1
|
Mederacke M, Conrad L, Doumpas N, Vetter R, Iber D. Geometric effects position renal vesicles during kidney development. Cell Rep 2023; 42:113526. [PMID: 38060445 DOI: 10.1016/j.celrep.2023.113526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 07/25/2023] [Accepted: 11/15/2023] [Indexed: 12/30/2023] Open
Abstract
During kidney development, reciprocal signaling between the epithelium and the mesenchyme coordinates nephrogenesis with branching morphogenesis of the collecting ducts. The mechanism that positions the renal vesicles, and thus the nephrons, relative to the branching ureteric buds has remained elusive. By combining computational modeling and experiments, we show that geometric effects concentrate the key regulator, WNT9b, at the junctions between parent and daughter branches where renal vesicles emerge, even when uniformly expressed in the ureteric epithelium. This curvature effect might be a general paradigm to create non-uniform signaling in development.
Collapse
Affiliation(s)
- Malte Mederacke
- Department of Biosystems Science and Engineering, ETH Zürich, Schanzenstrasse 44, 4056 Basel, Switzerland; Swiss Institute of Bioinformatics, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Lisa Conrad
- Department of Biosystems Science and Engineering, ETH Zürich, Schanzenstrasse 44, 4056 Basel, Switzerland; Swiss Institute of Bioinformatics, Mattenstrasse 26, 4058 Basel, Switzerland; Department for BioMedical Research (DBMR), University of Bern, Murtenstrasse 35, 3008 Bern, Switzerland
| | - Nikolaos Doumpas
- Department of Biosystems Science and Engineering, ETH Zürich, Schanzenstrasse 44, 4056 Basel, Switzerland; Swiss Institute of Bioinformatics, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Roman Vetter
- Department of Biosystems Science and Engineering, ETH Zürich, Schanzenstrasse 44, 4056 Basel, Switzerland; Swiss Institute of Bioinformatics, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Dagmar Iber
- Department of Biosystems Science and Engineering, ETH Zürich, Schanzenstrasse 44, 4056 Basel, Switzerland; Swiss Institute of Bioinformatics, Mattenstrasse 26, 4058 Basel, Switzerland.
| |
Collapse
|
2
|
Gu M, Chen P, Zeng D, Jiang X, Lv Q, Li Y, Zhang F, Wan S, Zhou Q, Lu Y, Wang X, Li L. Preeclampsia impedes foetal kidney development by delivering placenta-derived exosomes to glomerular endothelial cells. Cell Commun Signal 2023; 21:336. [PMID: 37996949 PMCID: PMC10666440 DOI: 10.1186/s12964-023-01286-y] [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: 05/30/2023] [Accepted: 08/19/2023] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND Foetal renal dysplasia is still the main cause of adult renal disease. Placenta-derived exosomes are an important communication tool, and they may play an important role in placental (both foetal and maternal) function. We hypothesize that in women with preeclampsia, foetal renal dysplasia is impeded by delivering placenta-derived exosomes to glomerular endothelial cells. METHODS In the present study, we established a PE trophoblast oxidative stress model to isolate exosomes from supernatants by ultracentrifugation (NO-exo and H/R-exo) and collected normal and PE umbilical cord blood plasma to isolate exosomes by ultracentrifugation combined with sucrose density gradient centrifugation (N-exo and PE-exo), then we investigated their effects on foetal kidney development by in vitro, ex vivo and in vivo models. RESULTS The PE trophoblast oxidative stress model was established successfully. After that, in in vitro studies, we found that H/R-exo and PE-exo could adversely affect glomerular endothelial cell proliferation, tubular formation, migration, and barrier functions. In ex vivo studies, H/R-exo and PE-exo both inhibited the growth and branch formation of kidney explants, along with the decrease of VE-cadherin and Occludin. In in vivo studies, we also found that H/R-exo and PE-exo could result in renal dysplasia, reduced glomerular number, and reduced barrier function in foetal mice. CONCLUSIONS In conclusion, we demonstrated that PE placenta-derived exosomes could lead to foetal renal dysplasia by delivering placenta-derived exosomes to foetal glomerular endothelial cells, which provides a novel understanding of the pathogenesis of foetal renal dysplasia. Video Abstract.
Collapse
Affiliation(s)
- Mengqi Gu
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Pengzheng Chen
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Dongmei Zeng
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Xiaotong Jiang
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Qingfeng Lv
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Yuchen Li
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Fengyuan Zhang
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Shuting Wan
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Qian Zhou
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Yuan Lu
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Xietong Wang
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China.
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
- The Laboratory of Medical Science and Technology Innovation Center (Institute of Translational Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences) of China, Jinan, 250117, Shandong, China.
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital, 328 Jingshi East Road, Jinan, 250025, Shandong, China.
| | - Lei Li
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China.
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
- The Laboratory of Medical Science and Technology Innovation Center (Institute of Translational Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences) of China, Jinan, 250117, Shandong, China.
| |
Collapse
|
3
|
Viola JM, Liu J, Huang A, Grindel SH, Prahl LS, Hughes AJ. Rho/ROCK activity tunes cell compartment segregation and differentiation in nephron-forming niches. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.08.566308. [PMID: 37986773 PMCID: PMC10659296 DOI: 10.1101/2023.11.08.566308] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Controlling the time and place of nephron formation in vitro would improve nephron density and connectivity in next-generation kidney replacement tissues. Recent developments in kidney organoid technology have paved the way to achieving self-sustaining nephrogenic niches in vitro. The physical and geometric structure of the niche are key control parameters in tissue engineering approaches. However, their relationship to nephron differentiation is unclear. Here we investigate the relationship between niche geometry, cell compartment mixing, and nephron differentiation by targeting the Rho/ROCK pathway, a master regulator of the actin cytoskeleton. We find that the ROCK inhibitor Y-27632 increases mixing between nephron progenitor and stromal compartments in native mouse embryonic kidney niches, and also increases nephrogenesis. Similar increases are also seen in reductionist mouse primary cell and human induced pluripotent stem cell (iPSC)-derived organoids perturbed by Y-27632, dependent on the presence of stromal cells. Our data indicate that niche organization is a determinant of nephron formation rate, bringing renewed focus to the spatial context of cell-cell interactions in kidney tissue engineering efforts.
Collapse
Affiliation(s)
- John M. Viola
- Department of Bioengineering, University of Pennsylvania, Philadelphia, 19104, PA, USA
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, 19104, PA, USA
| | - Jiageng Liu
- Department of Bioengineering, University of Pennsylvania, Philadelphia, 19104, PA, USA
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, 19104, PA, USA
| | - Aria Huang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, 19104, PA, USA
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, 19104, PA, USA
| | - Samuel H. Grindel
- Department of Bioengineering, University of Pennsylvania, Philadelphia, 19104, PA, USA
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, 19104, PA, USA
| | - Louis S. Prahl
- Department of Bioengineering, University of Pennsylvania, Philadelphia, 19104, PA, USA
| | - Alex J. Hughes
- Department of Bioengineering, University of Pennsylvania, Philadelphia, 19104, PA, USA
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, 19104, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, 19104, PA, USA
- Cell and Molecular Biology Graduate Group, University of Pennsylvania, Philadelphia, 19104, PA, USA
- Center for Soft and Living Matter, University of Pennsylvania, Philadelphia, 19104, PA, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, 19104, PA, USA
| |
Collapse
|
4
|
Prahl LS, Viola JM, Liu J, Hughes AJ. The developing murine kidney actively negotiates geometric packing conflicts to avoid defects. Dev Cell 2023; 58:110-120.e5. [PMID: 36693318 PMCID: PMC9924533 DOI: 10.1016/j.devcel.2022.12.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 10/17/2022] [Accepted: 12/20/2022] [Indexed: 01/24/2023]
Abstract
The physiological functions of several organs rely on branched epithelial tubule networks bearing specialized structures for secretion, gas exchange, or filtration. Little is known about conflicts in development between building enough tubules for adequate function and geometric constraints imposed by organ size. We show that the mouse embryonic kidney epithelium negotiates a physical packing conflict between increasing tubule tip numbers through branching and limited organ surface area. Through imaging of whole kidney explants, combined with computational and soft material modeling of tubule families, we identify six possible geometric packing phases, including two defective ones. Experiments in explants show that a radially oriented tension on tubule families is necessary and sufficient for them to switch to a vertical packing arrangement that increases surface tip density while avoiding defects. These results reveal developmental contingencies in response to physical limitations and create a framework for classifying congenital kidney defects.
Collapse
Affiliation(s)
- Louis S Prahl
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John M Viola
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jiageng Liu
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alex J Hughes
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell & Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
5
|
Hamon M, Cheng HM, Johnson M, Yanagawa N, Hauser PV. Effect of Hypoxia on Branching Characteristics and Cell Subpopulations during Kidney Organ Culture. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 9:bioengineering9120801. [PMID: 36551007 PMCID: PMC9774677 DOI: 10.3390/bioengineering9120801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/04/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022]
Abstract
During early developmental stages, embryonic kidneys are not fully vascularized and are potentially exposed to hypoxic conditions, which is known to influence cell proliferation and survival, ureteric bud branching, and vascularization of the developing kidney. To optimize the culture conditions of in vitro cultured kidneys and gain further insight into the effect of hypoxia on kidney development, we exposed mouse embryonic kidneys isolated at E11.5, E12.5, and E13.5 to hypoxic and normal culture conditions and compared ureteric bud branching patterns, the growth of the progenitor subpopulation hoxb7+, and the expression patterns of progenitor and differentiation markers. Branching patterns were quantified using whole organ confocal imaging and gradient-vector-based analysis. In our model, hypoxia causes an earlier expression of UB tip cell markers, and a delay in stalk cell marker gene expression. The metanephric mesenchyme (MM) exhibited a later expression of differentiation marker FGF8, marking a delay in nephron formation. Hypoxia further delayed the expression of stroma cell progenitor markers, a delay in cortical differentiation markers, as well as an earlier expression of medullary and ureteral differentiation markers. We conclude that standard conditions do not apply universally and that tissue engineering strategies need to optimize suitable culture conditions for each application. We also conclude that adapting culture conditions to specific aspects of organ development in tissue engineering can help to improve individual stages of tissue generation.
Collapse
Affiliation(s)
- Morgan Hamon
- Medical and Research Services, Greater Los Angeles Veterans Affairs Healthcare System at Sepulveda, North Hills, CA 91344, USA
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Correspondence: (M.H.); (P.V.H.)
| | - Hsiao-Min Cheng
- Medical and Research Services, Greater Los Angeles Veterans Affairs Healthcare System at Sepulveda, North Hills, CA 91344, USA
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Ming Johnson
- Medical and Research Services, Greater Los Angeles Veterans Affairs Healthcare System at Sepulveda, North Hills, CA 91344, USA
| | - Norimoto Yanagawa
- Medical and Research Services, Greater Los Angeles Veterans Affairs Healthcare System at Sepulveda, North Hills, CA 91344, USA
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Peter V. Hauser
- Medical and Research Services, Greater Los Angeles Veterans Affairs Healthcare System at Sepulveda, North Hills, CA 91344, USA
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Correspondence: (M.H.); (P.V.H.)
| |
Collapse
|
6
|
Kameda Y, Chuaychob S, Tanaka M, Liu Y, Okada R, Fujimoto K, Nakamura T, Yokokawa R. Three-dimensional tissue model in direct contact with an on-chip vascular bed enabled by removable membranes. LAB ON A CHIP 2022; 22:641-651. [PMID: 35018934 DOI: 10.1039/d1lc00751c] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Three-dimensional (3D) tissue culture is a powerful tool for understanding physiological events. However, 3D tissues still have limitations in their size, culture period, and maturity, which are caused by the lack of nutrients and oxygen supply through the vasculature. Here, we propose a new method for culturing a 3D tissue-a spheroid-directly on an 'on-chip vascular bed'. The method can be applied to any 3D tissue because the vascular bed is preformed, so that angiogenic factors from the tissue are not necessary to induce vasculature. The essential component of the assay system is the removable membrane that initially separates the 3D tissue culture well and the microchannel in which a uniform vascular bed is formed, and then allows the tissue to be settled directly onto the vascular bed following its removal. This in vitro system offers a new technique for evaluating the effects of vasculature on 3D tissues.
Collapse
Affiliation(s)
- Yoshikazu Kameda
- Department of Micro Engineering, Kyoto University, Kyoto Daigaku-Katsura, Nishikyo-ku, Kyoto 615-8540, Japan.
| | - Surachada Chuaychob
- Department of Micro Engineering, Kyoto University, Kyoto Daigaku-Katsura, Nishikyo-ku, Kyoto 615-8540, Japan.
| | - Miwa Tanaka
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo 135-8550, Japan
| | - Yang Liu
- Department of Micro Engineering, Kyoto University, Kyoto Daigaku-Katsura, Nishikyo-ku, Kyoto 615-8540, Japan.
| | - Ryu Okada
- Department of Micro Engineering, Kyoto University, Kyoto Daigaku-Katsura, Nishikyo-ku, Kyoto 615-8540, Japan.
| | - Kazuya Fujimoto
- Department of Micro Engineering, Kyoto University, Kyoto Daigaku-Katsura, Nishikyo-ku, Kyoto 615-8540, Japan.
| | - Takuro Nakamura
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo 135-8550, Japan
| | - Ryuji Yokokawa
- Department of Micro Engineering, Kyoto University, Kyoto Daigaku-Katsura, Nishikyo-ku, Kyoto 615-8540, Japan.
| |
Collapse
|
7
|
Serin N, Dihazi GH, Tayyeb A, Lenz C, Müller GA, Zeisberg M, Dihazi H. Calreticulin Deficiency Disturbs Ribosome Biogenesis and Results in Retardation in Embryonic Kidney Development. Int J Mol Sci 2021; 22:5858. [PMID: 34070742 PMCID: PMC8198291 DOI: 10.3390/ijms22115858] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/18/2021] [Accepted: 05/26/2021] [Indexed: 11/27/2022] Open
Abstract
Nephrogenesis is driven by complex signaling pathways that control cell growth and differentiation. The endoplasmic reticulum chaperone calreticulin (Calr) is well known for its function in calcium storage and in the folding of glycoproteins. Its role in kidney development is still not understood. We provide evidence for a pivotal role of Calr in nephrogenesis in this investigation. We show that Calr deficiency results in the disrupted formation of an intact nephrogenic zone and in retardation of nephrogenesis, as evidenced by the disturbance in the formation of comma-shaped and s-shaped bodies. Using proteomics and transcriptomics approaches, we demonstrated that in addition to an alteration in Wnt-signaling key proteins, embryonic kidneys from Calr-/- showed an overall impairment in expression of ribosomal proteins which reveals disturbances in protein synthesis and nephrogenesis. CRISPR/cas9 mediated knockout confirmed that Calr deficiency is associated with a deficiency of several ribosomal proteins and key proteins in ribosome biogenesis. Our data highlights a direct link between Calr expression and the ribosome biogenesis.
Collapse
Affiliation(s)
- Nazli Serin
- Clinic for Nephrology and Rheumatology, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany; (N.S.); (G.A.M.); (M.Z.)
- Department of Hematology and Oncology, University of Medical Center Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany
| | - Gry H. Dihazi
- Institute of Clinical Chemistry/UMG-Laboratories, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany; (G.H.D.); (C.L.)
| | - Asima Tayyeb
- School of Biological Sciences, University of the Punjab, Lahore 54590, Pakistan;
| | - Christof Lenz
- Institute of Clinical Chemistry/UMG-Laboratories, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany; (G.H.D.); (C.L.)
- Bioanalytical Mass Spectrometry, Max Planck Institute for Biophysical Chemistry, 37077 Göttingen, Germany
| | - Gerhard A. Müller
- Clinic for Nephrology and Rheumatology, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany; (N.S.); (G.A.M.); (M.Z.)
| | - Michael Zeisberg
- Clinic for Nephrology and Rheumatology, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany; (N.S.); (G.A.M.); (M.Z.)
| | - Hassan Dihazi
- Clinic for Nephrology and Rheumatology, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany; (N.S.); (G.A.M.); (M.Z.)
- Center for Biostructural Imaging of Neurodegeneration (BIN), University Medical Center Göttingen, 37075 Göttingen, Germany
| |
Collapse
|
8
|
Conrad L, Runser SVM, Fernando Gómez H, Lang CM, Dumond MS, Sapala A, Schaumann L, Michos O, Vetter R, Iber D. The biomechanical basis of biased epithelial tube elongation in lung and kidney development. Development 2021; 148:261770. [PMID: 33946098 PMCID: PMC8126414 DOI: 10.1242/dev.194209] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 03/16/2021] [Indexed: 01/16/2023]
Abstract
During lung development, epithelial branches expand preferentially in a longitudinal direction. This bias in outgrowth has been linked to a bias in cell shape and in the cell division plane. How this bias arises is unknown. Here, we show that biased epithelial outgrowth occurs independent of the surrounding mesenchyme, of preferential turnover of the extracellular matrix at the bud tips and of FGF signalling. There is also no evidence for actin-rich filopodia at the bud tips. Rather, we find epithelial tubes to be collapsed during early lung and kidney development, and we observe fluid flow in the narrow tubes. By simulating the measured fluid flow inside segmented narrow epithelial tubes, we show that the shear stress levels on the apical surface are sufficient to explain the reported bias in cell shape and outgrowth. We use a cell-based vertex model to confirm that apical shear forces, unlike constricting forces, can give rise to both the observed bias in cell shapes and tube elongation. We conclude that shear stress may be a more general driver of biased tube elongation beyond its established role in angiogenesis. This article has an associated 'The people behind the papers' interview.
Collapse
Affiliation(s)
- Lisa Conrad
- Department of Biosystems, Science and Engineering (D-BSSE), ETH Zurich, Mattenstraße 26, 4058 Basel, Switzerland.,Swiss Institute of Bioinformatics (SIB), Mattenstraße 26, 4058 Basel, Switzerland
| | - Steve Vincent Maurice Runser
- Department of Biosystems, Science and Engineering (D-BSSE), ETH Zurich, Mattenstraße 26, 4058 Basel, Switzerland.,Swiss Institute of Bioinformatics (SIB), Mattenstraße 26, 4058 Basel, Switzerland
| | - Harold Fernando Gómez
- Department of Biosystems, Science and Engineering (D-BSSE), ETH Zurich, Mattenstraße 26, 4058 Basel, Switzerland.,Swiss Institute of Bioinformatics (SIB), Mattenstraße 26, 4058 Basel, Switzerland
| | - Christine Michaela Lang
- Department of Biosystems, Science and Engineering (D-BSSE), ETH Zurich, Mattenstraße 26, 4058 Basel, Switzerland.,Swiss Institute of Bioinformatics (SIB), Mattenstraße 26, 4058 Basel, Switzerland
| | - Mathilde Sabine Dumond
- Department of Biosystems, Science and Engineering (D-BSSE), ETH Zurich, Mattenstraße 26, 4058 Basel, Switzerland.,Swiss Institute of Bioinformatics (SIB), Mattenstraße 26, 4058 Basel, Switzerland
| | - Aleksandra Sapala
- Department of Biosystems, Science and Engineering (D-BSSE), ETH Zurich, Mattenstraße 26, 4058 Basel, Switzerland.,Swiss Institute of Bioinformatics (SIB), Mattenstraße 26, 4058 Basel, Switzerland
| | - Laura Schaumann
- Department of Biosystems, Science and Engineering (D-BSSE), ETH Zurich, Mattenstraße 26, 4058 Basel, Switzerland.,Swiss Institute of Bioinformatics (SIB), Mattenstraße 26, 4058 Basel, Switzerland
| | - Odyssé Michos
- Department of Biosystems, Science and Engineering (D-BSSE), ETH Zurich, Mattenstraße 26, 4058 Basel, Switzerland.,Swiss Institute of Bioinformatics (SIB), Mattenstraße 26, 4058 Basel, Switzerland
| | - Roman Vetter
- Department of Biosystems, Science and Engineering (D-BSSE), ETH Zurich, Mattenstraße 26, 4058 Basel, Switzerland.,Swiss Institute of Bioinformatics (SIB), Mattenstraße 26, 4058 Basel, Switzerland
| | - Dagmar Iber
- Department of Biosystems, Science and Engineering (D-BSSE), ETH Zurich, Mattenstraße 26, 4058 Basel, Switzerland.,Swiss Institute of Bioinformatics (SIB), Mattenstraße 26, 4058 Basel, Switzerland
| |
Collapse
|
9
|
Abstract
Explants are three-dimensional tissue fragments maintained outside the organism. The goals of this article are to review the history of fish explant culture and discuss applications of this technique that may assist the modern zebrafish laboratory. Because most zebrafish workers do not have a background in tissue culture, the key variables of this method are deliberately explained in a general way. This is followed by a review of fish-specific explantation approaches, including presurgical husbandry, aseptic dissection technique, choice of media and additives, incubation conditions, viability assays, and imaging studies. Relevant articles since 1970 are organized in a table grouped by organ system. From these, I highlight several recent studies using explant culture to study physiological and embryological processes in teleosts, including circadian rhythms, hormonal regulation, and cardiac development.
Collapse
Affiliation(s)
- Elizabeth E. LeClair
- Department of Biological Sciences, College of Science and Health, DePaul University, Chicago, Illinois, USA
| |
Collapse
|
10
|
Folic acid supplementation alleviates reduced ureteric branching, nephrogenesis, and global DNA methylation induced by maternal nutrient restriction in rat embryonic kidney. PLoS One 2020; 15:e0230289. [PMID: 32251454 PMCID: PMC7135271 DOI: 10.1371/journal.pone.0230289] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 02/25/2020] [Indexed: 01/21/2023] Open
Abstract
We previously reported that maternal nutrient restriction (NR) inhibited ureteric branching, metanephric growth, and nephrogenesis in the rat. Here we examined whether folic acid, a methyl-group donor, rescues the inhibition of kidney development induced by NR and whether DNA methylation is involved in it. The offspring of dams given food ad libitum (CON) and those subjected to 50% food restriction (NR) were examined. NR significantly reduced ureteric tip number at embryonic day 14, which was attenuated by folic acid supplementation to nutrient restricted dams. At embryonic day 18, glomerular number, kidney weight, and global DNA methylation were reduced by NR, and maternal folic acid supplementation again alleviated them. Among DNA methyltransferases (DNMTs), DNMT1 was strongly expressed at embryonic day 15 in CON but was reduced in NR. In organ culture, an inhibitor of DNA methylation 5-aza-2 '-deoxycytidine as well as medium lacking methyl donors folic acid, choline, and methionine, significantly decreased ureteric tip number and kidney size mimicking the effect of NR. In conclusion, global DNA methylation is necessary for normal kidney development. Folic acid supplementation to nutrient restricted dams alleviated the impaired kidney development and DNA methylation in the offspring.
Collapse
|
11
|
Chang CH, Davies JA. In developing mouse kidneys, orientation of loop of Henle growth is adaptive and guided by long-range cues from medullary collecting ducts. J Anat 2019; 235:262-270. [PMID: 31099428 PMCID: PMC6637448 DOI: 10.1111/joa.13012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2019] [Indexed: 11/28/2022] Open
Abstract
The path taken by the loop of Henle, from renal cortex to medulla and back, is critical to the ability of the kidney to concentrate urine and recover water. Unlike most developing tubules, which navigate as blind‐ended cylinders, the loop of Henle extends as a sharply bent loop, the apex of which leads the double tubes behind it in a ‘V’ shape. Here, we show that, in normal kidney development, loops of Henle extend towards the centroid of the kidney with an accuracy that increases the longer they extend. Using cultured kidney rudiments, and manipulations that rotate or remove portions of the organ, we show that loop orientation depends on long‐range cues from the medulla rather than either the orientation of the parent nephron or local cues in the cortex. The loops appear to be attracted to the most mature branch point of the collecting duct system but, if this is removed, they will head towards the most mature collecting duct branch available to them. Our results demonstrate the adaptive nature of guidance of this unusual example of a growing epithelium, and set the stage for later work devoted to understanding the molecules and mechanisms that underlie it.
Collapse
Affiliation(s)
- C-Hong Chang
- Deanery of Biomedical Science, University of Edinburgh, Edinburgh, UK.,Yale University School of Medicine, Medicine, New Haven, CT, USA
| | - Jamie A Davies
- Deanery of Biomedical Science, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
12
|
Lawrence ML, Elhendawi M, Davies JA. Investigating Aspects of Renal Physiology and Pharmacology in Organ and Organoid Culture. Methods Mol Biol 2019; 1926:127-142. [PMID: 30742268 DOI: 10.1007/978-1-4939-9021-4_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Some aspects of renal physiology, in particular transport across tubular epithelia, are highly relevant to pharmacokinetics and to drug toxicity. The use of animals to model human renal physiology is limited, but human-derived renal organoids offer an alternative, relevant system in culture. Here, we explain how the activity of specific transport systems can be assessed in renal organoid and organ culture, using a system illustrated mainly for mouse but that can be extended to human organoids.
Collapse
Affiliation(s)
| | - Mona Elhendawi
- Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Faculty of Medicine, Clinical Pathology Department, Mansoura University, El-Mansoura, Egypt
| | - Jamie A Davies
- Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
13
|
Abstract
Kidney organogenesis has been a widely used classical model system to study inductive tissue interactions that guide differentiation of many organs. The basis for this is in the pioneering work done during the early 1950s when the conditions of how to support ex vivo growth and differentiation of developing kidneys were revealed. Importantly, culturing developing kidneys remains as an essential instrument to advance our understanding of molecular and cellular regulation of morphogenesis even today. Despite the fact that embryonic kidneys have been cultured for decades, it is not a trivial method and requires specific anatomical and developmental biology knowledge. This chapter outlines the general steps in organ culture and details the requirements for successful kidney explant differentiation.
Collapse
|
14
|
Sánchez N, Inostroza V, Pérez MC, Moya P, Ubilla A, Besa J, Llaguno E, Vera P-G C, Inzunza O, Gaete M. Tracking morphological complexities of organ development in culture. Mech Dev 2018; 154:179-192. [DOI: 10.1016/j.mod.2018.07.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 07/03/2018] [Accepted: 07/13/2018] [Indexed: 12/14/2022]
|
15
|
Fontana JM, Khodus GR, Unnersjö-Jess D, Blom H, Aperia A, Brismar H. Spontaneous calcium activity in metanephric mesenchymal cells regulates branching morphogenesis in the embryonic kidney. FASEB J 2018; 33:4089-4096. [PMID: 30496703 PMCID: PMC6404591 DOI: 10.1096/fj.201802054r] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The central role of calcium signaling during development of early vertebrates is well documented, but little is known about its role in mammalian embryogenesis. We have used immunofluorescence and time-lapse calcium imaging of cultured explanted embryonic rat kidneys to study the role of calcium signaling for branching morphogenesis. In mesenchymal cells, we recorded spontaneous calcium activity that was characterized by irregular calcium transients. The calcium signals were dependent on release of calcium from intracellular stores in the endoplasmic reticulum. Down-regulation of the calcium activity, both by blocking the sarco-endoplasmic reticulum Ca2+-ATPase and by chelating cytosolic calcium, resulted in retardation of branching morphogenesis and a reduced formation of primitive nephrons but had no effect on cell proliferation. We propose that spontaneous calcium activity contributes with a stochastic factor to the self-organizing process that controls branching morphogenesis, a major determinant of the ultimate number of nephrons in the kidney.-Fontana, J. M., Khodus, G. R., Unnersjö-Jess, D., Blom, H., Aperia, A., Brismar, H. Spontaneous calcium activity in metanephric mesenchymal cells regulates branching morphogenesis in the embryonic kidney.
Collapse
Affiliation(s)
- Jacopo M Fontana
- Department of Applied Physics, Science for Life Laboratory, Kungliga Tekniska Högskolan (KTH) Royal Institute of Technology, Solna, Sweden
| | - Georgiy R Khodus
- Department of Applied Physics, Science for Life Laboratory, Kungliga Tekniska Högskolan (KTH) Royal Institute of Technology, Solna, Sweden
| | - David Unnersjö-Jess
- Department of Applied Physics, Science for Life Laboratory, Kungliga Tekniska Högskolan (KTH) Royal Institute of Technology, Solna, Sweden
| | - Hans Blom
- Department of Applied Physics, Science for Life Laboratory, Kungliga Tekniska Högskolan (KTH) Royal Institute of Technology, Solna, Sweden
| | - Anita Aperia
- Department of Women's and Children's Health, Science for Life Laboratory, Karolinska Institutet, Solna, Sweden
| | - Hjalmar Brismar
- Department of Applied Physics, Science for Life Laboratory, Kungliga Tekniska Högskolan (KTH) Royal Institute of Technology, Solna, Sweden.,Department of Women's and Children's Health, Science for Life Laboratory, Karolinska Institutet, Solna, Sweden
| |
Collapse
|
16
|
Held M, Santeramo I, Wilm B, Murray P, Lévy R. Ex vivo live cell tracking in kidney organoids using light sheet fluorescence microscopy. PLoS One 2018; 13:e0199918. [PMID: 30048451 PMCID: PMC6062017 DOI: 10.1371/journal.pone.0199918] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 06/16/2018] [Indexed: 12/28/2022] Open
Abstract
Screening cells for their differentiation potential requires a combination of tissue culture models and imaging methods that allow for long-term tracking of the location and function of cells. Embryonic kidney re-aggregation in vitro assays have been established which allow for the monitoring of organotypic cell behaviour in re-aggregated and chimeric renal organoids. However, evaluation of cell integration is hampered by the high photonic load of standard fluorescence microscopy which poses challenges for imaging three-dimensional systems in real-time over a time course. Therefore, we employed light sheet microscopy, a technique that vastly reduces photobleaching and phototoxic effects. We have also developed a new method for culturing the re-aggregates which involves immersed culture, generating organoids which more closely reflect development in vivo. To facilitate imaging from various angles, we embedded the organoids in a freely rotatable hydrogel cylinder. Endpoint fixing and staining were performed to provide additional biomolecular information. We succeeded in imaging labelled cells within re-aggregated kidney organoids over 15 hours and tracking their fate while simultaneously monitoring the development of organotypic morphological structures. Our results show that Wt1-expressing embryonic kidney cells obtained from transgenic mice could integrate into re-aggregated chimeric kidney organoids and contribute to developing nephrons. Furthermore, the nascent proximal tubules that formed in the re-aggregated tissues using the new culture method displayed secretory function, as evidenced by their ability to secrete an organic anion mimic into the tubular lumen.
Collapse
Affiliation(s)
- Marie Held
- Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Ilaria Santeramo
- Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Bettina Wilm
- Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Patricia Murray
- Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Raphaël Lévy
- Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
17
|
Awazu M, Nagata M, Hida M. BMP7 dose-dependently stimulates proliferation and cadherin-11 expression via ERK and p38 in a murine metanephric mesenchymal cell line. Physiol Rep 2018; 5:5/16/e13378. [PMID: 28867673 PMCID: PMC5582263 DOI: 10.14814/phy2.13378] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 07/13/2017] [Accepted: 07/14/2017] [Indexed: 12/12/2022] Open
Abstract
BMP7 is expressed in ureteric buds and cap mesenchyme of the fetal kidney, mediating branching morphogenesis and survival and priming of metanephric mesenchyme. Although dose‐dependent effects of BMP7 in collecting duct cells have been reported, studies in metanephric mesenchymal cells are lacking. We examined the effects of BMP7 on MAP kinase activation, proliferation, and expression of cadherins in a metanephric mesenchymal cell line MS7 by thymidine incorporation, immunoblot analysis, and quantitative real‐time PCR. The levels of phosphorylated ERK (P‐ERK) and phosphorylated p38 (P‐p38) were not altered at 10 min, 1 h, and 6 h with low‐dose BMP7 (0.25 nmol/L), but were increased at 24 h. At 24 h, P‐ERK was increased with low‐dose BMP7, but not by intermediate‐ (1 nmol/L) or high‐dose (10 nmol/L) BMP7, whereas p38 was activated by intermediate‐dose BMP7. Cell proliferation of MS7 was significantly increased by low‐ and intermediate‐dose BMP7 and decreased by high‐dose BMP7. A p38 inhibitor SB203580 5 μmol/L or a MEK inhibitor PD98059 5 μmol/L abolished BMP7‐stimulated proliferation. Expression of cadherin‐11, an adhesion molecule known to promote cell migration and compaction, was upregulated by intermediate‐dose BMP7. BMP7‐induced cadherin‐11 expression was inhibited by cotreatment with SB203580 and PD98059. Finally, in metanephroi cultured with siRNA for cadherin‐11, the number and thickness of cap mesenchyme were reduced. In conclusion, BMP7 exerts differential effects depending on the concentration; it may expand mesenchymal cells in the stroma where BMP7 concentration is low and may upregulate cadherin‐11 promoting condensation around the tip of ureteric buds.
Collapse
Affiliation(s)
- Midori Awazu
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Michio Nagata
- Kidney and Vascular Pathology, University of Tsukuba, Ibaraki, Japan
| | - Mariko Hida
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
18
|
Lawrence ML, Smith JR, Davies JA. Functional transport of organic anions and cations in the murine mesonephros. Am J Physiol Renal Physiol 2018; 315:F130-F137. [PMID: 29561184 DOI: 10.1152/ajprenal.00021.2018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The mesonephros of mammals is a transient renal structure that contributes to various aspects of mammalian fetal development, including the male reproductive system, hematopoietic stem cells, and vascular endothelial cells. The mesonephros develops from the intermediate mesoderm and forms tubules that are segmented in a similar way to the nephrons of the permanent kidney (but lacking loops of Henle). Early studies have suggested that the mesonephros in marsupials and some placental mammals may perform an excretory function, but these studies have not directly shown active transport of organic anions and cations. Excretory function in the rodent mesonephros has not been investigated. Functional characterization of the earliest stages of mammalian renal development is important for our understanding of congenital disease and may help to inform the growing field of renal tissue engineering. Here, we use live uptake and efflux assays in vitro to show that the murine mesonephros is able to transport organic anions and cations through specific transporters from early in its development. Transcript analysis suggests that there are subtle differences between the transporters involved in uptake and efflux by the murine permanent metanephric tubules and by the mesonephric tubules. These data suggest that the mammalian mesonephros can provide an excretory function for the early developing embryo, in addition to the excretory function provided by the placenta.
Collapse
Affiliation(s)
- Melanie L Lawrence
- Deanery of Biomedical Science and Centre for Synthetic Biology, University of Edinburgh , United Kingdom
| | - James R Smith
- Centre for Inflammation Research, University of Edinburgh , Edinburgh , United Kingdom
| | - Jamie A Davies
- Deanery of Biomedical Science and Centre for Synthetic Biology, University of Edinburgh , United Kingdom
| |
Collapse
|
19
|
Elhendawi M, Davies JA. Sebinger Culture: A System Optimized for Morphological Maturation and Imaging of Cultured Mouse Metanephric Primordia. Bio Protoc 2018; 8:e2730. [PMID: 29546231 DOI: 10.21769/bioprotoc.2730] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Here, we present a detailed protocol on setting up embryonic renal organ cultures using a culture method that we have optimised for anatomical maturation and imaging. Our culture method places kidney rudiments on glass in a thin film of medium, which results in very flat cultures with all tubules in the same image plane. For reasons not yet understood, this technique results in improved renal maturation compared to traditional techniques. Typically, this protocol will result in an organ formed with distinct cortical and medullary regions as well as elongated, correctly positioned loops of Henle. This article describes our method and provides detailed advice. We have published qualitative and quantitative evaluations on the performance of the technique in Sebinger et al. (2010) and Chang and Davies (2012).
Collapse
Affiliation(s)
- Mona Elhendawi
- Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, UK.,Clinical Pathology Department, Faculty of Medicine, Mansoura University, El-Mansoura, Egypt
| | - Jamie A Davies
- Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
20
|
Abstract
The goal of this protocol is to describe a method for the dissection, isolation, and culture of mouse metanephric rudiments. During mammalian kidney development, the two progenitor tissues, the ureteric bud and the metanephric mesenchyme, communicate and reciprocally induce cellular mechanisms to eventually form the collecting system and the nephrons of the kidney. As mammalian embryos grow intrauterine and therefore are inaccessible to the observer, an organ culture has been developed. With this method, it is possible to study epithelial-mesenchymal interactions and cellular behavior during kidney organogenesis. Furthermore, the origin of congenital kidney and urogenital tract malformations can be investigated. After careful dissection, the metanephric rudiments are transferred onto a filter that floats on culture medium and can be kept in a cell culture incubator for several days. However, one must be aware that the conditions are artificial and could influence the metabolism in the tissue. Also, the penetration of test substances could be limited due to the extracellular matrix and basal membrane present in the explant. One main advantage of organ culture is that the experimenter can gain direct access to the organ. This technology is cheap, simple, and allows a large number of modifications, such as the addition of biologically active substances, the study of genetic variants, and the application of advanced imaging techniques.
Collapse
Affiliation(s)
- Bejan Aresh
- Department of Neuroscience, Uppsala University
| | - Christiane Peuckert
- Department of Organismal Biology, Evolutionary Biology Center, Uppsala University;
| |
Collapse
|
21
|
Saarela U, Akram SU, Desgrange A, Rak-Raszewska A, Shan J, Cereghini S, Ronkainen VP, Heikkilä J, Skovorodkin I, Vainio SJ. Novel fixed z-direction (FiZD) kidney primordia and an organoid culture system for time-lapse confocal imaging. Development 2017; 144:1113-1117. [PMID: 28219945 PMCID: PMC5358112 DOI: 10.1242/dev.142950] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 02/07/2017] [Indexed: 01/29/2023]
Abstract
Tissue, organ and organoid cultures provide suitable models for developmental studies, but our understanding of how the organs are assembled at the single-cell level still remains unclear. We describe here a novel fixed z-direction (FiZD) culture setup that permits high-resolution confocal imaging of organoids and embryonic tissues. In a FiZD culture a permeable membrane compresses the tissues onto a glass coverslip and the spacers adjust the thickness, enabling the tissue to grow for up to 12 days. Thus, the kidney rudiment and the organoids can adjust to the limited z-directional space and yet advance the process of kidney morphogenesis, enabling long-term time-lapse and high-resolution confocal imaging. As the data quality achieved was sufficient for computer-assisted cell segmentation and analysis, the method can be used for studying morphogenesis ex vivo at the level of the single constituent cells of a complex mammalian organogenesis model system. Summary: Time-lapse confocal imaging of organoids and embryonic tissues through fixed z-direction culture allows long-term single-cell resolution live imaging of tissue growth and morphogenesis.
Collapse
Affiliation(s)
- Ulla Saarela
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90220 Oulu, Finland.,Laboratory of Developmental Biology, Biocenter Oulu and InfoTech, 90220 Oulu, Finland.,Department of Medical Biochemistry and Molecular Medicine, Oulu Center for Cell Matrix Research, 90220 Oulu, Finland
| | - Saad Ullah Akram
- Laboratory of Developmental Biology, Biocenter Oulu and InfoTech, 90220 Oulu, Finland.,Center for Machine Vision Research, Department of Computer Science and Engineering, University of Oulu, 90014 Oulu, Finland
| | - Audrey Desgrange
- Sorbonne Universités, UPMC Univ Paris 06, IBPS - UMR7622 Developmental Biology, Paris F-75005, France.,Institut de Biologie Paris-Seine (IBPS) - CNRS UMR7622 Developmental Biology, F-75005 Paris, France
| | - Aleksandra Rak-Raszewska
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90220 Oulu, Finland.,Laboratory of Developmental Biology, Biocenter Oulu and InfoTech, 90220 Oulu, Finland.,Department of Medical Biochemistry and Molecular Medicine, Oulu Center for Cell Matrix Research, 90220 Oulu, Finland
| | - Jingdong Shan
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90220 Oulu, Finland.,Laboratory of Developmental Biology, Biocenter Oulu and InfoTech, 90220 Oulu, Finland.,Department of Medical Biochemistry and Molecular Medicine, Oulu Center for Cell Matrix Research, 90220 Oulu, Finland
| | - Silvia Cereghini
- Sorbonne Universités, UPMC Univ Paris 06, IBPS - UMR7622 Developmental Biology, Paris F-75005, France.,Institut de Biologie Paris-Seine (IBPS) - CNRS UMR7622 Developmental Biology, F-75005 Paris, France
| | | | - Janne Heikkilä
- Center for Machine Vision Research, Department of Computer Science and Engineering, University of Oulu, 90014 Oulu, Finland
| | - Ilya Skovorodkin
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90220 Oulu, Finland .,Laboratory of Developmental Biology, Biocenter Oulu and InfoTech, 90220 Oulu, Finland.,Department of Medical Biochemistry and Molecular Medicine, Oulu Center for Cell Matrix Research, 90220 Oulu, Finland
| | - Seppo J Vainio
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, 90220 Oulu, Finland .,Laboratory of Developmental Biology, Biocenter Oulu and InfoTech, 90220 Oulu, Finland.,Department of Medical Biochemistry and Molecular Medicine, Oulu Center for Cell Matrix Research, 90220 Oulu, Finland
| |
Collapse
|
22
|
Takasato M, Little MH. A strategy for generating kidney organoids: Recapitulating the development in human pluripotent stem cells. Dev Biol 2016; 420:210-220. [PMID: 27565022 PMCID: PMC6186756 DOI: 10.1016/j.ydbio.2016.08.024] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 06/19/2016] [Accepted: 08/21/2016] [Indexed: 02/06/2023]
Abstract
Directed differentiation of human pluripotent stem cells (hPSCs) can provide us any required tissue/cell types by recapitulating the development in vitro. The kidney is one of the most challenging organs to generate from hPSCs as the kidney progenitors are composed of at least 4 different cell types, including nephron, collecting duct, endothelial and interstitium progenitors, that are developmentally distinguished populations. Although the actual developmental process of the kidney during human embryogenesis has not been clarified yet, studies using model animals accumulated knowledge about the origins of kidney progenitors. The implications of these findings for the directed differentiation of hPSCs into the kidney include the mechanism of the intermediate mesoderm specification and its patterning along with anteroposterior axis. Using this knowledge, we previously reported successful generation of hPSCs-derived kidney organoids that contained all renal components and modelled human kidney development in vitro. In this review, we explain the developmental basis of the strategy behind this differentiation protocol and compare strategies of studies that also recently reported the induction of kidney cells from hPSCs. We also discuss the characterization of such kidney organoids and limitations and future applications of this technology.
Collapse
Affiliation(s)
- Minoru Takasato
- Murdoch Childrens Research Institute, Parkville, Victoria 3052, Australia; RIKEN Center for Developmental Biology, Kobe 650-0047, Japan.
| | - Melissa H Little
- Murdoch Childrens Research Institute, Parkville, Victoria 3052, Australia; Department of Pediatrics, University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
23
|
Rak-Raszewska A, Vainio S. Nephrogenesis in organoids to develop novel drugs and progenitor cell based therapies. Eur J Pharmacol 2016; 790:3-11. [DOI: 10.1016/j.ejphar.2016.07.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 06/21/2016] [Accepted: 07/06/2016] [Indexed: 11/25/2022]
|
24
|
Nieskens TTG, Wilmer MJ. Kidney-on-a-chip technology for renal proximal tubule tissue reconstruction. Eur J Pharmacol 2016; 790:46-56. [PMID: 27401035 DOI: 10.1016/j.ejphar.2016.07.018] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 07/01/2016] [Accepted: 07/08/2016] [Indexed: 12/11/2022]
Abstract
The renal proximal tubule epithelium is responsible for active secretion of endogenous and exogenous waste products from the body and simultaneous reabsorption of vital compounds from the glomerular filtrate. The complexity of this transport machinery makes investigation of processes such as tubular drug secretion a continuous challenge for researchers. Currently available renal cell culture models often lack sufficient physiological relevance and reliability. Introducing complex biological culture systems in a 3D microfluidic design improves the physiological relevance of in vitro renal proximal tubule epithelium models. Organ-on-a-chip technology provides a promising alternative, as it allows the reconstruction of a renal tubule structure. These microfluidic systems mimic the in vivo microenvironment including multi-compartmentalization and exposure to fluid shear stress. Increasing data supports that fluid shear stress impacts the phenotype and functionality of proximal tubule cultures, for which we provide an extensive background. In this review, we discuss recent developments of kidney-on-a-chip platforms with current and future applications. The improved proximal tubule functionality using 3D microfluidic systems is placed in perspective of investigating cellular signalling that can elucidate mechanistic aberrations involved in drug-induced kidney toxicity.
Collapse
Affiliation(s)
- Tom T G Nieskens
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Science, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Martijn J Wilmer
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Science, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
25
|
Miura T, Yokokawa R. Tissue culture on a chip: Developmental biology applications of self-organized capillary networks in microfluidic devices. Dev Growth Differ 2016; 58:505-15. [PMID: 27272910 DOI: 10.1111/dgd.12292] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Revised: 04/10/2016] [Accepted: 04/11/2016] [Indexed: 01/11/2023]
Abstract
Organ culture systems are used to elucidate the mechanisms of pattern formation in developmental biology. Various organ culture techniques have been used, but the lack of microcirculation in such cultures impedes the long-term maintenance of larger tissues. Recent advances in microfluidic devices now enable us to utilize self-organized perfusable capillary networks in organ cultures. In this review, we will overview past approaches to organ culture and current technical advances in microfluidic devices, and discuss possible applications of microfluidics towards the study of developmental biology.
Collapse
Affiliation(s)
- Takashi Miura
- Department of Anatomy and Cell Biology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-0054, Japan
| | - Ryuji Yokokawa
- Department of Micro Engineering, Kyoto University, Kyoto, 615-8540, Japan
| |
Collapse
|
26
|
Nishimura Y, Hsu HH, Wang PC. Detection of initial angiogenesis from dorsal aorta into metanephroi and elucidation of its role in kidney development. Regen Ther 2016; 4:27-35. [PMID: 31245485 PMCID: PMC6581801 DOI: 10.1016/j.reth.2016.01.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 12/13/2015] [Accepted: 01/06/2016] [Indexed: 12/13/2022] Open
Abstract
Reconstruction of blood vessels is considered the most difficult part for the complicated organs, therefore, blood vessel construction is regarded as a key point for kidney regeneration in vitro. Vasculogenesis and angiogenesis are the two mechanisms to form blood vessels in embryonic organs, and most studies resided in vaculogenesis. Angiogenesis resided mostly in adult diseases such as wound healing, growth of tumors, and psoriasis diseases. However, renal angiogenesis is simply attributed to the sprouting of pre-existing blood vessel from dorsal aorta into metanephroi, and its occurrence is considered to be at a late stage of metanephric development. Since no techniques are available for delicate detection, the initial angiogenesis from dorsal aorta into metanephroi as well as its role in kidney development still remained unclear. In this study, we developed a method to detect the initial angiogenesis of dorsal aorta into metanephroi, and firstly clarified that dorsal aorta angiogenesis occurred at an early stage of metanephric development. We also elucidated the role of dorsal aorta angiogenesis in promoting the early blood vessel formation, tubule formation and glomeruli maturation. It is suggested that blood flow and dynamic circulation of various factors at the early developing stage may be prerequisite to a successful construction of blood vessels in the complicated organs either in vitro or in vivo. These findings contribute to a better understanding of dorsal aorta angiogenesis during kidney development and shed light on its significant value for the application of tissue engineering to complicated organs.
Collapse
Affiliation(s)
| | | | - Pi-Chao Wang
- Division of Bioindustrial Sciences, Graduate School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba City, Ibaraki 305-8572, Japan
| |
Collapse
|
27
|
McClelland KS, Bowles J. Culturing murine embryonic organs: Pros, cons, tips and tricks. Differentiation 2016; 91:50-6. [DOI: 10.1016/j.diff.2016.01.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 01/17/2016] [Indexed: 11/26/2022]
|
28
|
Organ In Vitro Culture: What Have We Learned about Early Kidney Development? Stem Cells Int 2015; 2015:959807. [PMID: 26078765 PMCID: PMC4452498 DOI: 10.1155/2015/959807] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 01/07/2015] [Accepted: 01/08/2015] [Indexed: 12/15/2022] Open
Abstract
When Clifford Grobstein set out to study the inductive interaction between tissues in the developing embryo, he developed a method that remained important for the study of renal development until now. From the late 1950s on, in vitro cultivation of the metanephric kidney became a standard method. It provided an artificial environment that served as an open platform to study organogenesis. This review provides an introduction to the technique of organ culture, describes how the Grobstein assay and its variants have been used to study aspects of mesenchymal induction, and describes the search for natural and chemical inducers of the metanephric mesenchyme. The review also focuses on renal development, starting with ectopic budding of the ureteric bud, ureteric bud branching, and the generation of the nephron and presents the search for stem cells and renal progenitor cells that contribute to specific structures and tissues during renal development. It also presents the current use of Grobstein assay and its modifications in regenerative medicine and tissue engineering today. Together, this review highlights the importance of ex vivo kidney studies as a way to acquire new knowledge, which in the future can and will be implemented for developmental biology and regenerative medicine applications.
Collapse
|
29
|
Transport of organic anions and cations in murine embryonic kidney development and in serially-reaggregated engineered kidneys. Sci Rep 2015; 5:9092. [PMID: 25766625 PMCID: PMC4357899 DOI: 10.1038/srep09092] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 02/17/2015] [Indexed: 01/11/2023] Open
Abstract
Recent advances in renal tissue engineering have shown that dissociated, early renogenic tissue from the developing embryo can self-assemble into morphologically accurate kidney-like organs arranged around a central collecting duct tree. In order for such self-assembled kidneys to be useful therapeutically or as models for drug screening, it is necessary to demonstrate that they are functional. One of the main functional characteristics of mature kidneys is transport of organic anions and cations into and out of the proximal tubule. Here, we show that the transport function of embryonic kidneys allowed to develop in culture follows a developmental time-course that is comparable to embryonic kidney development in vivo. We also demonstrate that serially-reaggregated engineered kidneys can transport organic anions and cations through specific uptake and efflux channels. These results support the physiological relevance of kidneys grown in culture, a commonly used model for kidney development and research, and suggest that serially-reaggregated kidneys self-assembled from separated cells have some functional characteristics of intact kidneys.
Collapse
|
30
|
Bueters RRG, Kusters LJA, Klaasen A, van den Heuvel LP, Schreuder MF. Antibiotics and renal branching morphogenesis: comparison of toxicities. Pediatr Res 2014; 76:508-14. [PMID: 25167203 DOI: 10.1038/pr.2014.127] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 05/28/2014] [Indexed: 11/09/2022]
Abstract
BACKGROUND Many premature born neonates receive antibiotic drugs to treat infections, which are applied during active nephrogenesis. We studied the impact of clinical concentrations of gentamicin and alternatives, ceftazidime and meropenem, on ureteric branching. METHODS Mice metanephroi were dissected at embryonic day 13 and cultured in media with or without various concentrations of gentamicin, ceftazidime, or meropenem. Zero and 24 h kidney size were assessed by surface area measurements, and the ureteric tree was visualized by whole mount staining and confocal microscopy. Branching was evaluated by counting and gene expression levels of Wt1, Sox9, Bmp7, Fgf8, and Gdnf were investigated. RESULTS A concentration of 2,000 μmol/l ceftazidime impaired ureteric development. In addition, a 4.5-fold and a 2.5-fold downregulation was noted in Fgf8 and Gdnf, respectively. No adverse effects were noted after gentamicin or meropenem treatment. No relationship was noted between surface area expansion and ureteric bud formation, but surface area at explantation related to bud count after 24 h of culture. CONCLUSION Ceftazidime, but not gentamicin or meropenem reduced ureteric branching in mice and suggest a role for Fgf8 and Gdnf in its mechanism. Metanephros surface area measurements can be used to reduce intra- and inter-litter variation.
Collapse
Affiliation(s)
- Ruud R G Bueters
- Department of Pediatric Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lisanne J A Kusters
- Department of Pediatric Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Annelies Klaasen
- Department of Pediatric Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Michiel F Schreuder
- Department of Pediatric Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
31
|
The generation of kidney organoids by differentiation of human pluripotent cells to ureteric bud progenitor–like cells. Nat Protoc 2014; 9:2693-704. [DOI: 10.1038/nprot.2014.182] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
32
|
Lindström NO, Chang CH, Valerius MT, Hohenstein P, Davies JA. Node retraction during patterning of the urinary collecting duct system. J Anat 2014; 226:13-21. [PMID: 25292187 PMCID: PMC4299504 DOI: 10.1111/joa.12239] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2014] [Indexed: 11/28/2022] Open
Abstract
This report presents a novel mechanism for remodelling a branched epithelial tree. The mouse renal collecting duct develops by growth and repeated branching of an initially unbranched ureteric bud: this mechanism initially produces an almost fractal form with young branches connected to the centre of the kidney via a sequence of nodes (branch points) distributed widely throughout the developing organ. The collecting ducts of a mature kidney have a different form: from the nephrons in the renal cortex, long, straight lengths of collecting duct run almost parallel to one another through the renal medulla, and open together to the renal pelvis. Here we present time-lapse studies of E11.5 kidneys growing in culture: after about 5 days, the collecting duct trees show evidence of ‘node retraction’, in which the node of a ‘Y’-shaped branch moves downwards, shortening the stalk of the ‘Y’, lengthening its arms and narrowing their divergence angle so that the ‘Y’ becomes a ‘V’. Computer simulation suggests that node retraction can transform a spread tree, like that of an early kidney, into one with long, almost-parallel medullary rays similar to those seen in a mature real kidney.
Collapse
|
33
|
Davies JA, Chang CH, Lawrence ML, Mills CG, Mullins JJ. Engineered kidneys: principles, progress, and prospects. ACTA ACUST UNITED AC 2014. [DOI: 10.3402/arb.v1.24990] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
|
34
|
Blake J, Rosenblum ND. Renal branching morphogenesis: morphogenetic and signaling mechanisms. Semin Cell Dev Biol 2014; 36:2-12. [PMID: 25080023 DOI: 10.1016/j.semcdb.2014.07.011] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 07/14/2014] [Accepted: 07/17/2014] [Indexed: 12/28/2022]
Abstract
The human kidney is composed of an arborized network of collecting ducts, calyces and urinary pelvis that facilitate urine excretion and regulate urine composition. The renal collecting system is formed in utero, completed by the 34th week of gestation in humans, and dictates final nephron complement. The renal collecting system arises from the ureteric bud, a derivative of the intermediate-mesoderm derived nephric duct that responds to inductive signals from adjacent tissues via a process termed ureteric induction. The ureteric bud subsequently undergoes a series of iterative branching and remodeling events in a process called renal branching morphogenesis. Altered signaling that disrupts patterning of the nephric duct, ureteric induction, or renal branching morphogenesis leads to varied malformations of the renal collecting system collectively known as congenital anomalies of the kidney and urinary tract (CAKUT) and is the most frequently detected congenital renal aberration in infants. Here, we describe critical morphogenetic and cellular events that govern nephric duct specification, ureteric bud induction, renal branching morphogenesis, and cessation of renal branching morphogenesis. We also highlight salient molecular signaling pathways that govern these processes, and the investigative techniques used to interrogate them.
Collapse
Affiliation(s)
- Joshua Blake
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Canada; Department of Physiology, University of Toronto, Canada
| | - Norman D Rosenblum
- Division of Nephrology, Department of Paediatrics, The Hospital for Sick Children, Canada; Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Canada; Department of Physiology, University of Toronto, Canada.
| |
Collapse
|
35
|
Chen TL, Wang HL, Liu YH, Fang Y, Tan RZ, Zhou PH, Zhou Q, Lv XY. Highly effective ex vivo gene manipulation to study kidney development using self-complementary adenoassociated viruses. ScientificWorldJournal 2014; 2014:682189. [PMID: 25133251 PMCID: PMC4123690 DOI: 10.1155/2014/682189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 05/28/2014] [Accepted: 06/12/2014] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Ex vivo culture of intact embryonic kidney has become a powerful system for studying renal development. However, few methods have been available for gene manipulation and have impeded the identification and investigation of genes in this developmental process. RESULTS Here we systemically compared eight different serotypes of pseudotyped self-complementary adenoassociated viruses (scAAVs) transduction in cultured embryonic kidney with a modified culture procedure. We demonstrated that scAAV was highly effective in delivering genes into and expressing in compacted tissues. scAAV serotypes 2 and 8 exhibited higher efficiency of transduction compared to others. Expression kinetics assay revealed that scAAV can be used for gene manipulation at the study of UB branching and nephrogenesis. Repressing WT1 in cultured kidney using shRNA impairs tubule formation. We for the first time employed and validated scAAV as a gene delivery tool in cultured kidney. CONCLUSIONS These findings are expected to expedite the use of the ex vivo embryonic kidney cultures for kidney development research. For other ex vivo cultured organ models, scAAV could also be a promising tool for organogenesis study.
Collapse
Affiliation(s)
- Tie-Lin Chen
- Core Facility of Genetically Engineered Mice, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hong-Lian Wang
- Core Facility of Genetically Engineered Mice, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yun-Hong Liu
- Core Facility of Genetically Engineered Mice, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yin Fang
- Core Facility of Genetically Engineered Mice, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Rui-Zhi Tan
- Core Facility of Genetically Engineered Mice, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Pu-Hui Zhou
- College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Qin Zhou
- Core Facility of Genetically Engineered Mice, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiao-Yan Lv
- Departments of Dermatology, West China Hospital, Sichuan University, Sichuan 610041, China
| |
Collapse
|
36
|
Investigating the protective role of death receptor 3 (DR3) in renal injury using an organ culture model. Methods Mol Biol 2014; 1155:69-79. [PMID: 24788174 DOI: 10.1007/978-1-4939-0669-7_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
Death receptor 3 (DR3; also designated as Wsl-1, Apo3, LARD, TRAMP, TNFRSF25, and TR3) is a member of the tumor necrosis factor (TNF) receptor superfamily that has emerged as a major regulator of inflammation and autoimmune diseases. DR3 contains a homologous intracellular region called the death domain (DD) that can bind adaptor proteins, which also contain a DD, initiating cellular responses such as caspase activation and apoptotic cell death. However, in other circumstances DR3 can initiate induction of transcription genes and gene products that can prevent cell death from occurring. Our laboratory has reported an inducible expression of DR3 in human and mouse tubular epithelial cells in renal injury, but its function in these setting still remains unclear. To directly manipulate and evaluate the role of DR3 in vivo, I have used an in vitro organ culture (OC) model, which I have developed in our laboratory. In this chapter, I will describe in detail the OC model used to study the role of DR3 in renal injury and discuss its advantages and limitations. In my hands, the OC model has proven to be an efficient tool for studying human cell heterogeneity, basal and regulated receptor expression, signalling pathways, and various biological responses not readily achievable in traditional cell culture models. Various assays can be carried out on organ cultures including histology, biochemistry, cell biology, and molecular biology, which will not be described in detail in this chapter.
Collapse
|
37
|
ECM modulated early kidney development in embryonic organ culture. Biomaterials 2013; 34:6670-82. [DOI: 10.1016/j.biomaterials.2013.05.031] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 05/18/2013] [Indexed: 01/17/2023]
|
38
|
Chang CH, Davies JA. An improved method of renal tissue engineering, by combining renal dissociation and reaggregation with a low-volume culture technique, results in development of engineered kidneys complete with loops of Henle. Nephron Clin Pract 2012; 121:e79-85. [PMID: 23235540 DOI: 10.1159/000345514] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 10/23/2012] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Tissue engineering of functional kidney tissue is an important goal for clinical restoration of renal function in patients damaged by infectious, toxicological, or genetic disease. One promising approach is the use of the self-organizing abilities of embryonic kidney cells to arrange themselves, from a simply reaggregated cell suspension, into engineered organs similar to fetal kidneys. The previous state-of-the-art method for this results in the formation of a branched collecting duct tree, immature nephrons (S-shaped bodies) beside and connected to it, and supportive stroma. It does not, though, result in the significant formation of morphologically detectable loops of Henle - anatomical features of the nephron that are critical to physiological function. METHODS We have combined the best existing technique for renal tissue engineering from cell suspensions with a low-volume culture technique that allows intact kidney rudiments to make loops of Henle to test whether engineered kidneys can produce these loops. RESULTS The result is the formation of loops of Henle in engineered cultured 'fetal kidneys', very similar in both morphology and in number to those formed by intact organ rudiments. CONCLUSION This brings the engineering technique one important step closer to production of a fully realistic organ.
Collapse
Affiliation(s)
- C-Hong Chang
- University of Edinburgh Centre for Integrative Physiology, Edinburgh, UK.
| | | |
Collapse
|
39
|
Ofenbauer A, Sebinger DDR, Prewitz M, Gruber P, Werner C. Dewaxed ECM: A simple method for analyzing cell behaviour on decellularized extracellular matrices. J Tissue Eng Regen Med 2012; 9:1046-55. [PMID: 23172824 DOI: 10.1002/term.1658] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 08/11/2012] [Accepted: 10/21/2012] [Indexed: 01/20/2023]
Abstract
Decellularization techniques have been used on a wide variety of tissues to create cell-seedable scaffolds for tissue engineering. Finding a suitable decellularization protocol for a certain type of tissue can be laborious, especially when organ perfusion devices are needed. In this study, we report a quick and simple method for comparing decellularization protocols combining the use of paraffin slices and two-dimensional cell cultures. We developed three decellularization protocols for adult murine kidney that yielded decellularized extracellular matrices (ECMs) with varying histological properties. The resulting paraffin-embedded ECM slices were deparaffinized and reseeded with murine embryonic stem cells (mESCs). We analyzed cell attachment four days post seeding via determination of cell numbers, and used quantitative Real-Time PCR 13 days post seeding to measure gene expression levels of two genes associated with renal development, Pax2 and Pou3f3. The three decellularization protocols produced kidney-matrices that showed clearly distinguishable results. We demonstrated that formerly paraffin-embedded decellularized ECMs can effectively influence differentiation of stem cells. This method can be used to identify optimal decellularization protocols for recellularization of three-dimensional tissue-scaffolds with embryonic stem cells and other tissue-specific cell types.
Collapse
Affiliation(s)
- Andreas Ofenbauer
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials, Hohe Str. 6, 01069, Dresden, Germany
| | - David Daniel Raphael Sebinger
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials, Hohe Str. 6, 01069, Dresden, Germany
| | - Marina Prewitz
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials, Hohe Str. 6, 01069, Dresden, Germany
| | | | - Carsten Werner
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials, Hohe Str. 6, 01069, Dresden, Germany
| |
Collapse
|
40
|
Zhang X, Bush KT, Nigam SK. In vitro culture of embryonic kidney rudiments and isolated ureteric buds. Methods Mol Biol 2012; 886:13-21. [PMID: 22639247 DOI: 10.1007/978-1-61779-851-1_2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
In vitro culture of embryonic kidney rudiments has been utilized to study a variety of cellular processes and developmental mechanisms. Here, we describe two-dimensional (2D) culture of embryonic kidney rudiments on Transwell filters and three-dimensional (3D) cultures in collagen gels in detail, and 3D culture of isolated ureteric bud (UB) in Matrigel with BSN-conditioned media.
Collapse
Affiliation(s)
- Xing Zhang
- Pediatrics Department, University of California, San Diego, CA, USA
| | | | | |
Collapse
|
41
|
Kuzma-Kuzniarska M, Rak-Raszewska A, Kenny S, Edgar D, Wilm B, Fuente Mora C, Davies JA, Murray P. Integration potential of mouse and human bone marrow-derived mesenchymal stem cells. Differentiation 2011; 83:128-37. [PMID: 22364880 DOI: 10.1016/j.diff.2011.11.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Revised: 10/17/2011] [Accepted: 11/08/2011] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) are a multipotent cell population which has been described to exert renoprotective and regenerative effects in experimental models of kidney injury. Several lines of evidence indicate that MSCs also have the ability to contribute to nephrogenesis, suggesting that the cells can be employed in stem cell-based applications aimed at de novo renal tissue generation. In this study we re-evaluate the capacity of mouse and human bone marrow-derived MSCs to contribute to the development of renal tissue using a novel method of embryonic kidney culture. Although MSCs show expression of some genes involved in renal development, their contribution to nephrogenesis is very limited in comparison to other stem cell types tested. Furthermore, we found that both mouse and human MSCs have a detrimental effect on the ex vivo development of mouse embryonic kidney, this effect being mediated through a paracrine action. Stimulation with conditioned medium from a mouse renal progenitor population increases the ability of mouse MSCs to integrate into developing renal tissue and prevents the negative effects on kidney development, but does not appear to enhance their ability to undergo nephrogenesis.
Collapse
|
42
|
[The role of Pax2 in regulation of kidney development and kidney disease]. YI CHUAN = HEREDITAS 2011; 33:931-8. [PMID: 21951793 DOI: 10.3724/sp.j.1005.2011.00931] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Paired box2 (Pax2) gene plays a crucial role in kidney development and is expressed in the nephric duct, mesenchyme of pronephrons, mesonephrons, and metanephrons with special spatial and temporal characteristic. Research in animals indicate that Pax2 can interact with many important transcription factors such as Gdnf, Ret, SHH, Wnt4, and Fgf to organize the nephric linage specification, pro/mesonephric tubule formation and descent, emergence of the ureteric bud, branching morphogenesis, and nephron induction. Pax2 is associated with various congenital renal and ureter malformations, and the mutation is easist to detected in Renal-coloboma syndrome. In renal cell carcinoma, Wilms tumor and many acquired kidney diseases Pax2 is expressed abnormally, whose diagnose and therapy value will be the focus of further research. This paper reviews the molecular structure, expression and regulation of Pax2 in kidney development and diseases.
Collapse
|
43
|
Costantini F, Watanabe T, Lu B, Chi X, Srinivas S. Imaging kidney development. Cold Spring Harb Protoc 2011; 2011:pdb.top109. [PMID: 21536773 DOI: 10.1101/pdb.top109] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Development of the kidney involves interactions between several cell lineages and complex morphogenetic processes, such as branching of the ureteric bud (UB) to form the collecting duct system and condensation and differentiation of the mesenchymal progenitors to form the nephron epithelia. One of the advantages of the mouse kidney as an experimental system is that it can develop in culture, from the stage of initial branching of the UB (E11.5) for up to a week (although it achieves the size and degree of development of only an E13.5-E14.5 kidney in vivo). The availability of fluorescent proteins (FPs) has provided powerful tools for visualizing the morphogenesis of specific renal structures in organ cultures. Two categories of genetically modified mice that express FPs are useful for visualizing different cell lineages and developmental processes in these organ cultures: (1) transgenic mice that express a fluorescent reporter in the pattern of a specific gene; and (2) Cre reporter mice, which turn on an FP in cells with Cre recombinase activity (and their daughter cells), used in conjunction with cell type-specific Cre transgenic mice. Here, we describe some of the currently available Cre and FP transgenic lines that are useful for the study of kidney development.
Collapse
|
44
|
Ganeva V, Unbekandt M, Davies JA. An improved kidney dissociation and reaggregation culture system results in nephrons arranged organotypically around a single collecting duct system. Organogenesis 2011; 7:83-7. [PMID: 21386662 DOI: 10.4161/org.7.2.14881] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Methods for constructing engineered "tissues" from simple suspensions of cells are valuable for investigations into basic developmental biology and for tissue engineering. We recently published a method for producing embryonic renal tissues from suspensions of embryonic mouse renal cells. This method reproduced the anatomies and differentiation states of nephrons and stroma very well; it had the limitation, however, that what would, in normal development, be a single, highly branched collecting duct tree leading to a ureter developed, in the engineered system, as a multitude of very small collecting duct trees. These were isolated from each other and therefore would not be effective for draining urine to a common exit, were the tissue to be supplied with blood and physiologically active. Here, we report an improvement on the original method; it results in the formation of nephrons arranged around one single collecting duct tree as would happen in a normal kidney.
Collapse
Affiliation(s)
- Veronika Ganeva
- University of Edinburgh Centre for Integrative Physiology, UK
| | | | | |
Collapse
|
45
|
Ward HH, Romero E, Welford A, Pickett G, Bacallao R, Gattone VH, Ness SA, Wandinger-Ness A, Roitbak T. Adult human CD133/1(+) kidney cells isolated from papilla integrate into developing kidney tubules. Biochim Biophys Acta Mol Basis Dis 2011; 1812:1344-57. [PMID: 21255643 DOI: 10.1016/j.bbadis.2011.01.010] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Revised: 12/09/2010] [Accepted: 01/11/2011] [Indexed: 01/19/2023]
Abstract
Approximately 60,000 patients in the United States are waiting for a kidney transplant due to genetic, immunologic and environmentally caused kidney failure. Adult human renal stem cells could offer opportunities for autologous transplant and repair of damaged organs. Current data suggest that there are multiple progenitor types in the kidney with distinct localizations. In the present study, we characterize cells derived from human kidney papilla and show their capacity for tubulogenesis. In situ, nestin(+) and CD133/1(+) cells were found extensively intercalated between tubular epithelia in the loops of Henle of renal papilla, but not of the cortex. Populations of primary cells from the renal cortex and renal papilla were isolated by enzymatic digestion from human kidneys unsuited for transplant and immuno-enriched for CD133/1(+) cells. Isolated CD133/1(+) papillary cells were positive for nestin, as well as several human embryonic stem cell markers (SSEA4, Nanog, SOX2, and OCT4/POU5F1) and could be triggered to adopt tubular epithelial and neuronal-like phenotypes. Isolated papillary cells exhibited morphologic plasticity upon modulation of culture conditions and inhibition of asymmetric cell division. Labeled papillary cells readily associated with cortical tubular epithelia in co-culture and 3-dimensional collagen gel cultures. Heterologous organ culture demonstrated that CD133/1(+) progenitors from the papilla and cortex became integrated into developing kidney tubules. Tubular epithelia did not participate in tubulogenesis. Human renal papilla harbor cells with the hallmarks of adult kidney stem/progenitor cells that can be amplified and phenotypically modulated in culture while retaining the capacity to form new kidney tubules. This article is part of a Special Issue entitled: Polycystic Kidney Disease.
Collapse
Affiliation(s)
- Heather H Ward
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|