1
|
Afshar A, Khoradmehr A, Zare A, Basouli N, Keshtkar M, Nabipour I, Mahdipour M, Mahmoudpour M, Kaliyev AA, Mussin NM, Baspakova A, Tamadon A. Histological evaluation of renal progenitor/stem cells, renal mesenchymal stem-like cells, and endothelial progenitor cells in chronic kidney disease and end-stage renal disease, and molecular docking analysis of drug-receptor interactions. Tissue Cell 2024; 90:102527. [PMID: 39181089 DOI: 10.1016/j.tice.2024.102527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 08/02/2024] [Accepted: 08/17/2024] [Indexed: 08/27/2024]
Abstract
Chronic kidney disease (CKD) and end-stage renal disease (ESRD) are prevalent and debilitating conditions with a significant impact on patients' quality of life. In this study, we conducted a comprehensive investigation into the histological characteristics of renal progenitor/stem cells (RPCs), renal mesenchymal stem-like cells, and endothelial progenitor cells (EPCs) in CKD and ESRD patients. Additionally, we performed a molecular docking analysis to explore potential drug-receptor interactions involving common medications prescribed to CKD patients. Our histological examination revealed a noteworthy increase in the number of CD24- and CD133-positive cells in CKD and ESRD patients, representing RPCs. These cells are implicated in kidney repair and regeneration, underscoring their potential role in CKD management. Moreover, we observed an elevation in the number of EPCs within the kidneys of CKD and ESRD patients, suggesting a protective role of EPCs in kidney preservation. The molecular docking analysis unveiled intriguing insights into potential drug interventions. Notably, digoxin exhibited the highest in-silico binding affinity to numerous receptors associated with the functions of RPCs, renal mesenchymal stem-like cells, and EPCs, emphasizing the potential multifaceted effects of this cardiac glycoside in CKD patients. Other drugs, including apixaban, glimepiride, and glibenclamide, also displayed strong in-silico affinities to specific receptors, indicating their potential influence on various renal cell functions. In conclusion, this study provides valuable insights into the histological alterations in renal cell populations in CKD and ESRD patients and underscores the potential roles of RPCs and EPCs in kidney repair and preservation. The molecular docking analysis reveals the complex interactions between common drugs and renal cells, suggesting the need for further in-vitro and in-vivo research to fully understand these relationships. These findings contribute to our understanding of CKD and offer new avenues for research into potential therapeutic interventions.
Collapse
Affiliation(s)
- Alireza Afshar
- Student Research Committee, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Arezoo Khoradmehr
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | | | - Nahid Basouli
- Student Research Committee, Bushehr University of Medical Sciences, Bushehr, Iran
| | | | - Iraj Nabipour
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mahdi Mahdipour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Mahmoudpour
- The Persian Gulf Tropical Medicine Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Asset A Kaliyev
- General Surgery, West-Kazakhstan Medical University named after Marat Ospanov, Aktobe, Kazakhstan
| | - Nadiar M Mussin
- General Surgery, West-Kazakhstan Medical University named after Marat Ospanov, Aktobe, Kazakhstan
| | - Akmaral Baspakova
- Department for Scientific Work, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Amin Tamadon
- PerciaVista R&D Co., Shiraz, Iran; Department of Natural Sciences, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan; Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
2
|
Chen L. Renal Organoids from Whole Kidney Cells. Methods Mol Biol 2024; 2764:157-164. [PMID: 38393594 DOI: 10.1007/978-1-0716-3674-9_11] [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] [Indexed: 02/25/2024]
Abstract
Organoid technology, as a three-dimensional (3D) culture method, provides a feasible tool to self-organize multiple types of organ-specific cells, construct inherent anatomic structures, and display functional biological activities. For the purpose of renal regeneration, renal organoids are considered as predictive options to form functional kidney substitutions in vitro. Here, we describe an accessible and convenient way to generate renal organoids without differentiation procedures using whole kidney cells.
Collapse
Affiliation(s)
- Liang Chen
- Urology Department, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
3
|
Copur S, Yavuz F, Covic A, Kanbay M. A review on renal autologous cell transplantation: an investigational approach towards chronic kidney disease. Int Urol Nephrol 2023; 55:2539-2544. [PMID: 36971874 DOI: 10.1007/s11255-023-03574-5] [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: 12/30/2022] [Accepted: 03/22/2023] [Indexed: 03/29/2023]
Abstract
Chronic kidney disease is among the most common causes of mortality and morbidity in adult population with limited therapeutic approaches including various medications and kidney replacement therapies. Kidney transplantation is the gold standard therapeutic alternative for the management of chronic kidney disease; nonetheless, important drawbacks include the lack of adequate living or deceased donors, high rates of pre- and post-operative complications including surgical complications, infectious complications and medication-induced adverse effects. With the latest preclinical and in vitro studies demonstrating the potentiality of kidney cells obtained from diseased kidneys to convert into fully functional kidney cells lead to a novel therapeutic alternative referred as autologous selected renal cell transplantation. Even though the clinical studies investigating the efficiency and adverse effects of autologous selected renal cell transplantation are limited, it is no doubt promising. The need for future large-scale studies on chronic kidney disease patients from a diversity of etiologies is clear for the better establishment of the therapeutic potential of autologous selected renal cell transplantation. In this narrative review, our aim is to evaluate the role of renal autologous stem cell therapy in the management of chronic kidney disease.
Collapse
Affiliation(s)
- Sidar Copur
- Department of Medicine, Koc University School of Medicine, 34010, Istanbul, Turkey
| | - Furkan Yavuz
- Department of Medicine, Koc University School of Medicine, 34010, Istanbul, Turkey
| | - Adrian Covic
- Department of Nephrology, Grigore T. Popa University of Medicine, Iasi, Romania
| | - Mehmet Kanbay
- Department of Medicine, Division of Nephrology, Koc University School of Medicine, Istanbul, Turkey.
| |
Collapse
|
4
|
Kim JH, Yang H, Kim MW, Cho KS, Kim DS, Yim HE, Atala Z, Ko IK, Yoo JJ. The Delivery of the Recombinant Protein Cocktail Identified by Stem Cell-Derived Secretome Analysis Accelerates Kidney Repair After Renal Ischemia-Reperfusion Injury. Front Bioeng Biotechnol 2022; 10:848679. [PMID: 35646873 PMCID: PMC9130839 DOI: 10.3389/fbioe.2022.848679] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 04/27/2022] [Indexed: 12/14/2022] Open
Abstract
Recent advances in cell therapy have shown the potential to treat kidney diseases. As the treatment effects of the cell therapies are mainly attributed to secretomes released from the transplanted cells, the delivery of secretomes or conditioned medium (CM) has emerged as a promising treatment option for kidney disease. We previously demonstrated that the controlled delivery of human placental stem cells (hPSC)-derived CM using platelet-rich plasma (PRP) ameliorated renal damages and restored kidney function in an acute kidney injury (AKI) model in rats. The proteomics study of the hPSC-CM revealed that hPSC secrets several proteins that contribute to kidney tissue repair. Based on our results, this study proposed that the proteins expressed in the hPSC-CM and effective for kidney repair could be used as a recombinant protein cocktail to treat kidney diseases as an alternative to CM. In this study, we analyzed the secretome profile of hPSC-CM and identified five proteins (follistatin, uPAR, ANGPLT4, HGF, VEGF) that promote kidney repair. We investigated the feasibility of delivering the recombinant protein cocktail to improve structural and functional recovery after AKI. The pro-proliferative and anti-apoptotic effects of the protein cocktail on renal cells are demonstrated in vitro and in vivo. The intrarenal delivery of these proteins with PRP ameliorates the renal tubular damage and improved renal function in the AKI-induced rats, yielding similar therapeutic effects compared to the CM delivery. These results indicate that our strategy may provide a therapeutic solution to many challenges associated with kidney repair resulting from the lack of suitable off-the-shelf regenerative medicine products.
Collapse
Affiliation(s)
- Ji Hyun Kim
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Heejo Yang
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States.,Department of Urology, Soonchunhyang University College of Medicine, Cheonan, South Korea
| | - Michael W Kim
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Kang Su Cho
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States.,Department of Urology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Doo Sang Kim
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States.,Department of Urology, Soonchunhyang University College of Medicine, Cheonan, South Korea
| | - Hyung Eun Yim
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States.,Department of Pediatrics, Korea University College of Medicine, Seoul, South Korea
| | - Zachary Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - In Kap Ko
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - James J Yoo
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
5
|
Zhai Y, Li D, Wang Z, Shao L, Yin N, Li W. Cortex Mori Radicis attenuates streptozotocin-induced diabetic renal injury in mice via regulation of transient receptor potential canonical channel 6. Endocr Metab Immune Disord Drug Targets 2022; 22:862-873. [PMID: 35016601 DOI: 10.2174/1871530322666220110161458] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 11/09/2021] [Accepted: 12/14/2021] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Cortex Mori Radicis (CMR) has been reported to possess anti-pyretic, anti-convulsant, anti-allergic, anti-inflammatory, and anti-diabetic effects. In this study, we aimed to investigate the effect of CMR on streptozotocin (STZ)-induced diabetic renal injury in mice and explore the underlying mechanism. METHODS Mice were gavaged with different doses of CMR for continuous 7 days. Then, STZ (50 mg/kg) was applied to induce renal injury associated with type 1 diabetes. Firstly, blood glucose levels and metabolic parameters were evaluated, including weight, food intake, and excrement. HE and PAS staining were performed to present renal histological changes. Renal inflammation, fibrosis, and oxidative stress were assayed by real time PCR and ELISA, separately. Additionally, podocyte-related markers, such as nephrin and wilms' tumor-1 (WT-1) were detected by immunohistochemical staining and Western blot separately. Lastly, expression of transient receptor potential canonical channel 6 (TRPC6) and activation of MAPK signaling pathways were assayed. RESULTS CMR pretreatment significantly lowered the blood glucose levels, suppressed renal inflammation, fibrosis and oxidative stress, and relieved renal pathological injury, accompanying the inhibition of nephrin and WT-1 expression in STZ-induced diabetic mice. Moreover, CMR decreased the expression of TRPC6 and suppressed phosphorylation of ERK, but not P38 MAPK and JNK. Notably, the application of hyperforin, a specific activator of TRPC6, significantly abrogated the hypoglycemic effect of CMR and reversed the suppression of CMR on TRPC6 expression and ERK activation in the diabetic mice. CONCLUSION Our findings indicated that CMR attenuated early renal injury in STZ-induced diabetic mice through inhibiting ERK signaling via regulation of TRPC6, which suggests that CMR can be considered as a promising candidate for the management of diabetes-related renal complications.
Collapse
Affiliation(s)
- Yi Zhai
- Department of Cardiology, Liyuan Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, China
| | - Dan Li
- College of Acupuncture and Moxibustion, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Zhigang Wang
- Department of Pathogen Biology, School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Luyao Shao
- Department of Pathogen Biology, School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Nina Yin
- Department of Anatomy, School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Weihua Li
- Department of Cardiology, Liyuan Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, China
| |
Collapse
|
6
|
Aslam A, Shah S, Abbas G, Rehman AU, Malhi TH, Alotaibi NH, Alzarea AI, Rasool MF, Khurram H, Noureen S, Saeed Bokhari MT. Assessment of health-related quality of life in hypertensive hemodialysis patients. PeerJ 2022; 10:e12690. [PMID: 35036092 PMCID: PMC8734469 DOI: 10.7717/peerj.12690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 12/04/2021] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Globally, the prevalence of hypertension (HTN) with the coexistence of chronic kidney disease (CKD) is increasing, resulting in poor quality of life. The main objective of the study was to measure the health-related quality of life (HRQoL) of hypertensive hemodialysis patients. METHODS A multicenter follow-up study was carried out in six public and two private dialysis centers in Pakistan. A total of 517 hypertensive hemodialysis patients responded by completing the questionnaire at baseline and two subsequent phases. The quality of life of these patients was assessed using the EQ-5D-5L questionnaire (a standardized instrument for measuring generic health status). Statistical analysis was done using a multivariate linear regression model, Friedman test and Kruskal Wallis test. RESULTS The majority of patients (58.2%) had normal body mass index and about 60.5% of the patients were taking less salt due to HTN. Friedman test gave the statistically significant results (p ≤ 0.001) in systolic blood pressure (BP), diastolic BP and EQ-5D visual analogue scale (VAS) score between three phases (initial visit, first follow-up and second follow-up). A significant improvement was observed in self-care and usual activities from initial visit to first follow-up (p < 0.05). The most problematic dimension among the hypertensive patients with CKD was pain/discomfort (86.5%). CONCLUSIONS HTN with coexisting CKD in hemodialysis patients severely affected HRQoL. Pain/discomfort was the most problematic dimension among the participants.
Collapse
Affiliation(s)
- Ayesha Aslam
- Department of Neurology, King Edward Medical University, Lahore, Pakistan
| | - Shahid Shah
- Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Ghulam Abbas
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Anees ur Rehman
- Department of Pharmacy Practice, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| | - Tauqeer Hussain Malhi
- Department of Clinical Pharmacy, College of Pharmacy, Jouf University, Sakaka, Al-Jouf Province, Kingdom of Saudi Arabia
| | - Nasser Hadal Alotaibi
- Department of Clinical Pharmacy, College of Pharmacy, Jouf University, Sakaka, Al-Jouf Province, Kingdom of Saudi Arabia
| | - Abdulaziz Ibrahim Alzarea
- Department of Clinical Pharmacy, College of Pharmacy, Jouf University, Sakaka, Al-Jouf Province, Kingdom of Saudi Arabia
| | - Muhammad Fawad Rasool
- Department of Pharmacy Practice, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| | - Haris Khurram
- Department of Sciences and Humanities, National University of Computer and Emerging Sciences, Faisalabad, Pakistan
| | - Sibgha Noureen
- Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Muhammad Talha Saeed Bokhari
- Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| |
Collapse
|
7
|
Matchimakul P, Pongkan W, Kongtung P, Mektrirat R. Comparative quantitation of aquaporin-2 and arginine vasopressin receptor-2 localizations among chronic kidney disease and healthy kidney in dogs. Vet World 2021; 14:2773-2781. [PMID: 34903939 PMCID: PMC8654747 DOI: 10.14202/vetworld.2021.2773-2781] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 09/09/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND AND AIM Aquaporin-2 (AQP2) and arginine vasopressin receptor-2 (AVPR2) are proteins that control water homeostasis in principal cells. Chronic kidney disease (CKD) is defined as the impairment and irreversible loss of kidney function and/or structure, which causes water imbalances and polyuria. The study aimed to know the expression of AQPs and AVPR2 in the kidneys of a canine with CKD. MATERIALS AND METHODS The kidneys were collected from two dog carcasses from Small Animal Teaching Hospital, Faculty of Veterinary Medicine, Chiang Mai University. The kidney tissue was prepared for immunohistochemistry and investigated the expression and localization of tissue's AQP2 and AVPR2. For statistical analysis, the Mann-Whitney U-test was applied to the data. RESULTS By immunohistochemistry, AQP2 was expressed strongly in the basolateral and apical membranes of the principal cells, whereas AVPR2 was localized in the principal cell's basolateral membrane in both renal cortex and renal medulla. In the normal kidney, the semi-quantitative immunohistochemistry for the percentage of protein expression of AQP2 and AVPR2 was 5.062±0.4587 and 4.306±0.7695, respectively. In contrast, protein expression of AQP2 and AVPR2 in CKD was found to be 1.218±0.1719 and 0.8536±0.1396, respectively. The data shows that the percentage of AQP2 and AVPR2 expression was decreased, corresponding to a 4-fold and 5-fold in CKD (p<0.001). CONCLUSION Our findings revealed that CKD was a marked decrease in AQP2 and AVPR2 expression. The central role of specific AQP2 and AVPR2 in regulating water homeostasis will provide correlations in case of CKD with polyuria.
Collapse
Affiliation(s)
- Pitchaya Matchimakul
- Department of Veterinary Bioscience and Veterinary Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand
- Integrative Research Center for Veterinary Circulatory Sciences, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand
| | - Wanpitak Pongkan
- Department of Veterinary Bioscience and Veterinary Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand
- Integrative Research Center for Veterinary Circulatory Sciences, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand
| | - Piyamat Kongtung
- Central Laboratory, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand
| | - Raktham Mektrirat
- Department of Veterinary Bioscience and Veterinary Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand
- Integrative Research Center for Veterinary Circulatory Sciences, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand
| |
Collapse
|
8
|
Bioprinting of kidney in vitro models: cells, biomaterials, and manufacturing techniques. Essays Biochem 2021; 65:587-602. [PMID: 34096573 PMCID: PMC8365327 DOI: 10.1042/ebc20200158] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 05/03/2021] [Accepted: 05/20/2021] [Indexed: 12/19/2022]
Abstract
The number of patients with end-stage renal disease is continuously increasing worldwide. The only therapies for these patients are dialysis and organ transplantation, but the latter is limited due to the insufficient number of donor kidneys available. Research in kidney disease and alternative therapies are therefore of outmost importance. In vitro models that mimic human kidney functions are essential to provide better insights in disease and ultimately novel therapies. Bioprinting techniques have been increasingly used to create models with some degree of function, but their true potential is yet to be achieved. Bioprinted renal tissues and kidney-like constructs presents challenges, for example, choosing suitable renal cells and biomaterials for the formulation of bioinks. In addition, the fabrication of complex renal biological structures is still a major bottleneck. Advances in pluripotent stem cell-derived renal progenitors has contributed to in vivo-like rudiment structures with multiple renal cells, and these started to make a great impact on the achieved models. Natural- or synthetic-based biomaterial inks, such as kidney-derived extracellular matrix and gelatin-fibrin hydrogels, which show the potential to partially replicate in vivo-like microenvironments, have been largely investigated for bioprinting. As the field progresses, technological, biological and biomaterial developments will be required to yield fully functional in vitro tissues that can contribute to a better understanding of renal disease, to improve predictability in vitro of novel therapeutics, and to facilitate the development of alternative regenerative or replacement treatments. In this review, we resume the main advances on kidney in vitro models reported so far.
Collapse
|
9
|
Min S, Cleveland D, Ko IK, Kim JH, Yang HJ, Atala A, Yoo JJ. Accelerating neovascularization and kidney tissue formation with a 3D vascular scaffold capturing native vascular structure. Acta Biomater 2021; 124:233-243. [PMID: 33524561 DOI: 10.1016/j.actbio.2021.01.040] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 01/11/2021] [Accepted: 01/26/2021] [Indexed: 01/03/2023]
Abstract
Establishing an adequate vascularization of three-dimensional (3D) bioengineered tissues remains a critical challenge. We previously fabricated a vascular scaffold using the vascular corrosion casting technique, which provides a similar 3D geometry of native kidney vasculature. In this study, we functionalized the collagen vascular scaffold with a controlled release of vascular endothelial growth factor (VEGF vascular scaffold) to further promote vascularization. The VEGF vascular scaffold showed improved angiogenic capability in 2-dimensional (2D) and 3D in vitro settings. Implantation of the VEGF vascular scaffold seeded with human renal cells into a rat kidney demonstrated enhanced implant vascularization and reduced apoptosis of implanted human renal cells. Hybrid renal tubule-like structures composed of implanted human and migrated host renal cells were formed. This work highlights the critical role of early vascularization of the geometrically mimetic vascular scaffold using the VEGF incorporated vascular scaffold in reducing apoptosis of implanted cells as well as the formation of renal tissue structures.
Collapse
Affiliation(s)
- Sangil Min
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, NC, USA; Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - David Cleveland
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, NC, USA
| | - In Kap Ko
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, NC, USA
| | - Ji Hyun Kim
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, NC, USA
| | - Hee Jo Yang
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, NC, USA; Department of Urology, Soonchunhyang University College of Medicine, Cheonan, South Korea
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, NC, USA
| | - James J Yoo
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, NC, USA.
| |
Collapse
|
10
|
Ding B, Sun G, Liu S, Peng E, Wan M, Chen L, Jackson J, Atala A. Three-Dimensional Renal Organoids from Whole Kidney Cells: Generation, Optimization, and Potential Application in Nephrotoxicology In Vitro. Cell Transplant 2021; 29:963689719897066. [PMID: 32166969 PMCID: PMC7504083 DOI: 10.1177/0963689719897066] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The kidney function of patients with chronic kidney disease (CKD) is impaired
irreversibly. Organ transplantation is the only treatment to restore kidney function in
CKD patients. The assessment of new potential therapeutic procedures relies heavily on
experimental animal models, but it is limited by its human predictive capacity. In
addition, the frequently used two-dimensional in vitro human renal cell
models cannot replicate all the features of the in vivo situation. In this study, we
developed a three-dimensional (3D) in vitro human renal organoid model
from whole kidney cells as a promising drug screening tool. At present, the renal tissue
generated from human pluripotent stem cells (hPSCs) exhibits intrinsic tumorigenicity
properties. Here we first developed a 3D renal organoid culture system that originated
from adult differentiated cells without gene modification. Renal organoids composed of
multiple cell types were created under optimal experimental conditions and evaluated for
morphology, viability and erythropoietin production. As a novel screening tool for renal
toxicity, 3D organoids were exposed to three widely used drugs: aspirin, penicillin G and
cisplatin. The study results showed this 3D renal organoid model can be used as a drug
screening tool, a new in vitro 3D human kidney model, and provide hope
for potential regenerative therapies for CKD.
Collapse
Affiliation(s)
- Beichen Ding
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HB, China.,Department of Urinary Surgery, First Affiliated Hospital of Harbin Medical University, Harbin, HLJ, China
| | - Guoliang Sun
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HB, China
| | - Shiliang Liu
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HB, China
| | - Ejun Peng
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HB, China
| | - Meimei Wan
- Wake Forest Institute for Regenerative Medicine, Wake Forest University, Winston-Salem, NC, USA
| | - Liang Chen
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HB, China.,Wake Forest Institute for Regenerative Medicine, Wake Forest University, Winston-Salem, NC, USA
| | - John Jackson
- Wake Forest Institute for Regenerative Medicine, Wake Forest University, Winston-Salem, NC, USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest University, Winston-Salem, NC, USA
| |
Collapse
|
11
|
Sun G, Ding B, Wan M, Chen L, Jackson J, Atala A. Formation and optimization of three-dimensional organoids generated from urine-derived stem cells for renal function in vitro. Stem Cell Res Ther 2020; 11:309. [PMID: 32698872 PMCID: PMC7374873 DOI: 10.1186/s13287-020-01822-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 06/25/2020] [Accepted: 07/10/2020] [Indexed: 02/06/2023] Open
Abstract
Background Organoids play an important role in basic research, drug screening, and regenerative medicine. Here, we aimed to develop a novel kind of three-dimensional (3D) organoids generated from urine-derived stem cells (USCs) and to explore whether kidney-specific extracellular matrix (kECM) could enable such organoids for renal function in vitro. Methods USCs were isolated from human urine samples and cultured with kECM extraction to generate 3D organoids in vitro. Eight densities from 1000 to 8000 cells per organoids were prepared, and both ATP assay and Live/Dead staining were used to determine the optimal USC density in forming organoids and kECM additive concentration. The morphology and histology of as-made organoids were evaluated by hematoxylin and eosin (H.E.) staining, immunofluorescence staining and whole mount staining. Additionally, RT-qPCR was implemented to detect renal-related gene expression. Drug toxicity test was conducted to evaluate the potential application for drug screening. The renal organoids generated from whole adult kidney cells were used as a positive control in multiple assessments. Results The optimized cell density to generate ideal USC-derived organoids (USC-organoids) was 5000 cells/well, which was set as applying density in the following experiments. Besides, the optimal concentration of kECM was revealed to be 10%. On this condition, Live/Dead staining showed that USC-organoids were well self-organized without significant cell death. Moreover, H.E. staining showed that compact and viable organoids were generated without obvious necrosis inside organoids, which were very close to renal organoids morphologically. Furthermore, specific proximal tubule marker Aquaporin-1 (AQP1), kidney endocrine product erythropoietin (EPO), kidney glomerular markers Podocin and Synaptopodin were detected positively in USC-organoids with kECM. Nephrotoxicity testing showed that aspirin, penicillin G, and cisplatin could exert drug-induced toxicity on USC-organoids with kECM. Conclusions USC-organoids could be developed from USCs via an optimal procedure. Combining culture with kECM, USC-organoid properties including morphology, histology, and specific gene expression were identified to be similar with real renal organoids. Additionally, USC-organoids posed kECM in vitro showed the potential to be a drug screening tool which might take the place of renal organoids to some extent in the future.
Collapse
Affiliation(s)
- Guoliang Sun
- Department of Urology, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang Avenue, Qiaokou, Wuhan, 430030, HB, China
| | - Beichen Ding
- Department of Urology, First Affiliated Hospital of Harbin Medical University, Harbin, HLJ, China
| | - Meimei Wan
- Wake Forest Institute for Regenerative Medicine, Wake Forest University, Winston Salem, NC, USA
| | - Liang Chen
- Department of Urology, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang Avenue, Qiaokou, Wuhan, 430030, HB, China. .,Wake Forest Institute for Regenerative Medicine, Wake Forest University, Winston Salem, NC, USA.
| | - John Jackson
- Wake Forest Institute for Regenerative Medicine, Wake Forest University, Winston Salem, NC, USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest University, Winston Salem, NC, USA
| |
Collapse
|
12
|
Huling J, Min SI, Kim DS, Ko IK, Atala A, Yoo JJ. Kidney regeneration with biomimetic vascular scaffolds based on vascular corrosion casts. Acta Biomater 2019; 95:328-336. [PMID: 30953799 DOI: 10.1016/j.actbio.2019.04.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 03/19/2019] [Accepted: 04/01/2019] [Indexed: 10/27/2022]
Abstract
We have developed a biomimetic renal vascular scaffold based on a vascular corrosion casting technique. This study evaluated the feasibility of using this novel biomimetic scaffold for kidney regeneration in a rat kidney cortical defect model. Vascular corrosion casts were prepared from normal rat kidneys by perfusion with 10% polycaprolactone (PCL) solution, followed by tissue digestion. The corrosion PCL cast was coated with collagen, and PCL was removed from within the collagen coating, leaving only a hollow collagen-based biomimetic vascular scaffold. The fabricated scaffolds were pre-vascularized with MS1 endothelial cell coating, incorporated into 3D renal constructs, and subsequently implanted either with or without human renal cells in the renal cortex of nude rats. The implanted collagen-based vascular scaffold was easily identified and integrated into native kidney tissue. The biomimetic vascular scaffold coated with endothelial cells (MS1) showed significantly enhanced vascularization, as compared to the uncoated scaffold and hydrogel only groups (P < 0.001). Along with the improved vascularization effects, the MS1-coated scaffolds showed a significant renal cell infiltration from the neighboring host tissue, as compared to the other groups (P < 0.05). Moreover, addition of human renal cells to the MS1-coated scaffold resulted in further enhancement of vascularization and tubular structure regeneration within the implanted constructs. The biomimetic collagen vascular scaffolds coated with endothelial cells are able to enhance vascularization and facilitate the formation of renal tubules after 14 days when combined with human renal cells. This study shows the feasibility of bioengineering vascularized functional renal tissues for kidney regeneration. STATEMENT OF SIGNIFICANCE: Vascularization is one of the major hurdles affecting the survival and integration of implanted three-dimensional tissue constructs in vivo. A novel, biomimetic, collagen-based vascular scaffold that is structurally identical to native kidney tissue was developed and tested. This biomimetic vascularized scaffold system facilitates the development of new vessels and renal cell viability in vivo when implanted in a partial renal defect. The use of this scaffold system could address the challenges associated with vascularization, and may be an ideal treatment strategy for partial augmentation of renal function in patients with chronic kidney disease.
Collapse
|
13
|
George SK, Abolbashari M, Kim TH, Zhang C, Allickson J, Jackson JD, Lee SJ, Ko IK, Atala A, Yoo JJ. Effect of Human Amniotic Fluid Stem Cells on Kidney Function in a Model of Chronic Kidney Disease. Tissue Eng Part A 2019; 25:1493-1503. [PMID: 30829146 DOI: 10.1089/ten.tea.2018.0371] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Kidney disease is a major medical problem globally. Chronic kidney disease (CKD) is a progressive loss of kidney function. It causes accumulation of waste and fluid in the body, eventually resulting in kidney failure as well as damaging other organs. Although dialysis and kidney transplantation have been used as primary treatments for renal disease, dialysis does not restore full renal function, and there is a shortage of donor kidneys for transplantation. Recent advances in cell-based therapies have offered a means to augment and restore renal function. Various types of cells have been tested to evaluate their therapeutic effects on injured kidneys. Among various types of cells, amniotic fluid stem cells (AFSCs) share advantages of both embryonic and adult stem cells, such as pluripotent activity, remarkable plasticity, and immunomodulatory effects, which may allow their future therapeutic use as an "off-the-shelf" cell source. AFSC presents advantages of both conventional pluripotent and adult stem cells, such as pluripotent activity, remarkable plasticity, and immunomodulatory effects. This study demonstrates that administration of human-derived AFSC facilitates functional and structural improvement in a rat model of CKD, and suggests that cell therapy with AFSC has potential as a therapeutic strategy to recover renal function in patients with CKD. Impact Statement Patients with chronic kidney disease (CKD) have limited treatment options, and renal transplantation is the only definitive treatment method that restores kidney function. However, challenges associated with transplantation, including donor organ shortage, rejection, and life-long immunosuppression, remain a problem. Recently, stem cell-based therapies have been proposed as an alternative approach to augment and restore renal function. In this study, we used human-derived amniotic fluid stem cells (AFSCs) to treat CKD in a rat model and demonstrated that AFSC treatment facilitated positive effects in terms of improvements of renal function.
Collapse
Affiliation(s)
- Sunil K George
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Mehran Abolbashari
- Paul L. Foster School of Medicine, Texas Tech University, El Paso, Texas
| | - Tae-Hyoung Kim
- Department of Urology, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| | - Chao Zhang
- Department of Urology, Shanghai Changhai Hospital, the Second Military Medical University, Shanghai, China
| | - Julie Allickson
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - John D Jackson
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Sang Jin Lee
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - In Kap Ko
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - James J Yoo
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
14
|
Yim HE, Kim DS, Chung HC, Shing B, Moon KH, George SK, Kim MW, Atala Z, Kim JH, Ko IK, Yoo JJ. Controlled Delivery of Stem Cell-Derived Trophic Factors Accelerates Kidney Repair After Renal Ischemia-Reperfusion Injury in Rats. Stem Cells Transl Med 2019; 8:959-970. [PMID: 31144785 PMCID: PMC6708069 DOI: 10.1002/sctm.18-0222] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 03/04/2019] [Indexed: 12/20/2022] Open
Abstract
Renal disease is a worldwide health issue. Besides transplantation, current therapies revolve around dialysis, which only delays disease progression but cannot replace other renal functions, such as synthesizing erythropoietin. To address these limitations, cell‐based approaches have been proposed to restore damaged kidneys as an alternative to current therapies. Recent studies have shown that stem cell‐derived secretomes can enhance tissue regeneration. However, many growth factors undergo rapid degradation when they are injected into the body in a soluble form. Efficient delivery and controlled release of secreting factors at the sites of injury would improve the efficacy in tissue regeneration. Herein, we developed a gel‐based delivery system for controlled delivery of trophic factors in the conditioned medium (CM) secreted from human placental stem cells (HPSCs) and evaluated the effect of trophic factors on renal regeneration. CM treatment significantly enhanced cell proliferation and survival in vitro. Platelet‐rich plasma (PRP) was used as a delivery vehicle for CM. Analysis of the release kinetics demonstrated that CM delivery through the PRP gel resulted in a controlled release of the factors both in vitro and in vivo. In an acute kidney injury model in rats, functional and structural analysis showed that CM delivery using the PRP gel system into the injured kidney minimized renal tissue damage, leading to a more rapid functional recovery when compared with saline, CM, or vehicle only injection groups. These results suggest that controlled delivery of HPSC‐derived trophic factors may provide efficient repair of renal tissue injury. stem cells translational medicine2019;8:959&970
Collapse
Affiliation(s)
- Hyung Eun Yim
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina, USA.,Department of Pediatrics, College of Medicine, Korea University, Seoul, Korea
| | - Doo Sang Kim
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina, USA.,Department of Urology, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| | - Hyun Chul Chung
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina, USA.,Department of Urology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Brian Shing
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina, USA
| | - Kyung Hyun Moon
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina, USA.,Department of Urology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Sunil K George
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina, USA
| | - Michael W Kim
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina, USA
| | - Zachary Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina, USA
| | - Ji Hyun Kim
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina, USA
| | - In Kap Ko
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina, USA
| | - James J Yoo
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina, USA
| |
Collapse
|
15
|
Ali M, PR AK, Yoo JJ, Zahran F, Atala A, Lee SJ. A Photo-Crosslinkable Kidney ECM-Derived Bioink Accelerates Renal Tissue Formation. Adv Healthc Mater 2019; 8:e1800992. [PMID: 30725520 DOI: 10.1002/adhm.201800992] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 01/20/2019] [Indexed: 11/10/2022]
Abstract
3D bioprinting strategies in tissue engineering aim to fabricate clinically applicable tissue constructs that can replace the damaged or diseased tissues and organs. One of the main prerequisites in 3D bioprinting is finding an appropriate bioink that provides a tissue-specific microenvironment supporting the cellular growth and maturation. In this respect, decellularized extracellular matrix (dECM)-derived hydrogels have been considered as bioinks for the cell-based bioprinting due to their capability to inherit the intrinsic cues from native ECM. Herein, a photo-crosslinkable kidney ECM-derived bioink (KdECMMA) is developed that could provide a kidney-specific microenvironment for renal tissue bioprinting. Porcine whole kidneys are decellularized through a perfusion method, dissolved in an acid solution, and chemically modified by methacrylation. A KdECMMA-based bioink is formulated and evaluated for rheological properties and printability for the printing process. The results show that the bioprinted human kidney cells in the KdECMMA bioink are highly viable and mature with time. Moreover, the bioprinted renal constructs exhibit the structural and functional characteristics of the native renal tissue. The potential of the tissue-specific ECM-derived bioink is demonstrated for cell-based bioprinting that could enhance the cellular maturation and eventually tissue formation.
Collapse
Affiliation(s)
- Mohamed Ali
- Wake Forest Institute for Regenerative MedicineWake Forest School of MedicineMedical Center Boulevard Winston‐Salem NC 27157 USA
- Department of ChemistryFaculty of ScienceZagazig University Zagazig Sharkia 44519 Egypt
| | - Anil Kumar PR
- Wake Forest Institute for Regenerative MedicineWake Forest School of MedicineMedical Center Boulevard Winston‐Salem NC 27157 USA
| | - James J. Yoo
- Wake Forest Institute for Regenerative MedicineWake Forest School of MedicineMedical Center Boulevard Winston‐Salem NC 27157 USA
- School of Biomedical Engineering and SciencesWake Forest University‐Virginia Tech Winston‐Salem NC 27157 USA
| | - Faten Zahran
- Department of ChemistryFaculty of ScienceZagazig University Zagazig Sharkia 44519 Egypt
| | - Anthony Atala
- Wake Forest Institute for Regenerative MedicineWake Forest School of MedicineMedical Center Boulevard Winston‐Salem NC 27157 USA
- School of Biomedical Engineering and SciencesWake Forest University‐Virginia Tech Winston‐Salem NC 27157 USA
| | - Sang Jin Lee
- Wake Forest Institute for Regenerative MedicineWake Forest School of MedicineMedical Center Boulevard Winston‐Salem NC 27157 USA
- School of Biomedical Engineering and SciencesWake Forest University‐Virginia Tech Winston‐Salem NC 27157 USA
| |
Collapse
|
16
|
Jin LW, Pan M, Ye HY, Zheng Y, Chen Y, Huang WW, Xu XY, Zheng SB. Down-regulation of the long non-coding RNA XIST ameliorates podocyte apoptosis in membranous nephropathy via the miR-217-TLR4 pathway. Exp Physiol 2018; 104:220-230. [PMID: 30414341 DOI: 10.1113/ep087190] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 11/08/2018] [Indexed: 12/18/2022]
Abstract
NEW FINDINGS What is the central question of this study? What is the role of the long non-coding RNA X-inactive specific transcript (XIST), which is up-regulated in injured podocytes and membranous nephropathy, in the pathogenesis of membranous nephropathy? What is the main finding and its importance? XIST was up-regulated in kidney tissue with membranous nephropathy and in injured podocytes. Down-regulation of XIST inhibited podocyte apoptosis. XIST negatively regulated miR-217, and miR-217 modulated Toll-like receptor 4. Inhibition of XIST suppressed podocyte apoptosis induced by angiotensin II via miR-217. ABSTRACT Membranous nephropathy is often characterized by glomerular podocyte injury. Up-regulation of the long non-coding RNA (lncRNA) X-inactive specific transcript (XIST) has been verified in membranous nephropathy and in injured podocytes. Here the role of XIST in podocyte injury and membranous nephropathy was explored. Quantitative real-time PCR and western blot were performed to detect the expression of XIST and miR-217, and Toll-like receptor 4 (TLR4) protein, respectively. Podocyte apoptosis was evaluated with flow cytometry. Interaction between XIST and miR-217 was analysed by RNA immunoprecipitation and RNA pull-down assay. A dual luciferase reporter assay was used to examine the interplay between miR-217 and TLR4. Up-regulation of the lncRNA XIST and angiotensin II (Ang II) and kidney and podocyte injury were indicated in kidney tissue of patients with membranous nephropathy. Increase of XIST and apoptosis were induced by Ang II in podocytes. Down-regulation of XIST reversed podocyte apoptosis induced by Ang II. MiR-217 was negatively regulated by XIST. MiR-217 controlled TLR4 by targeting its 3'-untranslated region. XIST modulated TLR4 through miR-217 and inhibition of XIST reduced podocyte apoptosis induced by Ang II via regulating miR-217. Down-regulation of XIST ameliorates podocyte apoptosis via the miR-217-TLR4 pathway, which may improve membranous nephropathy.
Collapse
Affiliation(s)
- Ling-Wei Jin
- Department of Nephrology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Min Pan
- Department of Nephrology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Han-Yang Ye
- Department of Nephrology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Yu Zheng
- Department of Nephrology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Yan Chen
- Department of Nephrology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Wen-Wen Huang
- Department of Nephrology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Xiao-Yan Xu
- Department of Nephrology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Shu-Bei Zheng
- Department of Nephrology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| |
Collapse
|
17
|
Burmeister DM, McIntyre MK, Montgomery RK, Gómez BI, Dubick MA. Isolation and Characterization of Multipotent CD24+ Cells From the Renal Papilla of Swine. Front Med (Lausanne) 2018; 5:250. [PMID: 30283781 PMCID: PMC6156461 DOI: 10.3389/fmed.2018.00250] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 08/20/2018] [Indexed: 12/24/2022] Open
Abstract
Over 100,000 patients in the United States are currently waiting for a kidney transplant. With just over 10,000 cadaveric kidneys transplanted annually, it is of the utmost importance to optimize kidney viability upon transplantation. One exciting avenue may be xenotransplantation, which has rejuvenated interest after advanced gene editing techniques have been successfully used in swine. Simultaneously, acute kidney injury (AKI) is associated with high morbidity and mortality and currently lacks effective treatment. Animal models have been used extensively to address both of these issues, with recent emphasis on renal progenitor cells (RPCs). Due to anatomical similarities to humans we aimed to examine progenitor cells from the renal papillae of swine kidneys. To do this, RPCs were dissected from the renal papillae of healthy swine. Cell surface marker expression, proliferation, and differentiation of the RPCs were tested in vitro. Additionally, a mixed lymphocyte reaction was performed to examine immunomodulatory properties. RPCs displayed spindle shaped morphology with limited self-renewing capacity. Isolated RPCs were positive for CD24 and CD133 at early passages, but lost expression with subsequent passaging. Similarly, RPCs displayed myogenic, osteogenic, and adipogenic differentiation capacities at passage 2, but largely lost this by passage 6. Lastly, direct contact of RPCs with human lymphocytes increased release of IL6 and IL8. Taken together, RPCs from the papilla of porcine kidneys display transient stem cell properties that are lost with passaging, and either represent multiple types of progenitor cells, or a multipotent progenitor population. In instances of ischemic insult, augmentation of/with RPCs may potentiate regenerative properties of the kidney. While the use of swine for transplantation and ischemia studies confers obvious advantages, the populations of different progenitor cell populations within pig kidneys warrants further investigation. Ultimately, while gene editing techniques enhance the potential for xenotransplantation of organs or cells, the ultimate success of this strategy may be determined by the (dis)similarities of RPCs from different species.
Collapse
Affiliation(s)
- David M Burmeister
- Damage Control Resuscitation, United States Army Institute of Surgical Research, San Antonio, TX, United States
| | - Matthew K McIntyre
- Damage Control Resuscitation, United States Army Institute of Surgical Research, San Antonio, TX, United States
| | - Robbie K Montgomery
- Damage Control Resuscitation, United States Army Institute of Surgical Research, San Antonio, TX, United States
| | - Belinda I Gómez
- Damage Control Resuscitation, United States Army Institute of Surgical Research, San Antonio, TX, United States
| | - Michael A Dubick
- Damage Control Resuscitation, United States Army Institute of Surgical Research, San Antonio, TX, United States
| |
Collapse
|
18
|
Ding W, Yousefi K, Shehadeh LA. Isolation, Characterization, And High Throughput Extracellular Flux Analysis of Mouse Primary Renal Tubular Epithelial Cells. J Vis Exp 2018. [PMID: 29985358 PMCID: PMC6101965 DOI: 10.3791/57718] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Mitochondrial dysfunction in the renal tubular epithelial cells (TECs) can lead to renal fibrosis, a major cause of chronic kidney disease (CKD). Therefore, assessing mitochondrial function in primary TECs may provide valuable insight into the bioenergetic status of the cells, providing insight into the pathophysiology of CKD. While there are a number of complex protocols available for the isolation and purification of proximal tubules in different species, the field lacks a cost-effective method optimized for tubular cell isolation without the need for purification. Here, we provide an isolation protocol that allows for studies focusing on both primary mouse proximal and distal renal TECs. In addition to cost-effective reagents and minimal animal procedures required in this protocol, the isolated cells maintain high energy levels after isolation and can be sub-cultured up to four passages, allowing for continuous studies. Furthermore, using a high throughput extracellular flux analyzer, we assess the mitochondrial respiration directly in the isolated TECs in a 96-well plate for which we provide recommendations for the optimization of cell density and compound concentration. These observations suggest that this protocol can be used for renal tubular ex vivo studies with a consistent, well-standardized production of renal TECs. This protocol may have broader future applications to study mitochondrial dysfunction associated with renal disorders for drug discovery or drug characterization purposes.
Collapse
Affiliation(s)
- Wen Ding
- Department of Molecular and Cellular Pharmacology, University of Miami Leonard M. Miller School of Medicine; Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine
| | - Keyvan Yousefi
- Department of Molecular and Cellular Pharmacology, University of Miami Leonard M. Miller School of Medicine; Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine
| | - Lina A Shehadeh
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine; Department of Medicine, Division of Cardiology, University of Miami Leonard M. Miller School of Medicine; Vascular Biology Institute, University of Miami Leonard M. Miller School of Medicine; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Leonard M. Miller School of Medicine;
| |
Collapse
|
19
|
Manzoli V, Colter DC, Dhanaraj S, Fornoni A, Ricordi C, Pileggi A, Tomei AA. Engineering human renal epithelial cells for transplantation in regenerative medicine. Med Eng Phys 2017; 48:3-13. [DOI: 10.1016/j.medengphy.2017.03.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 03/03/2017] [Accepted: 03/26/2017] [Indexed: 12/16/2022]
|
20
|
Huling J, Yoo JJ. Comparing adult renal stem cell identification, characterization and applications. J Biomed Sci 2017; 24:32. [PMID: 28511675 PMCID: PMC5434527 DOI: 10.1186/s12929-017-0339-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 05/03/2017] [Indexed: 12/27/2022] Open
Abstract
Despite growing interest and effort, a consensus has yet to be reached in regards to the identification of adult renal stem cells. Organ complexity and low turnover of renal cells has made stem cell identification difficult and lead to the investigation of multiple possible populations. In this review, we summarize the work that has been done toward finding and characterizing an adult renal stem cell population. In addition to giving a general overview of what has been done, we aim to highlight the variation in methods and outcomes. The methods used to locate potential stem cell populations can vary widely, but even within the relatively standard practice of BrdU labeling of slowly dividing cells, there are significant differences in protocols and results. Additional diversity exists in cell marker profiles and apparent differentiation potential seen in potential stem cell sources. Cataloging the variety of methods and outcomes seen so far may help to streamline future investigation and stear the field toward consensus. But even without firmly defined populations, the application of renal stem cells holds tantalizing potential. Populations of highly proliferative, multipotent cells of renal origin show the ability to engraft in injured kidneys, mitigate functional loss and occasionally show the ability to generate nephrons de novo. The progress toward regenerative medicine applications is also summarized.
Collapse
Affiliation(s)
- Jennifer Huling
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, 27157, USA.
| | - James J Yoo
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, 27157, USA
| |
Collapse
|