1
|
Oliveira M, Sarker PP, Skovorodkin I, Kalantarifard A, Haskavuk T, Mac Intyre J, Nallukunnel Raju E, Nooranian S, Shioda H, Nishikawa M, Sakai Y, Vainio SJ, Elbuken C, Raykhel I. From ex ovo to in vitro: xenotransplantation and vascularization of mouse embryonic kidneys in a microfluidic chip. LAB ON A CHIP 2024; 24:4816-4826. [PMID: 39290081 PMCID: PMC11408908 DOI: 10.1039/d4lc00547c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 09/01/2024] [Indexed: 09/19/2024]
Abstract
Organoids are emerging as a powerful tool to investigate complex biological structures in vitro. Vascularization of organoids is crucial to recapitulate the morphology and function of the represented human organ, especially in the case of the kidney, whose primary function of blood filtration is closely associated with blood circulation. Current in vitro microfluidic approaches have only provided initial vascularization of kidney organoids, whereas in vivo transplantation to animal models is problematic due to ethical problems, with the exception of xenotransplantation onto a chicken chorioallantoic membrane (CAM). Although CAM can serve as a good environment for vascularization, it can only be used for a fixed length of time, limited by development of the embryo. Here, we propose a novel lab on a chip design that allows organoids of different origin to be cultured and vascularized on a CAM, as well as to be transferred to in vitro conditions when required. Mouse embryonic kidneys cultured on the CAM showed enhanced vascularization by intrinsic endothelial cells, and made connections with the chicken vasculature, as evidenced by blood flowing through them. After the chips were transferred to in vitro conditions, the vasculature inside the organoids was successfully maintained. To our knowledge, this is the first demonstration of the combination of in vivo and in vitro approaches applied to microfluidic chip design.
Collapse
Affiliation(s)
- Micaela Oliveira
- Microfluidics and Biosensor Research Group, Disease Networks Research Unit, Department of Biochemistry and Molecular Medicine, University of Oulu, Finland.
| | - Partha Protim Sarker
- Microfluidics and Biosensor Research Group, Disease Networks Research Unit, Department of Biochemistry and Molecular Medicine, University of Oulu, Finland.
- Developmental Biology Laboratory, Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.
| | - Ilya Skovorodkin
- Microfluidics and Biosensor Research Group, Disease Networks Research Unit, Department of Biochemistry and Molecular Medicine, University of Oulu, Finland.
- Developmental Biology Laboratory, Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.
| | - Ali Kalantarifard
- Microfluidics and Biosensor Research Group, Disease Networks Research Unit, Department of Biochemistry and Molecular Medicine, University of Oulu, Finland.
| | - Tugce Haskavuk
- Microfluidics and Biosensor Research Group, Disease Networks Research Unit, Department of Biochemistry and Molecular Medicine, University of Oulu, Finland.
- Developmental Biology Laboratory, Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.
| | - Jonatan Mac Intyre
- Microfluidics and Biosensor Research Group, Disease Networks Research Unit, Department of Biochemistry and Molecular Medicine, University of Oulu, Finland.
| | - Elizabath Nallukunnel Raju
- Microfluidics and Biosensor Research Group, Disease Networks Research Unit, Department of Biochemistry and Molecular Medicine, University of Oulu, Finland.
| | - Samin Nooranian
- Microfluidics and Biosensor Research Group, Disease Networks Research Unit, Department of Biochemistry and Molecular Medicine, University of Oulu, Finland.
| | - Hiroki Shioda
- Laboratory of Organs and Biosystems Engineering, Department of Chemical System Engineering, University of Tokyo, Tokyo, Japan
| | - Masaki Nishikawa
- Laboratory of Organs and Biosystems Engineering, Department of Chemical System Engineering, University of Tokyo, Tokyo, Japan
| | - Yasuyuki Sakai
- Laboratory of Organs and Biosystems Engineering, Department of Chemical System Engineering, University of Tokyo, Tokyo, Japan
| | - Seppo J Vainio
- Developmental Biology Laboratory, Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.
- Infotech Oulu, University of Oulu, Oulu, Finland
- Kvantum Institute, University of Oulu, Oulu, Finland
| | - Caglar Elbuken
- Microfluidics and Biosensor Research Group, Disease Networks Research Unit, Department of Biochemistry and Molecular Medicine, University of Oulu, Finland.
- VTT Technical Research Centre of Finland Ltd., Finland
| | - Irina Raykhel
- Developmental Biology Laboratory, Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.
- Laboratory of Organs and Biosystems Engineering, Department of Chemical System Engineering, University of Tokyo, Tokyo, Japan
| |
Collapse
|
2
|
Yoon B, Kim H, Jung SW, Park J. Single-cell lineage tracing approaches to track kidney cell development and maintenance. Kidney Int 2024; 105:1186-1199. [PMID: 38554991 DOI: 10.1016/j.kint.2024.01.045] [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: 09/08/2023] [Revised: 12/06/2023] [Accepted: 01/09/2024] [Indexed: 04/02/2024]
Abstract
The kidney is a complex organ consisting of various cell types. Previous studies have aimed to elucidate the cellular relationships among these cell types in developing and mature kidneys using Cre-loxP-based lineage tracing. However, this methodology falls short of fully capturing the heterogeneous nature of the kidney, making it less than ideal for comprehensively tracing cellular progression during kidney development and maintenance. Recent technological advancements in single-cell genomics have revolutionized lineage tracing methods. Single-cell lineage tracing enables the simultaneous tracing of multiple cell types within complex tissues and their transcriptomic profiles, thereby allowing the reconstruction of their lineage tree with cell state information. Although single-cell lineage tracing has been successfully applied to investigate cellular hierarchies in various organs and tissues, its application in kidney research is currently lacking. This review comprehensively consolidates the single-cell lineage tracing methods, divided into 4 categories (clustered regularly interspaced short palindromic repeat [CRISPR]/CRISPR-associated protein 9 [Cas9]-based, transposon-based, Polylox-based, and native barcoding methods), and outlines their technical advantages and disadvantages. Furthermore, we propose potential future research topics in kidney research that could benefit from single-cell lineage tracing and suggest suitable technical strategies to apply to these topics.
Collapse
Affiliation(s)
- Baul Yoon
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Hayoung Kim
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Su Woong Jung
- Division of Nephrology, Department of Internal Medicine, College of Medicine, Kyung Hee University, Seoul, Republic of Korea; Division of Nephrology, Department of Internal Medicine, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea.
| | - Jihwan Park
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea.
| |
Collapse
|
3
|
Creating a kidney organoid-vasculature interaction model using a novel organ-on-chip system. Sci Rep 2022; 12:20699. [PMID: 36450835 PMCID: PMC9712653 DOI: 10.1038/s41598-022-24945-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 11/22/2022] [Indexed: 12/02/2022] Open
Abstract
Kidney organoids derived from human induced pluripotent stem cells (iPSCs) have proven to be a valuable tool to study kidney development and disease. However, the lack of vascularization of these organoids often leads to insufficient oxygen and nutrient supply. Vascularization has previously been achieved by implantation into animal models, however, the vasculature arises largely from animal host tissue. Our aim is to transition from an in vivo implantation model towards an in vitro model that fulfils the advantages of vascularization whilst being fully human-cell derived. Our chip system supported culturing of kidney organoids, which presented nephron structures. We also showed that organoids cultured on chip showed increased maturation of endothelial populations based on a colocalization analysis of endothelial markers. Moreover, we observed migration and proliferation of human umbilical vein endothelial cells (HUVECs) cultured in the channels of the chip inside the organoid tissue, where these HUVECs interconnected with endogenous endothelial cells and formed structures presenting an open lumen resembling vessels. Our results establish for the first-time vascularization of kidney organoids in HUVEC co-culture conditions using a microfluidic organ-on-chip. Our model therefore provides a useful insight into kidney organoid vascularization in vitro and presents a tool for further studies of kidney development and drug testing, both for research purposes and pre-clinical applications.
Collapse
|
4
|
Potential Similarities in Sex Difference in Key Genes and Their Expression, Network, EQTL and Pathways between COVID-19 and Chronic Kidney Disease Based on Mouse Model. J Pers Med 2022; 12:jpm12071190. [PMID: 35887687 PMCID: PMC9323909 DOI: 10.3390/jpm12071190] [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: 04/18/2022] [Revised: 07/13/2022] [Accepted: 07/13/2022] [Indexed: 11/16/2022] Open
Abstract
COVID-19 and chronic kidney disease (CKD) share similarity in sex bias and key genes in the disease pathway of sex difference. We investigated the sex difference of molecular pathways of four key players of these two diseases using an existing large set of whole genome expression profiles from the kidneys of female and male mouse models. Our data show that there is little to no correlation at the whole genome expression level between female and male mice among these four genes. There are considerable sex differences among genes in upstream regulation, Ace2 complex interaction, and downstream pathways. Snap25 and Plcb4 may play important roles in the regulation of the expression level of Adam17, Tmprss2, and Cd146 in females. In males, Adh4 is a candidate gene for the regulation of Adam17, while Asl, Auts2, and Rabger1 are candidates for Tmprss2. Within the Ace2 complex, Cd146 directly influences the expression level of Adam17 and Ace2 in the female, while in the male Adam potentially has a stronger influence on Ace2 than that of Tmprss2. Among the top 100 most related genes, only one or two genes from four key genes and 11 from the control B-Actin were found to be the same between sexes. Among the top 10 sets of genes in the downstream pathway of Ace2, only two sets are the same between the sexes. We concluded that these known key genes and novel genes in CKD may play significant roles in the sex difference in the CKD and COVID-19 disease pathways.
Collapse
|
5
|
Xue B, Wang P, Yu W, Feng J, Li J, Zhao R, Yang Z, Yan X, Duan H. CD146 as a promising therapeutic target for retinal and choroidal neovascularization diseases. SCIENCE CHINA. LIFE SCIENCES 2022; 65:1157-1170. [PMID: 34729700 DOI: 10.1007/s11427-021-2020-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 10/20/2021] [Indexed: 11/26/2022]
Abstract
Blood vessel dysfunction causes several retinal diseases, including diabetic retinopathy, familial exudative vitreoretinopathy, macular degeneration and choroidal neovascularization in pathological myopia. Vascular endothelial growth factor (VEGF)-neutralizing proteins provide benefits in most of those diseases, yet unsolved haemorrhage and frequent intraocular injections still bothered patients. Here, we identified endothelial CD146 as a new target for retinal diseases. CD146 expression was activated in two ocular pathological angiogenesis models, a laser-induced choroid neovascularization model and an oxygen-induced retinopathy model. The absence of CD146 impaired hypoxia-induced cell migration and angiogenesis both in cell lines and animal model. Preventive or therapeutic treatment with anti-CD146 antibody AA98 significantly inhibited hypoxia-induced aberrant retinal angiogenesis in two retinal disease models. Mechanistically, under hypoxia condition, CD146 was involved in the activation of NFκB, Erk and Akt signalling pathways, which are partially independent of VEGF. Consistently, anti-CD146 therapy combined with anti-VEGF therapy showed enhanced impairment effect of hypoxia-induced angiogenesis in vitro and in vivo. Given the critical role of abnormal angiogenesis in retinal and choroidal diseases, our results provide novel insights into combinatorial therapy for neovascular fundus diseases.
Collapse
Affiliation(s)
- Bai Xue
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Ping Wang
- Key Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Wenzhen Yu
- Department of Ophthalmology, People's Hospital, Peking University, Beijing, 100044, China
| | - Jing Feng
- Key Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jie Li
- Department of Ophthalmology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Rulian Zhao
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Zhenglin Yang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China.
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, 610072, China.
| | - Xiyun Yan
- Key Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- Nanozyme Medical Center, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Hongxia Duan
- Key Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
6
|
Sharma A, Joshkon A, Ladjimi A, Traboulsi W, Bachelier R, Robert S, Foucault-Bertaud A, Leroyer AS, Bardin N, Somasundaram I, Blot-Chabaud M. Soluble CD146 as a Potential Target for Preventing Triple Negative Breast Cancer MDA-MB-231 Cell Growth and Dissemination. Int J Mol Sci 2022; 23:ijms23020974. [PMID: 35055160 PMCID: PMC8780963 DOI: 10.3390/ijms23020974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/07/2022] [Accepted: 01/08/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Triple Negative Breast Cancers (TNBC) are the most aggressive breast cancers and lead to poor prognoses. This is due to a high resistance to therapies, mainly because of the presence of Cancer Stem Cells (CSCs). Plasticity, a feature of CSCs, is acquired through the Epithelial to Mesenchymal Transition (EMT), a process that has been recently shown to be regulated by a key molecule, CD146. Of interest, CD146 is over-expressed in TNBC. Methods: The MDA-MB-231 TNBC cell line was used as a model to study the role of CD146 and its secreted soluble form (sCD146) in the development and dissemination of TNBC using in vitro and in vivo studies. Results: High expression of CD146 in a majority of MDA-MB-231 cells leads to an increased secretion of sCD146 that up-regulates the expression of EMT and CSC markers on the cells. These effects can be blocked with a specific anti-sCD146 antibody, M2J-1 mAb. M2J-1 mAb was able to reduce tumour development and dissemination in a model of cells xenografted in nude mice and an experimental model of metastasis, respectively, in part through its effects on CSC. Conclusion: We propose that M2J-1 mAb could be used as an additional therapeutic approach to fight TNBC.
Collapse
Affiliation(s)
- Akshita Sharma
- Department of Stem Cell and Regenerative Medicine, D.Y. Patil Universit, Kolhapur 416003, India; (A.S.); (I.S.)
| | - Ahmad Joshkon
- Faculty of Pharmacy, Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, 13005 Marseille, France; (A.J.); (A.L.); (W.T.); (R.B.); (S.R.); (A.F.-B.); (A.S.L.); (N.B.)
| | - Aymen Ladjimi
- Faculty of Pharmacy, Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, 13005 Marseille, France; (A.J.); (A.L.); (W.T.); (R.B.); (S.R.); (A.F.-B.); (A.S.L.); (N.B.)
| | - Waël Traboulsi
- Faculty of Pharmacy, Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, 13005 Marseille, France; (A.J.); (A.L.); (W.T.); (R.B.); (S.R.); (A.F.-B.); (A.S.L.); (N.B.)
| | - Richard Bachelier
- Faculty of Pharmacy, Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, 13005 Marseille, France; (A.J.); (A.L.); (W.T.); (R.B.); (S.R.); (A.F.-B.); (A.S.L.); (N.B.)
| | - Stéphane Robert
- Faculty of Pharmacy, Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, 13005 Marseille, France; (A.J.); (A.L.); (W.T.); (R.B.); (S.R.); (A.F.-B.); (A.S.L.); (N.B.)
| | - Alexandrine Foucault-Bertaud
- Faculty of Pharmacy, Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, 13005 Marseille, France; (A.J.); (A.L.); (W.T.); (R.B.); (S.R.); (A.F.-B.); (A.S.L.); (N.B.)
| | - Aurélie S. Leroyer
- Faculty of Pharmacy, Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, 13005 Marseille, France; (A.J.); (A.L.); (W.T.); (R.B.); (S.R.); (A.F.-B.); (A.S.L.); (N.B.)
| | - Nathalie Bardin
- Faculty of Pharmacy, Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, 13005 Marseille, France; (A.J.); (A.L.); (W.T.); (R.B.); (S.R.); (A.F.-B.); (A.S.L.); (N.B.)
| | - Indumathi Somasundaram
- Department of Stem Cell and Regenerative Medicine, D.Y. Patil Universit, Kolhapur 416003, India; (A.S.); (I.S.)
| | - Marcel Blot-Chabaud
- Faculty of Pharmacy, Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, 13005 Marseille, France; (A.J.); (A.L.); (W.T.); (R.B.); (S.R.); (A.F.-B.); (A.S.L.); (N.B.)
- Correspondence: ; Tel.: +33-4-91-83-56-85
| |
Collapse
|
7
|
Little MH, Howden SE, Lawlor KT, Vanslambrouck JM. Determining lineage relationships in kidney development and disease. Nat Rev Nephrol 2021; 18:8-21. [PMID: 34594045 DOI: 10.1038/s41581-021-00485-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2021] [Indexed: 12/17/2022]
Abstract
The lineage relationships of cells provide information about the origins of component cell types during development and repair as well as the source of aberrant cells during disease. Genetic approaches to lineage tracing applied in the mouse have revealed much about how the mammalian kidney forms, including the identification of key progenitors for the nephrons and stromal compartments. Inducible Cre systems have also facilitated lineage tracing studies in the postnatal animal that illustrate the changes in cellular fate that can occur during kidney injury. With the advent of single-cell transcriptional profiling and trajectory analyses, predictions of cellular relationships across development are now being made in model systems, such as the mouse, as well as in human fetal kidney. Importantly, these approaches provide predictions of lineage relationships rather than definitive evidence. Although genetic approaches to the study of lineage have not previously been possible in a human setting, the application of CRISPR-Cas9 gene editing of pluripotent stem cells is beginning to teach us about human lineage relationships.
Collapse
Affiliation(s)
- Melissa H Little
- Murdoch Children's Research Institute, Parkville, VIC, Australia. .,Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia. .,Department of Anatomy and Neuroscience, The University of Melbourne, Melbourne, VIC, Australia.
| | - Sara E Howden
- Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia
| | - Kynan T Lawlor
- Murdoch Children's Research Institute, Parkville, VIC, Australia
| | | |
Collapse
|
8
|
Rak-Raszewska A, Reint G, Geiger F, Naillat F, Vainio SJ. Deciphering the minimal quantity of mouse primary cells to undergo nephrogenesis ex vivo. Dev Dyn 2021; 251:536-550. [PMID: 34494340 DOI: 10.1002/dvdy.418] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 08/24/2021] [Accepted: 08/29/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Tissue organoids derived from primary cells have high potential for studying organ development and diseases in numerous organs. They recreate the morphological structure and mimic the functions of given organ while being compact in size, easy to produce, and suitable for use in various experimental setups. RESULTS In this study we established the number of cells that form mouse kidney rudiments at E11.5, and generated renal organoids of various sizes from the mouse primary cells of the metanephric mesenchyme (MM). We investigated the ability of renal organoids to undergo nephrogenesis upon Wnt/ β-catenin pathway-mediated tubule induction with a GSK-3 inhibitor (BIO) or by initiation through the ureteric bud (UB). We found that 5000 cells of MM cells are necessary to successfully form renal organoids with well-structured nephrons as judged by fluorescent microscopy, transmission electron microscopy (TEM), and quantitative Polymerase Chain Reaction (qPCR). These mouse organoids also recapitulated renal secretion function in the proximal tubules. CONCLUSIONS We show that a significant decrease of cells used to generate renal mouse organoids in a dissociation/re-aggregation assay, does not interfere with development, and goes toward 3Rs. This enables generation of more experimental samples with one mouse litter, limiting the number of animals used for studies.
Collapse
Affiliation(s)
- Aleksandra Rak-Raszewska
- Laboratory of Developmental Biology, Disease Networks Researtch Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Ganna Reint
- Laboratory of Developmental Biology, Disease Networks Researtch Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Fabienne Geiger
- Laboratory of Developmental Biology, Disease Networks Researtch Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Florence Naillat
- Laboratory of Developmental Biology, Disease Networks Researtch Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Seppo J Vainio
- Laboratory of Developmental Biology, Disease Networks Researtch Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.,Kvantum Institute, Infotech Oulu, University of Oulu, Oulu, Finland
| |
Collapse
|
9
|
D’Arcy C, Kiel C. Cell Adhesion Molecules in Normal Skin and Melanoma. Biomolecules 2021; 11:biom11081213. [PMID: 34439879 PMCID: PMC8391223 DOI: 10.3390/biom11081213] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 12/11/2022] Open
Abstract
Cell adhesion molecules (CAMs) of the cadherin, integrin, immunoglobulin, and selectin protein families are indispensable for the formation and maintenance of multicellular tissues, especially epithelia. In the epidermis, they are involved in cell–cell contacts and in cellular interactions with the extracellular matrix (ECM), thereby contributing to the structural integrity and barrier formation of the skin. Bulk and single cell RNA sequencing data show that >170 CAMs are expressed in the healthy human skin, with high expression levels in melanocytes, keratinocytes, endothelial, and smooth muscle cells. Alterations in expression levels of CAMs are involved in melanoma propagation, interaction with the microenvironment, and metastasis. Recent mechanistic analyses together with protein and gene expression data provide a better picture of the role of CAMs in the context of skin physiology and melanoma. Here, we review progress in the field and discuss molecular mechanisms in light of gene expression profiles, including recent single cell RNA expression information. We highlight key adhesion molecules in melanoma, which can guide the identification of pathways and strategies for novel anti-melanoma therapies.
Collapse
|
10
|
Li Q, Chen J, Yin M, Zhao J, Lu F, Wang Z, Yu X, Wang S, Zheng D, Wang H. High Level of Soluble CD146 In Cerebrospinal Fluid Might be a Biomarker of Severity of Anti-N-Methyl-D-Aspartate Receptor Encephalitis. Front Immunol 2021; 12:680424. [PMID: 34220828 PMCID: PMC8245058 DOI: 10.3389/fimmu.2021.680424] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 05/26/2021] [Indexed: 11/23/2022] Open
Abstract
Background Disruption of the blood–brain barrier (BBB) is an important pathophysiological process of anti-N-methyl-D-aspartate receptor (anti-NMDAR) encephalitis. A recent multi-center study showed that soluble (s) CD146 is a potential biomarker for monitoring early BBB damage and central nervous system inflammation, but little is known about sCD146 in anti-NMDAR encephalitis. Method Twenty-three anti-NMDAR encephalitis patients and seventeen controls with non-inflammatory neurological diseases were recruited. sCD146 and inflammatory cytokines in cerebrospinal fluid (CSF) and serum were detected by ELISA. Modified Rankin scale (mRS) scores were used to assess the neurological status of each patient. A follow-up review was completed three months after discharge. Results sCD146 levels in the CSF of patients with the acute stage anti-NMDAR encephalitis were significantly increased compared with controls and accompanied by increases in TNF-α, IL-6 and IL-10. CSF sCD146 was positively correlated with neuroinflammatory factors in the CSF and with mRS score. Three months after effective treatment, CSF sCD146 in patients was significantly decreased but remained significantly different compared with the controls. Conclusion Our data suggested that higher expression of CSF sCD146 correlated with more serious neurological damage. Therefore, levels of CSF sCD146 may represent a promising indicator for monitoring disease and optimizing clinical treatment decisions in the early stages of anti-NMDAR encephalitis.
Collapse
Affiliation(s)
- Qing Li
- Department of Geriatric Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Jinglong Chen
- Department of Geriatric Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Mengzhuo Yin
- Department of Geriatric Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Jun Zhao
- Department of Geriatric Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Fuchang Lu
- Department of Geriatric Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Zhanhang Wang
- Department of Neurology, Guangdong 999 Brain Hospital, Guangzhou, China
| | - Xiaoqi Yu
- Department of Geriatric Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Shuangyan Wang
- Department of Geriatric Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Dong Zheng
- Department of Neurology, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Honghao Wang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
11
|
Chen T, Ye B, Tan J, Yang H, He F, Khalil RA. CD146+Mesenchymal stem cells treatment improves vascularization, muscle contraction and VEGF expression, and reduces apoptosis in rat ischemic hind limb. Biochem Pharmacol 2021; 190:114530. [PMID: 33891966 DOI: 10.1016/j.bcp.2021.114530] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 03/12/2021] [Accepted: 03/17/2021] [Indexed: 01/09/2023]
Abstract
Peripheral arterial disease (PAD) is an increasingly common narrowing of the peripheral arteries that can lead to lower limb ischemia, muscle weakness and gangrene. Surgical vein or arterial grafts could improve PAD, but may not be suitable in elderly patients, prompting research into less invasive approaches. Mesenchymal stem cells (MSCs) have been proposed as potential therapy, but their effectiveness and underlying mechanisms in limb ischemia are unclear. We tested the hypothesis that treatment with naive MSCs (nMSCs) or MSCs expressing CD146 (CD146+MSCs) could improve vascularity and muscle function in rat model of hind-limb ischemia. Sixteen month old Sprague-Dawley rats were randomly assigned to 4 groups: sham-operated control, ischemia, ischemia + nMSCs and ischemia+CD146+MSCs. After 4 weeks of respective treatment, rat groups were assessed for ischemic clinical score, Tarlov score, muscle capillary density, TUNEL apoptosis assay, contractile force, and vascular endothelial growth factor (VEGF) mRNA expression. CD146+MSCs showed greater CD146 mRNA expression than nMSCs. Treatment with nMSCs or CD146+MSCs improved clinical and Tarlov scores, muscle capillary density, contractile force and VEGF mRNA expression in ischemic limbs as compared to non-treated ischemia group. The improvements in muscle vascularity and function were particularly greater in ischemia+CD146+MSCs than ischemia + nMSCs group. TUNEL positive apoptotic cells were least abundant in ischemia+CD146+MSCs compared with ischemia + nMSCs and non-treated ischemia groups. Thus, MSCs particularly those expressing CD146 improve vascularity, muscle function and VEGF expression and reduce apoptosis in rat ischemic limb, and could represent a promising approach to improve angiogenesis and muscle function in PAD.
Collapse
Affiliation(s)
- Tao Chen
- Department of Vascular Surgery, Ganzhou People's Hospital, the Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, Jiangxi, China; Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States.
| | - Bo Ye
- Department of Vascular Surgery, Ganzhou People's Hospital, the Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, Jiangxi, China
| | - Jing Tan
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Haifeng Yang
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Faming He
- Department of Vascular Surgery, Ganzhou People's Hospital, the Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, Jiangxi, China
| | - Raouf A Khalil
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
12
|
Abstract
Renin cells are essential for survival perfected throughout evolution to ensure normal development and defend the organism against a variety of homeostatic threats. During embryonic and early postnatal life, they are progenitors that participate in the morphogenesis of the renal arterial tree. In adult life, they are capable of regenerating injured glomeruli, control blood pressure, fluid-electrolyte balance, tissue perfusion, and in turn, the delivery of oxygen and nutrients to cells. Throughout life, renin cell descendants retain the plasticity or memory to regain the renin phenotype when homeostasis is threatened. To perform all of these functions and maintain well-being, renin cells must regulate their identity and fate. Here, we review the major mechanisms that control the differentiation and fate of renin cells, the chromatin events that control the memory of the renin phenotype, and the major pathways that determine their plasticity. We also examine how chronic stimulation of renin cells alters their fate leading to the development of a severe and concentric hypertrophy of the intrarenal arteries and arterioles. Lastly, we provide examples of additional changes in renin cell fate that contribute to equally severe kidney disorders.
Collapse
Affiliation(s)
- Maria Luisa S. Sequeira-Lopez
- Departments of Pediatrics an Biology, Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| | - R. Ariel Gomez
- Departments of Pediatrics an Biology, Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| |
Collapse
|
13
|
Dembele M, Delafosse M, Yousfi N, Debiec H, Ngo K, Plaisier E, Ronco P, Perry G. [Models of glomerular filtration barrier : New developments]. Med Sci (Paris) 2021; 37:242-248. [PMID: 33739271 DOI: 10.1051/medsci/2021010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
In this article, we present the latest innovations to generate in vitro models of the glomerular filtration barrier. There is currently a growing interest for such model systems that allow to reduce the use of animal models. Methodologies to improve their physiological relevance have taken advantage of the development of induced pluripotent stem cells and of bioengineering, particularly tissue engineering. Here, we first introduce the methods to overcome the limitations of the currently used glomerular cells based on the use of stem cells. The different approaches to obtain podocytes, the most important cells in the glomerulus, are presented. Finally, we emphasize the importance of the glomerular microenvironment in maintaining the glomerular cell phenotype, which can be achieved by co-culturing different glomerular cells, integration of biomaterials mimicking the extracellular matrix and introduction of flows with microfluidics.
Collapse
Affiliation(s)
- Mahamadou Dembele
- Inserm, Sorbonne Université, Maladies rénales fréqunentes et rares (CoRaKiD), UMRS 1155, Hôpital Tenon, Bâtiment recherche, 4 rue de la Chine, 75020 Paris, France
| | - Marion Delafosse
- Inserm, Sorbonne Université, Maladies rénales fréqunentes et rares (CoRaKiD), UMRS 1155, Hôpital Tenon, Bâtiment recherche, 4 rue de la Chine, 75020 Paris, France
| | - Nadhir Yousfi
- Inserm, Sorbonne Université, Maladies rénales fréqunentes et rares (CoRaKiD), UMRS 1155, Hôpital Tenon, Bâtiment recherche, 4 rue de la Chine, 75020 Paris, France
| | - Hanna Debiec
- Inserm, Sorbonne Université, Maladies rénales fréqunentes et rares (CoRaKiD), UMRS 1155, Hôpital Tenon, Bâtiment recherche, 4 rue de la Chine, 75020 Paris, France
| | - Kieu Ngo
- Sorbonne Université, CNRS, Laboratoire interfaces et systèmes électrochimiques, LISE, Campus Pierre et Marie Curie, 4 place Jussieu 75252 Paris, France
| | - Emmanuelle Plaisier
- Inserm, Sorbonne Université, Maladies rénales fréqunentes et rares (CoRaKiD), UMRS 1155, Hôpital Tenon, Bâtiment recherche, 4 rue de la Chine, 75020 Paris, France
| | - Pierre Ronco
- Inserm, Sorbonne Université, Maladies rénales fréqunentes et rares (CoRaKiD), UMRS 1155, Hôpital Tenon, Bâtiment recherche, 4 rue de la Chine, 75020 Paris, France
| | - Guillaume Perry
- Inserm, Sorbonne Université, Maladies rénales fréqunentes et rares (CoRaKiD), UMRS 1155, Hôpital Tenon, Bâtiment recherche, 4 rue de la Chine, 75020 Paris, France - CNRS, Sorbonne université, Laboratoire de génie électrique et électronique de Paris, GeePs, Campus Pierre et Marie Curie, 4 Place Jussieu 75252 Paris, France
| |
Collapse
|
14
|
Abed A, Leroyer AS, Kavvadas P, Authier F, Bachelier R, Foucault-Bertaud A, Bardin N, Cohen CD, Lindenmeyer MT, Genest M, Joshkon A, Jourde-Chiche N, Burtey S, Blot-Chabaud M, Dignat-George F, Chadjichristos CE. Endothelial-Specific Deletion of CD146 Protects Against Experimental Glomerulonephritis in Mice. Hypertension 2021; 77:1260-1272. [PMID: 33689459 DOI: 10.1161/hypertensionaha.119.14176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Ahmed Abed
- From the INSERM UMR-S1155, Tenon Hospital, Paris, France (A.A., P.K., F.A., M.G., C.E.C.).,Sorbonne Université, Paris, France (A.A., C.E.C.)
| | - Aurélie S Leroyer
- Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, France (A.S.L., R.B., A.F.-B., N.B., A.J., N.J.-C., S.B., M.B.-C., F.D.-G.)
| | - Panagiotis Kavvadas
- From the INSERM UMR-S1155, Tenon Hospital, Paris, France (A.A., P.K., F.A., M.G., C.E.C.)
| | - Florence Authier
- From the INSERM UMR-S1155, Tenon Hospital, Paris, France (A.A., P.K., F.A., M.G., C.E.C.)
| | - Richard Bachelier
- Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, France (A.S.L., R.B., A.F.-B., N.B., A.J., N.J.-C., S.B., M.B.-C., F.D.-G.)
| | - Alexandrine Foucault-Bertaud
- Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, France (A.S.L., R.B., A.F.-B., N.B., A.J., N.J.-C., S.B., M.B.-C., F.D.-G.)
| | - Nathalie Bardin
- Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, France (A.S.L., R.B., A.F.-B., N.B., A.J., N.J.-C., S.B., M.B.-C., F.D.-G.)
| | - Clemens D Cohen
- Nephrological Center, Medical Clinic and Policlinic IV, University of Munich, Germany (C.D.C.)
| | - Maja T Lindenmeyer
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Germany (M.T.L.)
| | - Magali Genest
- From the INSERM UMR-S1155, Tenon Hospital, Paris, France (A.A., P.K., F.A., M.G., C.E.C.)
| | - Ahmad Joshkon
- Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, France (A.S.L., R.B., A.F.-B., N.B., A.J., N.J.-C., S.B., M.B.-C., F.D.-G.)
| | - Noémie Jourde-Chiche
- Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, France (A.S.L., R.B., A.F.-B., N.B., A.J., N.J.-C., S.B., M.B.-C., F.D.-G.).,Department of Nephrology, Aix-Marseille University, AP-HM Hôpital de la Conception, Marseille, France (N.J.-C., S.B.)
| | - Stéphane Burtey
- Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, France (A.S.L., R.B., A.F.-B., N.B., A.J., N.J.-C., S.B., M.B.-C., F.D.-G.).,Department of Nephrology, Aix-Marseille University, AP-HM Hôpital de la Conception, Marseille, France (N.J.-C., S.B.)
| | - Marcel Blot-Chabaud
- Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, France (A.S.L., R.B., A.F.-B., N.B., A.J., N.J.-C., S.B., M.B.-C., F.D.-G.)
| | - Françoise Dignat-George
- Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, France (A.S.L., R.B., A.F.-B., N.B., A.J., N.J.-C., S.B., M.B.-C., F.D.-G.)
| | - Christos E Chadjichristos
- From the INSERM UMR-S1155, Tenon Hospital, Paris, France (A.A., P.K., F.A., M.G., C.E.C.).,Sorbonne Université, Paris, France (A.A., C.E.C.)
| |
Collapse
|
15
|
Joshkon A, Heim X, Dubrou C, Bachelier R, Traboulsi W, Stalin J, Fayyad-Kazan H, Badran B, Foucault-Bertaud A, Leroyer AS, Bardin N, Blot-Chabaud M. Role of CD146 (MCAM) in Physiological and Pathological Angiogenesis-Contribution of New Antibodies for Therapy. Biomedicines 2020; 8:biomedicines8120633. [PMID: 33352759 PMCID: PMC7767164 DOI: 10.3390/biomedicines8120633] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 02/07/2023] Open
Abstract
The fundamental role of cell adhesion molecules in mediating various biological processes as angiogenesis has been well-documented. CD146, an adhesion molecule of the immunoglobulin superfamily, and its soluble form, constitute major players in both physiological and pathological angiogenesis. A growing body of evidence shows soluble CD146 to be significantly elevated in the serum or interstitial fluid of patients with pathologies related to deregulated angiogenesis, as autoimmune diseases, obstetric and ocular pathologies, and cancers. To block the undesirable effects of this molecule, therapeutic antibodies have been developed. Herein, we review the multifaceted functions of CD146 in physiological and pathological angiogenesis and summarize the interest of using monoclonal antibodies for therapeutic purposes.
Collapse
Affiliation(s)
- Ahmad Joshkon
- Hematology Department, Center for CardioVascular and Nutrition Research C2VN, Faculty of Pharmacy, Timone Campus, Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut National de Recherche Pour L’agriculture, L’alimentation et L’environnement (INRAE), 13005 Marseille, France; (X.H.); (C.D.); (R.B.); (W.T.); (J.S.); (A.F.-B.); (A.S.L.); (N.B.); (M.B.-C.)
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Science, Lebanese University, Hadath 1104, Lebanon; (H.F.-K.); (B.B.)
- Correspondence:
| | - Xavier Heim
- Hematology Department, Center for CardioVascular and Nutrition Research C2VN, Faculty of Pharmacy, Timone Campus, Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut National de Recherche Pour L’agriculture, L’alimentation et L’environnement (INRAE), 13005 Marseille, France; (X.H.); (C.D.); (R.B.); (W.T.); (J.S.); (A.F.-B.); (A.S.L.); (N.B.); (M.B.-C.)
- Service d’immunologie, Pôle de Biologie, Hôpital de la Conception, Assistance Publique Hôpitaux de Marseille (AP-HM), 13005 Marseille, France
| | - Cléa Dubrou
- Hematology Department, Center for CardioVascular and Nutrition Research C2VN, Faculty of Pharmacy, Timone Campus, Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut National de Recherche Pour L’agriculture, L’alimentation et L’environnement (INRAE), 13005 Marseille, France; (X.H.); (C.D.); (R.B.); (W.T.); (J.S.); (A.F.-B.); (A.S.L.); (N.B.); (M.B.-C.)
| | - Richard Bachelier
- Hematology Department, Center for CardioVascular and Nutrition Research C2VN, Faculty of Pharmacy, Timone Campus, Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut National de Recherche Pour L’agriculture, L’alimentation et L’environnement (INRAE), 13005 Marseille, France; (X.H.); (C.D.); (R.B.); (W.T.); (J.S.); (A.F.-B.); (A.S.L.); (N.B.); (M.B.-C.)
| | - Wael Traboulsi
- Hematology Department, Center for CardioVascular and Nutrition Research C2VN, Faculty of Pharmacy, Timone Campus, Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut National de Recherche Pour L’agriculture, L’alimentation et L’environnement (INRAE), 13005 Marseille, France; (X.H.); (C.D.); (R.B.); (W.T.); (J.S.); (A.F.-B.); (A.S.L.); (N.B.); (M.B.-C.)
| | - Jimmy Stalin
- Hematology Department, Center for CardioVascular and Nutrition Research C2VN, Faculty of Pharmacy, Timone Campus, Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut National de Recherche Pour L’agriculture, L’alimentation et L’environnement (INRAE), 13005 Marseille, France; (X.H.); (C.D.); (R.B.); (W.T.); (J.S.); (A.F.-B.); (A.S.L.); (N.B.); (M.B.-C.)
| | - Hussein Fayyad-Kazan
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Science, Lebanese University, Hadath 1104, Lebanon; (H.F.-K.); (B.B.)
| | - Bassam Badran
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Science, Lebanese University, Hadath 1104, Lebanon; (H.F.-K.); (B.B.)
| | - Alexandrine Foucault-Bertaud
- Hematology Department, Center for CardioVascular and Nutrition Research C2VN, Faculty of Pharmacy, Timone Campus, Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut National de Recherche Pour L’agriculture, L’alimentation et L’environnement (INRAE), 13005 Marseille, France; (X.H.); (C.D.); (R.B.); (W.T.); (J.S.); (A.F.-B.); (A.S.L.); (N.B.); (M.B.-C.)
| | - Aurelie S. Leroyer
- Hematology Department, Center for CardioVascular and Nutrition Research C2VN, Faculty of Pharmacy, Timone Campus, Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut National de Recherche Pour L’agriculture, L’alimentation et L’environnement (INRAE), 13005 Marseille, France; (X.H.); (C.D.); (R.B.); (W.T.); (J.S.); (A.F.-B.); (A.S.L.); (N.B.); (M.B.-C.)
| | - Nathalie Bardin
- Hematology Department, Center for CardioVascular and Nutrition Research C2VN, Faculty of Pharmacy, Timone Campus, Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut National de Recherche Pour L’agriculture, L’alimentation et L’environnement (INRAE), 13005 Marseille, France; (X.H.); (C.D.); (R.B.); (W.T.); (J.S.); (A.F.-B.); (A.S.L.); (N.B.); (M.B.-C.)
- Service d’immunologie, Pôle de Biologie, Hôpital de la Conception, Assistance Publique Hôpitaux de Marseille (AP-HM), 13005 Marseille, France
| | - Marcel Blot-Chabaud
- Hematology Department, Center for CardioVascular and Nutrition Research C2VN, Faculty of Pharmacy, Timone Campus, Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut National de Recherche Pour L’agriculture, L’alimentation et L’environnement (INRAE), 13005 Marseille, France; (X.H.); (C.D.); (R.B.); (W.T.); (J.S.); (A.F.-B.); (A.S.L.); (N.B.); (M.B.-C.)
| |
Collapse
|
16
|
Single-Cell Transcriptomics Reveal Immune Mechanisms of the Onset and Progression of IgA Nephropathy. Cell Rep 2020; 33:108525. [PMID: 33357427 DOI: 10.1016/j.celrep.2020.108525] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 10/07/2020] [Accepted: 11/24/2020] [Indexed: 12/12/2022] Open
Abstract
IgA nephropathy (IgAN) is the leading cause of kidney failure due to an incomplete understanding of its pathogenesis. We perform single-cell RNA sequencing (RNA-seq) on kidneys and CD14+ peripheral blood mononuclear cells (PBMCs) collected from IgAN and normal samples. In IgAN, upregulation of JCHAIN in mesangial cells provides insight into the trigger mechanism for the dimerization and deposition of IgA1 in situ. The pathological mesangium also demonstrates a prominent inflammatory signature and increased cell-cell communication with other renal parenchymal cells and immune cells, suggesting disease progress from the mesangium to the entire kidney. Specific gene expression of kidney-resident macrophages and CD8+ T cells further indicates abnormal regulation associated with proliferation and inflammation. A transitional cell type among intercalated cells with fibrosis signatures is identified, suggesting an adverse outcome of interstitial fibrosis. Altogether, we systematically analyze the molecular events in the onset and progression of IgAN, providing a promising landscape for disease treatment.
Collapse
|
17
|
Molecular detection of maturation stages in the developing kidney. Dev Biol 2020; 470:62-73. [PMID: 33197428 DOI: 10.1016/j.ydbio.2020.11.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 10/31/2020] [Accepted: 11/03/2020] [Indexed: 12/21/2022]
Abstract
Recent advances in stem cell biology have enabled the generation of kidney organoids in vitro, and further maturation of these organoids is observed after experimental transplantation. However, the current organoids remain immature and their precise maturation stages are difficult to determine because of limited information on developmental stage-dependent gene expressions in the kidney in vivo. To establish relevant molecular coordinates, we performed single-cell RNA sequencing (scRNA-seq) on developing kidneys at different stages in the mouse. By selecting genes that exhibited upregulation at birth compared with embryonic day 15.5 as well as cell lineage-specific expression, we generated gene lists correlated with developmental stages in individual cell lineages. Application of these lists to transplanted embryonic kidneys revealed that most cell types, other than the collecting ducts, exhibited similar maturation to kidneys at the neonatal stage in vivo, revealing non-synchronous maturation across the cell lineages. Thus, our scRNA-seq data can serve as useful molecular coordinates to assess the maturation of developing kidneys and eventually of kidney organoids.
Collapse
|
18
|
Contribution of a GATA4-Expressing Hematopoietic Progenitor Lineage to the Adult Mouse Endothelium. Cells 2020; 9:cells9051257. [PMID: 32438714 PMCID: PMC7290801 DOI: 10.3390/cells9051257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/17/2020] [Accepted: 05/18/2020] [Indexed: 11/17/2022] Open
Abstract
Different sources have been claimed for the embryonic origin of the coronary endothelium. Recently, the potential of circulating cells as progenitors of the cardiac endothelium has also been suggested. In a previous study we have shown that circulating progenitors are recruited by the embryonic endocardium and incorporated into the coronary vessels. These progenitors derive from a mesodermal lineage characterized by the expression of Gata4 under control of the enhancer G2. Herein, we aim to trace this specific lineage throughout postnatal stages. We have found that more than 50% of the adult cardiac endothelium derives from the G2-GATA4 lineage. This percentage increases from embryos to adults probably due to differential proliferation and postnatal recruitment of circulating endothelial progenitors. In fact, injection of fetal liver or placental cells in the blood stream of neonates leads to incorporation of G2-GATA4 lineage cells to the coronary endothelium. On the other hand, labeling of the hematopoietic lineage by the stage E7.5 also resulted in positive coronary endothelial cells from both, embryos and adults. Our results suggest that early hematopoietic progenitors recruited by the embryonic ventricular endocardium can become the predominant source of definitive endothelium during the vascularization of the heart.
Collapse
|
19
|
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: 34] [Impact Index Per Article: 8.5] [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
|
20
|
Nishikawa M, Sakai Y, Yanagawa N. Design and strategy for manufacturing kidney organoids. Biodes Manuf 2020. [DOI: 10.1007/s42242-020-00060-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
21
|
Tan Z, Rak-Raszewska A, Skovorodkin I, Vainio SJ. Mouse Embryonic Stem Cell-Derived Ureteric Bud Progenitors Induce Nephrogenesis. Cells 2020; 9:E329. [PMID: 32023845 PMCID: PMC7072223 DOI: 10.3390/cells9020329] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/16/2020] [Accepted: 01/27/2020] [Indexed: 12/14/2022] Open
Abstract
Generation of kidney organoids from pluripotent stem cells (PSCs) is regarded as a potentially powerful way to study kidney development, disease, and regeneration. Direct differentiation of PSCs towards renal lineages is well studied; however, most of the studies relate to generation of nephron progenitor population from PSCs. Until now, differentiation of PSCs into ureteric bud (UB) progenitor cells has had limited success. Here, we describe a simple, efficient, and reproducible protocol to direct differentiation of mouse embryonic stem cells (mESCs) into UB progenitor cells. The mESC-derived UB cells were able to induce nephrogenesis when co-cultured with primary metanephric mesenchyme (pMM). In generated kidney organoids, the embryonic pMM developed nephron structures, and the mESC-derived UB cells formed numerous collecting ducts connected with the nephron tubules. Altogether, our study established an uncomplicated and reproducible platform to generate ureteric bud progenitors from mouse embryonic stem cells.
Collapse
Affiliation(s)
- Zenglai Tan
- Center for Cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, Laboratory of Developmental Biology, Infotech Oulu, University of Oulu, Aapistie 5A, 90220 Oulu, Finland; (A.R.-R.); (I.S.)
| | - Aleksandra Rak-Raszewska
- Center for Cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, Laboratory of Developmental Biology, Infotech Oulu, University of Oulu, Aapistie 5A, 90220 Oulu, Finland; (A.R.-R.); (I.S.)
| | - Ilya Skovorodkin
- Center for Cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, Laboratory of Developmental Biology, Infotech Oulu, University of Oulu, Aapistie 5A, 90220 Oulu, Finland; (A.R.-R.); (I.S.)
| | - Seppo J. Vainio
- Center for Cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, Laboratory of Developmental Biology, Infotech Oulu, University of Oulu, Aapistie 5A, 90220 Oulu, Finland; (A.R.-R.); (I.S.)
- Borealis Biobank of Northern Finland, Oulu Central Hospital, 90220 Oulu, Finland
| |
Collapse
|
22
|
Leroyer AS, Blin MG, Bachelier R, Bardin N, Blot-Chabaud M, Dignat-George F. CD146 (Cluster of Differentiation 146). Arterioscler Thromb Vasc Biol 2020; 39:1026-1033. [PMID: 31070478 DOI: 10.1161/atvbaha.119.312653] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
CD146 (cluster of differentiation 146) is an adhesion molecule that is expressed by different cells constituting vessels, particularly endothelial cells. The last 30 years of research in this field have shown that CD146 plays a key role in the control of several vessel functions. Three forms of CD146 have been described, including 2 transmembrane isoforms and a soluble protein that is detectable in the plasma. These CD146 forms mediate pleiotropic functions through homophilic and heterophilic interactions with proteins present on surrounding partners. Several studies used neutralizing antibodies, siRNA, or genetically modified mice to demonstrate the involvement of CD146 in the regulation of angiogenesis, vascular permeability, and leukocyte transmigration. In this review, we will focus on the current knowledge of the roles of CD146 in vascular homeostasis and diseases associated with endothelial dysfunction.
Collapse
Affiliation(s)
- Aurélie S Leroyer
- From the Aix-Marseille University, Center for CardioVascular and Nutrition Research, INSERM 1263, INRA 1260, France (A.S.L., M.G.B., R.B., N.B., M.B.-C., F.D.-G.)
| | - Muriel G Blin
- From the Aix-Marseille University, Center for CardioVascular and Nutrition Research, INSERM 1263, INRA 1260, France (A.S.L., M.G.B., R.B., N.B., M.B.-C., F.D.-G.)
| | - Richard Bachelier
- From the Aix-Marseille University, Center for CardioVascular and Nutrition Research, INSERM 1263, INRA 1260, France (A.S.L., M.G.B., R.B., N.B., M.B.-C., F.D.-G.)
| | - Nathalie Bardin
- From the Aix-Marseille University, Center for CardioVascular and Nutrition Research, INSERM 1263, INRA 1260, France (A.S.L., M.G.B., R.B., N.B., M.B.-C., F.D.-G.).,Assistance Publique-Hôpitaux de Marseille, Hôpital de la Conception, France (N.B., F.D.-G.)
| | - Marcel Blot-Chabaud
- From the Aix-Marseille University, Center for CardioVascular and Nutrition Research, INSERM 1263, INRA 1260, France (A.S.L., M.G.B., R.B., N.B., M.B.-C., F.D.-G.)
| | - Françoise Dignat-George
- From the Aix-Marseille University, Center for CardioVascular and Nutrition Research, INSERM 1263, INRA 1260, France (A.S.L., M.G.B., R.B., N.B., M.B.-C., F.D.-G.).,Assistance Publique-Hôpitaux de Marseille, Hôpital de la Conception, France (N.B., F.D.-G.)
| |
Collapse
|
23
|
Munro DAD, Wineberg Y, Tarnick J, Vink CS, Li Z, Pridans C, Dzierzak E, Kalisky T, Hohenstein P, Davies JA. Macrophages restrict the nephrogenic field and promote endothelial connections during kidney development. eLife 2019; 8:43271. [PMID: 30758286 PMCID: PMC6374076 DOI: 10.7554/elife.43271] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/29/2019] [Indexed: 12/17/2022] Open
Abstract
The origins and functions of kidney macrophages in the adult have been explored, but their roles during development remain largely unknown. Here we characterise macrophage arrival, localisation, heterogeneity, and functions during kidney organogenesis. Using genetic approaches to ablate macrophages, we identify a role for macrophages in nephron progenitor cell clearance as mouse kidney development begins. Throughout renal organogenesis, most kidney macrophages are perivascular and express F4/80 and CD206. These macrophages are enriched for mRNAs linked to developmental processes, such as blood vessel morphogenesis. Using antibody-mediated macrophage-depletion, we show macrophages support vascular anastomoses in cultured kidney explants. We also characterise a subpopulation of galectin-3+ (Gal3+) myeloid cells within the developing kidney. Our findings may stimulate research into macrophage-based therapies for renal developmental abnormalities and have implications for the generation of bioengineered kidney tissues. The kidneys clean our blood by filtering out waste products while ensuring that useful components, like nutrients, remain in the bloodstream. Blood enters the kidneys through a network of intricately arranged blood vessels, which associate closely with the ‘cleaning tubes’ that carry out filtration. Human kidneys start developing during the early phases of embryonic development. During this process, the newly forming blood vessels and cleaning tubes must grow in the right places for the adult kidney to work properly. Macrophages are cells of the immune system that clear away foreign, diseased, or damaged cells. They are also thought to encourage growth of the developing kidney, but how exactly they do this has remained unknown. Munro et al. therefore wanted to find out when macrophages first appeared in the embryonic kidney and how they might help control their development. Experiments using mice revealed that the first macrophages arrived in the kidney early during its development, alongside newly forming blood vessels. Further investigation using genetically modified mice that did not have macrophages revealed that these immune cells were needed at this stage to clear away misplaced kidney cells and help ‘set the scene’ for future development. At later stages, macrophages in the kidney interacted closely with growing blood vessels. As well as producing molecules linked with blood vessel formation, the macrophages wrapped around the vessels themselves, sometimes even eating cells lining the vessels and the blood cells carried within them. These observations suggested that macrophages actively shaped the network of blood vessels developing within the kidneys. Experiments removing macrophages from kidney tissue confirmed this: in normal kidneys, the blood vessels grew into a continuous network, but in kidneys lacking macrophages, far fewer connections formed between the vessels. This work sheds new light on how the complex structures in the adult kidney first arise and could be useful in future research. For example, adding macrophages to simplified, laboratory-grown ‘mini-kidneys’ could make them better models to study kidney growth, while patients suffering from kidney diseases might benefit from new drugs targeting macrophages.
Collapse
Affiliation(s)
- David AD Munro
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| | - Yishay Wineberg
- Department of Bioengineering, Bar-Ilan University, Ramat Gan, Israel.,Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan, Israel
| | - Julia Tarnick
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| | - Chris S Vink
- Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Zhuan Li
- Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Clare Pridans
- Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Elaine Dzierzak
- Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Tomer Kalisky
- Department of Bioengineering, Bar-Ilan University, Ramat Gan, Israel.,Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan, Israel
| | - Peter Hohenstein
- Leiden University Medical Center, Leiden University, Leiden, The Netherlands.,The Roslin Institute, The University of Edinburgh, Midlothian, United Kingdom
| | - Jamie A Davies
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
24
|
Homan KA, Gupta N, Kroll KT, Kolesky DB, Skylar-Scott M, Miyoshi T, Mau D, Valerius MT, Ferrante T, Bonventre JV, Lewis JA, Morizane R. Flow-enhanced vascularization and maturation of kidney organoids in vitro. Nat Methods 2019; 16:255-262. [PMID: 30742039 PMCID: PMC6488032 DOI: 10.1038/s41592-019-0325-y] [Citation(s) in RCA: 508] [Impact Index Per Article: 101.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 12/21/2018] [Indexed: 01/01/2023]
Abstract
Kidney organoids derived from human pluripotent stem cells have glomerular- and tubular-like compartments that are largely avascular and immature in static culture. Here we report an in vitro method for culturing kidney organoids under flow on millifluidic chips, which expands their endogenous pool of endothelial progenitor cells and generates vascular networks with perfusable lumens surrounded by mural cells. We found that vascularized kidney organoids cultured under flow had more mature podocyte and tubular compartments with enhanced cellular polarity and adult gene expression compared with that in static controls. Glomerular vascular development progressed through intermediate stages akin to those involved in the embryonic mammalian kidney's formation of capillary loops abutting foot processes. The association of vessels with these compartments was reduced after disruption of the endogenous VEGF gradient. The ability to induce substantial vascularization and morphological maturation of kidney organoids in vitro under flow opens new avenues for studies of kidney development, disease, and regeneration.
Collapse
Affiliation(s)
- Kimberly A Homan
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, USA
| | - Navin Gupta
- Renal Division, Brigham and Women's Hospital, Boston, MA, USA.,Harvard Stem Cell Institute, Cambridge, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Katharina T Kroll
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, USA
| | - David B Kolesky
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, USA
| | - Mark Skylar-Scott
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, USA
| | - Tomoya Miyoshi
- Renal Division, Brigham and Women's Hospital, Boston, MA, USA
| | - Donald Mau
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, USA
| | - M Todd Valerius
- Renal Division, Brigham and Women's Hospital, Boston, MA, USA.,Harvard Stem Cell Institute, Cambridge, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Thomas Ferrante
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, USA
| | - Joseph V Bonventre
- Renal Division, Brigham and Women's Hospital, Boston, MA, USA.,Harvard Stem Cell Institute, Cambridge, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Jennifer A Lewis
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, USA. .,Harvard Stem Cell Institute, Cambridge, MA, USA.
| | - Ryuji Morizane
- Renal Division, Brigham and Women's Hospital, Boston, MA, USA. .,Harvard Stem Cell Institute, Cambridge, MA, USA. .,Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
25
|
Murakami Y, Naganuma H, Tanigawa S, Fujimori T, Eto M, Nishinakamura R. Reconstitution of the embryonic kidney identifies a donor cell contribution to the renal vasculature upon transplantation. Sci Rep 2019; 9:1172. [PMID: 30718617 PMCID: PMC6362047 DOI: 10.1038/s41598-018-37793-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 12/14/2018] [Indexed: 01/08/2023] Open
Abstract
The kidney possesses a highly organised vasculature that is required for its filtration function. While recent advances in stem cell biology have enabled the in vitro generation of kidney tissues, at least partially, recapitulation of the complicated vascular architecture remains a huge challenge. Herein we develop a method to reconstitute both the kidney and its vascular architecture in vitro, using dissociated and sorted mouse embryonic kidney cells. Upon transplantation, arteriolar networks were re-established that ran through the interstitial space between branching ureteric buds and eventually entered glomeruli. Using this system, we found that donor-derived endothelial cells significantly contributed to the arterioles and glomerular capillaries formed after transplantation. Unexpectedly, the near-complete depletion of canonical endothelial cells from the donor embryonic kidney suggested the existence of unidentified donor-derived endothelial precursors that were negative for canonical endothelial markers, but still contributed significantly to the vasculature in the transplants. Thus, our protocol will serve as a useful platform for identification of renal endothelial precursors and induction of these precursors from pluripotent stem cells.
Collapse
Affiliation(s)
- Yoichi Murakami
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, 860-0811, Japan
| | - Hidekazu Naganuma
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, 860-0811, Japan
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Shunsuke Tanigawa
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, 860-0811, Japan
| | - Toshihiko Fujimori
- Division of Embryology, National Institute for Basic Biology, Aichi, 444-8787, Japan
| | - Masatoshi Eto
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Ryuichi Nishinakamura
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, 860-0811, Japan.
| |
Collapse
|
26
|
Menon R, Otto EA, Kokoruda A, Zhou J, Zhang Z, Yoon E, Chen YC, Troyanskaya O, Spence JR, Kretzler M, Cebrián C. Single-cell analysis of progenitor cell dynamics and lineage specification in the human fetal kidney. Development 2018; 145:145/16/dev164038. [PMID: 30166318 PMCID: PMC6124540 DOI: 10.1242/dev.164038] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 07/30/2018] [Indexed: 12/11/2022]
Abstract
The mammalian kidney develops through reciprocal interactions between the ureteric bud and the metanephric mesenchyme to give rise to the entire collecting system and the nephrons. Most of our knowledge of the developmental regulators driving this process arises from the study of gene expression and functional genetics in mice and other animal models. In order to shed light on human kidney development, we have used single-cell transcriptomics to characterize gene expression in different cell populations, and to study individual cell dynamics and lineage trajectories during development. Single-cell transcriptome analyses of 6414 cells from five individual specimens identified 11 initial clusters of specific renal cell types as defined by their gene expression profile. Further subclustering identifies progenitors, and mature and intermediate stages of differentiation for several renal lineages. Other lineages identified include mesangium, stroma, endothelial and immune cells. Novel markers for these cell types were revealed in the analysis, as were components of key signaling pathways driving renal development in animal models. Altogether, we provide a comprehensive and dynamic gene expression profile of the developing human kidney at the single-cell level. Summary: New markers for specific cell types in the developing human kidney are identified and computational approaches infer developmental trajectories and interrogate the complex network of signaling pathways and cellular transitions.
Collapse
Affiliation(s)
- Rajasree Menon
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Edgar A Otto
- Department of Internal Medicine, Division of Nephrology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Austin Kokoruda
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jian Zhou
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA.,Graduate Program in Quantitative and Computational Biology, Princeton University, Princeton, NJ 08544, USA
| | - Zidong Zhang
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA.,Graduate Program in Quantitative and Computational Biology, Princeton University, Princeton, NJ 08544, USA
| | - Euisik Yoon
- Department of Electrical Engineering and Computer Science, Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yu-Chih Chen
- Department of Electrical Engineering and Computer Science, Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Olga Troyanskaya
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA.,Flatiron Institute, Simons Foundation, New York, NY 10010, USA.,Department of Computer Science, Princeton University, Princeton, NJ
| | - Jason R Spence
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI 48109, USA .,Department of Cell and Developmental Biology, Division of Gastroenterology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Matthias Kretzler
- Department of Internal Medicine, Division of Nephrology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Cristina Cebrián
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
27
|
Abstract
The nephron is a multifunctional filtration device equipped with an array of sophisticated sensors. For appropriate physiological function in the human and mouse, nephrons must be stereotypically arrayed in large numbers, and this essential structural property that defines the kidney is determined during its fetal development. This review explores the process of nephron determination in the fetal kidney, providing an overview of the foundational literature in the field as well as exploring new developments in this dynamic research area. Mechanisms that ensure that a large number of nephrons can be formed from a small initial number of progenitor cells are central to this process, and the question of how the nephron progenitor cell population balances epithelial differentiation with renewal in the progenitor state is a subject of particular interest. Key growth factor signaling pathways and transcription factor networks are discussed.
Collapse
Affiliation(s)
- Leif Oxburgh
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine 04074, USA;
| |
Collapse
|
28
|
Shaw I, Rider S, Mullins J, Hughes J, Péault B. Pericytes in the renal vasculature: roles in health and disease. Nat Rev Nephrol 2018; 14:521-534. [DOI: 10.1038/s41581-018-0032-4] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
29
|
Development of the renal vasculature. Semin Cell Dev Biol 2018; 91:132-146. [PMID: 29879472 DOI: 10.1016/j.semcdb.2018.06.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 06/01/2018] [Accepted: 06/04/2018] [Indexed: 12/17/2022]
Abstract
The kidney vasculature has a unique and complex architecture that is central for the kidney to exert its multiple and essential physiological functions with the ultimate goal of maintaining homeostasis. An appropriate development and coordinated assembly of the different vascular cell types and their association with the corresponding nephrons is crucial for the generation of a functioning kidney. In this review we provide an overview of the renal vascular anatomy, histology, and current knowledge of the embryological origin and molecular pathways involved in its development. Understanding the cellular and molecular mechanisms involved in renal vascular development is the first step to advance the field of regenerative medicine.
Collapse
|
30
|
Czerniecki SM, Cruz NM, Harder JL, Menon R, Annis J, Otto EA, Gulieva RE, Islas LV, Kim YK, Tran LM, Martins TJ, Pippin JW, Fu H, Kretzler M, Shankland SJ, Himmelfarb J, Moon RT, Paragas N, Freedman BS. High-Throughput Screening Enhances Kidney Organoid Differentiation from Human Pluripotent Stem Cells and Enables Automated Multidimensional Phenotyping. Cell Stem Cell 2018; 22:929-940.e4. [PMID: 29779890 PMCID: PMC5984728 DOI: 10.1016/j.stem.2018.04.022] [Citation(s) in RCA: 302] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 02/05/2018] [Accepted: 04/24/2018] [Indexed: 12/21/2022]
Abstract
Organoids derived from human pluripotent stem cells are a potentially powerful tool for high-throughput screening (HTS), but the complexity of organoid cultures poses a significant challenge for miniaturization and automation. Here, we present a fully automated, HTS-compatible platform for enhanced differentiation and phenotyping of human kidney organoids. The entire 21-day protocol, from plating to differentiation to analysis, can be performed automatically by liquid-handling robots, or alternatively by manual pipetting. High-content imaging analysis reveals both dose-dependent and threshold effects during organoid differentiation. Immunofluorescence and single-cell RNA sequencing identify previously undetected parietal, interstitial, and partially differentiated compartments within organoids and define conditions that greatly expand the vascular endothelium. Chemical modulation of toxicity and disease phenotypes can be quantified for safety and efficacy prediction. Screening in gene-edited organoids in this system reveals an unexpected role for myosin in polycystic kidney disease. Organoids in HTS formats thus establish an attractive platform for multidimensional phenotypic screening.
Collapse
Affiliation(s)
- Stefan M Czerniecki
- Department of Medicine, Division of Nephrology, University of Washington School of Medicine, Seattle, WA 98109, USA; Kidney Research Institute, University of Washington School of Medicine, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine and Quellos High Throughput Screening Core, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Nelly M Cruz
- Department of Medicine, Division of Nephrology, University of Washington School of Medicine, Seattle, WA 98109, USA; Kidney Research Institute, University of Washington School of Medicine, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine and Quellos High Throughput Screening Core, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Jennifer L Harder
- Department of Internal Medicine, Division of Nephrology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Rajasree Menon
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - James Annis
- Institute for Stem Cell and Regenerative Medicine and Quellos High Throughput Screening Core, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Edgar A Otto
- Department of Internal Medicine, Division of Nephrology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Ramila E Gulieva
- Department of Medicine, Division of Nephrology, University of Washington School of Medicine, Seattle, WA 98109, USA; Kidney Research Institute, University of Washington School of Medicine, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine and Quellos High Throughput Screening Core, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Laura V Islas
- Department of Medicine, Division of Nephrology, University of Washington School of Medicine, Seattle, WA 98109, USA; Kidney Research Institute, University of Washington School of Medicine, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine and Quellos High Throughput Screening Core, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Yong Kyun Kim
- Department of Medicine, Division of Nephrology, University of Washington School of Medicine, Seattle, WA 98109, USA; Kidney Research Institute, University of Washington School of Medicine, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine and Quellos High Throughput Screening Core, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Linh M Tran
- Department of Medicine, Division of Nephrology, University of Washington School of Medicine, Seattle, WA 98109, USA; Kidney Research Institute, University of Washington School of Medicine, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine and Quellos High Throughput Screening Core, University of Washington School of Medicine, Seattle, WA 98109, USA; Department of Medicine, Division of Hematology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Timothy J Martins
- Institute for Stem Cell and Regenerative Medicine and Quellos High Throughput Screening Core, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Jeffrey W Pippin
- Department of Medicine, Division of Nephrology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Hongxia Fu
- Institute for Stem Cell and Regenerative Medicine and Quellos High Throughput Screening Core, University of Washington School of Medicine, Seattle, WA 98109, USA; Department of Medicine, Division of Hematology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Matthias Kretzler
- Department of Internal Medicine, Division of Nephrology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Stuart J Shankland
- Department of Medicine, Division of Nephrology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Jonathan Himmelfarb
- Department of Medicine, Division of Nephrology, University of Washington School of Medicine, Seattle, WA 98109, USA; Kidney Research Institute, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Randall T Moon
- Institute for Stem Cell and Regenerative Medicine and Quellos High Throughput Screening Core, University of Washington School of Medicine, Seattle, WA 98109, USA; Department of Pharmacology, University of Washington School of Medicine and Howard Hughes Medical Institute, Seattle, WA 98109, USA
| | - Neal Paragas
- Department of Medicine, Division of Nephrology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Benjamin S Freedman
- Department of Medicine, Division of Nephrology, University of Washington School of Medicine, Seattle, WA 98109, USA; Kidney Research Institute, University of Washington School of Medicine, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine and Quellos High Throughput Screening Core, University of Washington School of Medicine, Seattle, WA 98109, USA; Department of Pathology, University of Washington School of Medicine, Seattle, WA 98109, USA.
| |
Collapse
|
31
|
Zhou J, Plagge A, Murray P. Functional comparison of distinct Brachyury+ states in a renal differentiation assay. Biol Open 2018; 7:bio.031799. [PMID: 29666052 PMCID: PMC5992531 DOI: 10.1242/bio.031799] [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: 12/21/2022] Open
Abstract
Mesodermal populations can be generated in vitro from mouse embryonic stem cells (mESCs) using three-dimensional (3-D) aggregates called embryoid bodies or two-dimensional (2-D) monolayer culture systems. Here, we investigated whether Brachyury-expressing mesodermal cells generated using 3-D or 2-D culture systems are equivalent or, instead, have different properties. Using a Brachyury-GFP/E2-Crimson reporter mESC line, we isolated Brachyury-GFP + mesoderm cells using flow-activated cell sorting and compared their gene expression profiles and ex vivo differentiation patterns. Quantitative real-time polymerase chain reaction analysis showed significant up-regulation of Cdx2, Foxf1 and Hoxb1 in the Brachyury-GFP+ cells isolated from the 3-D system compared with those isolated from the 2-D system. Furthermore, using an ex vivo mouse kidney rudiment assay, we found that, irrespective of their source, Brachyury-GFP+ cells failed to integrate into developing nephrons, which are derived from the intermediate mesoderm. However, Brachyury-GFP+ cells isolated under 3-D conditions appeared to differentiate into endothelial-like cells within the kidney rudiments, whereas the Brachyury-GFP+ isolated from the 2-D conditions only did so to a limited degree. The high expression of Foxf1 in the 3-D Brachyury-GFP+ cells combined with their tendency to differentiate into endothelial-like cells suggests that these mesodermal cells may represent lateral plate mesoderm.
Collapse
Affiliation(s)
- Jing Zhou
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool L69 3BX, UK
| | - Antonius Plagge
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool L69 3BX, UK
| | - Patricia Murray
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool L69 3BX, UK
| |
Collapse
|
32
|
Munro DAD, Davies JA. Vascularizing the Kidney in the Embryo and Organoid: Questioning Assumptions about Renal Vasculogenesis. J Am Soc Nephrol 2018; 29:1593-1595. [PMID: 29748327 DOI: 10.1681/asn.2018020179] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- David A D Munro
- Deanery of Biomedical Sciences, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Jamie A Davies
- Deanery of Biomedical Sciences, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
33
|
Renal Subcapsular Transplantation of PSC-Derived Kidney Organoids Induces Neo-vasculogenesis and Significant Glomerular and Tubular Maturation In Vivo. Stem Cell Reports 2018; 10:751-765. [PMID: 29503086 PMCID: PMC5918682 DOI: 10.1016/j.stemcr.2018.01.041] [Citation(s) in RCA: 275] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 01/30/2018] [Accepted: 01/30/2018] [Indexed: 01/10/2023] Open
Abstract
Human pluripotent stem cell (hPSC)-derived kidney organoids may facilitate disease modeling and the generation of tissue for renal replacement. Long-term application, however, will require transferability between hPSC lines and significant improvements in organ maturation. A key question is whether time or a patent vasculature is required for ongoing morphogenesis. Here, we show that hPSC-derived kidney organoids, derived in fully defined medium conditions and in the absence of any exogenous vascular endothelial growth factor, develop host-derived vascularization. In vivo imaging of organoids under the kidney capsule confirms functional glomerular perfusion as well as connection to pre-existing vascular networks in the organoids. Wide-field electron microscopy demonstrates that transplantation results in formation of a glomerular basement membrane, fenestrated endothelial cells, and podocyte foot processes. Furthermore, compared with non-transplanted organoids, polarization and segmental specialization of tubular epithelium are observed. These data demonstrate that functional vascularization is required for progressive morphogenesis of human kidney organoids.
Collapse
|
34
|
Krause M, Rak-Raszewska A, Naillat F, Saarela U, Schmidt C, Ronkainen VP, Bart G, Ylä-Herttuala S, Vainio SJ. Exosomes as secondary inductive signals involved in kidney organogenesis. J Extracell Vesicles 2018; 7:1422675. [PMID: 29410779 PMCID: PMC5795705 DOI: 10.1080/20013078.2017.1422675] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 12/22/2017] [Indexed: 12/16/2022] Open
Abstract
The subfraction of extracellular vesicles, called exosomes, transfers biological molecular information not only between cells but also between tissues and organs as nanolevel signals. Owing to their unique properties such that they contain several RNA species and proteins implicated in kidney development, exosomes are putative candidates to serve as developmental programming units in embryonic induction and tissue interactions. We used the mammalian metanephric kidney and its nephron-forming mesenchyme containing the nephron progenitor/stem cells as a model to investigate if secreted exosomes could serve as a novel type of inductive signal in a process defined as embryonic induction that controls organogenesis. As judged by several characteristic criteria, exosomes were enriched and purified from a cell line derived from embryonic kidney ureteric bud (UB) and from primary embryonic kidney UB cells, respectively. The cargo of the UB-derived exosomes was analysed by qPCR and proteomics. Several miRNA species that play a role in Wnt pathways and enrichment of proteins involved in pathways regulating the organization of the extracellular matrix as well as tissue homeostasis were identified. When labelled with fluorescent dyes, the uptake of the exosomes by metanephric mesenchyme (MM) cells and the transfer of their cargo to the cells can be observed. Closer inspection revealed that besides entering the cytoplasm, the exosomes were competent to also reach the nucleus. Furthermore, fluorescently labelled exosomal RNA enters into the cytoplasm of the MM cells. Exposure of the embryonic kidney-derived exosomes to the whole MM in an ex vivo organ culture setting did not lead to an induction of nephrogenesis but had an impact on the overall organization of the tissue. We conclude that the exosomes provide a novel signalling system with an apparent role in secondary embryonic induction regulating organogenesis.
Collapse
Affiliation(s)
- Mirja Krause
- Biocenter Oulu, Laboratory of Developmental Biology, InfoTech Oulu, Center for Cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
- The Ritchie Centre, Hudson Institute of Medical Research Core, Clayton, Australia
| | - Aleksandra Rak-Raszewska
- Biocenter Oulu, Laboratory of Developmental Biology, InfoTech Oulu, Center for Cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Florence Naillat
- Biocenter Oulu, Laboratory of Developmental Biology, InfoTech Oulu, Center for Cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Ulla Saarela
- Biocenter Oulu, Laboratory of Developmental Biology, InfoTech Oulu, Center for Cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Christina Schmidt
- Biocenter Oulu, Laboratory of Developmental Biology, InfoTech Oulu, Center for Cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Veli-Pekka Ronkainen
- Biocenter Oulu, Tissue Imaging Center, Light Microscopy Facility, Faculty of Biochemistry and Molecular Medicine, Developmental Biology Lab, University of Oulu, Oulu, Finland
| | - Geneviève Bart
- Biocenter Oulu, Laboratory of Developmental Biology, InfoTech Oulu, Center for Cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Seppo Ylä-Herttuala
- Department of Biotechnology and Molecular Medicine, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Seppo J. Vainio
- Biocenter Oulu, Laboratory of Developmental Biology, InfoTech Oulu, Center for Cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| |
Collapse
|
35
|
Turunen S, Kaisto S, Skovorodkin I, Mironov V, Kalpio T, Vainio S, Rak-Raszewska A. 3D bioprinting of the kidney—hype or hope? ACTA ACUST UNITED AC 2018. [DOI: 10.3934/celltissue.2018.3.119] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
36
|
Patel M, Velagapudi C, Burns H, Doss R, Lee MJ, Mariappan MM, Wagner B, Arar M, Barnes VL, Abboud HE, Barnes JL. Mouse Metanephric Mesenchymal Cell-Derived Angioblasts Undergo Vasculogenesis in Three-Dimensional Culture. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 188:768-784. [PMID: 29269120 DOI: 10.1016/j.ajpath.2017.10.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 09/25/2017] [Accepted: 10/30/2017] [Indexed: 10/18/2022]
Abstract
In vitro models for the investigation of renal vascular development are limited. We previously showed that isolated metanephric mesenchymal (MM) and ureteric bud (UB) cells grown in three-dimensional (3D) matrices formed organoids that consisted of primitive vascular structures surrounding a polarized epithelium. Here, we examined the potential of two principal effectors of vasculogenesis, vascular endothelial growth factor A (VEGF-A), and platelet-derived growth factor B chain (PDGF-BB), to stimulate MM cell differentiation. The results showed that MM cells possess angioblast characteristics by expressing phenotypic markers for endothelial and mesenchymal cells. UB cells synthesize VEGF-A and PDGF-BB proteins and RNA, whereas the MM cells express the respective cognate receptors, supporting their role in directional induction of vasculogenesis. VEGF-A stimulated proliferation of MM cells in monolayer and in 3D sponges but did not affect MM cell migration, organization, or vasculogenesis. However, PDGF-BB stimulated MM cell proliferation, migration, and vasculogenesis in monolayer and organization of the cells into primitive capillary-like assemblies in 3D sea sponge scaffolds in vitro. A role for PDGF-BB in vasculogenesis in the 3D MM/UB co-culture system was validated by direct interference with PDGF-BB or PDGF receptor-β cell interactions to implicate PDGF-BB as a primary effector of MM cell vasculogenesis. Thus, MM cells resemble early renal angioblasts that may provide an ideal platform for the investigation of renal vasculogenesis in vitro.
Collapse
Affiliation(s)
- Mandakini Patel
- Department of Medicine, Division of Nephrology, Audie Murphy Memorial Veterans Administration Hospital, South Texas Veterans Health Care System, San Antonio, Texas
| | - Chakradhar Velagapudi
- Department of Medicine, Division of Nephrology, Audie Murphy Memorial Veterans Administration Hospital, South Texas Veterans Health Care System, San Antonio, Texas
| | | | | | | | | | - Brent Wagner
- Department of Medicine, Division of Nephrology, Audie Murphy Memorial Veterans Administration Hospital, South Texas Veterans Health Care System, San Antonio, Texas; The Medical Research Service, Audie Murphy Memorial Veterans Administration Hospital, South Texas Veterans Health Care System, San Antonio, Texas
| | - Mazen Arar
- Department of Pediatrics, The University of Texas Health Science Center, San Antonio, Texas
| | | | - Hanna E Abboud
- Department of Medicine, Division of Nephrology, Audie Murphy Memorial Veterans Administration Hospital, South Texas Veterans Health Care System, San Antonio, Texas; The Medical Research Service, Audie Murphy Memorial Veterans Administration Hospital, South Texas Veterans Health Care System, San Antonio, Texas
| | - Jeffrey L Barnes
- Department of Medicine, Division of Nephrology, Audie Murphy Memorial Veterans Administration Hospital, South Texas Veterans Health Care System, San Antonio, Texas; Probetex, Inc., San Antonio, Texas; The Medical Research Service, Audie Murphy Memorial Veterans Administration Hospital, South Texas Veterans Health Care System, San Antonio, Texas.
| |
Collapse
|
37
|
Xu Q, Junttila S, Scherer A, Giri KR, Kivelä O, Skovorodkin I, Röning J, Quaggin SE, Marti HP, Shan J, Samoylenko A, Vainio SJ. Renal carcinoma/kidney progenitor cell chimera organoid as a novel tumorigenesis gene discovery model. Dis Model Mech 2017; 10:1503-1515. [PMID: 29084770 PMCID: PMC5769601 DOI: 10.1242/dmm.028332] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 10/16/2017] [Indexed: 12/13/2022] Open
Abstract
Three-dimensional (3D) organoids provide a new way to model various diseases, including cancer. We made use of recently developed kidney-organ-primordia tissue-engineering technologies to create novel renal organoids for cancer gene discovery. We then tested whether our novel assays can be used to examine kidney cancer development. First, we identified the transcriptomic profiles of quiescent embryonic mouse metanephric mesenchyme (MM) and of MM in which the nephrogenesis program had been induced ex vivo. The transcriptome profiles were then compared to the profiles of tumor biopsies from renal cell carcinoma (RCC) patients, and control samples from the same kidneys. Certain signature genes were identified that correlated in the developmentally induced MM and RCC, including components of the caveolar-mediated endocytosis signaling pathway. An efficient siRNA-mediated knockdown (KD) of Bnip3, Gsn, Lgals3, Pax8, Cav1, Egfr or Itgb2 gene expression was achieved in mouse RCC (Renca) cells. The live-cell imaging analysis revealed inhibition of cell migration and cell viability in the gene-KD Renca cells in comparison to Renca controls. Upon siRNA treatment, the transwell invasion capacity of Renca cells was also inhibited. Finally, we mixed E11.5 MM with yellow fluorescent protein (YFP)-expressing Renca cells to establish chimera organoids. Strikingly, we found that the Bnip3-, Cav1- and Gsn-KD Renca-YFP+ cells as a chimera with the MM in 3D organoid rescued, in part, the RCC-mediated inhibition of the nephrogenesis program during epithelial tubules formation. Altogether, our research indicates that comparing renal ontogenesis control genes to the genes involved in kidney cancer may provide new growth-associated gene screens and that 3D RCC-MM chimera organoids can serve as a novel model with which to investigate the behavioral roles of cancer cells within the context of emergent complex tissue structures. Editor’s Choice: Chimeras between embryonic kidney cells and renal carcinoma cells serve as a novel model to assay the roles of co-regulated genes in kidney development and renal carcinogenesis.
Collapse
Affiliation(s)
- Qi Xu
- Biocenter Oulu, Laboratory of Developmental Biology, InfoTech Oulu, Center for Cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, Oulu University, FI-90014 Oulu, Finland
| | - Sanna Junttila
- Biocenter Oulu, Laboratory of Developmental Biology, InfoTech Oulu, Center for Cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, Oulu University, FI-90014 Oulu, Finland
| | | | - Khem Raj Giri
- Biocenter Oulu, Laboratory of Developmental Biology, InfoTech Oulu, Center for Cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, Oulu University, FI-90014 Oulu, Finland
| | - Oona Kivelä
- Biocenter Oulu, Laboratory of Developmental Biology, InfoTech Oulu, Center for Cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, Oulu University, FI-90014 Oulu, Finland.,ValiFinn, FI-90220 Oulu, Finland
| | - Ilya Skovorodkin
- Biocenter Oulu, Laboratory of Developmental Biology, InfoTech Oulu, Center for Cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, Oulu University, FI-90014 Oulu, Finland
| | - Juha Röning
- Department of Computer Science and Engineering, University of Oulu, FI-90014 Oulu, Finland
| | - Susan E Quaggin
- Biocenter Oulu, Laboratory of Developmental Biology, InfoTech Oulu, Center for Cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, Oulu University, FI-90014 Oulu, Finland.,Feinberg Cardiovascular Research Institute, Division of Medicine-Nephrology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Hans-Peter Marti
- Department of Clinical Medicine, University of Bergen, N-5020 Bergen, Norway
| | - Jingdong Shan
- Biocenter Oulu, Laboratory of Developmental Biology, InfoTech Oulu, Center for Cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, Oulu University, FI-90014 Oulu, Finland
| | - Anatoly Samoylenko
- Biocenter Oulu, Laboratory of Developmental Biology, InfoTech Oulu, Center for Cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, Oulu University, FI-90014 Oulu, Finland
| | - Seppo J Vainio
- Biocenter Oulu, Laboratory of Developmental Biology, InfoTech Oulu, Center for Cell Matrix Research, Faculty of Biochemistry and Molecular Medicine, Oulu University, FI-90014 Oulu, Finland
| |
Collapse
|
38
|
Long Noncoding RNA uc001pwg.1 Is Downregulated in Neointima in Arteriovenous Fistulas and Mediates the Function of Endothelial Cells Derived from Pluripotent Stem Cells. Stem Cells Int 2017; 2017:4252974. [PMID: 29387090 PMCID: PMC5745761 DOI: 10.1155/2017/4252974] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 09/19/2017] [Accepted: 10/02/2017] [Indexed: 11/17/2022] Open
Abstract
Recent studies indicate important roles for long noncoding RNAs (lncRNAs) as essential regulators of gene expression. However, the specific roles of lncRNAs in stenotic lesions of arteriovenous fistula (AVF) failure are still largely unknown. We first analyzed the expression profiles of lncRNAs in human stenosed and nonstenotic uremic veins using RNA-sequencing methodology. A total of 19 lncRNAs were found to be differentially expressed in stenotic lesions. Among these, uc001pwg.1 was one of the most significantly downregulated lncRNAs and enriched in both control vein segments and human umbilical vein endothelial cells (HUVECs). Further studies revealed that uc001pwg.1 overexpression could increase nitric oxide synthase (eNOS) phosphorylation and nitric oxide (NO) production in endothelial cells (ECs) derived from human-induced pluripotent stem cells (HiPSCs). Mechanistically, uc001pwg.1 improves endothelial function via mediating MCAM expression. This study represents the first effort of identifying a novel candidate lncRNA for modulating the function of iPSC-ECs, which may facilitate the improvement of stem cell-based therapies for AVF failure.
Collapse
|
39
|
Troy/TNFRSF19 marks epithelial progenitor cells during mouse kidney development that continue to contribute to turnover in adult kidney. Proc Natl Acad Sci U S A 2017; 114:E11190-E11198. [PMID: 29237753 DOI: 10.1073/pnas.1714145115] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
During kidney development, progressively committed progenitor cells give rise to the distinct segments of the nephron, the functional unit of the kidney. Similar segment-committed progenitor cells are thought to be involved in the homeostasis of adult kidney. However, markers for most segment-committed progenitor cells remain to be identified. Here, we evaluate Troy/TNFRSF19 as a segment-committed nephron progenitor cell marker. Troy is expressed in the ureteric bud during embryonic development. During postnatal nephrogenesis, Troy+ cells are present in the cortex and papilla and display an immature tubular phenotype. Tracing of Troy+ cells during nephrogenesis demonstrates that Troy+ cells clonally give rise to tubular structures that persist for up to 2 y after induction. Troy+ cells have a 40-fold higher capacity than Troy- cells to form organoids, which is considered a stem cell property in vitro. In the adult kidney, Troy+ cells are present in the papilla and these cells continue to contribute to collecting duct formation during homeostasis. The number of Troy-derived cells increases after folic acid-induced injury. Our data show that Troy marks a renal stem/progenitor cell population in the developing kidney that in adult kidney contributes to homeostasis, predominantly of the collecting duct, and regeneration.
Collapse
|
40
|
Sequeira-Lopez MLS, Torban E. New insights into precursors of renal endothelium. Kidney Int 2017; 90:244-246. [PMID: 27418087 DOI: 10.1016/j.kint.2016.03.043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 03/23/2016] [Indexed: 02/01/2023]
Abstract
The kidney vasculature is extremely complex, yet, despite recent progress, our understanding of how the renal vascular system develops is limited. By using advanced tissue engineering techniques and in vivo and in vitro depletion of specific populations of endothelial cell precursors, Halt et al. have identified a CD146-expressing precursor as an important player in the development of the renal vasculature.
Collapse
Affiliation(s)
| | - Elena Torban
- McGill University and McGill University Health Center Research Institute, Department of Medicine/Nephrology, Montreal, Quebec, Canada.
| |
Collapse
|
41
|
Mukherjee E, Maringer K, Papke E, Bushnell D, Schaefer C, Kramann R, Ho J, Humphreys BD, Bates C, Sims-Lucas S. Endothelial marker-expressing stromal cells are critical for kidney formation. Am J Physiol Renal Physiol 2017; 313:F611-F620. [PMID: 28539333 PMCID: PMC6148306 DOI: 10.1152/ajprenal.00136.2017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 05/05/2017] [Accepted: 05/22/2017] [Indexed: 11/22/2022] Open
Abstract
Kidneys are highly vascularized and contain many distinct vascular beds. However, the origins of renal endothelial cells and roles of the developing endothelia in the formation of the kidney are unclear. We have shown that the Foxd1-positive renal stroma gives rise to endothelial marker-expressing progenitors that are incorporated within a subset of peritubular capillaries; however, the significance of these cells is unclear. The purpose of this study was to determine whether deletion of Flk1 in the Foxd1 stroma was important for renal development. To that end, we conditionally deleted Flk1 (critical for endothelial cell development) in the renal stroma by breeding-floxed Flk1 mice (Flk1fl/fl ) with Foxd1cre mice to generate Foxd1cre; Flk1fl/fl (Flk1ST-/- ) mice. We then performed FACsorting, histological, morphometric, and metabolic analyses of Flk1ST-/- vs. control mice. We confirmed decreased expression of endothelial markers in the renal stroma of Flk1ST-/- kidneys via flow sorting and immunostaining, and upon interrogation of embryonic and postnatal Flk1ST-/- mice, we found they had dilated peritubular capillaries. Three-dimensional reconstructions showed reduced ureteric branching and fewer nephrons in developing Flk1ST-/- kidneys vs. CONTROLS Juvenile Flk1ST-/- kidneys displayed renal papillary hypoplasia and a paucity of collecting ducts. Twenty-four-hour urine collections revealed that postnatal Flk1ST-/- mice had urinary-concentrating defects. Thus, while lineage-tracing revealed that the renal cortical stroma gave rise to a small subset of endothelial progenitors, these Flk1-expressing stromal cells are critical for patterning the peritubular capillaries. Also, loss of Flk1 in the renal stroma leads to nonautonomous-patterning defects in ureteric lineages.
Collapse
Affiliation(s)
- Elina Mukherjee
- Rangos Research Center, Children's Hospital of Pittsburgh of University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Katherine Maringer
- Rangos Research Center, Children's Hospital of Pittsburgh of University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Emily Papke
- Rangos Research Center, Children's Hospital of Pittsburgh of University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Daniel Bushnell
- Rangos Research Center, Children's Hospital of Pittsburgh of University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Caitlin Schaefer
- Rangos Research Center, Children's Hospital of Pittsburgh of University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Rafael Kramann
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule, Aachen University, Aachen, Germany
| | - Jacqueline Ho
- Rangos Research Center, Children's Hospital of Pittsburgh of University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Benjamin D Humphreys
- Renal Division, Washington University School of Medicine, St. Louis, Missouri; and
| | - Carlton Bates
- Rangos Research Center, Children's Hospital of Pittsburgh of University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Sunder Sims-Lucas
- Rangos Research Center, Children's Hospital of Pittsburgh of University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania;
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
42
|
Cycles of vascular plexus formation within the nephrogenic zone of the developing mouse kidney. Sci Rep 2017; 7:3273. [PMID: 28607473 PMCID: PMC5468301 DOI: 10.1038/s41598-017-03808-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 05/04/2017] [Indexed: 12/25/2022] Open
Abstract
The renal vasculature is required for blood filtration, blood pressure regulation, and pH maintenance, as well as other specialised kidney functions. Yet, despite its importance, many aspects of its development are poorly understood. To provide a detailed spatiotemporal analysis of kidney vascularisation, we collected images of embryonic mouse kidneys at various developmental time-points. Here we describe the first stages of kidney vascularisation and demonstrate that polygonal networks of vessels (endothelial plexuses) form in cycles at the periphery of the kidney. We show that kidney vascularisation initiates at E11, when vessels connected to the embryonic circulation form a ring around the ureteric bud. From E13.5, endothelial plexuses organise around populations of cap mesenchymal and ureteric bud cells in a cyclical, predictable manner. Specifically, as the ureteric bud bifurcates, endothelia form across the bifurcation site as the cap mesenchyme splits. The plexuses are vascular, carry erythrocytes, are enclosed within a basement membrane, and can always be traced back to the renal artery. Our results are a major step towards understanding how the global architecture of the renal vasculature is achieved.
Collapse
|
43
|
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
|