1
|
Khare V, Cherqui S. Targeted gene therapy for rare genetic kidney diseases. Kidney Int 2024; 106:1051-1061. [PMID: 39222842 DOI: 10.1016/j.kint.2024.07.034] [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: 02/27/2024] [Revised: 05/13/2024] [Accepted: 07/01/2024] [Indexed: 09/04/2024]
Abstract
Chronic kidney disease is one of the leading causes of mortality worldwide because of kidney failure and the associated challenges of its treatment including dialysis and kidney transplantation. About one-third of chronic kidney disease cases are linked to inherited monogenic factors, making them suitable for potential gene therapy interventions. However, the intricate anatomical structure of the kidney poses a challenge, limiting the effectiveness of targeted gene delivery to the renal system. In this review, we explore the progress made in the field of targeted gene therapy approaches and their implications for rare genetic kidney disorders, examining preclinical studies and prospects for clinical application. In vivo gene therapy is most commonly used for kidney-targeted gene delivery and involves administering viral and nonviral vectors through various routes such as systemic, renal vein, and renal arterial injections. Small nucleic acids have also been used in preclinical and clinical studies for treating certain kidney disorders. Unexpectedly, hematopoietic stem and progenitor cells have been used as an ex vivo gene therapy vehicle for kidney gene delivery, highlighting their ability to differentiate into macrophages within the kidney, forming tunneling nanotubes that can deliver genetic material and organelles to adjacent kidney cells, even across the basement membrane to target the proximal tubular cells. As gene therapy technologies continue to advance and our understanding of kidney biology deepens, there is hope for patients with genetic kidney disorders to eventually avoid kidney transplantation.
Collapse
Affiliation(s)
- Veenita Khare
- Department of Pediatrics, Division of Genetics, University of California, San Diego, La Jolla, California, USA
| | - Stephanie Cherqui
- Department of Pediatrics, Division of Genetics, University of California, San Diego, La Jolla, California, USA.
| |
Collapse
|
2
|
Doan TNA, Cowley JM, Phillips AL, Briffa JF, Leemaqz SY, Burton RA, Romano T, Wlodek ME, Bianco-Miotto T. Imprinted gene alterations in the kidneys of growth restricted offspring may be mediated by a long non-coding RNA. Epigenetics 2024; 19:2294516. [PMID: 38126131 PMCID: PMC10761017 DOI: 10.1080/15592294.2023.2294516] [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: 07/31/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023] Open
Abstract
Altered epigenetic mechanisms have been previously reported in growth restricted offspring whose mothers experienced environmental insults during pregnancy in both human and rodent studies. We previously reported changes in the expression of the DNA methyltransferase Dnmt3a and the imprinted genes Cdkn1c (Cyclin-dependent kinase inhibitor 1C) and Kcnq1 (Potassium voltage-gated channel subfamily Q member 1) in the kidney tissue of growth restricted rats whose mothers had uteroplacental insufficiency induced on day 18 of gestation, at both embryonic day 20 (E20) and postnatal day 1 (PN1). To determine the mechanisms responsible for changes in the expression of these imprinted genes, we investigated DNA methylation of KvDMR1, an imprinting control region (ICR) that includes the promoter of the antisense long non-coding RNA Kcnq1ot1 (Kcnq1 opposite strand/antisense transcript 1). Kcnq1ot1 expression decreased by 51% in growth restricted offspring compared to sham at PN1. Interestingly, there was a negative correlation between Kcnq1ot1 and Kcnq1 in the E20 growth restricted group (Spearman's ρ = 0.014). No correlation was observed between Kcnq1ot1 and Cdkn1c expression in either group at any time point. Additionally, there was a 11.25% decrease in the methylation level at one CpG site within KvDMR1 ICR. This study, together with others in the literature, supports that long non-coding RNAs may mediate changes seen in tissues of growth restricted offspring.
Collapse
Affiliation(s)
- Thu N. A. Doan
- School of Agriculture, Food and Wine, & Waite Research Institute, University of Adelaide, Adelaide, South Australia, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - James M. Cowley
- School of Agriculture, Food and Wine, & Waite Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Aaron L. Phillips
- School of Agriculture, Food and Wine, & Waite Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Jessica F. Briffa
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, Victoria, Australia
| | - Shalem Y. Leemaqz
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
- SAHMRI Women and Kids, South Australian Health & Medical Research Institute, Adelaide, South Australia, Australia
- College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | - Rachel A. Burton
- School of Agriculture, Food and Wine, & Waite Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Tania Romano
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Mary E. Wlodek
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, Victoria, Australia
- Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, Victoria, Australia
| | - Tina Bianco-Miotto
- School of Agriculture, Food and Wine, & Waite Research Institute, University of Adelaide, Adelaide, South Australia, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
3
|
Yuan F, Lerman LO. Targeted therapeutic strategies for the kidney. Expert Opin Ther Targets 2024; 28:979-989. [PMID: 39491501 PMCID: PMC11617265 DOI: 10.1080/14728222.2024.2421756] [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: 09/20/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
INTRODUCTION Kidney diseases impose a significant burden with high incidence and mortality rates. Current treatment options for kidney diseases are limited, necessitating urgent development of novel and effective therapeutic strategies to delay or reverse disease progression. Targeted therapies for the kidney hold promise in significantly enhancing treatment outcomes, offering hope to patients afflicted with renal disorders. AREAS COVERED This review summarized advances in kidney-targeted therapies including genes, peptides and proteins, cell-based, nanoparticles, and localized delivery routes. We also explored the potential clinical applications, prospects, and challenges of targeted therapies for renal disorders. EXPERT OPINION Advances in targeted therapies for renal conditions have enhanced therapeutic outcomes. Clinical application of kidney-targeted therapies is currently limited by renal structure and the scarcity of robust biomarkers. Bridging the gap from basic and pre-clinical research targeting the kidney to achieving clinical translation remains a formidable challenge.
Collapse
Affiliation(s)
- Fei Yuan
- Division of Nephrology and Hypertension, Mayo Clinic; Rochester, MN, USA
- Department of Urology, National Children’s Medical Center, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lilach O. Lerman
- Division of Nephrology and Hypertension, Mayo Clinic; Rochester, MN, USA
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
4
|
Porter CM, Qian GC, Grindel SH, Hughes AJ. Highly parallel production of designer organoids by mosaic patterning of progenitors. Cell Syst 2024; 15:649-661.e9. [PMID: 38981488 PMCID: PMC11257788 DOI: 10.1016/j.cels.2024.06.004] [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: 11/14/2023] [Revised: 04/09/2024] [Accepted: 06/17/2024] [Indexed: 07/11/2024]
Abstract
Organoids derived from human stem cells are a promising approach for disease modeling, regenerative medicine, and fundamental research. However, organoid variability and limited control over morphological outcomes remain as challenges. One open question is the extent to which engineering control over culture conditions can guide organoids to specific compositions. Here, we extend a DNA "velcro" cell patterning approach, precisely controlling the number and ratio of human induced pluripotent stem cell-derived progenitors contributing to nephron progenitor (NP) organoids and mosaic NP/ureteric bud (UB) tip cell organoids within arrays of microwells. We demonstrate long-term control over organoid size and morphology, decoupled from geometric constraints. We then show emergent trends in organoid tissue proportions that depend on initial progenitor cell composition. These include higher nephron and stromal cell representation in mosaic NP/UB organoids vs. NP-only organoids and a "goldilocks" initial cell ratio in mosaic organoids that optimizes the formation of proximal tubule structures.
Collapse
Affiliation(s)
- Catherine M Porter
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA; Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Soft and Living Matter, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Precision Engineering for Health (CPE4H), University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Grace C Qian
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA; Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Samuel H Grindel
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA; Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Soft and Living Matter, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Precision Engineering for Health (CPE4H), University of Pennsylvania, Philadelphia, PA 19104, USA; Materials Research Science and Engineering Center (MRSEC), University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alex J Hughes
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA; Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Soft and Living Matter, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Precision Engineering for Health (CPE4H), University of Pennsylvania, Philadelphia, PA 19104, USA; Materials Research Science and Engineering Center (MRSEC), University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
5
|
Sakolish C, Moyer HL, Tsai HHD, Ford LC, Dickey AN, Wright FA, Han G, Bajaj P, Baltazar MT, Carmichael PL, Stanko JP, Ferguson SS, Rusyn I. Analysis of reproducibility and robustness of a renal proximal tubule microphysiological system OrganoPlate 3-lane 40 for in vitro studies of drug transport and toxicity. Toxicol Sci 2023; 196:52-70. [PMID: 37555834 PMCID: PMC10613961 DOI: 10.1093/toxsci/kfad080] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2023] Open
Abstract
Microphysiological systems are an emerging area of in vitro drug development, and their independent evaluation is important for wide adoption and use. The primary goal of this study was to test reproducibility and robustness of a renal proximal tubule microphysiological system, OrganoPlate 3-lane 40, as an in vitro model for drug transport and toxicity studies. This microfluidic model was compared with static multiwell cultures and tested using several human renal proximal tubule epithelial cell (RPTEC) types. The model was characterized in terms of the functional transport for various tubule-specific proteins, epithelial permeability of small molecules (cisplatin, tenofovir, and perfluorooctanoic acid) versus large molecules (fluorescent dextrans, 60-150 kDa), and gene expression response to a nephrotoxic xenobiotic. The advantages offered by OrganoPlate 3-lane 40 as compared with multiwell cultures are the presence of media flow, albeit intermittent, and increased throughput compared with other microfluidic models. However, OrganoPlate 3-lane 40 model appeared to offer only limited (eg, MRP-mediated transport) advantages in terms of either gene expression or functional transport when compared with the multiwell plate culture conditions. Although OrganoPlate 3-lane 40 can be used to study cellular uptake and direct toxic effects of small molecules, it may have limited utility for drug transport studies. Overall, this study offers refined experimental protocols and comprehensive comparative data on the function of RPETCs in traditional multiwell culture and microfluidic OrganoPlate 3-lane 40, information that will be invaluable for the prospective end-users of in vitro models of the human proximal tubule.
Collapse
Affiliation(s)
- Courtney Sakolish
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas 77843, USA
| | - Haley L Moyer
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas 77843, USA
| | - Han-Hsuan D Tsai
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas 77843, USA
| | - Lucie C Ford
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas 77843, USA
| | - Allison N Dickey
- Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina 27695, USA
| | - Fred A Wright
- Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina 27695, USA
- Department of Statistics, North Carolina State University, Raleigh, North Carolina 27695, USA
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina 27695, USA
| | - Gang Han
- Department of Epidemiology and Biostatistics, Texas A&M University, College Station, Texas 77843, USA
| | - Piyush Bajaj
- Global Investigative Toxicology, Preclinical Safety, Sanofi, Cambridge, Massachusetts 02141, USA
| | - Maria T Baltazar
- Safety & Environmental Assurance Centre (SEAC), Unilever, Bedfordshire MK44 1LQ, UK
| | - Paul L Carmichael
- Safety & Environmental Assurance Centre (SEAC), Unilever, Bedfordshire MK44 1LQ, UK
| | - Jason P Stanko
- Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709, USA
| | - Stephen S Ferguson
- Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709, USA
| | - Ivan Rusyn
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas 77843, USA
| |
Collapse
|
6
|
Xu J, Zhou X, Zhang T, Zhang B, Xu PX. Smarca4 deficiency induces Pttg1 oncogene upregulation and hyperproliferation of tubular and interstitial cells during kidney development. Front Cell Dev Biol 2023; 11:1233317. [PMID: 37727504 PMCID: PMC10506413 DOI: 10.3389/fcell.2023.1233317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/17/2023] [Indexed: 09/21/2023] Open
Abstract
Kidney formation and nephrogenesis are controlled by precise spatiotemporal gene expression programs, which are coordinately regulated by cell-cycle, cell type-specific transcription factors and epigenetic/chromatin regulators. However, the roles of epigenetic/chromatin regulators in kidney development and disease remain poorly understood. In this study, we investigated the impact of deleting the chromatin remodeling factor Smarca4 (Brg1), a human Wilms tumor-associated gene, in Wnt4-expressing cells. Smarca4 deficiency led to severe tubular defects and a shortened medulla. Through unbiased single-cell RNA sequencing analyses, we identified multiple types of Wnt4 Cre-labeled interstitial cells, along with nephron-related cells. Smarca4 deficiency increased interstitial cells but markedly reduced tubular cells, resulting in cells with mixed identity and elevated expression of cell-cycle regulators and genes associated with extracellular matrix and epithelial-to-mesenchymal transition/fibrosis. We found that Smarca4 loss induced a significant upregulation of the oncogene Pttg1 and hyperproliferation of Wnt4 Cre-labeled cells. These changes in the cellular state could hinder the cellular transition into characteristic tubular structures, eventually leading to fibrosis. In conclusion, our findings shed light on novel cell types and genes associated with Wnt4 Cre-labeled cells and highlight the critical role of Smarca4 in regulating tubular cell differentiation and the expression of the cancer-causing gene Pttg1 in the kidney. These findings may provide valuable insights into potential therapeutic strategies for renal cell carcinoma resulting from SMARCA4 deficiency.
Collapse
Affiliation(s)
- Jinshu Xu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Xianxiao Zhou
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ting Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Pin-Xian Xu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
7
|
Syed Mohamed SMD, Welsh GI, Roy I. Renal tissue engineering for regenerative medicine using polymers and hydrogels. Biomater Sci 2023; 11:5706-5726. [PMID: 37401545 DOI: 10.1039/d3bm00255a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2023]
Abstract
Chronic Kidney Disease (CKD) is a growing worldwide problem, leading to end-stage renal disease (ESRD). Current treatments for ESRD include haemodialysis and kidney transplantation, but both are deemed inadequate since haemodialysis does not address all other kidney functions, and there is a shortage of suitable donor organs for transplantation. Research in kidney tissue engineering has been initiated to take a regenerative medicine approach as a potential treatment alternative, either to develop effective cell therapy for reconstruction or engineer a functioning bioartificial kidney. Currently, renal tissue engineering encompasses various materials, mainly polymers and hydrogels, which have been chosen to recreate the sophisticated kidney architecture. It is essential to address the chemical and mechanical aspects of the materials to ensure they can support cell development to restore functionality and feasibility. This paper reviews the types of polymers and hydrogels that have been used in kidney tissue engineering applications, both natural and synthetic, focusing on the processing and formulation used in creating bioactive substrates and how these biomaterials affect the cell biology of the kidney cells used.
Collapse
Affiliation(s)
| | - Gavin I Welsh
- Renal Bristol, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol BS1 3NY, UK
| | - Ipsita Roy
- Department of Materials Science and Engineering, Faculty of Engineering, University of Sheffield, Sheffield S37HQ, UK.
| |
Collapse
|
8
|
Peek JL, Wilson MH. Cell and gene therapy for kidney disease. Nat Rev Nephrol 2023:10.1038/s41581-023-00702-3. [PMID: 36973494 DOI: 10.1038/s41581-023-00702-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2023] [Indexed: 03/29/2023]
Abstract
Kidney disease is a leading cause of morbidity and mortality across the globe. Current interventions for kidney disease include dialysis and renal transplantation, which have limited efficacy or availability and are often associated with complications such as cardiovascular disease and immunosuppression. There is therefore a pressing need for novel therapies for kidney disease. Notably, as many as 30% of kidney disease cases are caused by monogenic disease and are thus potentially amenable to genetic medicine, such as cell and gene therapy. Systemic disease that affects the kidney, such as diabetes and hypertension, might also be targetable by cell and gene therapy. However, although there are now several approved gene and cell therapies for inherited diseases that affect other organs, none targets the kidney. Promising recent advances in cell and gene therapy have been made, including in the kidney research field, suggesting that this form of therapy might represent a potential solution for kidney disease in the future. In this Review, we describe the potential for cell and gene therapy in treating kidney disease, focusing on recent genetic studies, key advances and emerging technologies, and we describe several crucial considerations for renal genetic and cell therapies.
Collapse
Affiliation(s)
- Jennifer L Peek
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Matthew H Wilson
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA.
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Veterans Affairs, Tennessee Valley Health Services, Nashville, TN, USA.
| |
Collapse
|
9
|
Yeh H. Applications of Transcriptomics in the Research of Antibody-Mediated Rejection in Kidney Transplantation: Progress and Perspectives. Organogenesis 2022; 18:2131357. [PMID: 36259540 PMCID: PMC9586696 DOI: 10.1080/15476278.2022.2131357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
Antibody-mediated rejection (ABMR) is the major cause of chronic allograft dysfunction and loss in kidney transplantation. The immunological mechanisms of ABMR that have been featured in the latest studies indicate a highly complex interplay between various immune and nonimmune cell types. Clinical diagnostic standards have long been criticized for being arbitrary and the lack of accuracy. Transcriptomic approaches, including microarray and RNA sequencing of allograft biopsies, enable the identification of differential gene expression and the continuous improvement of diagnostics. Given that conventional bulk transcriptomic approaches only reflect the average gene expression but not the status at the single-cell level, thereby ignoring the heterogeneity of the transcriptome across individual cells, single-cell RNA sequencing is rising as a powerful tool to provide a high-resolution transcriptome map of immune cells, which allows the elucidation of the pathogenesis and may facilitate the development of novel strategies for clinical treatment of ABMR.
Collapse
Affiliation(s)
- Hsuan Yeh
- Division of Renal-Electrolyte, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA,CONTACT Hsuan Yeh S976 Scaife Hall 3550 Terrace Street Pittsburgh, PA 15261
| |
Collapse
|
10
|
Safi W, Marco A, Moya D, Prado P, Garreta E, Montserrat N. Assessing kidney development and disease using kidney organoids and CRISPR engineering. Front Cell Dev Biol 2022; 10:948395. [PMID: 36120564 PMCID: PMC9479189 DOI: 10.3389/fcell.2022.948395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/06/2022] [Indexed: 11/26/2022] Open
Abstract
The differentiation of human pluripotent stem cells (hPSCs) towards organoids is one of the biggest scientific advances in regenerative medicine. Kidney organoids have not only laid the groundwork for various organ-like tissue systems but also provided insights into kidney embryonic development. Thus, several protocols for the differentiation of renal progenitors or mature cell types have been established. Insights into the interplay of developmental pathways in nephrogenesis and determination of different cell fates have enabled the in vitro recapitulation of nephrogenesis. Here we first provide an overview of kidney morphogenesis and patterning in the mouse model in order to dissect signalling pathways that are key to define culture conditions sustaining renal differentiation from hPSCs. Secondly, we also highlight how genome editing approaches have provided insights on the specific role of different genes and molecular pathways during renal differentiation from hPSCs. Based on this knowledge we further review how CRISPR/Cas9 technology has enabled the recapitulation and correction of cellular phenotypes associated with human renal disease. Last, we also revise how the field has positively benefited from emerging technologies as single cell RNA sequencing and discuss current limitations on kidney organoid technology that will take advantage from bioengineering solutions to help standardizing the use of this model systems to study kidney development and disease.
Collapse
Affiliation(s)
- Wajima Safi
- Pluripotency for Organ Regeneration. Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Technology (BIST), Barcelona, Spain
- *Correspondence: Wajima Safi, ; Elena Garreta, ; Nuria Montserrat,
| | - Andrés Marco
- Pluripotency for Organ Regeneration. Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Technology (BIST), Barcelona, Spain
| | | | - Patricia Prado
- Pluripotency for Organ Regeneration. Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Technology (BIST), Barcelona, Spain
| | - Elena Garreta
- Pluripotency for Organ Regeneration. Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Technology (BIST), Barcelona, Spain
- *Correspondence: Wajima Safi, ; Elena Garreta, ; Nuria Montserrat,
| | - Nuria Montserrat
- Pluripotency for Organ Regeneration. Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Technology (BIST), Barcelona, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina, Madrid, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
- *Correspondence: Wajima Safi, ; Elena Garreta, ; Nuria Montserrat,
| |
Collapse
|
11
|
Karp S, Pollak MR, Subramanian B. Disease Modeling with Kidney Organoids. MICROMACHINES 2022; 13:1384. [PMID: 36144007 PMCID: PMC9506184 DOI: 10.3390/mi13091384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/16/2022] [Accepted: 08/23/2022] [Indexed: 06/16/2023]
Abstract
Kidney diseases often lack optimal treatments, causing millions of deaths each year. Thus, developing appropriate model systems to study human kidney disease is of utmost importance. Some of the most promising human kidney models are organoids or small organ-resembling tissue collectives, derived from human-induced pluripotent stem cells (hiPSCs). However, they are more akin to a first-trimester fetal kidney than an adult kidney. Therefore, new strategies are needed to advance their maturity. They have great potential for disease modeling and eventually auxiliary therapy if they can reach the maturity of an adult kidney. In this review, we will discuss the current state of kidney organoids in terms of their similarity to the human kidney and use as a disease modeling system thus far. We will then discuss potential pathways to advance the maturity of kidney organoids to match an adult kidney for more accurate human disease modeling.
Collapse
|
12
|
Feng X, Jin X, Zhou R, Jiang Q, Wang Y, Zhang X, Shang K, Zhang J, Yu C, Shou J. Deep learning approach identified a gene signature predictive of the severity of renal damage caused by chronic cadmium accumulation. JOURNAL OF HAZARDOUS MATERIALS 2022; 433:128795. [PMID: 35405588 DOI: 10.1016/j.jhazmat.2022.128795] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 03/07/2022] [Accepted: 03/23/2022] [Indexed: 06/14/2023]
Abstract
Epidemiology studies have indicated that environmental cadmium exposure, even at low levels, will result in chronic cadmium accumulation in the kidney with profound adverse consequences and that the diabetic population is more susceptible. However, the underlying mechanisms are yet not fully understood. In the present study, we applied an animal model to study chronic cadmium exposure-induced renal injury and performed whole transcriptome profiling studies. Repetitive CdCl2 exposure resulted in cadmium accumulation and remarkable renal injuries in the animals. The diabetic ob/ob mice manifested increased severity of renal injury compared with the wild type C57BL/6 J littermate controls. RNA-Seq data showed that cadmium treatment induced dramatic gene expression changes in a dose-dependent manner. Among the differentially expressed genes include the apoptosis hallmark genes which significantly demarcated the treatment effects. Pathway enrichment and network analyses revealed biological oxidation (mainly glucuronidation) as one of the major stress responses induced by cadmium treatment. We next implemented a deep learning algorithm in conjunction with cloud computing and discovered a gene signature that can predict the degree of renal injury induced by cadmium treatment. The present study provided, for the first time, a comprehensive mechanistic understanding of chronic cadmium-induced nephrotoxicity in normal and diabetic populations at the whole genome level.
Collapse
Affiliation(s)
- Xuefang Feng
- Department of Nephrology, Yangpu Hospital, School of Medicine, Tongji University, Shanghai 200090, China
| | - Xian Jin
- EnnovaBio Pharmaceuticals, Shanghai 201203, China; Ennovabio (ZheJiang) Pharmaceuticals, Shaoxing, Zhejiang 312366, China
| | - Rong Zhou
- Department of Nephrology, Yangpu Hospital, School of Medicine, Tongji University, Shanghai 200090, China
| | - Qian Jiang
- Department of Nephrology, Yangpu Hospital, School of Medicine, Tongji University, Shanghai 200090, China
| | - Yanan Wang
- Department of Nephrology, Yangpu Hospital, School of Medicine, Tongji University, Shanghai 200090, China
| | - Xing Zhang
- EnnovaBio Pharmaceuticals, Shanghai 201203, China
| | - Ke Shang
- EnnovaBio Pharmaceuticals, Shanghai 201203, China; Ennovabio (ZheJiang) Pharmaceuticals, Shaoxing, Zhejiang 312366, China
| | - Jianhua Zhang
- EnnovaBio Pharmaceuticals, Shanghai 201203, China; Ennovabio (ZheJiang) Pharmaceuticals, Shaoxing, Zhejiang 312366, China
| | - Chen Yu
- Department of Nephrology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China.
| | - Jianyong Shou
- EnnovaBio Pharmaceuticals, Shanghai 201203, China; Ennovabio (ZheJiang) Pharmaceuticals, Shaoxing, Zhejiang 312366, China.
| |
Collapse
|
13
|
Abstract
PURPOSE OF REVIEW The aim of this study was to summarize recent findings in kidney gene therapy while proposing cystinuria as a model kidney disease target for genome engineering therapeutics. RECENT FINDINGS Despite the advances of gene therapy for treating diseases of other organs, the kidney lags behind. Kidney-targeted gene delivery remains an obstacle to gene therapy of kidney disease. Nanoparticle and adeno-associated viral vector technologies offer emerging hope for kidney gene therapy. Cystinuria represents a model potential target for kidney gene therapy due to its known genetic and molecular basis, targetability, and capacity for phenotypic rescue. SUMMARY Although gene therapy for kidney disease remains a major challenge, new and evolving technologies may actualize treatment for cystinuria and other kidney diseases.
Collapse
Affiliation(s)
- Jennifer L. Peek
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, TN 37232
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232
| | - Matthew H. Wilson
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN 37232
- Department of Veterans Affairs, Tennessee Valley Health Services, Nashville, TN, 37212
| |
Collapse
|
14
|
Voggel J, Mohr J, Nüsken KD, Dötsch J, Nüsken E, Alejandre Alcazar MA. Translational insights into mechanisms and preventive strategies after renal injury in neonates. Semin Fetal Neonatal Med 2022; 27:101245. [PMID: 33994314 DOI: 10.1016/j.siny.2021.101245] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Adverse perinatal circumstances can cause acute kidney injury (AKI) and contribute to chronic kidney disease (CKD). Accumulating evidence indicate that a wide spectrum of perinatal conditions interferes with normal kidney development and ultimately leads to aberrant kidney structure and function later in life. The present review addresses the lack of mechanistic knowledge with regard to perinatal origins of CKD and provides a comprehensive overview of pre- and peri-natal insults, including genetic predisposition, suboptimal nutritional supply, obesity and maternal metabolic disorders as well as placental insufficiency leading to intrauterine growth restriction (IUGR), prematurity, infections, inflammatory processes, and the need for life-saving treatments (e.g. oxygen supplementation, mechanical ventilation, medications) in neonates. Finally, we discuss future preventive, therapeutic, and regenerative directions. In summary, this review highlights the perinatal vulnerability of the kidney and the early origins of increased susceptibility toward AKI and CKD during postnatal life. Promotion of kidney health and prevention of disease require the understanding of perinatal injury in order to optimize perinatal micro- and macro-environments and enable normal kidney development.
Collapse
Affiliation(s)
- Jenny Voggel
- University of Cologne, Faculty of Medicine, University Hospital Cologne, Department of Pediatric and Adolescent Medicine, Germany; University of Cologne, Faculty of Medicine, University Hospital Cologne, Center for Molecular Medicine Cologne (CMMC), Germany
| | - Jasmine Mohr
- University of Cologne, Faculty of Medicine, University Hospital Cologne, Translational Experimental Pediatrics - Experimental Pulmonology, Department of Pediatric and Adolescent Medicine, Germany; University of Cologne, Faculty of Medicine, University Hospital Cologne, Center for Molecular Medicine Cologne (CMMC), Germany
| | - Kai-Dietrich Nüsken
- University of Cologne, Faculty of Medicine, University Hospital Cologne, Department of Pediatric and Adolescent Medicine, Germany
| | - Jörg Dötsch
- University of Cologne, Faculty of Medicine, University Hospital Cologne, Department of Pediatric and Adolescent Medicine, Germany
| | - Eva Nüsken
- University of Cologne, Faculty of Medicine, University Hospital Cologne, Department of Pediatric and Adolescent Medicine, Germany
| | - Miguel A Alejandre Alcazar
- University of Cologne, Faculty of Medicine, University Hospital Cologne, Translational Experimental Pediatrics - Experimental Pulmonology, Department of Pediatric and Adolescent Medicine, Germany; University of Cologne, Faculty of Medicine, University Hospital Cologne, Center for Molecular Medicine Cologne (CMMC), Germany; Excellence Cluster on Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Faculty of Medicine, University Hospital Cologne Cologne, Germany; Institute for Lung Health, University of Giessen and Marburg Lung Centre (UGMLC), Member of the German Centre for Lung Research (DZL), Gießen, Germany.
| |
Collapse
|
15
|
Abstract
The kidney is a highly complex organ in the human body. Although creating an in vitro model of the human kidney is challenging, tremendous advances have been made in recent years. Kidney organoids are in vitro kidney models that are generated from stem cells in three-dimensional (3D) cultures. They exhibit remarkable degree of similarities with the native tissue in terms of cell type, morphology, and function. The establishment of 3D kidney organoids facilitates a mechanistic study of cell communications, and these organoids can be used for drug screening, disease modeling, and regenerative medicine applications. This review discusses the cellular complexity during in vitro kidney generation. We intend to highlight recent progress in kidney organoids and the applications of these relatively new technologies.
Collapse
|
16
|
Neumann EK, Patterson NH, Rivera ES, Allen JL, Brewer M, deCaestecker MP, Caprioli RM, Fogo AB, Spraggins JM. Highly multiplexed immunofluorescence of the human kidney using co-detection by indexing. Kidney Int 2022; 101:137-143. [PMID: 34619231 PMCID: PMC8741652 DOI: 10.1016/j.kint.2021.08.033] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 08/24/2021] [Accepted: 08/26/2021] [Indexed: 01/03/2023]
Abstract
The human kidney is composed of many cell types that vary in their abundance and distribution from normal to diseased organ. As these cell types perform unique and essential functions, it is important to confidently label each within a single tissue to accurately assess tissue architecture and microenvironments. Towards this goal, we demonstrate the use of co-detection by indexing (CODEX) multiplexed immunofluorescence for visualizing 23 antigens within the human kidney. Using CODEX, many of the major cell types and substructures, such as collecting ducts, glomeruli, and thick ascending limb, were visualized within a single tissue section. Of these antibodies, 19 were conjugated in-house, demonstrating the flexibility and utility of this approach for studying the human kidney using custom and commercially available antibodies. We performed a pilot study that compared both fresh frozen and formalin-fixed paraffin-embedded healthy non-neoplastic and diabetic nephropathy kidney tissues. The largest cellular differences between the two groups was observed in cells labeled with aquaporin 1, cytokeratin 7, and α-smooth muscle actin. Thus, our data show the power of CODEX multiplexed immunofluorescence for surveying the cellular diversity of the human kidney and the potential for applications within pathology, histology, and building anatomical atlases.
Collapse
Affiliation(s)
- Elizabeth K. Neumann
- Department of Biochemistry, Vanderbilt University, Nashville, TN, USA 37232,Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN, USA 37232
| | - Nathan Heath Patterson
- Department of Biochemistry, Vanderbilt University, Nashville, TN, USA 37232,Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN, USA 37232
| | - Emilio S. Rivera
- Department of Biochemistry, Vanderbilt University, Nashville, TN, USA 37232,Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN, USA 37232
| | - Jamie L. Allen
- Department of Biochemistry, Vanderbilt University, Nashville, TN, USA 37232,Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN, USA 37232
| | - Maya Brewer
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN USA 37232
| | - Mark P. deCaestecker
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN USA 37232
| | - Richard M. Caprioli
- Department of Biochemistry, Vanderbilt University, Nashville, TN, USA 37232,Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN, USA 37232.,Department of Chemistry, Vanderbilt University, Nashville, TN, USA 37232
| | - Agnes B. Fogo
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN USA 37232,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN USA 37232.,Departments of Medicine and Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA 37232
| | - Jeffrey M. Spraggins
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN, USA 37232.,Department of Chemistry, Vanderbilt University, Nashville, TN, USA 37232,Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA 37232
| |
Collapse
|
17
|
Chan K, Li X. Current Epigenetic Insights in Kidney Development. Genes (Basel) 2021; 12:genes12081281. [PMID: 34440455 PMCID: PMC8391601 DOI: 10.3390/genes12081281] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/07/2021] [Accepted: 08/19/2021] [Indexed: 12/31/2022] Open
Abstract
The kidney is among the best characterized developing tissues, with the genes and signaling pathways that regulate embryonic and adult kidney patterning and development having been extensively identified. It is now widely understood that DNA methylation and histone modification patterns are imprinted during embryonic development and must be maintained in adult cells for appropriate gene transcription and phenotypic stability. A compelling question then is how these epigenetic mechanisms play a role in kidney development. In this review, we describe the major genes and pathways that have been linked to epigenetic mechanisms in kidney development. We also discuss recent applications of single-cell RNA sequencing (scRNA-seq) techniques in the study of kidney development. Additionally, we summarize the techniques of single-cell epigenomics, which can potentially be used to characterize epigenomes at single-cell resolution in embryonic and adult kidneys. The combination of scRNA-seq and single-cell epigenomics will help facilitate the further understanding of early cell lineage specification at the level of epigenetic modifications in embryonic and adult kidney development, which may also be used to investigate epigenetic mechanisms in kidney diseases.
Collapse
Affiliation(s)
- Katrina Chan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA;
| | - Xiaogang Li
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
- Correspondence: ; Tel.: +1-507-266-0110
| |
Collapse
|
18
|
Schumacher A, Rookmaaker MB, Joles JA, Kramann R, Nguyen TQ, van Griensven M, LaPointe VLS. Defining the variety of cell types in developing and adult human kidneys by single-cell RNA sequencing. NPJ Regen Med 2021; 6:45. [PMID: 34381054 PMCID: PMC8357940 DOI: 10.1038/s41536-021-00156-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 07/22/2021] [Indexed: 01/14/2023] Open
Abstract
The kidney is among the most complex organs in terms of the variety of cell types. The cellular complexity of human kidneys is not fully unraveled and this challenge is further complicated by the existence of multiple progenitor pools and differentiation pathways. Researchers disagree on the variety of renal cell types due to a lack of research providing a comprehensive picture and the challenge to translate findings between species. To find an answer to the number of human renal cell types, we discuss research that used single-cell RNA sequencing on developing and adult human kidney tissue and compares these findings to the literature of the pre-single-cell RNA sequencing era. We find that these publications show major steps towards the discovery of novel cell types and intermediate cell stages as well as complex molecular signatures and lineage pathways throughout development. The variety of cell types remains variable in the single-cell literature, which is due to the limitations of the technique. Nevertheless, our analysis approaches an accumulated number of 41 identified cell populations of renal lineage and 32 of non-renal lineage in the adult kidney, and there is certainly much more to discover. There is still a need for a consensus on a variety of definitions and standards in single-cell RNA sequencing research, such as the definition of what is a cell type. Nevertheless, this early-stage research already proves to be of significant impact for both clinical and regenerative medicine, and shows potential to enhance the generation of sophisticated in vitro kidney tissue.
Collapse
Affiliation(s)
- A Schumacher
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Cell Biology-Inspired Tissue Engineering, Maastricht University, Maastricht, The Netherlands
| | - M B Rookmaaker
- Department of Nephrology, University Medical Center, Utrecht, The Netherlands
| | - J A Joles
- Department of Nephrology, University Medical Center, Utrecht, The Netherlands
| | - R Kramann
- Institute of Experimental Medicine and Systems Biology, RWTH Aachen, Aachen, Germany
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, The Netherlands
| | - T Q Nguyen
- Department of Pathology, University Medical Center, Utrecht, The Netherlands
| | - M van Griensven
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Cell Biology-Inspired Tissue Engineering, Maastricht University, Maastricht, The Netherlands
| | - V L S LaPointe
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Cell Biology-Inspired Tissue Engineering, Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
19
|
Kim SR, Puranik AS, Jiang K, Chen X, Zhu XY, Taylor I, Khodadadi-Jamayran A, Lerman A, Hickson LJ, Childs BG, Textor SC, Tchkonia T, Niewold TB, Kirkland JL, Lerman LO. Progressive Cellular Senescence Mediates Renal Dysfunction in Ischemic Nephropathy. J Am Soc Nephrol 2021; 32:1987-2004. [PMID: 34135081 PMCID: PMC8455278 DOI: 10.1681/asn.2020091373] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 03/29/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Peripheral vascular diseases may induce chronic ischemia and cellular injury distal to the arterial obstruction. Cellular senescence involves proliferation arrest in response to stress, which can damage neighboring cells. Renal artery stenosis (RAS) induces stenotic-kidney dysfunction and injury, but whether these arise from cellular senescenceand their temporal pattern remain unknown. METHODS Chronic renal ischemia was induced in transgenic INK-ATTAC and wild type C57BL/6 mice by unilateral RAS, and kidney function (in vivo micro-MRI) and tissue damage were assessed. Mouse healthy and stenotic kidneys were analyzed using unbiased single-cell RNA-sequencing. To demonstrate translational relevance, cellular senescence was studied in human stenotic kidneys. RESULTS Using intraperitoneal AP20187 injections starting 1, 2, or 4 weeks after RAS, selective clearance of cells highly expressing p16Ink4a attenuated cellular senescence and improved stenotic-kidney function; however, starting treatment immediately after RAS induction was unsuccessful. Broader clearance of senescent cells, using the oral senolytic combination dasatinib and quercetin, in C57BL/6 RAS mice was more effective in clearing cells positive for p21 (Cdkn1a) and alleviating renal dysfunction and damage. Unbiased, single-cell RNA sequencing in freshly dissociated cells from healthy and stenotic mouse kidneys identified stenotic-kidney epithelial cells undergoing both mesenchymal transition and senescence. As in mice, injured human stenotic kidneys exhibited cellular senescence, suggesting this process is conserved. CONCLUSIONS Maladaptive tubular cell senescence, involving upregulated p16 (Cdkn2a), p19 (Cdkn2d), and p21 (Cdkn1a) expression, is associated with renal dysfunction and injury in chronic ischemia. These findings support development of senolytic strategies to delay chronic ischemic renal injury.
Collapse
Affiliation(s)
- Seo Rin Kim
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota,Department of Nephrology and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Amrutesh S. Puranik
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota,Colton Center for Autoimmunity, Division of Rheumatology, New York University Langone Medical Center, New York, New York
| | - Kai Jiang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Xiaojun Chen
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota,Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xiang-Yang Zhu
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Ian Taylor
- FlowJo, BD Life Sciences, Ashland, Oregon
| | | | - Amir Lerman
- Department of Cardiology, Mayo Clinic, Rochester, Minnesota
| | - LaTonya J. Hickson
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Bennett G. Childs
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Stephen C. Textor
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Tamara Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - Timothy B. Niewold
- Colton Center for Autoimmunity, Division of Rheumatology, New York University Langone Medical Center, New York, New York
| | - James L. Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - Lilach O. Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
20
|
Chen WY, Evangelista EA, Yang J, Kelly EJ, Yeung CK. Kidney Organoid and Microphysiological Kidney Chip Models to Accelerate Drug Development and Reduce Animal Testing. Front Pharmacol 2021; 12:695920. [PMID: 34381363 PMCID: PMC8350564 DOI: 10.3389/fphar.2021.695920] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/13/2021] [Indexed: 01/17/2023] Open
Abstract
Kidneys are critical for the elimination of many drugs and metabolites via the urine, filtering waste and maintaining proper fluid and electrolyte balance. Emerging technologies incorporating engineered three-dimensional (3D) in vitro cell culture models, such as organoids and microphysiological systems (MPS) culture platforms, have been developed to replicate nephron function, leading to enhanced efficacy, safety, and toxicity evaluation of new drugs and environmental exposures. Organoids are tiny, self-organized three-dimensional tissue cultures derived from stem cells that can include dozens of cell types to replicate the complexity of an organ. In contrast, MPS are highly controlled fluidic culture systems consisting of isolated cell type(s) that can be used to deconvolute mechanism and pathophysiology. Both systems, having their own unique benefits and disadvantages, have exciting applications in the field of kidney disease modeling and therapeutic discovery and toxicology. In this review, we discuss current uses of both hPSC-derived organoids and MPS as pre-clinical models for studying kidney diseases and drug induced nephrotoxicity. Examples such as the use of organoids to model autosomal dominant polycystic kidney disease, and the use of MPS to predict renal clearance and nephrotoxic concentrations of novel drugs are briefly discussed. Taken together, these novel platforms allow investigators to elaborate critical scientific questions. While much work needs to be done, utility of these 3D cell culture technologies has an optimistic outlook and the potential to accelerate drug development while reducing the use of animal testing.
Collapse
Affiliation(s)
- Wei-Yang Chen
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, WA, United States
| | - Eric A Evangelista
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, WA, United States
| | - Jade Yang
- Department of Pharmaceutics, University of Washington School of Pharmacy, Seattle, WA, United States
| | - Edward J Kelly
- Department of Pharmaceutics, University of Washington School of Pharmacy, Seattle, WA, United States
- Kidney Research Institute, University of Washington School of Medicine, Seattle, WA, United States
| | - Catherine K Yeung
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, WA, United States
- Kidney Research Institute, University of Washington School of Medicine, Seattle, WA, United States
| |
Collapse
|
21
|
Fritsche E, Haarmann-Stemmann T, Kapr J, Galanjuk S, Hartmann J, Mertens PR, Kämpfer AAM, Schins RPF, Tigges J, Koch K. Stem Cells for Next Level Toxicity Testing in the 21st Century. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2006252. [PMID: 33354870 DOI: 10.1002/smll.202006252] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/13/2020] [Indexed: 06/12/2023]
Abstract
The call for a paradigm change in toxicology from the United States National Research Council in 2007 initiates awareness for the invention and use of human-relevant alternative methods for toxicological hazard assessment. Simple 2D in vitro systems may serve as first screening tools, however, recent developments infer the need for more complex, multicellular organotypic models, which are superior in mimicking the complexity of human organs. In this review article most critical organs for toxicity assessment, i.e., skin, brain, thyroid system, lung, heart, liver, kidney, and intestine are discussed with regards to their functions in health and disease. Embracing the manifold modes-of-action how xenobiotic compounds can interfere with physiological organ functions and cause toxicity, the need for translation of such multifaceted organ features into the dish seems obvious. Currently used in vitro methods for toxicological applications and ongoing developments not yet arrived in toxicity testing are discussed, especially highlighting the potential of models based on embryonic stem cells and induced pluripotent stem cells of human origin. Finally, the application of innovative technologies like organs-on-a-chip and genome editing point toward a toxicological paradigm change moves into action.
Collapse
Affiliation(s)
- Ellen Fritsche
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
- Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, 40225, Germany
| | | | - Julia Kapr
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Saskia Galanjuk
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Julia Hartmann
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Peter R Mertens
- Department of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke-University Magdeburg, Magdeburg, 39106, Germany
| | - Angela A M Kämpfer
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Roel P F Schins
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Julia Tigges
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Katharina Koch
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| |
Collapse
|
22
|
Zahmatkesh E, Khoshdel-Rad N, Mirzaei H, Shpichka A, Timashev P, Mahmoudi T, Vosough M. Evolution of organoid technology: Lessons learnt in Co-Culture systems from developmental biology. Dev Biol 2021; 475:37-53. [PMID: 33684433 DOI: 10.1016/j.ydbio.2021.03.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/25/2021] [Accepted: 03/01/2021] [Indexed: 02/07/2023]
Abstract
In recent years, the development of 3D organoids has opened new avenues of investigation into development, physiology, and regenerative medicine. Organoid formation and the process of organogenesis share common developmental pathways; thus, our knowledge of developmental biology can help model the complexity of different organs to refine organoids into a more sophisticated platform. The developmental process is strongly dependent on complex networks and communication of cell-cell and cell-matrix interactions among different cell populations and their microenvironment, during embryogenesis. These interactions affect cell behaviors such as proliferation, survival, migration, and differentiation. Co-culture systems within the organoid technology were recently developed and provided the highly physiologically relevant systems. Supportive cells including various types of endothelial and stromal cells provide the proper microenvironment, facilitate organoid assembly, and improve vascularization and maturation of organoids. This review discusses the role of the co-culture systems in organoid generation, with a focus on how knowledge of developmental biology has directed and continues to shape the development of more evolved 3D co-culture system-derived organoids.
Collapse
Affiliation(s)
- Ensieh Zahmatkesh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Regenrative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Niloofar Khoshdel-Rad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Regenrative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Anastasia Shpichka
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov University, Moscow, Russia.
| | - Peter Timashev
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov University, Moscow, Russia; Institute for Regenerative Medicine, Sechenov University, Moscow, Russia; Chemistry Department, Lomonosov Moscow State University, Moscow, Russia; Department of Polymers and Composites, N.N.Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Moscow, Russia.
| | - Tokameh Mahmoudi
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Massoud Vosough
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Regenrative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
23
|
Tissue Chips and Microphysiological Systems for Disease Modeling and Drug Testing. MICROMACHINES 2021; 12:mi12020139. [PMID: 33525451 PMCID: PMC7911320 DOI: 10.3390/mi12020139] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/23/2021] [Accepted: 01/26/2021] [Indexed: 12/15/2022]
Abstract
Tissue chips (TCs) and microphysiological systems (MPSs) that incorporate human cells are novel platforms to model disease and screen drugs and provide an alternative to traditional animal studies. This review highlights the basic definitions of TCs and MPSs, examines four major organs/tissues, identifies critical parameters for organization and function (tissue organization, blood flow, and physical stresses), reviews current microfluidic approaches to recreate tissues, and discusses current shortcomings and future directions for the development and application of these technologies. The organs emphasized are those involved in the metabolism or excretion of drugs (hepatic and renal systems) and organs sensitive to drug toxicity (cardiovascular system). This article examines the microfluidic/microfabrication approaches for each organ individually and identifies specific examples of TCs. This review will provide an excellent starting point for understanding, designing, and constructing novel TCs for possible integration within MPS.
Collapse
|
24
|
Yang S, Chen Z, Cheng Y, Liu T, Pu Y, Liang G. Environmental toxicology wars: Organ-on-a-chip for assessing the toxicity of environmental pollutants. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 268:115861. [PMID: 33120150 DOI: 10.1016/j.envpol.2020.115861] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 10/13/2020] [Accepted: 10/14/2020] [Indexed: 05/07/2023]
Abstract
Environmental pollution is a widespread problem, which has seriously threatened human health and led to an increase of human diseases. Therefore, it is critical to evaluate environmental pollutants quickly and efficiently. Because of obvious inter-species differences between animals and humans, and lack of physiologically-relevant microenvironment, animal models and in vitro two-dimensional (2D) models can not accurately describe toxicological effects and predicting actual in vivo responses. To make up the limitations of conventional environmental toxicology screening, organ-on-a-chip (OOC) systems are increasingly developing. OOC systems can provide a well-organized architecture with comparable to the complex microenvironment in vivo and generate realistic responses to environmental pollutants. The feasibility, adjustability and reliability of OCC systems make it possible to offer new opportunities for environmental pollutants screening, which can study their metabolism, collective response, and fate in vivo. Further progress can address the challenges to make OCC systems better investigate and evaluate environmental pollutants with high predictive power.
Collapse
Affiliation(s)
- Sheng Yang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, PR China, 210009.
| | - Zaozao Chen
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, PR China, 210096.
| | - Yanping Cheng
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, PR China, 210009.
| | - Tong Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, PR China, 210009.
| | - Yuepu Pu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, PR China, 210009.
| | - Geyu Liang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, PR China, 210009.
| |
Collapse
|
25
|
The Renal Extracellular Matrix as a Supportive Scaffold for Kidney Tissue Engineering: Progress and Future Considerations. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1345:103-118. [PMID: 34582017 DOI: 10.1007/978-3-030-82735-9_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
During the past decades, diverse methods have been used toward renal tissue engineering in order to replace renal function. The goals of all these techniques included the recapitulation of renal filtration, re-absorptive, and secretary functions, and replacement of endocrine/metabolic activities. It is also imperative to develop a reliable, up scalable, and timely manufacturing process. Decellularization of the kidney with intact ECM is crucial for in-vivo compatibility and targeted clinical application. Contemporarily there is an increasing interest and research in the field of regenerative medicine including stem cell therapy and tissue bioengineering in search for new and reproducible sources of kidneys. In this chapter, we sought to determine the most effective method of renal decellularization and recellularization with emphasis on biologic composition and support of stem cell growth. Current barriers and limitations of bioengineered strategies will be also discussed, and strategies to overcome these are suggested.
Collapse
|
26
|
Lin J, Susztak K. Complexities of Understanding Function from CKD-Associated DNA Variants. Clin J Am Soc Nephrol 2020; 15:1028-1040. [PMID: 32513823 PMCID: PMC7341770 DOI: 10.2215/cjn.15771219] [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] [Indexed: 11/23/2022]
Abstract
Genome-wide association studies (GWASs) have facilitated the unbiased discovery of hundreds of genomic loci associated with CKD and kidney function. The vast majority of disease-associated DNA variants are noncoding. Those that are causal in CKD pathogenesis likely modulate transcription of target genes in a cell type-specific manner. To gain novel biological insights into mechanisms driving the development of CKD, the causal variants (which are usually not the most significant variant reported in a GWAS), their target genes, and causal cell types need to be identified. This functional validation requires a large number of new data sets, complex bioinformatics analyses, and experimental cellular and in vivo studies. Here, we review the basic principles and some of the current approaches being leveraged to assign functional significance to a genotype-phenotype association.
Collapse
Affiliation(s)
- Jennie Lin
- Division of Nephrology and Hypertension, Feinberg Cardiovascular and Renal Research Institute, Department of Medicine, Northwestern University, Chicago, Illinois
- Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| | - Katalin Susztak
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Genetics, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
27
|
Xie R, Korolj A, Liu C, Song X, Lu RXZ, Zhang B, Ramachandran A, Liang Q, Radisic M. h-FIBER: Microfluidic Topographical Hollow Fiber for Studies of Glomerular Filtration Barrier. ACS CENTRAL SCIENCE 2020; 6:903-912. [PMID: 32607437 PMCID: PMC7318083 DOI: 10.1021/acscentsci.9b01097] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Indexed: 05/07/2023]
Abstract
Kidney-on-a-chip devices may revolutionize the discovery of new therapies. However, fabricating a 3D glomerulus remains a challenge, due to a requirement for a microscale soft material with complex topography to support cell culture in a native configuration. Here, we describe the use of microfluidic spinning to recapitulate complex concave and convex topographies over multiple length scales, required for biofabrication of a biomimetic 3D glomerulus. We produced a microfluidic extruded topographic hollow fiber (h-FIBER), consisting of a vessel-like perfusable tubular channel for endothelial cell cultivation, and a glomerulus-like knot with microconvex topography on its surface for podocyte cultivation. Meter long h-FIBERs were produced in microfluidics within minutes, followed by chemically induced inflation for generation of topographical cues on the 3D scaffold surface. The h-FIBERs were assembled into a hot-embossed plastic 96-well plate. Long-term perfusion, podocyte barrier formation, endothelialization, and permeability tests were easily performed by a standard pipetting technique on the platform. Following long-term culture (1 month), a functional filtration barrier, measured by the transfer of albumin from the blood vessel side to the ultrafiltrate side, suggested the establishment of an engineered glomerulus.
Collapse
Affiliation(s)
- Ruoxiao Xie
- MOE
Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology,
Beijing Key Lab of Microanalytical Methods & Instrumentation,
Department of Chemistry, Centre for Synthetic and Systems Biology, Tsinghua University, Beijing 100084, P. R. China
- Institute
for Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario M5S 3G9, Canada
| | - Anastasia Korolj
- Institute
for Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario M5S 3G9, Canada
- Department
of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5T 3A1, Canada
| | - Chuan Liu
- Institute
for Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario M5S 3G9, Canada
| | - Xin Song
- Department
of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5T 3A1, Canada
| | - Rick Xing Ze Lu
- Institute
for Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario M5S 3G9, Canada
| | - Boyang Zhang
- Institute
for Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario M5S 3G9, Canada
| | - Arun Ramachandran
- Department
of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5T 3A1, Canada
| | - Qionglin Liang
- MOE
Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology,
Beijing Key Lab of Microanalytical Methods & Instrumentation,
Department of Chemistry, Centre for Synthetic and Systems Biology, Tsinghua University, Beijing 100084, P. R. China
| | - Milica Radisic
- Institute
for Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario M5S 3G9, Canada
- Department
of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5T 3A1, Canada
| |
Collapse
|
28
|
Li K, Chen Y, Zhang J, Guan Y, Sun C, Li X, Xie X, Zhang D, Yu X, Liu T, Zhang X, Kong F, Zhao S. Microenvironment derived from metanephros transplantation inhibits the progression of acute kidney injury in glycerol-induced rat models. Ren Fail 2020; 42:89-97. [PMID: 31900008 PMCID: PMC6968583 DOI: 10.1080/0886022x.2019.1708393] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Background Embryonic metanephros is the mammalian renal anlagen, which is considered as a potential source for the regeneration of functional whole kidneys. Some studies reported that metanephros implanted into unilateral nephrectomized animals can develop into kidney tissue. However, kidneys are nephrotoxic in renal failure patients, and whether metanephros can grow in nephrotoxic has not been reported. This study aims to investigate the growth of metanephros in acute nephrotoxic environment and analyze the therapeutic effect of metanephros microenvironment on acute kidney injury (AKI). Methods AKI was induced in 200 g Wistar rats by giving intramuscular injections of 50% glycerol (10 mL/kg) in their hind limbs. 45 rats were divided randomly into three groups (control, glycerin, and metanephros). Metanephros group was transplanted two metanephroi (embryonic day 15) into the renal capsule of AKI rats. Glycerin group was AKI rats without transplantation. Control group was untreated. Results Mature glomeruli and tubules were detected in the grafts in metanephros group, which means that metanephroi can grow into tissues with mature kidney structure under acute nephrotoxic. Then, we assessed the renal function of host rats and found that there were fewer tubular necrosis in metanephros group than glycerin group, and the serum creatinine and urea nitrogen were significantly lower in metanephros group than glycerin group. Conclusion These results suggested that embryonic metanephroi can grow into tissues with mature kidney structure under acute nephrotoxic, and the graft microenvironment was effective in inhibiting the progression of AKI, which provides a new approach for the treatment of acute renal injury.
Collapse
Affiliation(s)
- Kailin Li
- Department of Central Research Lab, The Second Hospital of Shandong University, Jinan, China.,Key Laboratory for Kidney Regeneration of Shandong Province, Jinan, China
| | - Yuan Chen
- Department of Central Research Lab, The Second Hospital of Shandong University, Jinan, China
| | - Jianye Zhang
- Department of Urology, The Second Hospital of Shandong University, Jinan, China
| | - Yong Guan
- Key Laboratory for Kidney Regeneration of Shandong Province, Jinan, China.,Shandong Provincial Hospital, Jinan, China
| | - Chao Sun
- Department of Central Research Lab, The Second Hospital of Shandong University, Jinan, China.,Key Laboratory for Kidney Regeneration of Shandong Province, Jinan, China
| | - Xian Li
- The Second Hospital of Shandong University, Jinan, China
| | - Xiaoshuai Xie
- The Second Hospital of Shandong University, Jinan, China
| | - Denglu Zhang
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xin Yu
- Department of Urology, The Second Hospital of Shandong University, Jinan, China
| | - Tongyan Liu
- The Second Hospital of Shandong University, Jinan, China
| | - Xufeng Zhang
- The Second Hospital of Shandong University, Jinan, China
| | - Feng Kong
- Department of Central Research Lab, The Second Hospital of Shandong University, Jinan, China.,Key Laboratory for Kidney Regeneration of Shandong Province, Jinan, China.,Shandong Provincial Hospital, Jinan, China.,Karolinska Institutet Collaborative Laboratory for Stem Cell Ressearch, Shandong University, Jinan, China
| | - Shengtian Zhao
- Key Laboratory for Kidney Regeneration of Shandong Province, Jinan, China.,Shandong Provincial Hospital, Jinan, China.,Karolinska Institutet Collaborative Laboratory for Stem Cell Ressearch, Shandong University, Jinan, China
| |
Collapse
|
29
|
Khoshdel Rad N, Aghdami N, Moghadasali R. Cellular and Molecular Mechanisms of Kidney Development: From the Embryo to the Kidney Organoid. Front Cell Dev Biol 2020; 8:183. [PMID: 32266264 PMCID: PMC7105577 DOI: 10.3389/fcell.2020.00183] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/04/2020] [Indexed: 12/27/2022] Open
Abstract
Development of the metanephric kidney is strongly dependent on complex signaling pathways and cell-cell communication between at least four major progenitor cell populations (ureteric bud, nephron, stromal, and endothelial progenitors) in the nephrogenic zone. In recent years, the improvement of human-PSC-derived kidney organoids has opened new avenues of research on kidney development, physiology, and diseases. Moreover, the kidney organoids provide a three-dimensional (3D) in vitro model for the study of cell-cell and cell-matrix interactions in the developing kidney. In vitro re-creation of a higher-order and vascularized kidney with all of its complexity is a challenging issue; however, some progress has been made in the past decade. This review focuses on major signaling pathways and transcription factors that have been identified which coordinate cell fate determination required for kidney development. We discuss how an extensive knowledge of these complex biological mechanisms translated into the dish, thus allowed the establishment of 3D human-PSC-derived kidney organoids.
Collapse
Affiliation(s)
- Niloofar Khoshdel Rad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Nasser Aghdami
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - 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
|
30
|
|
31
|
Lee SJ, Wang HJ, Kim TH, Choi JS, Kulkarni G, Jackson JD, Atala A, Yoo JJ. In Situ Tissue Regeneration of Renal Tissue Induced by Collagen Hydrogel Injection. Stem Cells Transl Med 2019; 7:241-250. [PMID: 29380564 PMCID: PMC5788870 DOI: 10.1002/sctm.16-0361] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 11/17/2017] [Indexed: 12/12/2022] Open
Abstract
Host stem/progenitor cells can be mobilized and recruited to a target location using biomaterials, and these cells may be used for in situ tissue regeneration. The objective of this study was to investigate whether host biologic resources could be used to regenerate renal tissue in situ. Collagen hydrogel was injected into the kidneys of normal mice, and rat kidneys that had sustained ischemia/reperfusion injury. After injection, the kidneys of both animal models were examined up to 4 weeks for host tissue response. The infiltrating host cells present within the injection regions expressed renal stem/progenitor cell markers, PAX‐2, CD24, and CD133, as well as mesenchymal stem cell marker, CD44. The regenerated renal structures were identified by immunohistochemistry for renal cell specific markers, including synaptopodin and CD31 for glomeruli and cytokeratin and neprilysin for tubules. Quantitatively, the number of glomeruli found in the injected regions was significantly higher when compared to normal regions of renal cortex. This phenomenon occurred in normal and ischemic injured kidneys. Furthermore, the renal function after ischemia/reperfusion injury was recovered after collagen hydrogel injection. These results demonstrate that introduction of biomaterials into the kidney is able to facilitate the regeneration of glomerular and tubular structures in normal and injured kidneys. Such an approach has the potential to become a simple and effective treatment for patients with renal failure. Stem Cells Translational Medicine2018;7:241–250
Collapse
Affiliation(s)
- Sang Jin Lee
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Hung-Jen Wang
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA.,Department of Urology, Chang Gung Memorial Hospital, Kaohsiung Medical Center, Chang Gung University Collagen of Medicine, Kaohsiung City, Taiwan, Republic of China
| | - Tae-Hyoung Kim
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA.,Department of Urology, Chung-Ang University Hospital, Seoul, South Korea
| | - Jin San Choi
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Gauri Kulkarni
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - John D Jackson
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - James J Yoo
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
32
|
O'Hara RE, Arsenault MG, Esparza Gonzalez BP, Patriquen A, Hartwig S. Three Optimized Methods for In Situ Quantification of Progenitor Cell Proliferation in Embryonic Kidneys Using BrdU, EdU, and PCNA. Can J Kidney Health Dis 2019; 6:2054358119871936. [PMID: 31523438 PMCID: PMC6734617 DOI: 10.1177/2054358119871936] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 07/02/2019] [Indexed: 01/02/2023] Open
Abstract
Background: Nephron progenitor cells derived from the metanephric mesenchyme undergo a complex balance of self-renewal and differentiation throughout kidney development to give rise to the mature nephron. Cell proliferation is an important index of progenitor population dynamics. However, accurate and reproducible in situ quantification of cell proliferation within progenitor populations can be technically difficult to achieve due to the complexity and harsh tissue treatment required of certain protocols. Objective: To optimize and compare the performance of the 3 most accurate S phase–specific labeling methods used for in situ detection and quantification of nephron progenitor and ureteric bud cell proliferation in the developing kidney, namely, 5-bromo-2’-deoxyuridine (BrdU), 5-ethynyl-2’-deoxyuridine (EdU), and proliferating cell nuclear antigen (PCNA). Methods: Protocols for BrdU, EdU, and PCNA were optimized for fluorescence labeling on paraformaldehyde-fixed, paraffin-embedded mouse kidney tissue sections, with co-labeling of nephron progenitor cells and ureteric bud with Six2 and E-cadherin antibodies, respectively. Image processing and analysis, including quantification of proliferating cells, were carried out using free ImageJ software. Results: All 3 methods detect similar ratios of nephron progenitor and ureteric bud proliferating cells. The BrdU staining protocol is the lengthiest and most complex protocol to perform, requires tissue denaturation, and is most subject to interexperimental signal variability. In contrast, bound PCNA and EdU protocols are relatively more straightforward, consistently yield clear results, and far more easily lend themselves to co-staining; however, the bound PCNA protocol requires substantive additional postexperimental analysis to distinguish the punctate nuclear PCNA staining pattern characteristic of proliferating cells. Conclusions: All 3 markers exhibit distinct advantages and disadvantages in quantifying cell proliferation in kidney progenitor populations, with EdU and PCNA protocols being favored due to greater technical ease and reproducibility of results associated with these methods.
Collapse
Affiliation(s)
- Rosalie E O'Hara
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, Canada
| | - Michel G Arsenault
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, Canada
| | - Blanca P Esparza Gonzalez
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, Canada
| | - Ashley Patriquen
- Diagnostic Services, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, Canada
| | - Sunny Hartwig
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, Canada
| |
Collapse
|
33
|
Vanslambrouck JM, Wilson SB, Tan KS, Soo JYC, Scurr M, Spijker HS, Starks LT, Neilson A, Cui X, Jain S, Little MH, Howden SE. A Toolbox to Characterize Human Induced Pluripotent Stem Cell-Derived Kidney Cell Types and Organoids. J Am Soc Nephrol 2019; 30:1811-1823. [PMID: 31492807 DOI: 10.1681/asn.2019030303] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 07/25/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The generation of reporter lines for cell identity, lineage, and physiologic state has provided a powerful tool in advancing the dissection of mouse kidney morphogenesis at a molecular level. Although use of this approach is not an option for studying human development in vivo, its application in human induced pluripotent stem cells (iPSCs) is now feasible. METHODS We used CRISPR/Cas9 gene editing to generate ten fluorescence reporter iPSC lines designed to identify nephron progenitors, podocytes, proximal and distal nephron, and ureteric epithelium. Directed differentiation to kidney organoids was performed according to published protocols. Using immunofluorescence and live confocal microscopy, flow cytometry, and cell sorting techniques, we investigated organoid patterning and reporter expression characteristics. RESULTS Each iPSC reporter line formed well patterned kidney organoids. All reporter lines showed congruence of endogenous gene and protein expression, enabling isolation and characterization of kidney cell types of interest. We also demonstrated successful application of reporter lines for time-lapse imaging and mouse transplantation experiments. CONCLUSIONS We generated, validated, and applied a suite of fluorescence iPSC reporter lines for the study of morphogenesis within human kidney organoids. This fluorescent iPSC reporter toolbox enables the visualization and isolation of key populations in forming kidney organoids, facilitating a range of applications, including cellular isolation, time-lapse imaging, protocol optimization, and lineage-tracing approaches. These tools offer promise for enhancing our understanding of this model system and its correspondence with human kidney morphogenesis.
Collapse
Affiliation(s)
| | - Sean B Wilson
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Ker Sin Tan
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Joanne Y-C Soo
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Michelle Scurr
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - H Siebe Spijker
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | - Lakshi T Starks
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Amber Neilson
- Department of Genetics, Genome Engineering and iPSC Center and
| | - Xiaoxia Cui
- Department of Genetics, Genome Engineering and iPSC Center and
| | - Sanjay Jain
- Department of Medicine, Kidney Translational Research Center, Washington University School of Medicine, St. Louis, Missouri; and
| | - Melissa Helen Little
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia; .,Faculty of Medicine, Dentistry and Health Sciences, Department of Paediatrics and.,Department of Anatomy and Neuroscience, University of Melbourne, Victoria, Australia
| | - Sara E Howden
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Faculty of Medicine, Dentistry and Health Sciences, Department of Paediatrics and
| |
Collapse
|
34
|
Miyazaki T, Gharib SA, Hsu YWA, Xu K, Khodakivskyi P, Kobayashi A, Paragas J, Klose AD, Francis KP, Dubikovskaya E, Page-McCaw PS, Barasch J, Paragas N. Cell-specific image-guided transcriptomics identifies complex injuries caused by ischemic acute kidney injury in mice. Commun Biol 2019; 2:326. [PMID: 31508501 PMCID: PMC6718519 DOI: 10.1038/s42003-019-0571-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 08/09/2019] [Indexed: 01/20/2023] Open
Abstract
The kidney's inherent complexity has made identifying cell-specific pathways challenging, particularly when temporally associating them with the dynamic pathophysiology of acute kidney injury (AKI). Here, we combine renal cell-specific luciferase reporter mice using a chemoselective luciferin to guide the acquisition of cell-specific transcriptional changes in C57BL/6 background mice. Hydrogen peroxide generation, a common mechanism of tissue damage, was tracked using a peroxy-caged-luciferin to identify optimum time points for immunoprecipitation of labeled ribosomes for RNA-sequencing. Together, these tools revealed a profound impact of AKI on mitochondrial pathways in the collecting duct. In fact, targeting the mitochondria with an antioxidant, ameliorated not only hydrogen peroxide generation, but also significantly reduced oxidative stress and the expression of the AKI biomarker, LCN2. This integrative approach of coupling physiological imaging with transcriptomics and drug testing revealed how the collecting duct responds to AKI and opens new venues for cell-specific predictive monitoring and treatment.
Collapse
Affiliation(s)
- Tomoaki Miyazaki
- 1Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA 98195 USA
- 2Division of Nephrology, Department of Medicine, Showa University, Yokohama, Japan
| | - Sina A Gharib
- 3Computational Medicine Core, Center for Lung Biology, University of Washington, Seattle, WA 98195 USA
| | - Yun-Wei A Hsu
- 1Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA 98195 USA
| | - Katherine Xu
- 4Renal Division, Department of Medicine, Columbia University, New York, NY 10027 USA
| | - Pavlo Khodakivskyi
- 5Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology of Lausanne (EPFL), Lausanne, Switzerland
| | - Akio Kobayashi
- 1Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA 98195 USA
| | | | | | | | - Elena Dubikovskaya
- 5Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology of Lausanne (EPFL), Lausanne, Switzerland
| | - Patrick S Page-McCaw
- 9Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232 USA
| | - Jonathan Barasch
- 4Renal Division, Department of Medicine, Columbia University, New York, NY 10027 USA
| | - Neal Paragas
- 1Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA 98195 USA
| |
Collapse
|
35
|
Brovold M, Almeida JI, Pla-Palacín I, Sainz-Arnal P, Sánchez-Romero N, Rivas JJ, Almeida H, Dachary PR, Serrano-Aulló T, Soker S, Baptista PM. Naturally-Derived Biomaterials for Tissue Engineering Applications. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1077:421-449. [PMID: 30357702 PMCID: PMC7526297 DOI: 10.1007/978-981-13-0947-2_23] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Naturally-derived biomaterials have been used for decades in multiple regenerative medicine applications. From the simplest cell microcarriers made of collagen or alginate, to highly complex decellularized whole-organ scaffolds, these biomaterials represent a class of substances that is usually first in choice at the time of electing a functional and useful biomaterial. Hence, in this chapter we describe the several naturally-derived biomaterials used in tissue engineering applications and their classification, based on composition. We will also describe some of the present uses of the generated tissues like drug discovery, developmental biology, bioprinting and transplantation.
Collapse
Affiliation(s)
- Matthew Brovold
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA
| | - Joana I Almeida
- Health Research Institute of Aragón (IIS Aragón), Zaragoza, Spain
| | - Iris Pla-Palacín
- Health Research Institute of Aragón (IIS Aragón), Zaragoza, Spain
| | - Pilar Sainz-Arnal
- Health Research Institute of Aragón (IIS Aragón), Zaragoza, Spain
- Aragon Health Sciences Institute (IACS), Zaragoza, Spain
| | | | - Jesus J Rivas
- Health Research Institute of Aragón (IIS Aragón), Zaragoza, Spain
| | - Helen Almeida
- Health Research Institute of Aragón (IIS Aragón), Zaragoza, Spain
| | - Pablo Royo Dachary
- Instituto de Investigación Sanitária de Aragón (IIS Aragón), Zaragoza, Spain
- Liver Transplant Unit, Gastroenterology Department, Lozano Blesa University Hospital, Zaragoza, Spain
| | - Trinidad Serrano-Aulló
- Instituto de Investigación Sanitária de Aragón (IIS Aragón), Zaragoza, Spain
- Liver Transplant Unit, Gastroenterology Department, Lozano Blesa University Hospital, Zaragoza, Spain
| | - Shay Soker
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA.
| | - Pedro M Baptista
- Instituto de Investigación Sanitária de Aragón (IIS Aragón), Zaragoza, Spain.
- Center for Biomedical Research Network Liver and Digestive Diseases (CIBERehd), Zaragoza, Spain.
- Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Madrid, Spain.
- Biomedical and Aerospace Engineering Department, Universidad Carlos III de Madrid, Madrid, Spain.
- Fundación ARAID, Zaragoza, Spain.
| |
Collapse
|
36
|
Ciampi O, Bonandrini B, Derosas M, Conti S, Rizzo P, Benedetti V, Figliuzzi M, Remuzzi A, Benigni A, Remuzzi G, Tomasoni S. Engineering the vasculature of decellularized rat kidney scaffolds using human induced pluripotent stem cell-derived endothelial cells. Sci Rep 2019; 9:8001. [PMID: 31142801 PMCID: PMC6541625 DOI: 10.1038/s41598-019-44393-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 05/15/2019] [Indexed: 01/18/2023] Open
Abstract
Generating new kidneys using tissue engineering technologies is an innovative strategy for overcoming the shortage of donor organs for transplantation. Here we report how to efficiently engineer the kidney vasculature of decellularized rat kidney scaffolds by using human induced pluripotent stem cell (hiPSCs)-derived endothelial cells (hiPSC-ECs). In vitro, hiPSC-ECs responded to flow stress by acquiring an alignment orientation, and attached to and proliferated on the acellular kidney sections, maintaining their phenotype. The hiPSC-ECs were able to self-organize into chimeric kidney organoids to form vessel-like structures. Ex vivo infusion of hiPSC-ECs through the renal artery and vein of acellular kidneys resulted in the uniform distribution of the cells in all the vasculature compartments, from glomerular capillaries to peritubular capillaries and small vessels. Ultrastructural analysis of repopulated scaffolds through transmission and scanning electron microscopy demonstrated the presence of continuously distributed cells along the vessel wall, which was also confirmed by 3D reconstruction of z-stack images showing the continuity of endothelial cell coverage inside the vessels. Notably, the detection of fenestrae in the endothelium of glomerular capillaries but not in the vascular capillaries was clear evidence of site-specific endothelial cell specialisation.
Collapse
Affiliation(s)
- Osele Ciampi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, 24126, Bergamo, Italy
| | - Barbara Bonandrini
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, 24126, Bergamo, Italy.,Department of Chemistry, Materials and Chemical Engineering Giulio Natta, Politecnico di Milano, 20133, Milan, Italy
| | - Manuela Derosas
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, 24126, Bergamo, Italy
| | - Sara Conti
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, 24126, Bergamo, Italy
| | - Paola Rizzo
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, 24126, Bergamo, Italy
| | - Valentina Benedetti
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, 24126, Bergamo, Italy
| | - Marina Figliuzzi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, 24126, Bergamo, Italy
| | - Andrea Remuzzi
- Department of Industrial Engineering, Bergamo University, 24044, Dalmine, Bergamo, Italy
| | - Ariela Benigni
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, 24126, Bergamo, Italy
| | - Giuseppe Remuzzi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, 24126, Bergamo, Italy.,L. Sacco Department of Biomedical and Clinical Sciences, University of Milan, 20122, Milan, Italy
| | - Susanna Tomasoni
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, 24126, Bergamo, Italy.
| |
Collapse
|
37
|
Rinschen MM, Limbutara K, Knepper MA, Payne DM, Pisitkun T. From Molecules to Mechanisms: Functional Proteomics and Its Application to Renal Tubule Physiology. Physiol Rev 2019; 98:2571-2606. [PMID: 30182799 DOI: 10.1152/physrev.00057.2017] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Classical physiological studies using electrophysiological, biophysical, biochemical, and molecular techniques have created a detailed picture of molecular transport, bioenergetics, contractility and movement, and growth, as well as the regulation of these processes by external stimuli in cells and organisms. Newer systems biology approaches are beginning to provide deeper and broader understanding of these complex biological processes and their dynamic responses to a variety of environmental cues. In the past decade, advances in mass spectrometry-based proteomic technologies have provided invaluable tools to further elucidate these complex cellular processes, thereby confirming, complementing, and advancing common views of physiology. As one notable example, the application of proteomics to study the regulation of kidney function has yielded novel insights into the chemical and physical processes that tightly control body fluids, electrolytes, and metabolites to provide optimal microenvironments for various cellular and organ functions. Here, we systematically review, summarize, and discuss the most significant key findings from functional proteomic studies in renal epithelial physiology. We also identify further improvements in technological and bioinformatics methods that will be essential to advance precision medicine in nephrology.
Collapse
Affiliation(s)
- Markus M Rinschen
- Department II of Internal Medicine, University Hospital Cologne , Cologne , Germany ; Center for Molecular Medicine Cologne, University of Cologne , Cologne , Germany ; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne , Cologne , Germany ; Division of Nephrology, Faculty of Medicine, Chulalongkorn University , Bangkok , Thailand ; Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, National Institutes of Health , Bethesda, Maryland ; and Center of Excellence in Systems Biology, Research Affairs, Faculty of Medicine, Chulalongkorn University , Bangkok , Thailand
| | - Kavee Limbutara
- Department II of Internal Medicine, University Hospital Cologne , Cologne , Germany ; Center for Molecular Medicine Cologne, University of Cologne , Cologne , Germany ; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne , Cologne , Germany ; Division of Nephrology, Faculty of Medicine, Chulalongkorn University , Bangkok , Thailand ; Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, National Institutes of Health , Bethesda, Maryland ; and Center of Excellence in Systems Biology, Research Affairs, Faculty of Medicine, Chulalongkorn University , Bangkok , Thailand
| | - Mark A Knepper
- Department II of Internal Medicine, University Hospital Cologne , Cologne , Germany ; Center for Molecular Medicine Cologne, University of Cologne , Cologne , Germany ; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne , Cologne , Germany ; Division of Nephrology, Faculty of Medicine, Chulalongkorn University , Bangkok , Thailand ; Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, National Institutes of Health , Bethesda, Maryland ; and Center of Excellence in Systems Biology, Research Affairs, Faculty of Medicine, Chulalongkorn University , Bangkok , Thailand
| | - D Michael Payne
- Department II of Internal Medicine, University Hospital Cologne , Cologne , Germany ; Center for Molecular Medicine Cologne, University of Cologne , Cologne , Germany ; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne , Cologne , Germany ; Division of Nephrology, Faculty of Medicine, Chulalongkorn University , Bangkok , Thailand ; Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, National Institutes of Health , Bethesda, Maryland ; and Center of Excellence in Systems Biology, Research Affairs, Faculty of Medicine, Chulalongkorn University , Bangkok , Thailand
| | - Trairak Pisitkun
- Department II of Internal Medicine, University Hospital Cologne , Cologne , Germany ; Center for Molecular Medicine Cologne, University of Cologne , Cologne , Germany ; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne , Cologne , Germany ; Division of Nephrology, Faculty of Medicine, Chulalongkorn University , Bangkok , Thailand ; Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, National Institutes of Health , Bethesda, Maryland ; and Center of Excellence in Systems Biology, Research Affairs, Faculty of Medicine, Chulalongkorn University , Bangkok , Thailand
| |
Collapse
|
38
|
Hariharan K, Stachelscheid H, Rossbach B, Oh SJ, Mah N, Schmidt-Ott K, Kurtz A, Reinke P. Parallel generation of easily selectable multiple nephronal cell types from human pluripotent stem cells. Cell Mol Life Sci 2019; 76:179-192. [PMID: 30310934 PMCID: PMC11105784 DOI: 10.1007/s00018-018-2929-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 09/21/2018] [Accepted: 09/26/2018] [Indexed: 02/05/2023]
Abstract
Human pluripotent stem cells (hPSCs) provide a source for the generation of defined kidney cells and renal organoids applicable in regenerative medicine, disease modeling, and drug screening. These applications require the provision of hPSC-derived renal cells by reproducible, scalable, and efficient methods. We established a chemically defined protocol by application of Activin A, BMP4, and Retinoic acid followed by GDNF, which steered hPSCs to the renal lineage and resulted in populations of SIX2+/CITED1+ metanephric mesenchyme- (MM) and of HOXB7+/GRHL2+ ureteric bud (UB)-like cells already by 6 days. Transcriptome analysis corroborated that the PSC-derived cell types at day 8 resemble their renal vesicle and ureteric epithelial counterpart in vivo, forming tubular and glomerular renal cells 6 days later. We demonstrate that starting from hPSCs, our in vitro protocol generates a pool of nephrogenic progenitors at the renal vesicle stage, which can be further directed into specialized nephronal cell types including mesangial-, proximal tubular-, distal tubular, collecting duct epithelial cells, and podocyte precursors after 14 days. This simple and rapid method to produce renal cells from a common precursor pool in 2D culture provides the basis for scaled-up production of tailored renal cell types, which are applicable for drug testing or cell-based regenerative therapies.
Collapse
Affiliation(s)
- Krithika Hariharan
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Harald Stachelscheid
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- Berlin Institute of Health (BIH), Stem Cell Core, Berlin, Germany
| | - Bella Rossbach
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Su-Jun Oh
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Nancy Mah
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Kai Schmidt-Ott
- Department of Nephrology and Intensive Care, Charité University Medicine Berlin, Berlin, Germany
- Max Delbrueck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Andreas Kurtz
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.
| | - Petra Reinke
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- Department of Nephrology and Intensive Care, Charité University Medicine Berlin, Berlin, Germany
| |
Collapse
|
39
|
Hortu I, Ozceltik G, Sahin C, Akman L, Yildirim N, Erbas O. Granulocyte Colony-Stimulating Factor Prevents Ischemia/Reperfusion-Induced Ovarian Injury in Rats: Evaluation of Histological and Biochemical Parameters. Reprod Sci 2018; 26:1389-1394. [PMID: 30497339 DOI: 10.1177/1933719118816839] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Granulocyte colony-stimulating factor (G-CSF) is a glycoprotein commonly used in the field of medicine to treat neutropenia. Granulocyte colony-stimulating factor has also crucial roles in ameliorating the ischemia/reperfusion (I/R) injury in particular tissues. In this study, we aimed to investigate the protective effect of G-CSF on ovarian damage in experimental ovarian I/R injury. Thirty adult female rats were used. Rats were separated randomly into 5 groups; Group 1: sham group (abdominal wall was opened and closed surgically), Group 2: torsion group with 3-hour ischemia using vascular clips. Group 3: torsion + G-CSF group with 3-hour ischemia 30 minutes after the administration intraperitoneal (i.p.) of 100 µg/kg of G-CSF. Group 4: torsion-detorsion group with 3 hour ischemia and 3 hour reperfusion. Group 5: torsion-detorsion + G-CSF group with 3 hour ischemia followed by 100 µg/kg of G-CSF i.p. administration 30 minutes prior to 3 hour of detorsion/reperfusion. Ovarian tissue damage was scored on histopathology. Ovarian tissue malondialdehyde (MDA) was measured biochemically. In comparison with the sham group, both the torsion and torsion-detorsion groups had significantly higher scores for follicular degeneration, vascular congestion, edema, hemorrhage, and leukocyte infiltration (P < .05). When compared group torsion-detorsion + G-CSF to group torsion-detorsion, parameters aforementioned significantly decreased in group torsion-detorsion + G-CSF (P < .05). Granulocyte colony-stimulating factor has also decreased MDA levels notably both in the torsion + G-CSF and torsion-detorsion + G-CSF groups (P < .05, P < .01). Our experimental study suggests that G-CSF can be a novel agent for the treatment of ovarian I/R injury.
Collapse
Affiliation(s)
- Ismet Hortu
- Department of Obstetrics and Gynecology, Ege University School of Medicine, Izmir, Turkey
| | - Gokay Ozceltik
- Department of Obstetrics and Gynecology, Ege University School of Medicine, Izmir, Turkey
| | - Cagdas Sahin
- Department of Obstetrics and Gynecology, Ege University School of Medicine, Izmir, Turkey
| | - Levent Akman
- Department of Obstetrics and Gynecology, Ege University School of Medicine, Izmir, Turkey
| | - Nuri Yildirim
- Department of Obstetrics and Gynecology, Ege University School of Medicine, Izmir, Turkey
| | - Oytun Erbas
- Department of Physiology, Istanbul Bilim University School of Medicine, Istanbul, Turkey
| |
Collapse
|
40
|
Roufosse C, Simmonds N, Clahsen-van Groningen M, Haas M, Henriksen KJ, Horsfield C, Loupy A, Mengel M, Perkowska-Ptasińska A, Rabant M, Racusen LC, Solez K, Becker JU. A 2018 Reference Guide to the Banff Classification of Renal Allograft Pathology. Transplantation 2018; 102:1795-1814. [PMID: 30028786 PMCID: PMC7597974 DOI: 10.1097/tp.0000000000002366] [Citation(s) in RCA: 492] [Impact Index Per Article: 70.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 05/07/2018] [Accepted: 05/23/2018] [Indexed: 12/24/2022]
Abstract
The Banff Classification of Allograft Pathology is an international consensus classification for the reporting of biopsies from solid organ transplants. Since its initial conception in 1991 for renal transplants, it has undergone review every 2 years, with attendant updated publications. The rapid expansion of knowledge in the field has led to numerous revisions of the classification. The resultant dispersal of relevant content makes it difficult for novices and experienced pathologists to faithfully apply the classification in routine diagnostic work and in clinical trials. This review shall provide a complete and simple illustrated reference guide of the Banff Classification of Kidney Allograft Pathology based on all publications including the 2017 update. It is intended as a concise desktop reference for pathologists and clinicians, providing definitions, Banff Lesion Scores and Banff Diagnostic Categories. An online website reference guide hosted by the Banff Foundation for Allograft Pathology (www.banfffoundation.org) is being developed, which will be updated with future refinement of the Banff Classification from 2019 onward.
Collapse
Affiliation(s)
- Candice Roufosse
- Department of Medicine, Imperial College, London, United Kingdom
- North West London Pathology, London, United Kingdom
| | - Naomi Simmonds
- Department of Histopathology, Guy's and St. Thomas' National Health Service Foundation Trust, London, United Kingdom
| | | | - Mark Haas
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA
| | | | - Catherine Horsfield
- Department of Histopathology, Guy's and St. Thomas' National Health Service Foundation Trust, London, United Kingdom
| | - Alexandre Loupy
- Paris Translational Research Center for Organ Transplantation, Paris, France
| | - Michael Mengel
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Canada
| | | | - Marion Rabant
- Department of Pathology, Necker Hospital University Paris Descartes, Paris, France
| | | | - Kim Solez
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Canada
| | - Jan U. Becker
- Institute of Pathology, University Hospital of Cologne, Cologne, Germany
| |
Collapse
|
41
|
Yeo WS, Zhang YC. Bioengineering in renal transplantation: technological advances and novel options. Pediatr Nephrol 2018; 33:1105-1111. [PMID: 28589209 DOI: 10.1007/s00467-017-3706-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 05/07/2017] [Accepted: 05/11/2017] [Indexed: 01/03/2023]
Abstract
End-stage kidney disease (ESKD) is one of the most prevalent diseases in the world with significant morbidity and mortality. Current modes of renal replacement therapy include dialysis and renal transplantation. Although dialysis is an acceptable mode of renal replacement therapy, it does have its shortcomings, which include poorer life expectancy compared with renal transplantation, risk of infections and vascular thrombosis, lack of vascular access and absence of biosynthetic functions of the kidney. Renal transplantation, in contrast, is the preferred option of renal replacement therapy, with improved morbidity and mortality rates and quality of life, compared with dialysis. Renal transplantation, however, may not be available to all patients with ESKD. Some of the key factors limiting the availability and efficiency of renal transplantation include shortage of donor organs and the constant risk of rejection with complications associated with over-immunosuppression respectively. This review focuses chiefly on the potential roles of bioengineering in overcoming limitations in renal transplantation via the development of cell-based bioartificial dialysis devices as bridging options before renal transplantation, and the development of new sources of organs utilizing cell and organ engineering.
Collapse
Affiliation(s)
- Wee-Song Yeo
- Division of Pediatric Nephrology, Dialysis and Renal Transplantation, Shaw-National Kidney Foundation, National University Hospital Children's Kidney Centre, Khoo Teck Puat-National University, Children's Medical Institute, National University Health System, NUHS Tower Block, 1E Kent Ridge Road, Singapore, 119228, Singapore.
| | - Yao-Chun Zhang
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
42
|
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
|
43
|
Zheng CX, Sui BD, Hu CH, Qiu XY, Zhao P, Jin Y. Reconstruction of structure and function in tissue engineering of solid organs: Toward simulation of natural development based on decellularization. J Tissue Eng Regen Med 2018; 12:1432-1447. [PMID: 29701314 DOI: 10.1002/term.2676] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 10/13/2017] [Accepted: 04/16/2018] [Indexed: 12/21/2022]
Abstract
Failure of solid organs, such as the heart, liver, and kidney, remains a major cause of the world's mortality due to critical shortage of donor organs. Tissue engineering, which uses elements including cells, scaffolds, and growth factors to fabricate functional organs in vitro, is a promising strategy to mitigate the scarcity of transplantable organs. Within recent years, different construction strategies that guide the combination of tissue engineering elements have been applied in solid organ tissue engineering and have achieved much progress. Most attractively, construction strategy based on whole-organ decellularization has become a popular and promising approach, because the overall structure of extracellular matrix can be well preserved. However, despite the preservation of whole structure, the current constructs derived from decellularization-based strategy still perform partial functions of solid organs, due to several challenges, including preservation of functional extracellular matrix structure, implementation of functional recellularization, formation of functional vascular network, and realization of long-term functional integration. This review overviews the status quo of solid organ tissue engineering, including both advances and challenges. We have also put forward a few techniques with potential to solve the challenges, mainly focusing on decellularization-based construction strategy. We propose that the primary concept for constructing tissue-engineered solid organs is fabricating functional organs based on intact structure via simulating the natural development and regeneration processes.
Collapse
Affiliation(s)
- Chen-Xi Zheng
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, China.,Research and Development Center for Tissue Engineering, Fourth Military Medical University, Shaanxi, China
| | - Bing-Dong Sui
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, China.,Research and Development Center for Tissue Engineering, Fourth Military Medical University, Shaanxi, China
| | - Cheng-Hu Hu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, China.,Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi, China
| | - Xin-Yu Qiu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, China.,Research and Development Center for Tissue Engineering, Fourth Military Medical University, Shaanxi, China
| | - Pan Zhao
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, China.,Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi, China
| | - Yan Jin
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, China.,Research and Development Center for Tissue Engineering, Fourth Military Medical University, Shaanxi, China
| |
Collapse
|
44
|
Rahman MS, Spitzhorn LS, Wruck W, Hagenbeck C, Balan P, Graffmann N, Bohndorf M, Ncube A, Guillot PV, Fehm T, Adjaye J. The presence of human mesenchymal stem cells of renal origin in amniotic fluid increases with gestational time. Stem Cell Res Ther 2018; 9:113. [PMID: 29695308 PMCID: PMC5918774 DOI: 10.1186/s13287-018-0864-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 03/19/2018] [Accepted: 04/10/2018] [Indexed: 12/17/2022] Open
Abstract
Background Established therapies for managing kidney dysfunction such as kidney dialysis and transplantation are limited due to the shortage of compatible donated organs and high costs. Stem cell-based therapies are currently under investigation as an alternative treatment option. As amniotic fluid is composed of fetal urine harboring mesenchymal stem cells (AF-MSCs), we hypothesized that third-trimester amniotic fluid could be a novel source of renal progenitor and differentiated cells. Methods Human third-trimester amniotic fluid cells (AFCs) were isolated and cultured in distinct media. These cells were characterized as renal progenitor cells with respect to cell morphology, cell surface marker expression, transcriptome and differentiation into chondrocytes, osteoblasts and adipocytes. To test for renal function, a comparative albumin endocytosis assay was performed using AF-MSCs and commercially available renal cells derived from kidney biopsies. Comparative transcriptome analyses of first, second and third trimester-derived AF-MSCs were conducted to monitor expression of renal-related genes. Results Regardless of the media used, AFCs showed expression of pluripotency-associated markers such as SSEA4, TRA-1-60, TRA-1-81 and C-Kit. They also express the mesenchymal marker Vimentin. Immunophenotyping confirmed that third-trimester AFCs are bona fide MSCs. AF-MSCs expressed the master renal progenitor markers SIX2 and CITED1, in addition to typical renal proteins such as PODXL, LHX1, BRN1 and PAX8. Albumin endocytosis assays demonstrated the functionality of AF-MSCs as renal cells. Additionally, upregulated expression of BMP7 and downregulation of WT1, CD133, SIX2 and C-Kit were observed upon activation of WNT signaling by treatment with the GSK-3 inhibitor CHIR99201. Transcriptome analysis and semiquantitative PCR revealed increasing expression levels of renal-specific genes (e.g., SALL1, HNF4B, SIX2) with gestational time. Moreover, AF-MSCs shared more genes with human kidney cells than with native MSCs and gene ontology terms revealed involvement of biological processes associated with kidney morphogenesis. Conclusions Third-trimester amniotic fluid contains AF-MSCs of renal origin and this novel source of kidney progenitors may have enormous future potentials for disease modeling, renal repair and drug screening. Electronic supplementary material The online version of this article (10.1186/s13287-018-0864-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Md Shaifur Rahman
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich Heine University, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Lucas-Sebastian Spitzhorn
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich Heine University, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Wasco Wruck
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich Heine University, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Carsten Hagenbeck
- Department of Obstetrics and Gynaecology, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Percy Balan
- Department of Obstetrics and Gynaecology, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Nina Graffmann
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich Heine University, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Martina Bohndorf
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich Heine University, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Audrey Ncube
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich Heine University, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Pascale V Guillot
- Institute for Women's Health, Maternal and Fetal Medicine Department, University College London, London, WC1E 6HX, UK
| | - Tanja Fehm
- Department of Obstetrics and Gynaecology, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - James Adjaye
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich Heine University, Moorenstraße 5, 40225, Düsseldorf, Germany.
| |
Collapse
|
45
|
Herrera GA, Teng J, Zeng C, Xu H, Liang M, Alexander JS, Liu B, Boyer C, Turbat-Herrera EA. Phenotypic plasticity of mesenchymal stem cells is crucial for mesangial repair in a model of immunoglobulin light chain-associated mesangial damage. Ultrastruct Pathol 2018; 42:262-288. [PMID: 29668344 DOI: 10.1080/01913123.2018.1449772] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Mesangiopathies produced by glomerulopathic monoclonal immunoglobulin light chains (GLCs) acting on the glomerular mesangium produce two characteristic lesions: AL-amyloidosis (AL-Am) and light chain deposition disease (LCDD). In both cases, the pathology is centered in the mesangium, where initial and progressive damage occurs. In AL-Am the mesangial matrix is destroyed and replaced by amyloid fibrils and in LCDD, the mesangial matrix is increased and remodeled. The collagen IV rich matrix is replaced by tenascin. In both conditions, mesangial cells (MCs) become apoptotic as a direct effect of the GLCs. MCs were incubated in-vitro with GLCs and animal kidneys were perfused ex-vivo via the renal artery with GLCs, producing expected lesions, and then mesenchymal stem cells (MSCs) were added to both platforms. Each of the two platforms provided unique information that when put together created a comprehensive evaluation of the processes involved. A "cocktail" with growth and differentiating factors was used to study its effect on mesangial repair. MSCs displayed remarkable phenotypic plasticity during the repair process. The first role of the MSCs after migrating to the affected areas was to dispose of the amyloid fibrils (in AL-Am), the altered mesangial matrix (in LCDD) and apoptotic MCs/debris. To accomplish this task, MSCs transformed into facultative macrophages acquiring an abundance of lysosomes and endocytotic capabilities required to engage in phagocytic functions. Once the mesangial cleaning was completed, MSCs transformed into functional MCs restoring the mesangium to normal. "Cocktail" made the repair process more efficient.
Collapse
Affiliation(s)
- Guillermo A Herrera
- a Departments of Pathology and Translational Pathobiology and Cell Biology and Anatomy , Louisiana State Health Sciences Center , Shreveport , LA , USA
| | - Jiamin Teng
- b Department of Pathology and Translational Pathobiology , Louisiana State Health Sciences Center , Shreveport , LA , USA
| | - Chun Zeng
- b Department of Pathology and Translational Pathobiology , Louisiana State Health Sciences Center , Shreveport , LA , USA
| | - Hongzhi Xu
- b Department of Pathology and Translational Pathobiology , Louisiana State Health Sciences Center , Shreveport , LA , USA
| | - Man Liang
- b Department of Pathology and Translational Pathobiology , Louisiana State Health Sciences Center , Shreveport , LA , USA
| | - J Steven Alexander
- c Department of Molecular and Cellular Physiology , Louisiana State Health Sciences Center , Shreveport , LA , USA
| | - Bing Liu
- b Department of Pathology and Translational Pathobiology , Louisiana State Health Sciences Center , Shreveport , LA , USA
| | - Chris Boyer
- c Department of Molecular and Cellular Physiology , Louisiana State Health Sciences Center , Shreveport , LA , USA
| | - Elba A Turbat-Herrera
- d Departments of Pathology and Translational Pathobiology , Medicine, and Cell Biology and Anatomy, Louisiana State Health Sciences Center , Shreveport , LA , USA
| |
Collapse
|
46
|
Arbra CA, Nadig SN, Dennis SG, Pattanaik S, Bainbridge HA, Rhett JM, Fann SA, Atkinson C, Yost MJ. Microdissection of Primary Renal Tissue Segments and Incorporation with Novel Scaffold-free Construct Technology. J Vis Exp 2018. [PMID: 29658916 DOI: 10.3791/57358] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Kidney transplantation is now a mainstream therapy for end-stage renal disease. However, with approximately 96,000 people on the waiting list and only one-fourth of these patients achieving transplantation, there is a dire need for alternatives for those with failing organs. In order to decrease the harmful consequences of dialysis along with the overall healthcare costs it incurs, active investigation is ongoing in search of alternative solutions to organ transplantation. Implantable tissue-engineered renal cellular constructs are one such feasible approach to replacing lost renal functionality. Here, described for the first time, is the microdissection of murine kidneys for isolation of living corticomedullary renal segments. These segments are capable of rapid incorporation within scaffold-free endothelial-fibroblast constructs which may enable rapid connection with host vasculature once implanted. Adult mouse kidneys were procured from living donors, followed by stereoscope microdissection to obtain renal segments 200 - 300 µm in diameter. Multiple renal constructs were fabricated using primary renal segments harvested from only one kidney. This method demonstrates a procedure which could salvage functional renal tissue from organs that would otherwise be discarded.
Collapse
Affiliation(s)
- Chase A Arbra
- Department of Surgery, Medical University of South Carolina
| | - Satish N Nadig
- Department of Surgery, Medical University of South Carolina
| | | | | | | | | | - Stephen A Fann
- Department of Surgery, Medical University of South Carolina
| | - Carl Atkinson
- Department of Surgery, Medical University of South Carolina
| | - Michael J Yost
- Department of Surgery, Medical University of South Carolina;
| |
Collapse
|
47
|
Solez K, Fung KC, Saliba KA, Sheldon VLC, Petrosyan A, Perin L, Burdick JF, Fissell WH, Demetris AJ, Cornell LD. The bridge between transplantation and regenerative medicine: Beginning a new Banff classification of tissue engineering pathology. Am J Transplant 2018; 18:321-327. [PMID: 29194964 PMCID: PMC5817246 DOI: 10.1111/ajt.14610] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 11/21/2017] [Accepted: 11/24/2017] [Indexed: 01/25/2023]
Abstract
The science of regenerative medicine is arguably older than transplantation-the first major textbook was published in 1901-and a major regenerative medicine meeting took place in 1988, three years before the first Banff transplant pathology meeting. However, the subject of regenerative medicine/tissue engineering pathology has never received focused attention. Defining and classifying tissue engineering pathology is long overdue. In the next decades, the field of transplantation will enlarge at least tenfold, through a hybrid of tissue engineering combined with existing approaches to lessening the organ shortage. Gradually, transplantation pathologists will become tissue-(re-) engineering pathologists with enhanced skill sets to address concerns involving the use of bioengineered organs. We outline ways of categorizing abnormalities in tissue-engineered organs through traditional light microscopy or other modalities including biomarkers. We propose creating a new Banff classification of tissue engineering pathology to standardize and assess de novo bioengineered solid organs transplantable success in vivo. We recommend constructing a framework for a classification of tissue engineering pathology now with interdisciplinary consensus discussions to further develop and finalize the classification at future Banff Transplant Pathology meetings, in collaboration with the human cell atlas project. A possible nosology of pathologic abnormalities in tissue-engineered organs is suggested.
Collapse
Affiliation(s)
- K. Solez
- Department of Laboratory Medicine and PathologyFaculty of Medicine and DentistryUniversity of AlbertaEdmontonABCanada
| | - K. C. Fung
- Department of Laboratory Medicine and PathologyFaculty of Medicine and DentistryUniversity of AlbertaEdmontonABCanada
| | - K. A. Saliba
- Department of Laboratory Medicine and PathologyFaculty of Medicine and DentistryUniversity of AlbertaEdmontonABCanada
| | - V. L. C. Sheldon
- Medical Anthropology ProgramDepartment of AnthropologyFaculty of Arts and SciencesUniversity of TorontoTorontoOntarioCanada
| | - A. Petrosyan
- Division of Urology GOFARR Laboratory for Organ Regenerative Research and Cell TherapeuticsChildren's Hospital Los AngelesSaban Research InstituteUniversity of Southern CaliforniaLos AngelesCAUSA
| | - L. Perin
- Division of Urology GOFARR Laboratory for Organ Regenerative Research and Cell TherapeuticsChildren's Hospital Los AngelesSaban Research InstituteUniversity of Southern CaliforniaLos AngelesCAUSA
| | - J. F. Burdick
- Department of SurgeryJohns Hopkins School of MedicineBaltimoreMDUSA
| | - W. H. Fissell
- Department of MedicineVanderbilt University Medical CenterNashvilleTNUSA
| | - A. J. Demetris
- Department of PathologyUniversity of PittsburghUPMC‐MontefiorePittsburghPAUSA
| | - L. D. Cornell
- Department of Laboratory Medicine and PathologyMayo ClinicRochesterMNUSA
| |
Collapse
|
48
|
Skowron B, Baranowska A, Kaszuba-Zwoińska J, Więcek G, Malska-Woźniak A, Heczko P, Strus M. Experimental model for acute kidney injury caused by uropathogenic Escherichia coli. POSTEP HIG MED DOSW 2017; 71:520-529. [PMID: 28665281 DOI: 10.5604/01.3001.0010.3833] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Acute kidney injury (AKI) is the rapid deterioration of renal function, diagnosed on the basis of an increase in serum creatinine and abnormal urinary parameters. AKI is associated with increased risk of mortality or chronic kidney disease (CKD). The aim of the study was to develop an experimental model for AKI resulting from Escherichia coli-induced pyelonephritis. E. coli was isolated from a patient with clinical symptoms of urinary tract infection (UTI). MATERIAL/METHODS The study included three groups of female Wistar rats (groups 1, 2 and 3), in which pyelonephritis was induced by transurethral inoculation with highly virulent E. coli (105, 107 and 109 cfu/ml, respectively). Urine and blood samples for analysis were obtained prior to the inoculation (day 0), as well as 7, 14 and 21 days thereafter. RESULTS Aside from a microbiological examination of urine samples, daily urine output, serum creatinine (CreaS), creatinine clearance (CrCl), interleukin 6 (IL-6), fractional excretion of sodium (FENa) and fractional excretion of urea (FEUrea) were determined. A histopathological examination of kidney and urinary bladder specimens was conducted as well. While UTI-related pyelonephritis developed irrespective of E. coli inoculum size, AKI was observed only following transurethral administration of E. coli at the intermediate and high dose, i.e. 107 and 109 cfu/ml, respectively (group 2 and 3). DISCUSSION An increase in CreaS and abnormal diuresis were accompanied by changes in parameters specific for various forms of AKI, i.e. FENa and FEUrea. Based on these changes, administration of E. coli at 107 cfu/ml was demonstrated to induce renal AKI, whereas inoculation with 109 cfu/ml seemed to cause not only ascending pyelonephritis, but perhaps also bacteremia and urosepsis (prerenal component of AKI).
Collapse
Affiliation(s)
- Beata Skowron
- Department of Pathophysiology, Jagiellonian University Medical College, Krakow, Poland
| | - Agnieszka Baranowska
- Department of Pathophysiology, Jagiellonian University Medical College, Krakow, Poland
| | | | - Grażyna Więcek
- Department of Microbiology, Jagiellonian University Medical College, Krakow, Poland
| | - Anna Malska-Woźniak
- Department of Microbiology, Jagiellonian University Medical College, Krakow, Poland
| | - Piotr Heczko
- Department of Microbiology, Jagiellonian University Medical College, Krakow, Poland
| | - Magdalena Strus
- Department of Microbiology, Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
49
|
Destefani AC, Sirtoli GM, Nogueira BV. Advances in the Knowledge about Kidney Decellularization and Repopulation. Front Bioeng Biotechnol 2017; 5:34. [PMID: 28620603 PMCID: PMC5451511 DOI: 10.3389/fbioe.2017.00034] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 05/03/2017] [Indexed: 12/15/2022] Open
Abstract
End-stage renal disease (ESRD) is characterized by the progressive deterioration of renal function that may compromise different tissues and organs. The major treatment indicated for patients with ESRD is kidney transplantation. However, the shortage of available organs, as well as the high rate of organ rejection, supports the need for new therapies. Thus, the implementation of tissue bioengineering to organ regeneration has emerged as an alternative to traditional organ transplantation. Decellularization of organs with chemical, physical, and/or biological agents generates natural scaffolds, which can serve as basis for tissue reconstruction. The recellularization of these scaffolds with different cell sources, such as stem cells or adult differentiated cells, can provide an organ with functionality and no immune response after in vivo transplantation on the host. Several studies have focused on improving these techniques, but until now, there is no optimal decellularization method for the kidney available yet. Herein, an overview of the current literature for kidney decellularization and whole-organ recellularization is presented, addressing the pros and cons of the actual techniques already developed, the methods adopted to evaluate the efficacy of the procedures, and the challenges to be overcome in order to achieve an optimal protocol.
Collapse
Affiliation(s)
- Afrânio Côgo Destefani
- Tissue Engineering Core—LUCCAR, Morphology, Federal University of Espírito Santo (UFES), Vitória, Brazil
- Health Sciences Center, Federal University of Espírito Santo (UFES), Vitória, Brazil
- Health Sciences Center, Postgraduate Program in Biotechnology/RENORBIO, Vitória, Brazil
| | - Gabriela Modenesi Sirtoli
- Tissue Engineering Core—LUCCAR, Morphology, Federal University of Espírito Santo (UFES), Vitória, Brazil
- Health Sciences Center, Federal University of Espírito Santo (UFES), Vitória, Brazil
| | - Breno Valentim Nogueira
- Tissue Engineering Core—LUCCAR, Morphology, Federal University of Espírito Santo (UFES), Vitória, Brazil
- Health Sciences Center, Federal University of Espírito Santo (UFES), Vitória, Brazil
- Health Sciences Center, Postgraduate Program in Biotechnology/RENORBIO, Vitória, Brazil
| |
Collapse
|
50
|
Tögel F, Valerius MT, Freedman BS, Iatrino R, Grinstein M, Bonventre JV. Repair after nephron ablation reveals limitations of neonatal neonephrogenesis. JCI Insight 2017; 2:e88848. [PMID: 28138555 DOI: 10.1172/jci.insight.88848] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The neonatal mouse kidney retains nephron progenitor cells in a nephrogenic zone for 3 days after birth. We evaluated whether de novo nephrogenesis can be induced postnatally beyond 3 days. Given the long-term implications of nephron number for kidney health, it would be useful to enhance nephrogenesis in the neonate. We induced nephron reduction by cryoinjury with or without contralateral nephrectomy during the neonatal period or after 1 week of age. There was no detectable compensatory de novo nephrogenesis, as determined by glomerular counting and lineage tracing. Contralateral nephrectomy resulted in additional adaptive healing, with little or no fibrosis, but did not also stimulate de novo nephrogenesis. In contrast, injury initiated at 1 week of age led to healing with fibrosis. Thus, despite the presence of progenitor cells and ongoing nephron maturation in the newborn mouse kidney, de novo nephrogenesis is not inducible by acute nephron reduction. This indicates that additional nephron progenitors cannot be recruited after birth despite partial renal ablation providing a reparative stimulus and suggests that nephron number in the mouse is predetermined at birth.
Collapse
Affiliation(s)
- Florian Tögel
- Renal Division, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - M Todd Valerius
- Renal Division, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Benjamin S Freedman
- Renal Division, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Rossella Iatrino
- Renal Division, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Mor Grinstein
- Center for Regenerative Medicine and Department of Orthopaedic Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Joseph V Bonventre
- Renal Division, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| |
Collapse
|