zebrafish provides a unique experimental animal model that can provide a greater molecular and genetic understanding of renal progenitors, which may contribute to the development of potential regenerative therapies for human renal afflictions.
Collapse
|
37
|
Edeling M, Ragi G, Huang S, Pavenstädt H, Susztak K. Developmental signalling pathways in renal fibrosis: the roles of Notch, Wnt and Hedgehog. Nat Rev Nephrol 2016; 12:426-39. [PMID: 27140856 DOI: 10.1038/nrneph.2016.54] [Citation(s) in RCA: 318] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Kidney fibrosis is a common histological manifestation of functional decline in the kidney. Fibrosis is a reactive process that develops in response to excessive epithelial injury and inflammation, leading to myofibroblast activation and an accumulation of extracellular matrix. Here, we describe how three key developmental signalling pathways - Notch, Wnt and Hedgehog (Hh) - are reactivated in response to kidney injury and contribute to the fibrotic response. Although transient activation of these pathways is needed for repair of injured tissue, their sustained activation is thought to promote fibrosis. Excessive Wnt and Notch expression prohibit epithelial differentiation, whereas increased Wnt and Hh expression induce fibroblast proliferation and myofibroblastic transdifferentiation. Notch, Wnt and Hh are fundamentally different signalling pathways, but their choreographed activation seems to be just as important for fibrosis as it is for embryonic kidney development. Decreasing the activity of Notch, Wnt or Hh signalling could potentially provide a new therapeutic strategy to ameliorate the development of fibrosis in chronic kidney disease.
Collapse
Affiliation(s)
- Maria Edeling
- Renal Electrolyte and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, 415 Curie Boulevard, 415 Clinical Research Building, Philadelphia, Pennsylvania 19104, USA.,Department of Molecular Nephrology, Internal Medicine D, University Hospital Albert-Schweitzer-Straße 33, Münster 48149, Germany
| | - Grace Ragi
- Renal Electrolyte and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, 415 Curie Boulevard, 415 Clinical Research Building, Philadelphia, Pennsylvania 19104, USA
| | - Shizheng Huang
- Renal Electrolyte and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, 415 Curie Boulevard, 415 Clinical Research Building, Philadelphia, Pennsylvania 19104, USA
| | - Hermann Pavenstädt
- Department of Molecular Nephrology, Internal Medicine D, University Hospital Albert-Schweitzer-Straße 33, Münster 48149, Germany
| | - Katalin Susztak
- Renal Electrolyte and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, 415 Curie Boulevard, 415 Clinical Research Building, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
38
|
Liu X, Liu H, Sun L, Chen Z, Nie H, Sun A, Liu G, Guan G. The role of long-term label-retaining cells in the regeneration of adult mouse kidney after ischemia/reperfusion injury. Stem Cell Res Ther 2016; 7:68. [PMID: 27137761 PMCID: PMC4852428 DOI: 10.1186/s13287-016-0324-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 03/22/2016] [Accepted: 04/12/2016] [Indexed: 12/17/2022] Open
Abstract
Background Label-retaining cells (LRCs) have been recognized as rare stem and progenitor-like cells, but their complex biological features in renal repair at the cellular level have never been reported. This study was conducted to evaluate whether LRCs in kidney are indeed renal stem/progenitor cells and to delineate their potential role in kidney regeneration. Methods We utilized a long-term pulse chase of 5-bromo-2'-deoxyuridine (BrdU)-labeled cells in C57BL/6J mice to identify renal LRCs. We tracked the precise morphological characteristics and locations of BrdU+LRCs by both immunohistochemistry and immunofluorescence. To examine whether these BrdU+LRCs contribute to the repair of acute kidney injury, we analyzed biological characteristics of BrdU+LRCs in mice after ischemia/reperfusion (I/R) injury. Results The findings revealed that the nuclei of BrdU+ LRCs exhibited different morphological characteristics in normal adult kidneys, including nuclei in pairs or scattered, fragmented or intact, strongly or weakly positive. Only 24.3 ± 1.5 % of BrdU+ LRCs co-expressed with Ki67 and 9.1 ± 1.4 % of BrdU+ LRCs were positive for TUNEL following renal I/R injury. Interestingly, we found that newly regenerated cells formed a niche-like structure and LRCs in pairs tended to locate in this structure, but the number of those LRCs was very low. We found a few scattered LRCs co-expressed Lotus tetragonolobus agglutinin (LTA) in the early phase of injury, suggesting differentiation of those LRCs in mouse kidney. Conclusions Our findings suggest that LRCs are not a simple type of slow-cycling cells in adult kidneys, indicating a limited role of these cells in the regeneration of I/R injured kidney. Thus, LRCs cannot reliably be considered stem/progenitor cells in the regeneration of adult mouse kidney. When researchers use this technique to study the cellular basis of renal repair, these complex features of renal LRCs and the purity of real stem cells among renal LRCs should be considered.
Collapse
Affiliation(s)
- Xiangchun Liu
- Department of Nephrology, The Second Hospital of Shandong University,Shandong University, Jinan, PR. China
| | - Haiying Liu
- Department of Nephrology, The Second Hospital of Shandong University,Shandong University, Jinan, PR. China.
| | - Lina Sun
- Department of Nephrology, The Second Hospital of Shandong University,Shandong University, Jinan, PR. China
| | - Zhixin Chen
- Department of Nephrology, The Second Hospital of Shandong University,Shandong University, Jinan, PR. China
| | - Huibin Nie
- Department of Nephrology, The Second Hospital of Shandong University,Shandong University, Jinan, PR. China
| | - Aili Sun
- Department of Nephrology, The Second Hospital of Shandong University,Shandong University, Jinan, PR. China
| | - Gang Liu
- Department of Nephrology, The Second Hospital of Shandong University,Shandong University, Jinan, PR. China
| | - Guangju Guan
- Department of Nephrology, The Second Hospital of Shandong University,Shandong University, Jinan, PR. China.
| |
Collapse
|
39
|
Dissecting Stages of Human Kidney Development and Tumorigenesis with Surface Markers Affords Simple Prospective Purification of Nephron Stem Cells. Sci Rep 2016; 6:23562. [PMID: 27020553 PMCID: PMC4810363 DOI: 10.1038/srep23562] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 03/08/2016] [Indexed: 01/16/2023] Open
Abstract
When assembling a nephron during development a multipotent stem cell pool becomes
restricted as differentiation ensues. A faulty differentiation arrest in this
process leads to transformation and initiation of a Wilms’ tumor.
Mapping these transitions with respective surface markers affords accessibility to
specific cell subpopulations. NCAM1 and CD133 have been previously suggested to mark
human renal progenitor populations. Herein, using cell sorting, RNA sequencing,
in vitro studies with serum-free media and in vivo
xenotransplantation we demonstrate a sequential map that links human kidney
development and tumorigenesis; In nephrogenesis,
NCAM1+CD133− marks
SIX2+ multipotent renal stem cells transiting to
NCAM1+CD133+ differentiating segment-specific
SIX2− epithelial progenitors and
NCAM1−CD133+ differentiated nephron
cells. In tumorigenesis, NCAM1+CD133−
marks SIX2+ blastema that includes the ALDH1+ WT
cancer stem/initiating cells, while NCAM1+CD133+ and
NCAM1−CD133+ specifying early and late
epithelial differentiation, are severely restricted in tumor initiation capacity and
tumor self-renewal. Thus, negative selection for CD133 is required for defining
NCAM1+ nephron stem cells in normal and malignant
nephrogenesis.
Collapse
|
40
|
Drummond BE, Wingert RA. Insights into kidney stem cell development and regeneration using zebrafish. World J Stem Cells 2016; 8:22-31. [PMID: 26981168 PMCID: PMC4766248 DOI: 10.4252/wjsc.v8.i2.22] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Revised: 11/28/2015] [Accepted: 01/11/2016] [Indexed: 02/06/2023] Open
Abstract
Kidney disease is an escalating global health problem, for which the formulation of therapeutic approaches using stem cells has received increasing research attention. The complexity of kidney anatomy and function, which includes the diversity of renal cell types, poses formidable challenges in the identification of methods to generate replacement structures. Recent work using the zebrafish has revealed their high capacity to regenerate the integral working units of the kidney, known as nephrons, following acute injury. Here, we discuss these findings and explore the ways that zebrafish can be further utilized to gain a deeper molecular appreciation of renal stem cell biology, which may uncover important clues for regenerative medicine.
Collapse
|
41
|
Yang J, Zhu F, Wang X, Yao W, Wang M, Pei G, Hu Z, Guo Y, Zhao Z, Wang P, Mou J, Sun J, Zeng R, Xu G, Liao W, Yao Y. Continuous AMD3100 Treatment Worsens Renal Fibrosis through Regulation of Bone Marrow Derived Pro-Angiogenic Cells Homing and T-Cell-Related Inflammation. PLoS One 2016; 11:e0149926. [PMID: 26900858 PMCID: PMC4763993 DOI: 10.1371/journal.pone.0149926] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 02/05/2016] [Indexed: 11/18/2022] Open
Abstract
AMD3100 is a small molecule inhibitor of chemokine receptor type 4 (CXCR4), which is located in the cell membranes of CD34+ cells and a variety of inflammatory cells and has been reported to reduce organ fibrosis in the lung, liver and myocardium. However, the effect of AMD3100 on renal fibrosis is unknown. This study investigated the impact of AMD3100 on renal fibrosis. C57bl/6 mice were subjected to unilateral ureteral obstruction (UUO) surgery with or without AMD3100 administration. Tubular injury, collagen deposition and fibrosis were detected and analyzed by histological staining, immunocytochemistry and Western Blot. Bone marrow derived pro-angiogenic cells (CD45+, CD34+ and CD309+ cells) and capillary density (CD31+) were measured by flow cytometry (FACS) and immunofluorescence (IF). Inflammatory cells, chemotactic factors and T cell proliferation were characterized. We found that AMD3100 treatment did not alleviate renal fibrosis but, rather, increased tissue damage and renal fibrosis. Continuous AMD3100 administration did not improve bone marrow derived pro-angiogenic cells mobilization but, rather, inhibited the migration of bone marrow derived pro-angiogenic cells into the fibrotic kidney. Additionally, T cell infiltration was significantly increased in AMD3100-treated kidneys compared to un-treated kidneys. Thus, treatment of UUO mice with AMD3100 led to an increase in T cell infiltration, suggesting that AMD3100 aggravated renal fibrosis.
Collapse
Affiliation(s)
- Juan Yang
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, Hubei, China
| | - Fengming Zhu
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, Hubei, China
| | - Xiaohui Wang
- Department of Nephrology, Fifth Hospital of Wuhan, 122 Xianzheng Street, Hanyang district, Wuhan 430050, Hubei, China
| | - Weiqi Yao
- Wuhan Hamilton Biotechnology-Co.LTD., B6-4, Wuhan institute of biotechnology, #666 Gaoxin Road, Wuhan 430073, Hubei, China
| | - Meng Wang
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, Hubei, China
| | - Guangchang Pei
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, Hubei, China
| | - Zhizhi Hu
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, Hubei, China
| | - Yujiao Guo
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, Hubei, China
| | - Zhi Zhao
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, Hubei, China
| | - Pengge Wang
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, Hubei, China
| | - Jingyi Mou
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, Hubei, China
| | - Jie Sun
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, Hubei, China
| | - Rui Zeng
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, Hubei, China
| | - Gang Xu
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, Hubei, China
| | - Wenhui Liao
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, Hubei, China
- * E-mail: (WL); (YY)
| | - Ying Yao
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, Hubei, China
- * E-mail: (WL); (YY)
| |
Collapse
|
42
|
Katari R, Edgar L, Wong T, Boey A, Mancone S, Igel D, Callese T, Voigt M, Tamburrini R, Zambon JP, Perin L, Orlando G. Tissue-Engineering Approaches to Restore Kidney Function. Curr Diab Rep 2015; 15:69. [PMID: 26275443 DOI: 10.1007/s11892-015-0643-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Kidney transplantation for the treatment of chronic kidney disease has established outcome and quality of life. However, its implementation is severely limited by a chronic shortage of donor organs; consequently, most candidates remain on dialysis and on the waiting list while accruing further morbidity and mortality. Furthermore, those patients that do receive kidney transplants are committed to a life-long regimen of immunosuppressive drugs that also carry significant adverse risk profiles. The disciplines of tissue engineering and regenerative medicine have the potential to produce alternative therapies which circumvent the obstacles posed by organ shortage and immunorejection. This review paper describes some of the most promising tissue-engineering solutions currently under investigation for the treatment of acute and chronic kidney diseases. The various stem cell therapies, whole embryo transplantation, and bioengineering with ECM scaffolds are outlined and summarized.
Collapse
Affiliation(s)
- Ravi Katari
- Section of Transplantation, Department of Surgery, Wake Forest School of Medicine, Winston Salem, NC, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
|
44
|
Morales EE, Wingert RA. Renal stem cell reprogramming: Prospects in regenerative medicine. World J Stem Cells 2014; 6:458-466. [PMID: 25258667 PMCID: PMC4172674 DOI: 10.4252/wjsc.v6.i4.458] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Revised: 08/21/2014] [Accepted: 09/01/2014] [Indexed: 02/06/2023] Open
Abstract
Stem cell therapy is a promising future enterprise for renal replacement in patients with acute and chronic kidney disease, conditions which affect millions worldwide and currently require patients to undergo lifelong medical treatments through dialysis and/or organ transplant. Reprogramming differentiated renal cells harvested from the patient back into a pluripotent state would decrease the risk of tissue rejection and provide a virtually unlimited supply of cells for regenerative medicine treatments, making it an exciting area of current research in nephrology. Among the major hurdles that need to be overcome before stem cell therapy for the kidney can be applied in a clinical setting are ensuring the fidelity and relative safety of the reprogrammed cells, as well as achieving feasible efficiency in the reprogramming processes that are utilized. Further, improved knowledge about the genetic control of renal lineage development is vital to identifying predictable and efficient reprogramming approaches, such as the expression of key modulators or the regulation of gene activity through small molecule mimetics. Here, we discuss several recent advances in induced pluripotent stem cell technologies. We also explore strategies that have been successful in renal progenitor generation, and explore what these methods might mean for the development of cell-based regenerative therapies for kidney disease.
Collapse
|
45
|
Dziedzic K, Pleniceanu O, Dekel B. Kidney stem cells in development, regeneration and cancer. Semin Cell Dev Biol 2014; 36:57-65. [PMID: 25128731 DOI: 10.1016/j.semcdb.2014.08.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 08/03/2014] [Accepted: 08/05/2014] [Indexed: 12/17/2022]
Abstract
The generation of nephrons during development depends on differentiation via a mesenchymal to epithelial transition (MET) of self-renewing, tissue-specific stem cells confined to a specific anatomic niche of the nephrogenic cortex. These cells may transform to generate oncogenic stem cells and drive pediatric renal cancer. Once nephron epithelia are formed the view of post-MET tissue renal growth and maintenance by adult tissue-specific epithelial stem cells becomes controversial. Recently, genetic lineage tracing that followed clonal evolution of single kidney cells showed that the need for new cells is constantly driven by fate-restricted unipotent clonal expansions in varying kidney segments arguing against a multipotent adult stem cell model. Lineage-restriction was similarly maintained in kidney organoids grown in culture. Importantly, kidney cells in which Wnt was activated were traced to give significant clonal progeny indicating a clonogenic hierarchy. In vivo nephron epithelia may be endowed with the capacity akin to that of unipotent epithelial stem/progenitor such that under specific stimuli can clonally expand/self renew by local proliferation of mature differentiated cells. Finding ways to ex vivo preserve and expand the observed in vivo kidney-forming capacity inherent to both the fetal and adult kidneys is crucial for taking renal regenerative medicine forward. Some of the strategies used to achieve this are sorting human fetal nephron stem/progenitor cells, growing adult nephrospheres or reprogramming differentiated kidney cells toward expandable renal progenitors.
Collapse
Affiliation(s)
- Klaudyna Dziedzic
- Pediatric Stem Cell Research Institute, Sheba Medical Center, Tel Hashomer, Israel; Sackler School of Medicine, Tel Aviv University, Israel
| | - Oren Pleniceanu
- Pediatric Stem Cell Research Institute, Sheba Medical Center, Tel Hashomer, Israel; Sackler School of Medicine, Tel Aviv University, Israel
| | - Benjamin Dekel
- Pediatric Stem Cell Research Institute, Sheba Medical Center, Tel Hashomer, Israel; Sackler School of Medicine, Tel Aviv University, Israel.
| |
Collapse
|
46
|
Shukrun R, Pode-Shakked N, Pleniceanu O, Omer D, Vax E, Peer E, Pri-Chen S, Jacob J, Hu Q, Harari-Steinberg O, Huff V, Dekel B. Wilms' tumor blastemal stem cells dedifferentiate to propagate the tumor bulk. Stem Cell Reports 2014; 3:24-33. [PMID: 25068119 PMCID: PMC4110791 DOI: 10.1016/j.stemcr.2014.05.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 05/18/2014] [Accepted: 05/19/2014] [Indexed: 12/15/2022] Open
Abstract
An open question remains in cancer stem cell (CSC) biology whether CSCs are by definition at the top of the differentiation hierarchy of the tumor. Wilms’ tumor (WT), composed of blastema and differentiated renal elements resembling the nephrogenic zone of the developing kidney, is a valuable model for studying this question because early kidney differentiation is well characterized. WT neural cell adhesion molecule 1-positive (NCAM1+) aldehyde dehydrogenase 1-positive (ALDH1+) CSCs have been recently isolated and shown to harbor early renal progenitor traits. Herein, by generating pure blastema WT xenografts, composed solely of cells expressing the renal developmental markers SIX2 and NCAM1, we surprisingly show that sorted ALDH1+ WT CSCs do not correspond to earliest renal stem cells. Rather, gene expression and proteomic comparative analyses disclose a cell type skewed more toward epithelial differentiation than the bulk of the blastema. Thus, WT CSCs are likely to dedifferentiate to propagate WT blastema. The Wilms’ tumor (WT) blastema can be exclusively propagated in mice Gene and protein analyses place the WT CSC at a specific developmental stage WT CSCs do not correspond to the earliest renal stem cells WT CSCs are likely to dedifferentiate to propagate WT blastema
Collapse
Affiliation(s)
- Rachel Shukrun
- Pediatric Stem Cell Research Institute, Sheba Medical Center, Tel Hashomer 5262000, Israel
- Sheba Centers for Regenerative Medicine and Cancer Research, Sheba Medical Center, Ramat-Gan, Tel Hashomer 5262000, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Naomi Pode-Shakked
- Pediatric Stem Cell Research Institute, Sheba Medical Center, Tel Hashomer 5262000, Israel
- Sheba Centers for Regenerative Medicine and Cancer Research, Sheba Medical Center, Ramat-Gan, Tel Hashomer 5262000, Israel
- Dr. Pinchas Borenstein Talpiot Medical Leadership Program 2013, Sheba Medical Center, Tel Hashomer 5262000, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Oren Pleniceanu
- Pediatric Stem Cell Research Institute, Sheba Medical Center, Tel Hashomer 5262000, Israel
- Sheba Centers for Regenerative Medicine and Cancer Research, Sheba Medical Center, Ramat-Gan, Tel Hashomer 5262000, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Dorit Omer
- Pediatric Stem Cell Research Institute, Sheba Medical Center, Tel Hashomer 5262000, Israel
- Sheba Centers for Regenerative Medicine and Cancer Research, Sheba Medical Center, Ramat-Gan, Tel Hashomer 5262000, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Einav Vax
- Pediatric Stem Cell Research Institute, Sheba Medical Center, Tel Hashomer 5262000, Israel
- Sheba Centers for Regenerative Medicine and Cancer Research, Sheba Medical Center, Ramat-Gan, Tel Hashomer 5262000, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Eyal Peer
- Sheba Centers for Regenerative Medicine and Cancer Research, Sheba Medical Center, Ramat-Gan, Tel Hashomer 5262000, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Sara Pri-Chen
- The Maurice and Gabriela Goldschleger Eye Research Institute, Sheba Medical Center, Tel Hashomer 5262000, Israel
| | - Jasmine Jacob
- Sheba Centers for Regenerative Medicine and Cancer Research, Sheba Medical Center, Ramat-Gan, Tel Hashomer 5262000, Israel
| | - Qianghua Hu
- Department of Genetics, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Orit Harari-Steinberg
- Pediatric Stem Cell Research Institute, Sheba Medical Center, Tel Hashomer 5262000, Israel
- Sheba Centers for Regenerative Medicine and Cancer Research, Sheba Medical Center, Ramat-Gan, Tel Hashomer 5262000, Israel
| | - Vicki Huff
- Department of Genetics, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Benjamin Dekel
- Pediatric Stem Cell Research Institute, Sheba Medical Center, Tel Hashomer 5262000, Israel
- Sheba Centers for Regenerative Medicine and Cancer Research, Sheba Medical Center, Ramat-Gan, Tel Hashomer 5262000, Israel
- The Maurice and Gabriela Goldschleger Eye Research Institute, Sheba Medical Center, Tel Hashomer 5262000, Israel
- Division of Pediatric Nephrology, Sheba Medical Center, Tel Hashomer 5262000, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Corresponding author
| |
Collapse
|
47
|
Rinkevich Y, Montoro DT, Contreras-Trujillo H, Harari-Steinberg O, Newman AM, Tsai JM, Lim X, Van-Amerongen R, Bowman A, Januszyk M, Pleniceanu O, Nusse R, Longaker MT, Weissman IL, Dekel B. In vivo clonal analysis reveals lineage-restricted progenitor characteristics in mammalian kidney development, maintenance, and regeneration. Cell Rep 2014; 7:1270-83. [PMID: 24835991 DOI: 10.1016/j.celrep.2014.04.018] [Citation(s) in RCA: 165] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2013] [Revised: 03/02/2014] [Accepted: 04/09/2014] [Indexed: 12/18/2022] Open
Abstract
The mechanism and magnitude by which the mammalian kidney generates and maintains its proximal tubules, distal tubules, and collecting ducts remain controversial. Here, we use long-term in vivo genetic lineage tracing and clonal analysis of individual cells from kidneys undergoing development, maintenance, and regeneration. We show that the adult mammalian kidney undergoes continuous tubulogenesis via expansions of fate-restricted clones. Kidneys recovering from damage undergo tubulogenesis through expansions of clones with segment-specific borders, and renal spheres developing in vitro from individual cells maintain distinct, segment-specific fates. Analysis of mice derived by transfer of color-marked embryonic stem cells (ESCs) into uncolored blastocysts demonstrates that nephrons are polyclonal, developing from expansions of singly fated clones. Finally, we show that adult renal clones are derived from Wnt-responsive precursors, and their tracing in vivo generates tubules that are segment specific. Collectively, these analyses demonstrate that fate-restricted precursors functioning as unipotent progenitors continuously maintain and self-preserve the mouse kidney throughout life.
Collapse
Affiliation(s)
- Yuval Rinkevich
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Daniel T Montoro
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Humberto Contreras-Trujillo
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Orit Harari-Steinberg
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Sackler School of Medicine, Tel Aviv University, Tel Aviv 52621, Israel
| | - Aaron M Newman
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jonathan M Tsai
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xinhong Lim
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Renee Van-Amerongen
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Angela Bowman
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael Januszyk
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Oren Pleniceanu
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Sackler School of Medicine, Tel Aviv University, Tel Aviv 52621, Israel
| | - Roel Nusse
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Developmental Biology, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael T Longaker
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Irving L Weissman
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Ludwig Center for Cancer Stem Cell Research, Stanford University, Stanford, CA 94305, USA
| | - Benjamin Dekel
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Sackler School of Medicine, Tel Aviv University, Tel Aviv 52621, Israel.
| |
Collapse
|
48
|
Urbach A, Yermalovich A, Zhang J, Spina CS, Zhu H, Perez-Atayde AR, Shukrun R, Charlton J, Sebire N, Mifsud W, Dekel B, Pritchard-Jones K, Daley GQ. Lin28 sustains early renal progenitors and induces Wilms tumor. Genes Dev 2014; 28:971-82. [PMID: 24732380 PMCID: PMC4018495 DOI: 10.1101/gad.237149.113] [Citation(s) in RCA: 137] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Accepted: 03/25/2014] [Indexed: 11/25/2022]
Abstract
Wilms Tumor, the most common pediatric kidney cancer, evolves from the failure of terminal differentiation of the embryonic kidney. Here we show that overexpression of the heterochronic regulator Lin28 during kidney development in mice markedly expands nephrogenic progenitors by blocking their final wave of differentiation, ultimately resulting in a pathology highly reminiscent of Wilms tumor. Using lineage-specific promoters to target Lin28 to specific cell types, we observed Wilms tumor only when Lin28 is aberrantly expressed in multiple derivatives of the intermediate mesoderm, implicating the cell of origin as a multipotential renal progenitor. We show that withdrawal of Lin28 expression reverts tumorigenesis and markedly expands the numbers of glomerulus-like structures and that tumor formation is suppressed by enforced expression of Let-7 microRNA. Finally, we demonstrate overexpression of the LIN28B paralog in a significant percentage of human Wilms tumor. Our data thus implicate the Lin28/Let-7 pathway in kidney development and tumorigenesis.
Collapse
Affiliation(s)
- Achia Urbach
- Stem Cell Transplantation Program, Division of Pediatric Hematology/Oncology, Children’s Hospital Boston, Boston, Massachusetts 02115, USA
- Howard Hughes Medical Institute, Boston, Massachusetts 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, USA
- Harvard Stem Cell Institute, Boston, Massachusetts 02115, USA
| | - Alena Yermalovich
- Stem Cell Transplantation Program, Division of Pediatric Hematology/Oncology, Children’s Hospital Boston, Boston, Massachusetts 02115, USA
- Howard Hughes Medical Institute, Boston, Massachusetts 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, USA
- Harvard Stem Cell Institute, Boston, Massachusetts 02115, USA
| | - Jin Zhang
- Stem Cell Transplantation Program, Division of Pediatric Hematology/Oncology, Children’s Hospital Boston, Boston, Massachusetts 02115, USA
- Howard Hughes Medical Institute, Boston, Massachusetts 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, USA
- Harvard Stem Cell Institute, Boston, Massachusetts 02115, USA
| | - Catherine S. Spina
- Stem Cell Transplantation Program, Division of Pediatric Hematology/Oncology, Children’s Hospital Boston, Boston, Massachusetts 02115, USA
- Howard Hughes Medical Institute, Boston, Massachusetts 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, USA
- Harvard Stem Cell Institute, Boston, Massachusetts 02115, USA
| | - Hao Zhu
- Children’s Research Institute
- Department of Pediatrics
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | | | - Rachel Shukrun
- Pediatric Stem Cell Research Institute
- Division of Pediatric Nephrology, Sheba Medical Center, Sackler School of Medicine, Tel Aviv University, Tel Aviv 52621, Israel
| | - Jocelyn Charlton
- Institute of Child Health, University College London, London WC1H 0AJ, United Kingdom
| | - Neil Sebire
- Department of Histopathology, Camelia Botnar Laboratories, Great Ormond Street Hospital for Children, London WC1N 3JH, United Kingdom
| | - William Mifsud
- Department of Histopathology, Camelia Botnar Laboratories, Great Ormond Street Hospital for Children, London WC1N 3JH, United Kingdom
| | - Benjamin Dekel
- Pediatric Stem Cell Research Institute
- Division of Pediatric Nephrology, Sheba Medical Center, Sackler School of Medicine, Tel Aviv University, Tel Aviv 52621, Israel
| | - Kathy Pritchard-Jones
- Institute of Child Health, University College London, London WC1H 0AJ, United Kingdom
| | - George Q. Daley
- Stem Cell Transplantation Program, Division of Pediatric Hematology/Oncology, Children’s Hospital Boston, Boston, Massachusetts 02115, USA
- Howard Hughes Medical Institute, Boston, Massachusetts 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, USA
- Harvard Stem Cell Institute, Boston, Massachusetts 02115, USA
| |
Collapse
|
49
|
Kang M, Han YM. Differentiation of human pluripotent stem cells into nephron progenitor cells in a serum and feeder free system. PLoS One 2014; 9:e94888. [PMID: 24728509 PMCID: PMC3984279 DOI: 10.1371/journal.pone.0094888] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Accepted: 03/20/2014] [Indexed: 12/13/2022] Open
Abstract
Objectives Kidney disease is emerging as a critical medical problem worldwide. Because of limited treatment options for the damaged kidney, stem cell treatment is becoming an alternative therapeutic approach. Of many possible human stem cell sources, pluripotent stem cells are most attractive due to their self-renewal and pluripotent capacity. However, little is known about the derivation of renal lineage cells from human pluripotent stem cells (hPSCs). In this study, we developed a novel protocol for differentiation of nephron progenitor cells (NPCs) from hPSCs in a serum- and feeder-free system. Materials and Methods We designed step-wise protocols for differentiation of human pluripotent stem cells toward primitive streak, intermediate mesoderm and NPCs by recapitulating normal nephrogenesis. Expression of key marker genes was examined by RT-PCR, real time RT-PCR and immunocytochemistry. Each experiment was independently performed three times to confirm its reproducibility. Results After modification of culture period and concentration of exogenous factors, hPSCs can differentiate into NPCs that markedly express specific marker genes such as SIX2, GDNF, HOXD11, WT1 and CITED1 in addition to OSR1, PAX2, SALL1 and EYA1. Moreover, NPCs possess the potential of bidirectional differentiation into both renal tubular epithelial cells and glomerular podocytes in defined culture conditions. In particular, approximately 70% of SYN-positive cells were obtained from hPSC-derived NPCs after podocytes induction. NPCs can also form in vitro tubule-like structures in three dimensional culture systems. Conclusions Our novel protocol for hPSCs differentiation into NPCs can be useful for producing alternative sources of cell replacement therapy and disease modeling for human kidney diseases.
Collapse
Affiliation(s)
- Minyong Kang
- Graduate Schools of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Yong-Mahn Han
- Graduate Schools of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
- Department of Biological Sciences, KAIST, Daejeon, Republic of Korea
- * E-mail:
| |
Collapse
|
50
|
Cirio MC, de Groh ED, de Caestecker MP, Davidson AJ, Hukriede NA. Kidney regeneration: common themes from the embryo to the adult. Pediatr Nephrol 2014; 29:553-64. [PMID: 24005792 PMCID: PMC3944192 DOI: 10.1007/s00467-013-2597-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 07/26/2013] [Accepted: 07/29/2013] [Indexed: 02/07/2023]
Abstract
The vertebrate kidney has an inherent ability to regenerate following acute damage. Successful regeneration of the injured kidney requires the rapid replacement of damaged tubular epithelial cells and reconstitution of normal tubular function. Identifying the cells that participate in the regeneration process as well as the molecular mechanisms involved may reveal therapeutic targets for the treatment of kidney disease. Renal regeneration is associated with the expression of genetic pathways that are necessary for kidney organogenesis, suggesting that the regenerating tubular epithelium may be "reprogrammed" to a less-differentiated, progenitor state. This review will highlight data from various vertebrate models supporting the hypothesis that nephrogenic genes are reactivated as part of the process of kidney regeneration following acute kidney injury (AKI). Emphasis will be placed on the reactivation of developmental pathways and how our understanding of the resulting regeneration process may be enhanced by lessons learned in the embryonic kidney.
Collapse
Affiliation(s)
- M. Cecilia Cirio
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Eric D. de Groh
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Mark P. de Caestecker
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Alan J. Davidson
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Neil A. Hukriede
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA, United States of America
| |
Collapse
|