1
|
Boegelein L, Schreiber P, Philipp A, Nusshag C, Essbauer S, Zeier M, Krautkrämer E. Replication kinetics of pathogenic Eurasian orthohantaviruses in human mesangial cells. Virol J 2024; 21:241. [PMID: 39354507 PMCID: PMC11446005 DOI: 10.1186/s12985-024-02517-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 09/23/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND Eurasian pathogenic orthohantaviruses cause hemorrhagic fever with renal syndrome (HFRS) characterized by acute kidney injury (AKI). The virulence of orthohantaviruses varies enormously and direct infection of different renal cell types contribute to pathogenesis. Glomerular mesangial cells play an essential role in the interplay between kidney cells and proper kidney function. Therefore, we analyzed the replication competence of different orthohantavirus species in primary mesangial cells and a mesangial cell line. METHODS We tested the suitability of the mesangial cell line CIHGM-1 (conditionally immortalized human glomerular mesangial cells) as cell culture model for orthohantavirus kidney infection by comparison with primary human renal mesangial cells (HRMCs). We analyzed infection with high pathogenic Hantaan virus (HTNV), moderate pathogenic Puumala virus (PUUV) and non-/low-pathogenic Tula virus (TULV). RESULTS Effective viral spread was observed for PUUV only, whereas infection with HTNV and TULV was abortive. However, in contrast to TULV, HTNV exhibits an initially high infection rate and declines afterwards. This replication pattern was observed in HRMCs and CIHGM-1 cells. Viability or adhesion was neither impaired for PUUV-infected CIHGM-1 nor HRMCs. A loss of migration capacity was observed in PUUV-infected CIHGM-1 cells, but not in HRMCs. CONCLUSIONS The identification of differences in the replication competence of pathogenic orthohantavirus strains in renal mesangial cells is of special interest and may provide useful insights in the virus-specific mechanisms of orthohantavirus induced AKI. The use of CIHGM-1 cells will facilitate the research in a relevant cell culture system.
Collapse
Affiliation(s)
- Lukas Boegelein
- Department of Nephrology, University of Heidelberg, Im Neuenheimer Feld 162, D-69120, Heidelberg, Germany
| | - Pamela Schreiber
- Department of Nephrology, University of Heidelberg, Im Neuenheimer Feld 162, D-69120, Heidelberg, Germany
| | - Alexandra Philipp
- Department of Nephrology, University of Heidelberg, Im Neuenheimer Feld 162, D-69120, Heidelberg, Germany
| | - Christian Nusshag
- Department of Nephrology, University of Heidelberg, Im Neuenheimer Feld 162, D-69120, Heidelberg, Germany
| | - Sandra Essbauer
- Department Virology and Intracellular Agents, Bundeswehr Institute of Microbiology, German Centre for Infection Research, Munich Partner Site, D-80937, Munich, Germany
| | - Martin Zeier
- Department of Nephrology, University of Heidelberg, Im Neuenheimer Feld 162, D-69120, Heidelberg, Germany
| | - Ellen Krautkrämer
- Department of Nephrology, University of Heidelberg, Im Neuenheimer Feld 162, D-69120, Heidelberg, Germany.
| |
Collapse
|
2
|
Huang Y, Geng J, Wang M, Liu W, Hu H, Shi W, Li M, Huo G, Huang G, Xu A. A simple protocol to establish a conditionally immortalized mouse podocyte cell line. Sci Rep 2024; 14:11591. [PMID: 38773220 PMCID: PMC11109129 DOI: 10.1038/s41598-024-62547-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/17/2024] [Indexed: 05/23/2024] Open
Abstract
Podocytes are specialized terminally differentiated cells in the glomerulus that are the primary target cells in many glomerular diseases. However, the current podocyte cell lines suffer from prolonged in vitro differentiation and limited survival time, which impede research progress. Therefore, it is necessary to establish a cell line that exhibits superior performance and characteristics. We propose a simple protocol to obtain an immortalized mouse podocyte cell (MPC) line from suckling mouse kidneys. Primary podocytes were cultured in vitro and infected with the SV40 tsA58 gene to obtain immortalized MPCs. The podocytes were characterized using Western blotting and quantitative real-time PCR. Podocyte injury was examined using the Cell Counting Kit-8 assay and flow cytometry. First, we successfully isolated an MPC line and identified 39 °C as the optimal differentiation temperature. Compared to undifferentiated MPCs, the expression of WT1 and synaptopodin was upregulated in differentiated MPCs. Second, the MPCs ceased proliferating at a nonpermissive temperature after day 4, and podocyte-specific proteins were expressed normally after at least 15 passages. Finally, podocyte injury models were induced to simulate podocyte injury in vitro. In summary, we provide a simple and popularized protocol to establish a conditionally immortalized MPC, which is a powerful tool for the study of podocytes.
Collapse
Affiliation(s)
- Yujiao Huang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Jie Geng
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Mengdan Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Wenbin Liu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Haikun Hu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Wei Shi
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Mei Li
- Dongfang Hospital of Beijing University of Chinese Medicine, Beijing, 100078, China
| | - Guiyang Huo
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Guangrui Huang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Anlong Xu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102488, China.
| |
Collapse
|
3
|
Koch B, Shehata M, Müller-Ruttloff C, Gouda SA, Wetzstein N, Patyna S, Scholz A, Schmid T, Dietrich U, Münch C, Ziebuhr J, Geiger H, Martinez-Sobrido L, Baer PC, Mostafa A, Pleschka S. Influenza A virus replicates productively in primary human kidney cells and induces factors and mechanisms related to regulated cell death and renal pathology observed in virus-infected patients. Front Cell Infect Microbiol 2024; 14:1363407. [PMID: 38590437 PMCID: PMC10999593 DOI: 10.3389/fcimb.2024.1363407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 02/29/2024] [Indexed: 04/10/2024] Open
Abstract
Introduction Influenza A virus (IAV) infection can cause the often-lethal acute respiratory distress syndrome (ARDS) of the lung. Concomitantly, acute kidney injury (AKI) is frequently noticed during IAV infection, correlating with an increased mortality. The aim of this study was to elucidate the interaction of IAV with human kidney cells and, thereby, to assess the mechanisms underlying IAV-mediated AKI. Methods To investigate IAV effects on nephron cells we performed infectivity assays with human IAV, as well as with human isolates of either low or highly pathogenic avian IAV. Also, transcriptome and proteome analysis of IAV-infected primary human distal tubular kidney cells (DTC) was performed. Furthermore, the DTC transcriptome was compared to existing transcriptomic data from IAV-infected lung and trachea cells. Results We demonstrate productive replication of all tested IAV strains on primary and immortalized nephron cells. Comparison of our transcriptome and proteome analysis of H1N1-type IAV-infected human primary distal tubular cells (DTC) with existing data from H1N1-type IAV-infected lung and primary trachea cells revealed enrichment of specific factors responsible for regulated cell death in primary DTC, which could be targeted by specific inhibitors. Discussion IAV not only infects, but also productively replicates on different human nephron cells. Importantly, multi-omics analysis revealed regulated cell death as potential contributing factor for the clinically observed kidney pathology in influenza.
Collapse
Affiliation(s)
- Benjamin Koch
- Department of Internal Medicine 4, Nephrology, University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Mahmoud Shehata
- Center of Scientific Excellence for Influenza Viruses, National Research Centre (NRC), Cairo, Egypt
- Institute of Medical Virology, Justus Liebig University Giessen, Giessen, Germany
| | - Christin Müller-Ruttloff
- Institute of Medical Virology, Justus Liebig University Giessen, Giessen, Germany
- German Center for Infection Research (DZIF), Partner Site Giessen, Giessen, Germany
| | - Shady A. Gouda
- Institute for Biochemistry II, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Nils Wetzstein
- Department of Internal Medicine 2, Infectious Diseases, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Sammy Patyna
- Department of Internal Medicine 4, Nephrology, University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Anica Scholz
- Institute of Biochemistry I, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Tobias Schmid
- Institute of Biochemistry I, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Ursula Dietrich
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
| | - Christian Münch
- Institute for Biochemistry II, Goethe University Frankfurt, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt am Main, Germany
- Cardio-Pulmonary Institute, Frankfurt am Main, Germany
| | - John Ziebuhr
- Institute of Medical Virology, Justus Liebig University Giessen, Giessen, Germany
- German Center for Infection Research (DZIF), Partner Site Giessen, Giessen, Germany
| | - Helmut Geiger
- Department of Internal Medicine 4, Nephrology, University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Luis Martinez-Sobrido
- Texas Biomedical Research Institute, Disease Intervention & Prevention (DIP) and Host Pathogen Interactions (HPI) Programs, San Antonio, TX, United States
| | - Patrick C. Baer
- Department of Internal Medicine 4, Nephrology, University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Ahmed Mostafa
- Center of Scientific Excellence for Influenza Viruses, National Research Centre (NRC), Cairo, Egypt
- Texas Biomedical Research Institute, Disease Intervention & Prevention (DIP) and Host Pathogen Interactions (HPI) Programs, San Antonio, TX, United States
| | - Stephan Pleschka
- Institute of Medical Virology, Justus Liebig University Giessen, Giessen, Germany
- German Center for Infection Research (DZIF), Partner Site Giessen, Giessen, Germany
| |
Collapse
|
4
|
Attar S, Browning VE, Liu Y, Nichols EK, Tsue AF, Shechner DM, Shendure J, Lieberman JA, Akilesh S, Beliveau BJ. Programmable peroxidase-assisted signal amplification enables flexible detection of nucleic acid targets in cellular and histopathological specimens. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.30.526264. [PMID: 36778496 PMCID: PMC9915481 DOI: 10.1101/2023.01.30.526264] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
In situ hybridization (ISH) is a powerful tool for investigating the spatial arrangement of nucleic acid targets in fixed samples. ISH is typically visualized using fluorophores to allow high sensitivity and multiplexing or with colorimetric labels to facilitate co-visualization with histopathological stains. Both approaches benefit from signal amplification, which makes target detection effective, rapid, and compatible with a broad range of optical systems. Here, we introduce a unified technical platform, termed 'pSABER', for the amplification of ISH signals in cell and tissue systems. pSABER decorates the in situ target with concatemeric binding sites for a horseradish peroxidase-conjugated oligonucleotide which can then catalyze the massive localized deposition of fluorescent or colorimetric substrates. We demonstrate that pSABER effectively labels DNA and RNA targets, works robustly in cultured cells and challenging formalin fixed paraffin embedded (FFPE) specimens. Furthermore, pSABER can achieve 25-fold signal amplification over conventional signal amplification by exchange reaction (SABER) and can be serially multiplexed using solution exchange. Therefore, by linking nucleic acid detection to robust signal amplification capable of diverse readouts, pSABER will have broad utility in research and clinical settings.
Collapse
|
5
|
Barreiro K, Lay AC, Leparc G, Tran VDT, Rosler M, Dayalan L, Burdet F, Ibberson M, Coward RJM, Huber TB, Krämer BK, Delic D, Holthofer H. An in vitro approach to understand contribution of kidney cells to human urinary extracellular vesicles. J Extracell Vesicles 2023; 12:e12304. [PMID: 36785873 PMCID: PMC9925963 DOI: 10.1002/jev2.12304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/26/2022] [Accepted: 01/05/2023] [Indexed: 02/15/2023] Open
Abstract
Extracellular vesicles (EV) are membranous particles secreted by all cells and found in body fluids. Established EV contents include a variety of RNA species, proteins, lipids and metabolites that are considered to reflect the physiological status of their parental cells. However, to date, little is known about cell-type enriched EV cargo in complex EV mixtures, especially in urine. To test whether EV secretion from distinct human kidney cells in culture differ and can recapitulate findings in normal urine, we comprehensively analysed EV components, (particularly miRNAs, long RNAs and protein) from conditionally immortalised human kidney cell lines (podocyte, glomerular endothelial, mesangial and proximal tubular cells) and compared to EV secreted in human urine. EV from cell culture media derived from immortalised kidney cells were isolated by hydrostatic filtration dialysis (HFD) and characterised by electron microscopy (EM), nanoparticle tracking analysis (NTA) and Western blotting (WB). RNA was isolated from EV and subjected to miRNA and RNA sequencing and proteins were profiled by tandem mass tag proteomics. Representative sets of EV miRNAs, RNAs and proteins were detected in each cell type and compared to human urinary EV isolates (uEV), EV cargo database, kidney biopsy bulk RNA sequencing and proteomics, and single-cell transcriptomics. This revealed that a high proportion of the in vitro EV signatures were also found in in vivo datasets. Thus, highlighting the robustness of our in vitro model and showing that this approach enables the dissection of cell type specific EV cargo in biofluids and the potential identification of cell-type specific EV biomarkers of kidney disease.
Collapse
Affiliation(s)
- Karina Barreiro
- Institute for Molecular Medicine Finland (FIMM)University of HelsinkiHelsinkiFinland
| | - Abigail C. Lay
- Bristol RenalBristol Medical SchoolFaculty of Health SciencesUniversity of BristolBristolUK
| | - German Leparc
- Boehringer Ingelheim Pharma GmbH & Co. KG BiberachBiberachGermany
| | - Van Du T. Tran
- Vital‐IT GroupSIB Swiss Institute of BioinformaticsLausanneSwitzerland
| | - Marcel Rosler
- Boehringer Ingelheim Pharma GmbH & Co. KG BiberachBiberachGermany
| | - Lusyan Dayalan
- Bristol RenalBristol Medical SchoolFaculty of Health SciencesUniversity of BristolBristolUK
| | - Frederic Burdet
- Vital‐IT GroupSIB Swiss Institute of BioinformaticsLausanneSwitzerland
| | - Mark Ibberson
- Vital‐IT GroupSIB Swiss Institute of BioinformaticsLausanneSwitzerland
| | - Richard J. M. Coward
- Bristol RenalBristol Medical SchoolFaculty of Health SciencesUniversity of BristolBristolUK
| | - Tobias B. Huber
- III Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Bernhard K. Krämer
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology)University Medical Centre MannheimUniversity of HeidelbergMannheimGermany
| | - Denis Delic
- Boehringer Ingelheim Pharma GmbH & Co. KG BiberachBiberachGermany
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology)University Medical Centre MannheimUniversity of HeidelbergMannheimGermany
| | - Harry Holthofer
- Institute for Molecular Medicine Finland (FIMM)University of HelsinkiHelsinkiFinland
- III Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| |
Collapse
|
6
|
Nusshag C, Boegelein L, Schreiber P, Essbauer S, Osberghaus A, Zeier M, Krautkrämer E. Expression Profile of Human Renal Mesangial Cells Is Altered by Infection with Pathogenic Puumala Orthohantavirus. Viruses 2022; 14:v14040823. [PMID: 35458553 PMCID: PMC9025590 DOI: 10.3390/v14040823] [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: 03/21/2022] [Revised: 04/10/2022] [Accepted: 04/14/2022] [Indexed: 11/24/2022] Open
Abstract
Acute kidney injury (AKI) with proteinuria is a hallmark of infections with Eurasian orthohantaviruses. Different kidney cells are identified as target cells of hantaviruses. Mesangial cells may play a central role in the pathogenesis of AKI by regulation of inflammatory mediators and signaling cascades. Therefore, we examined the characteristics of hantavirus infection on human renal mesangial cells (HRMCs). Receptor expression and infection with pathogenic Puumala virus (PUUV) and low-pathogenic Tula virus (TULV) were explored. To analyze changes in protein expression in infected mesangial cells, we performed a proteome profiler assay analyzing 38 markers of kidney damage. We compared the proteome profile of in vitro-infected HRMCs with the profile detected in urine samples of 11 patients with acute hantavirus infection. We observed effective productive infection of HRMCs with pathogenic PUUV, but only poor abortive infection for low-pathogenic TULV. PUUV infection resulted in the deregulation of proteases, adhesion proteins, and cytokines associated with renal damage. The urinary proteome profile of hantavirus patients demonstrated also massive changes, which in part correspond to the alterations observed in the in vitro infection of HRMCs. The direct infection of mesangial cells may induce a local environment of signal mediators that contributes to AKI in hantavirus infection.
Collapse
Affiliation(s)
- Christian Nusshag
- Department of Nephrology, University of Heidelberg, D-69120 Heidelberg, Germany; (C.N.); (L.B.); (P.S.); (A.O.); (M.Z.)
| | - Lukas Boegelein
- Department of Nephrology, University of Heidelberg, D-69120 Heidelberg, Germany; (C.N.); (L.B.); (P.S.); (A.O.); (M.Z.)
| | - Pamela Schreiber
- Department of Nephrology, University of Heidelberg, D-69120 Heidelberg, Germany; (C.N.); (L.B.); (P.S.); (A.O.); (M.Z.)
| | - Sandra Essbauer
- Bundeswehr Institute of Microbiology, Department Virology and Intracellular Agents, German Centre for Infection Research, Munich Partner Site, D-80937 Munich, Germany;
| | - Anja Osberghaus
- Department of Nephrology, University of Heidelberg, D-69120 Heidelberg, Germany; (C.N.); (L.B.); (P.S.); (A.O.); (M.Z.)
| | - Martin Zeier
- Department of Nephrology, University of Heidelberg, D-69120 Heidelberg, Germany; (C.N.); (L.B.); (P.S.); (A.O.); (M.Z.)
| | - Ellen Krautkrämer
- Department of Nephrology, University of Heidelberg, D-69120 Heidelberg, Germany; (C.N.); (L.B.); (P.S.); (A.O.); (M.Z.)
- Correspondence:
| |
Collapse
|
7
|
Loustau T, Abou-Faycal C, Erne W, zur Wiesch PA, Ksouri A, Imhof T, Mörgelin M, Li C, Mathieu M, Salomé N, Crémel G, Dhaouadi S, Bouhaouala-Zahar B, Koch M, Orend G. Modulating tenascin-C functions by targeting the MAtrix REgulating MOtif, “MAREMO”. Matrix Biol 2022; 108:20-38. [DOI: 10.1016/j.matbio.2022.02.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 01/31/2022] [Accepted: 02/23/2022] [Indexed: 12/11/2022]
|
8
|
Tuffin J, Chesor M, Kuzmuk V, Johnson T, Satchell SC, Welsh GI, Saleem MA. GlomSpheres as a 3D co-culture spheroid model of the kidney glomerulus for rapid drug-screening. Commun Biol 2021; 4:1351. [PMID: 34857869 PMCID: PMC8640035 DOI: 10.1038/s42003-021-02868-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 10/28/2021] [Indexed: 01/28/2023] Open
Abstract
The glomerulus is the filtration unit of the kidney. Injury to any component of this specialised structure leads to impaired filtration and eventually fibrosis and chronic kidney disease. Current two and three dimensional (2D and 3D) models that attempt to recreate structure and interplay between glomerular cells are imperfect. Most 2D models are simplistic and unrepresentative, and 3D organoid approaches are currently difficult to reproduce at scale and do not fit well with current industrial drug-screening approaches. Here we report a rapidly generated and highly reproducible 3D co-culture spheroid model (GlomSpheres), better demonstrating the specialised physical and molecular structure of a glomerulus. Co-cultured using a magnetic spheroid formation approach, conditionally immortalised (CI) human podocytes and glomerular endothelial cells (GEnCs) deposited mature, organized isoforms of collagen IV and Laminin. We demonstrate a dramatic upregulation of key podocyte (podocin, nephrin and podocalyxin) and GEnC (pecam-1) markers. Electron microscopy revealed podocyte foot process interdigitation and endothelial vessel formation. Incubation with pro-fibrotic agents (TGF-β1, Adriamycin) induced extracellular matrix (ECM) dysregulation and podocyte loss, which were attenuated by the anti-fibrotic agent Nintedanib. Incubation with plasma from patients with kidney disease induced acute podocyte loss and ECM dysregulation relative to patient matched remission plasma, and Nintedanib reduced podocyte loss. Finally, we developed a rapid imaging approach to demonstrate the model's usefulness in higher throughput pharmaceutical screening. GlomSpheres therefore represent a robust, scalable, replacement for 2D in vitro glomerular disease models.
Collapse
Affiliation(s)
- Jack Tuffin
- Bristol Renal, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS1 3NY, UK.
| | - Musleeha Chesor
- Bristol Renal, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS1 3NY, UK.,Faculty of Medicine, Princess of Naradhiwas University, Narathiwat, Thailand
| | - Valeryia Kuzmuk
- Bristol Renal, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS1 3NY, UK
| | | | - Simon C Satchell
- Bristol Renal, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS1 3NY, UK
| | - Gavin I Welsh
- Bristol Renal, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS1 3NY, UK
| | - Moin A Saleem
- Bristol Renal, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS1 3NY, UK
| |
Collapse
|
9
|
PaintSHOP enables the interactive design of transcriptome- and genome-scale oligonucleotide FISH experiments. Nat Methods 2021; 18:937-944. [PMID: 34226720 PMCID: PMC8349872 DOI: 10.1038/s41592-021-01187-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 05/18/2021] [Indexed: 12/31/2022]
Abstract
Fluorescence in situ hybridization (FISH) allows researchers to visualize the spatial position and quantity of nucleic acids in fixed samples. Recently, considerable progress has been made in developing oligonucleotide (oligo)-based FISH methods that have enabled researchers to study the three-dimensional organization of the genome at super-resolution and visualize the spatial patterns of gene expression for thousands of genes in individual cells. However, there are few existing computational tools to support the bioinformatics workflows necessary to carry out these experiments utilizing oligo FISH probes. Here, we introduce Paint Server and Homology Optimization Pipeline (PaintSHOP), an interactive platform for the design of oligo FISH experiments. PaintSHOP enables researchers to identify probes for their experimental targets efficiently, to incorporate additional necessary sequences such as primer pairs, and to easily generate files documenting library design. PaintSHOP democratizes and standardizes the process of designing complex probe sets for the oligo FISH community. Paint Server and Homology Optimization Pipeline (PaintSHOP), an interactive platform for the design of oligo FISH experiments, democratizes and standardizes the process of designing complex probe sets for the oligo FISH community.
Collapse
|
10
|
Iwakura T, Marschner JA, Zhao ZB, Świderska MK, Anders HJ. Electric cell-substrate impedance sensing in kidney research. Nephrol Dial Transplant 2021; 36:216-223. [PMID: 31598727 DOI: 10.1093/ndt/gfz191] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 08/18/2019] [Indexed: 12/20/2022] Open
Abstract
Electric cell-substrate impedance sensing (ECIS) is a quantitative, label-free, non-invasive analytical method allowing continuous monitoring of the behaviour of adherent cells by online recording of transcellular impedance. ECIS offers a wide range of practical applications to study cell proliferation, migration, differentiation, toxicity and monolayer barrier integrity. All of these applications are relevant for basic kidney research, e.g. on endothelial cells, tubular and glomerular epithelial cells. This review gives an overview on the fundamental principles of the ECIS technology. We name strengths and remaining hurdles for practical applications, present an ECIS array reuse protocol, and review its past, present and potential future contributions to preclinical kidney research.
Collapse
Affiliation(s)
- Takamasa Iwakura
- Renal Division, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU Munich, München, Germany.,Internal Medicine I, Division of Nephrology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Julian A Marschner
- Renal Division, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU Munich, München, Germany
| | - Zhi Bo Zhao
- Renal Division, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU Munich, München, Germany
| | - Monika Katarzyna Świderska
- Renal Division, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU Munich, München, Germany
| | - Hans-Joachim Anders
- Renal Division, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU Munich, München, Germany
| |
Collapse
|
11
|
Yang W, Chen L, Jhuang Y, Lin Y, Hung P, Ko Y, Tsai M, Lee Y, Hsu L, Yeh C, Hsu H, Huang C. Injection of hybrid 3D spheroids composed of podocytes, mesenchymal stem cells, and vascular endothelial cells into the renal cortex improves kidney function and replenishes glomerular podocytes. Bioeng Transl Med 2021; 6:e10212. [PMID: 34027096 PMCID: PMC8126810 DOI: 10.1002/btm2.10212] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 01/07/2021] [Accepted: 01/11/2021] [Indexed: 12/12/2022] Open
Abstract
Podocytes are highly differentiated epithelial cells that are crucial for maintaining the glomerular filtration barrier in the kidney. Podocyte injury followed by depletion is the major cause of pathological progression of kidney diseases. Although cell therapy has been considered a promising alternative approach to kidney transplantation for the treatment of kidney injury, the resultant therapeutic efficacy in terms of improved renal function is limited, possibly owing to significant loss of engrafted cells. Herein, hybrid three-dimensional (3D) cell spheroids composed of podocytes, mesenchymal stem cells, and vascular endothelial cells were designed to mimic the glomerular microenvironment and as a cell delivery vehicle to replenish the podocyte population by cell transplantation. After creating a native glomerulus-like condition, the expression of multiple genes encoding growth factors and basement membrane factors that are strongly associated with podocyte maturation and functionality was significantly enhanced. Our in vivo results demonstrated that intrarenal transplantation of podocytes in the form of hybrid 3D cell spheroids improved engraftment efficiency and replenished glomerular podocytes. Moreover, the proteinuria of the experimental mice with hypertensive nephropathy was effectively reduced. These data clearly demonstrated the potential of hybrid 3D cell spheroids for repairing injured kidneys.
Collapse
Affiliation(s)
- Wen‐Yu Yang
- Institute of Biomedical EngineeringNational Tsing Hua UniversityHsinchuTaiwan
- Department of Biomedical Engineering and Environmental ScienceNational Tsing Hua UniversityHsinchuTaiwan
| | - Li‐Chi Chen
- Institute of Biomedical EngineeringNational Tsing Hua UniversityHsinchuTaiwan
| | - Ya‐Ting Jhuang
- Kidney Research Center, Department of NephrologyLinkou Chang Gung Memorial HospitalTaoyuanTaiwan
| | - Yu‐Jie Lin
- Institute of Biomedical EngineeringNational Tsing Hua UniversityHsinchuTaiwan
| | - Pei‐Yu Hung
- Kidney Research Center, Department of NephrologyLinkou Chang Gung Memorial HospitalTaoyuanTaiwan
| | - Yi‐Ching Ko
- Kidney Research Center, Department of NephrologyLinkou Chang Gung Memorial HospitalTaoyuanTaiwan
| | - Meng‐Yu Tsai
- Institute of Biomedical EngineeringNational Tsing Hua UniversityHsinchuTaiwan
| | - Yun‐Wei Lee
- Institute of Biomedical EngineeringNational Tsing Hua UniversityHsinchuTaiwan
| | - Li‐Wen Hsu
- Bioresource Collection and Research CenterFood Industry Research and Development InstituteHsinchuTaiwan
| | - Chih‐Kuang Yeh
- Department of Biomedical Engineering and Environmental ScienceNational Tsing Hua UniversityHsinchuTaiwan
| | - Hsiang‐Hao Hsu
- Kidney Research Center, Department of NephrologyLinkou Chang Gung Memorial HospitalTaoyuanTaiwan
- College of Medicine, School of MedicineChang Gung UniversityTaoyuanTaiwan
| | - Chieh‐Cheng Huang
- Institute of Biomedical EngineeringNational Tsing Hua UniversityHsinchuTaiwan
| |
Collapse
|
12
|
McWilliam SJ, Wright RD, Welsh GI, Tuffin J, Budge KL, Swan L, Wilm T, Martinas IR, Littlewood J, Oni L. The complex interplay between kidney injury and inflammation. Clin Kidney J 2021; 14:780-788. [PMID: 33777361 PMCID: PMC7986351 DOI: 10.1093/ckj/sfaa164] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 06/10/2020] [Indexed: 12/11/2022] Open
Abstract
Acute kidney injury (AKI) has gained significant attention following patient safety alerts about the increased risk of harm to patients, including increased mortality and hospitalization. Common causes of AKI include hypovolaemia, nephrotoxic medications, ischaemia and acute glomerulonephritis, although in reality it may be undetermined or multifactorial. A period of inflammation either as a contributor to the kidney injury or resulting from the injury is almost universally seen. This article was compiled following a workshop exploring the interplay between injury and inflammation. AKI is characterized by some degree of renal cell death through either apoptosis or necrosis, together with a strong inflammatory response. Studies interrogating the resolution of renal inflammation identify a whole range of molecules that are upregulated and confirm that the kidneys are able to intrinsically regenerate after an episode of AKI, provided the threshold of damage is not too high. Kidneys are unable to generate new nephrons, and dysfunctional or repeated episodes will lead to further nephron loss that is ultimately associated with the development of renal fibrosis and chronic kidney disease (CKD). The AKI to CKD transition is a complex process mainly facilitated by maladaptive repair mechanisms. Early biomarkers mapping out this process would allow a personalized approach to identifying patients with AKI who are at high risk of developing fibrosis and subsequent CKD. This review article highlights this process and explains how laboratory models of renal inflammation and injury assist with understanding the underlying disease process and allow interrogation of medications aimed at targeting the mechanistic interplay.
Collapse
Affiliation(s)
- Stephen J McWilliam
- Department of Paediatric Pharmacology, Alder Hey Children’s Hospital, Liverpool, UK
- Department of Women and Children’s Health, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Rachael D Wright
- Department of Women and Children’s Health, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Gavin I Welsh
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | - Jack Tuffin
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | - Kelly L Budge
- Department of Women and Children’s Health, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Laura Swan
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Thomas Wilm
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Ioana-Roxana Martinas
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - James Littlewood
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
- Department of Nephrology, Royal Liverpool University Hospital, Liverpool, UK
| | - Louise Oni
- Department of Women and Children’s Health, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
- Department of Paediatric Nephrology, Alder Hey Children’s NHS Foundation Trust Hospital, Liverpool, UK
| |
Collapse
|
13
|
Preston R, Naylor RW, Stewart G, Bierzynska A, Saleem MA, Lowe M, Lennon R. A role for OCRL in glomerular function and disease. Pediatr Nephrol 2020; 35:641-648. [PMID: 31811534 PMCID: PMC7056711 DOI: 10.1007/s00467-019-04317-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 07/15/2019] [Accepted: 07/23/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND Lowe syndrome and Dent-2 disease are caused by mutations in the OCRL gene, which encodes for an inositol 5-phosphatase. The renal phenotype associated with OCRL mutations typically comprises a selective proximal tubulopathy, which can manifest as Fanconi syndrome in the most extreme cases. METHODS Here, we report a 12-year-old male with nephrotic-range proteinuria and focal segmental glomerulosclerosis on renal biopsy. As a glomerular pathology was suspected, extensive investigation of tubular function was not performed. RESULTS Surprisingly, whole exome sequencing identified a genetic variant in OCRL (c1467-2A>G) that introduced a novel splice mutation leading to skipping of exon 15. In situ hybridisation of adult human kidney tissue and zebrafish larvae showed OCRL expression in the glomerulus, supporting a role for OCRL in glomerular function. In cultured podocytes, we found that OCRL associated with the linker protein IPIP27A and CD2AP, a protein that is important for maintenance of the podocyte slit diaphragm. CONCLUSION Taken together, this work suggests a previously under-appreciated role for OCRL in glomerular function and highlights the importance of investigating tubular function in patients with persistent proteinuria.
Collapse
Affiliation(s)
- Rebecca Preston
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Richard W Naylor
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Graham Stewart
- Renal Department, Ninewells Hospital, Dundee, DD1 9SY, UK
| | | | - Moin A Saleem
- Children's and Academic Renal Unit, University of Bristol, Bristol, UK
| | - Martin Lowe
- Division of Molecular and Cellular Function, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, M13 9PT, UK.
| | - Rachel Lennon
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, M13 9PT, UK.
- Department of Paediatric Nephrology, Royal Manchester Children's Hospital, Manchester Academic Health Science Centre, Manchester University Hospital NHS Foundation Trust, Manchester, UK.
| |
Collapse
|
14
|
Wright RD, Dimou P, Northey SJ, Beresford MW. Mesangial cells are key contributors to the fibrotic damage seen in the lupus nephritis glomerulus. JOURNAL OF INFLAMMATION-LONDON 2019; 16:22. [PMID: 31807119 PMCID: PMC6857320 DOI: 10.1186/s12950-019-0227-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 11/05/2019] [Indexed: 12/21/2022]
Abstract
Background Lupus nephritis (LN) affects up to 80% of juvenile-onset systemic lupus erythematosus patients. Mesangial cells (MCs) comprise a third of the glomerular cells and are key contributors to fibrotic changes within the kidney. This project aims to identify the roles of MCs in an in vitro model of LN. Methods Conditionally immortalised MCs were treated with pro-inflammatory cytokines or with patient sera in an in vitro model of LN and assessed for their roles in inflammation and fibrosis. Results MCs were shown to produce pro-inflammatory cytokines in response to a model of the inflammatory environment in LN. Further the cells expressed increased levels of mRNA for extracellular matrix (ECM) proteins (COL1A1, COL1A2, COL4A1 and LAMB1), matrix metalloproteinase enzymes (MMP9) and tissue inhibitors of matrix metalloproteinases (TIMP1). Treatment of MCs with serum from patients with active LN was able to induce a similar, albeit milder phenotype. Treatment of MCs with cytokines or patient sera was able to induce secretion of TGF-β1, a known inducer of fibrotic changes. Inhibition of TGF-β1 actions through SB-431542 (an activin A receptor type II-like kinase (ALK5) inhibitor) was able to reduce these responses suggesting that the release of TGF-β1 plays a role in these changes. Conclusions MCs contribute to the inflammatory environment in LN by producing cytokines involved in leukocyte recruitment, activation and maturation. Further the cells remodel the ECM via protein deposition and enzymatic degradation. This occurs through the actions of TGF-β1 on its receptor, ALK5. This may represent a potential therapeutic target for treatment of LN-associated fibrosis.
Collapse
Affiliation(s)
- Rachael D Wright
- 1Department of Women's and Children's Health, Institute of Translational Medicine, University of Liverpool, member of Liverpool Health Partners, Liverpool, UK.,2Department of Women and Children's Health, Institute in the Park, Alder Hey Children's NHS Foundation Trust, Eaton Road, Liverpool, L12 2AP UK
| | - Paraskevi Dimou
- 1Department of Women's and Children's Health, Institute of Translational Medicine, University of Liverpool, member of Liverpool Health Partners, Liverpool, UK
| | - Sarah J Northey
- 1Department of Women's and Children's Health, Institute of Translational Medicine, University of Liverpool, member of Liverpool Health Partners, Liverpool, UK
| | - Michael W Beresford
- 1Department of Women's and Children's Health, Institute of Translational Medicine, University of Liverpool, member of Liverpool Health Partners, Liverpool, UK.,3Department of Paediatric Rheumatology, Alder Hey Children's NHS Foundation Trust, member of Liverpool Health Partners, Liverpool, UK
| |
Collapse
|
15
|
Grigorieva IV, Oszwald A, Grigorieva EF, Schachner H, Neudert B, Ostendorf T, Floege J, Lindenmeyer MT, Cohen CD, Panzer U, Aigner C, Schmidt A, Grosveld F, Thakker RV, Rees AJ, Kain R. A Novel Role for GATA3 in Mesangial Cells in Glomerular Development and Injury. J Am Soc Nephrol 2019; 30:1641-1658. [PMID: 31405951 DOI: 10.1681/asn.2018111143] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 05/01/2019] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND GATA3 is a dual-zinc finger transcription factor that regulates gene expression in many developing tissues. In the kidney, GATA3 is essential for ureteric bud branching, and mice without it fail to develop kidneys. In humans, autosomal dominant GATA3 mutations can cause renal aplasia as part of the hypoparathyroidism, renal dysplasia, deafness (HDR) syndrome that includes mesangioproliferative GN. This suggests that GATA3 may have a previously unrecognized role in glomerular development or injury. METHODS To determine GATA3's role in glomerular development or injury, we assessed GATA3 expression in developing and mature kidneys from Gata3 heterozygous (+/-) knockout mice, as well as injured human and rodent kidneys. RESULTS We show that GATA3 is expressed by FOXD1 lineage stromal progenitor cells, and a subset of these cells mature into mesangial cells (MCs) that continue to express GATA3 in adult kidneys. In mice, we uncover that GATA3 is essential for normal glomerular development, and mice with haploinsufficiency of Gata3 have too few MC precursors and glomerular abnormalities. Expression of GATA3 is maintained in MCs of adult kidneys and is markedly increased in rodent models of mesangioproliferative GN and in IgA nephropathy, suggesting that GATA3 plays a critical role in the maintenance of glomerular homeostasis. CONCLUSIONS These results provide new insights on the role GATA3 plays in MC development and response to injury. It also shows that GATA3 may be a novel and robust nuclear marker for identifying MCs in tissue sections.
Collapse
Affiliation(s)
| | | | | | | | | | - Tammo Ostendorf
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule Aachen University, Aachen, Germany
| | - Jürgen Floege
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule Aachen University, Aachen, Germany
| | - Maja T Lindenmeyer
- Nephrological Center, Medical Clinic and Policlinic IV, University of Munich, Munich, Germany
| | - Clemens D Cohen
- Nephrological Center, Medical Clinic and Policlinic IV, University of Munich, Munich, Germany
| | - Ulf Panzer
- III. Medical Clinic, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christof Aigner
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna, Vienna, Austria
| | - Alice Schmidt
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna, Vienna, Austria
| | - Frank Grosveld
- Department of Cell Biology, Dr. Molewaterplein 50, Rotterdam, The Netherlands; and
| | - Rajesh V Thakker
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, University of Oxford, Oxford, UK
| | | | | |
Collapse
|
16
|
Sieber KB, Batorsky A, Siebenthall K, Hudkins KL, Vierstra JD, Sullivan S, Sur A, McNulty M, Sandstrom R, Reynolds A, Bates D, Diegel M, Dunn D, Nelson J, Buckley M, Kaul R, Sampson MG, Himmelfarb J, Alpers CE, Waterworth D, Akilesh S. Integrated Functional Genomic Analysis Enables Annotation of Kidney Genome-Wide Association Study Loci. J Am Soc Nephrol 2019; 30:421-441. [PMID: 30760496 PMCID: PMC6405142 DOI: 10.1681/asn.2018030309] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 12/26/2018] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Linking genetic risk loci identified by genome-wide association studies (GWAS) to their causal genes remains a major challenge. Disease-associated genetic variants are concentrated in regions containing regulatory DNA elements, such as promoters and enhancers. Although researchers have previously published DNA maps of these regulatory regions for kidney tubule cells and glomerular endothelial cells, maps for podocytes and mesangial cells have not been available. METHODS We generated regulatory DNA maps (DNase-seq) and paired gene expression profiles (RNA-seq) from primary outgrowth cultures of human glomeruli that were composed mainly of podocytes and mesangial cells. We generated similar datasets from renal cortex cultures, to compare with those of the glomerular cultures. Because regulatory DNA elements can act on target genes across large genomic distances, we also generated a chromatin conformation map from freshly isolated human glomeruli. RESULTS We identified thousands of unique regulatory DNA elements, many located close to transcription factor genes, which the glomerular and cortex samples expressed at different levels. We found that genetic variants associated with kidney diseases (GWAS) and kidney expression quantitative trait loci were enriched in regulatory DNA regions. By combining GWAS, epigenomic, and chromatin conformation data, we functionally annotated 46 kidney disease genes. CONCLUSIONS We demonstrate a powerful approach to functionally connect kidney disease-/trait-associated loci to their target genes by leveraging unique regulatory DNA maps and integrated epigenomic and genetic analysis. This process can be applied to other kidney cell types and will enhance our understanding of genome regulation and its effects on gene expression in kidney disease.
Collapse
Affiliation(s)
| | - Anna Batorsky
- Altius Institute for Biomedical Sciences, Seattle, Washington
| | | | | | - Jeff D Vierstra
- Altius Institute for Biomedical Sciences, Seattle, Washington
| | | | - Aakash Sur
- Phase Genomics Inc., Seattle, Washington
- Department of Biomedical and Health Informatics, and
| | - Michelle McNulty
- Division of Pediatric Nephrology, Department of Pediatrics, University of Michigan School of Medicine, Ann Arbor, Michigan; and
| | | | - Alex Reynolds
- Altius Institute for Biomedical Sciences, Seattle, Washington
| | - Daniel Bates
- Altius Institute for Biomedical Sciences, Seattle, Washington
| | - Morgan Diegel
- Altius Institute for Biomedical Sciences, Seattle, Washington
| | - Douglass Dunn
- Altius Institute for Biomedical Sciences, Seattle, Washington
| | - Jemma Nelson
- Altius Institute for Biomedical Sciences, Seattle, Washington
| | - Michael Buckley
- Altius Institute for Biomedical Sciences, Seattle, Washington
| | - Rajinder Kaul
- Altius Institute for Biomedical Sciences, Seattle, Washington
| | - Matthew G Sampson
- Division of Pediatric Nephrology, Department of Pediatrics, University of Michigan School of Medicine, Ann Arbor, Michigan; and
| | - Jonathan Himmelfarb
- Division of Nephrology, Department of Medicine, University of Washington, Seattle, Washington
- Kidney Research Institute, Seattle, Washington
| | - Charles E Alpers
- Department of Anatomic Pathology
- Kidney Research Institute, Seattle, Washington
| | | | - Shreeram Akilesh
- Department of Anatomic Pathology,
- Kidney Research Institute, Seattle, Washington
| |
Collapse
|
17
|
Popik W, Correa H, Khatua A, Aronoff DM, Alcendor DJ. Mesangial cells, specialized renal pericytes and cytomegalovirus infectivity: Implications for HCMV pathology in the glomerular vascular unit and post-transplant renal disease. ACTA ACUST UNITED AC 2018; 5. [PMID: 29977613 PMCID: PMC6027753 DOI: 10.15761/jts.1000248] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background Human Cytomegalovirus (HCMV) infection is problematic after kidney transplantation. Human mesangial cells along with human glomerular endothelial cells and podocytes constitute the renal glomerular vascular unit (GVU). HCMV infection of the GVU is poorly understood. Methods GVU cells infectivity was analysed by microscopy and immunofluorescence. Cytokines profiles were measured by Luminex assays. Renal tissue analysis for HCMV infection was performed by immunohistochemistry. Results Mesangial cells and glomerular endothelial cells but not podocytes were permissive for both lab adapted and clinical strains of HCMV. Luminex analysis of cytokines expressed by mesangial cells exposed to the SBCMV clinical strain was examined. A Tricell infection model of the GVU maintains >90% viability with a unique cytokine profile. Finally, we show αSMA stained mesangial cells permissive for HCMV in renal tissue from a transplant patient. Conclusions HCMV infection of mesangial cells induces angiogenic and proinflammatory cytokines that could contribute to glomerular inflammation.
Collapse
Affiliation(s)
- Waldemar Popik
- Department of Internal Medicine and 4Department of Microbiology and Immunology, Center for AIDS Health Disparities Research, Meharry Medical College, School of Medicine, 1005 Dr. D.B. Todd Jr. Blvd., Nashville, Tennessee 37208-3599 USA
| | - Hernan Correa
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee, USA
| | - Atanu Khatua
- Meharry Medical College, School of Medicine, Centre for AIDS Health Disparities Research, 1005 Dr. D.B. Todd Jr. Blvd., Nashville, Tennessee 37208-3599, USA
| | - David M Aronoff
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee, USA.,Division of Infectious Diseases, Department of Medicine, and Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Centre, Nashville, Tennessee 37232, USA
| | - Donald J Alcendor
- Meharry Medical College, School of Medicine, Centre for AIDS Health Disparities Research, 1005 Dr. D.B. Todd Jr. Blvd., Nashville, Tennessee 37208-3599, USA
| |
Collapse
|
18
|
Cheng L, Gou S, Qiu H, Ma L, Fu P. Complement regulatory proteins in kidneys of patients with anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis. Clin Exp Immunol 2018; 191:116-124. [PMID: 28940198 PMCID: PMC5721235 DOI: 10.1111/cei.13051] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2017] [Indexed: 02/05/2023] Open
Abstract
The complement system activation is involved in the development of anti-neutrophil cytoplasmic antibody-associated vasculitis (AAV). The study aimed to investigate the expression of complement regulatory proteins (CRPs) CD46, CD55 and CD59 in kidneys of 51 AVV patients. The expression of CD46, CD55 and CD59 in kidneys was detected by immunohistochemistry and double immunofluorescence staining. The immunohistochemical examination revealed that expression of the three CRPs could be detected in the glomeruli and tubules of both AAV patients and normal controls. The expression levels of the three CRPs in glomeruli of patients with AAV were significantly lower than those of normal controls. The scores of CD46 and CD55 expression in the tubules of AAV patients were significantly lower than those of normal controls, while there was no significant difference between the scores of CD59 expression in tubules of AAV patients and those of normal controls. Among AAV patients, the expression level of CD46 in glomeruli correlated inversely with the proportion of normal glomeruli, while it correlated with tubular atrophy in renal interstitium (r = -0·305, P = 0·026; r = 0·330, P = 0·023, respectively). The expression levels of CD55 and CD59 in glomeruli correlated with the proportion of total crescents (r = 0·384, P = 0·006; r = 0·351, P = 0·011, respectively). Double immunofluorescence staining indicated that all three CRPs were expressed on endothelial cells, podocytes and mesangial cells in glomeruli. The expression levels of the three CRPs were dysregulated in kidneys of patients with AAV. The expression levels of CD46, CD55 and CD59 were associated with the severity of renal injury of AAV patients.
Collapse
Affiliation(s)
- L. Cheng
- Division of NephrologyKidney Research Institute, West China Hospital of Sichuan UniversityChengduChina
| | - S.‐J. Gou
- Division of NephrologyKidney Research Institute, West China Hospital of Sichuan UniversityChengduChina
| | - H.‐Y. Qiu
- Division of NephrologyKidney Research Institute, West China Hospital of Sichuan UniversityChengduChina
| | - L. Ma
- Division of NephrologyKidney Research Institute, West China Hospital of Sichuan UniversityChengduChina
| | - P. Fu
- Division of NephrologyKidney Research Institute, West China Hospital of Sichuan UniversityChengduChina
| |
Collapse
|
19
|
Umehara K, Sun Y, Hiura S, Hamada K, Itoh M, Kitamura K, Oshima M, Iwama A, Saito K, Anzai N, Chiba K, Akita H, Furihata T. A New Conditionally Immortalized Human Fetal Brain Pericyte Cell Line: Establishment and Functional Characterization as a Promising Tool for Human Brain Pericyte Studies. Mol Neurobiol 2017; 55:5993-6006. [PMID: 29128907 DOI: 10.1007/s12035-017-0815-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 10/27/2017] [Indexed: 12/30/2022]
Abstract
While pericytes wrap around microvascular endothelial cells throughout the human body, their highest coverage rate is found in the brain. Brain pericytes actively contribute to various brain functions, including the development and stabilization of the blood-brain barrier (BBB), tissue regeneration, and brain inflammation. Accordingly, detailed characterization of the functional nature of brain pericytes is important for understanding the mechanistic basis of brain physiology and pathophysiology. Herein, we report on the development of a new human brain pericyte cell line, hereafter referred to as the human brain pericyte/conditionally immortalized clone 37 (HBPC/ci37). Developed via the cell conditionally immortalization method, these cells exhibited excellent proliferative ability at 33 °C. However, when cultured at 37 °C, HBPC/ci37 cells showed a differentiated phenotype that was marked by morphological alterations and increases in several pericyte-enriched marker mRNA levels, such as platelet-derived growth factor receptor β. It was also found that HBPC/ci37 cells possessed the facilitative ability of in vitro BBB formation and differentiation into a neuronal lineage. Furthermore, HBPC/ci37 cells exhibited the typical "reactive" features of brain pericytes in response to pro-inflammatory cytokines. To summarize, our results clearly demonstrate that HBPC/ci37 cells possess the ability to perform several key brain pericyte functions while also showing the capacity for extensive and continuous proliferation. Based on these findings, it can be expected that, as a unique human brain pericyte model, HBPC/ci37 cells have the potential to contribute to significant advances in the understanding of human brain pericyte physiology and pathophysiology.
Collapse
Affiliation(s)
- Kenta Umehara
- Laboratory of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, 260-8675, Japan
| | - Yuchen Sun
- Laboratory of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, 260-8675, Japan
| | - Satoshi Hiura
- Department of Biochemistry, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, 260-8675, Japan
| | - Koki Hamada
- Department of Biochemistry, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, 260-8675, Japan
| | - Motoyuki Itoh
- Department of Biochemistry, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, 260-8675, Japan
| | - Keita Kitamura
- Laboratory of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, 260-8675, Japan
| | - Motohiko Oshima
- Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Atsushi Iwama
- Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Kosuke Saito
- Laboratory of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, 260-8675, Japan
- Division of Medical Safety Science, National Institute of Health Sciences, Tokyo, 158-8501, Japan
| | - Naohiko Anzai
- Department of Pharmacology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Kan Chiba
- Laboratory of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, 260-8675, Japan
| | - Hidetaka Akita
- Laboratory of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, 260-8675, Japan
| | - Tomomi Furihata
- Laboratory of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, 260-8675, Japan.
- Department of Pharmacology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan.
| |
Collapse
|
20
|
Li HY, Oh YS, Choi JW, Jung JY, Jun HS. Blocking lysophosphatidic acid receptor 1 signaling inhibits diabetic nephropathy in db/db mice. Kidney Int 2017; 91:1362-1373. [PMID: 28111010 DOI: 10.1016/j.kint.2016.11.010] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 11/07/2016] [Accepted: 11/10/2016] [Indexed: 01/03/2023]
Abstract
Lysophosphatidic acid (LPA) is known to regulate various biological responses by binding to LPA receptors. The serum level of LPA is elevated in diabetes, but the involvement of LPA in the development of diabetes and its complications remains unknown. Therefore, we studied LPA signaling in diabetic nephropathy and the molecular mechanisms involved. The expression of autotaxin, an LPA synthesis enzyme, and LPA receptor 1 was significantly increased in both mesangial cells (SV40 MES13) maintained in high-glucose media and the kidney cortex of diabetic db/db mice. Increased urinary albumin excretion, increased glomerular tuft area and volume, and mesangial matrix expansion were observed in db/db mice and reduced by treatment with ki16425, a LPA receptor 1/3 antagonist. Transforming growth factor (TGF)β expression and Smad-2/3 phosphorylation were upregulated in SV40 MES13 cells by LPA stimulation or in the kidney cortex of db/db mice, and this was blocked by ki16425 treatment. LPA receptor 1 siRNA treatment inhibited LPA-induced TGFβ expression, whereas cells overexpressing LPA receptor 1 showed enhanced LPA-induced TGFβ expression. LPA treatment of SV40 MES13 cells increased phosphorylated glycogen synthase kinase (GSK)3β at Ser9 and induced translocation of sterol regulatory element-binding protein (SREBP)1 into the nucleus. Blocking GSK3β phosphorylation inhibited SREBP1 activation and consequently blocked LPA-induced TGFβ expression in SV40 MES13 cells. Phosphorylated GSK3β and nuclear SREBP1 accumulation were increased in the kidney cortex of db/db mice and ki16425 treatment blocked these pathways. Thus, LPA receptor 1 signaling increased TGFβ expression via GSK3β phosphorylation and SREBP1 activation, contributing to the development of diabetic nephropathy.
Collapse
Affiliation(s)
- Hui Ying Li
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea; Department of Internal Medicine, Yanbian University Hospital, Yanji, Jilin Province, China
| | - Yoon Sin Oh
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea; Gachon Medical Research Institute, Gil Hospital, Incheon, Korea.
| | - Ji-Woong Choi
- College of Pharmacy, Gachon University, Incheon, Korea
| | - Ji Yong Jung
- Gachon Medical Research Institute, Gil Hospital, Incheon, Korea; Division of Nephrology, Department of Internal Medicine, Gachon University School of Medicine, Incheon, Korea
| | - Hee-Sook Jun
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea; Gachon Medical Research Institute, Gil Hospital, Incheon, Korea; College of Pharmacy, Gachon University, Incheon, Korea.
| |
Collapse
|
21
|
Establishment of a Conditionally Immortalized Wilms Tumor Cell Line with a Homozygous WT1 Deletion within a Heterozygous 11p13 Deletion and UPD Limited to 11p15. PLoS One 2016; 11:e0155561. [PMID: 27213811 PMCID: PMC4876997 DOI: 10.1371/journal.pone.0155561] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 04/29/2016] [Indexed: 01/27/2023] Open
Abstract
We describe a stromal predominant Wilms tumor with focal anaplasia and a complex, tumor specific chromosome 11 aberration: a homozygous deletion of the entire WT1 gene within a heterozygous 11p13 deletion and an additional region of uniparental disomy (UPD) limited to 11p15.5-p15.2 including the IGF2 gene. The tumor carried a heterozygous p.T41A mutation in CTNNB1. Cells established from the tumor carried the same chromosome 11 aberration, but a different, homozygous p.S45Δ CTNNB1 mutation. Uniparental disomy (UPD) 3p21.3pter lead to the homozygous CTNNB1 mutation. The tumor cell line was immortalized using the catalytic subunit of human telomerase (hTERT) in conjunction with a novel thermolabile mutant (U19dl89-97tsA58) of SV40 large T antigen (LT). This cell line is cytogenetically stable and can be grown indefinitely representing a valuable tool to study the effect of a complete lack of WT1 in tumor cells. The origin/fate of Wilms tumors with WT1 mutations is currently poorly defined. Here we studied the expression of several genes expressed in early kidney development, e.g. FOXD1, PAX3, SIX1, OSR1, OSR2 and MEIS1 and show that these are expressed at similar levels in the parental and the immortalized Wilms10 cells. In addition the limited potential for muscle/ osteogenic/ adipogenic differentiation similar to all other WT1 mutant cell lines is also observed in the Wilms10 tumor cell line and this is retained in the immortalized cells. In summary these Wilms10 cells are a valuable model system for functional studies of WT1 mutant cells.
Collapse
|
22
|
Keir LS, Firth R, May C, Ni L, Welsh GI, Saleem MA. Generating conditionally immortalised podocyte cell lines from wild-type mice. Nephron Clin Pract 2015; 129:128-36. [PMID: 25720381 DOI: 10.1159/000369816] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 11/10/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Understanding podocyte biology is key to deciphering the pathogenesis of numerous glomerular diseases. However, cultivation of primary podocytes results in dedifferentiation with loss of specialised architecture. Human conditionally immortalised podocytes partly overcome this problem, utilising a temperature-sensitive transgene. Conditionally immortalised murine podocytes exist, but are derived from the Immortomouse. METHODS Using retroviral temperature-sensitive SV40 transfection, we created a conditionally immortalised podocyte cell line from wild-type mice. RESULTS These cells develop characteristic mature podocyte morphology and robustly express slit diaphragm proteins. Functionally, these cells demonstrate comparable responses in motility and glucose uptake to human conditionally immortalised podocytes. CONCLUSION Podocyte-specific transgenic mice are extensively used to study glomerular disease and this technique could be used to make podocyte cell lines from any mouse, allowing study at the cellular level. This will help characterise these disease models and add to the laboratory resources used to study podocytopathies and glomerular disease.
Collapse
|
23
|
Sampson MG, Hodgin JB, Kretzler M. Defining nephrotic syndrome from an integrative genomics perspective. Pediatr Nephrol 2015; 30:51-63; quiz 59. [PMID: 24890338 PMCID: PMC4241380 DOI: 10.1007/s00467-014-2857-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 05/06/2014] [Accepted: 05/14/2014] [Indexed: 12/15/2022]
Abstract
Nephrotic syndrome (NS) is a clinical condition with a high degree of morbidity and mortality, caused by failure of the glomerular filtration barrier, resulting in massive proteinuria. Our current diagnostic, prognostic and therapeutic decisions in NS are largely based upon clinical or histological patterns such as "focal segmental glomerulosclerosis" or "steroid sensitive". Yet these descriptive classifications lack the precision to explain the physiologic origins and clinical heterogeneity observed in this syndrome. A more precise definition of NS is required to identify mechanisms of disease and capture various clinical trajectories. An integrative genomics approach to NS applies bioinformatics and computational methods to comprehensive experimental, molecular and clinical data for holistic disease definition. A unique aspect is analysis of data together to discover NS-associated molecules, pathways, and networks. Integrating multidimensional datasets from the outset highlights how molecular lesions impact the entire individual. Data sets integrated range from genetic variation to gene expression, to histologic changes, to progression of chronic kidney disease (CKD). This review will introduce the tenets of integrative genomics and suggest how it can increase our understanding of NS from molecular and pathophysiological perspectives. A diverse group of genome-scale experiments are presented that have sought to define molecular signatures of NS. Finally, the Nephrotic Syndrome Study Network (NEPTUNE) will be introduced as an international, prospective cohort study of patients with NS that utilizes an integrated systems genomics approach from the outset. A major NEPTUNE goal is to achieve comprehensive disease definition from a genomics perspective and identify shared molecular drivers of disease.
Collapse
Affiliation(s)
- Matthew G. Sampson
- Division of Nephrology, Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA,to whom correspondence should be addressed: Matthew Sampson, Division of Nephrology, University of Michigan School of Medicine, 8220D MSRB III, West Medical Center Drive, Ann Arbor, MI 48109, kidneyomics.org, , Telephone and Fax: 734-647-9361. Matthias Kretzler, Medicine/Nephrology and Computational Medicine and Bioinformatics, University of Michigan, 1560 MSRB II, 1150 W. Medical Center Dr.-SPC5676, Ann Arbor, MI 48109-5676, 734-615-5757, fax: 734-763-0982,
| | - Jeffrey B. Hodgin
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Matthias Kretzler
- Division of Nephrology, Department of Internal Medicine and Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA,to whom correspondence should be addressed: Matthew Sampson, Division of Nephrology, University of Michigan School of Medicine, 8220D MSRB III, West Medical Center Drive, Ann Arbor, MI 48109, kidneyomics.org, , Telephone and Fax: 734-647-9361. Matthias Kretzler, Medicine/Nephrology and Computational Medicine and Bioinformatics, University of Michigan, 1560 MSRB II, 1150 W. Medical Center Dr.-SPC5676, Ann Arbor, MI 48109-5676, 734-615-5757, fax: 734-763-0982,
| |
Collapse
|
24
|
Conditionally immortalized human pancreatic stellate cell lines demonstrate enhanced proliferation and migration in response to IGF-I. Exp Cell Res 2014; 330:300-310. [PMID: 25304103 DOI: 10.1016/j.yexcr.2014.09.033] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 09/18/2014] [Accepted: 09/27/2014] [Indexed: 01/11/2023]
Abstract
Pancreatic stellate cells (PSCs) play a key role in the dense desmoplastic stroma associated with pancreatic ductal adenocarcinoma. Studies on human PSCs have been minimal due to difficulty in maintaining primary PSC in culture. We have generated the first conditionally immortalized human non-tumor (NPSC) and tumor-derived (TPSC) pancreatic stellate cells via transformation with the temperature-sensitive SV40 large T antigen and human telomerase (hTERT). These cells proliferate at 33°C. After transfer to 37°C, the SV40LT is switched off and the cells regain their primary PSC phenotype and growth characteristics. NPSC contained cytoplasmic vitamin A-storing lipid droplets, while both NPSC and TPSC expressed the characteristic markers αSMA, vimentin, desmin and GFAP. Proteome array analysis revealed that of the 55 evaluated proteins, 27 (49%) were upregulated ≥3-fold in TPSC compared to NPSC, including uPA, pentraxin-3, endoglin and endothelin-1. Two insulin-like growth factor binding proteins (IGFBPs) were inversely expressed. Although discordant IGFBP-2 and IGFBP-3 levels, IGF-I was found to stimulate proliferation of both NPSC and TPSC. Both basal and IGF-I stimulated motility was significantly enhanced in TPSC compared to NPSC. In conclusion, these cells provide a unique resource that will facilitate further study of the active stroma compartment associated with pancreatic cancer.
Collapse
|
25
|
Ma L, Shelness GS, Snipes JA, Murea M, Antinozzi PA, Cheng D, Saleem MA, Satchell SC, Banas B, Mathieson PW, Kretzler M, Hemal AK, Rudel LL, Petrovic S, Weckerle A, Pollak MR, Ross MD, Parks JS, Freedman BI. Localization of APOL1 protein and mRNA in the human kidney: nondiseased tissue, primary cells, and immortalized cell lines. J Am Soc Nephrol 2014; 26:339-48. [PMID: 25012173 DOI: 10.1681/asn.2013091017] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Although APOL1 gene variants are associated with nephropathy in African Americans, little is known about APOL1 protein synthesis, uptake, and localization in kidney cells. To address these questions, we examined APOL1 protein and mRNA localization in human kidney and human kidney-derived cell lines. Indirect immunofluorescence microscopy performed on nondiseased nephrectomy cryosections from persons with normal kidney function revealed that APOL1 protein was markedly enriched in podocytes (colocalized with synaptopodin and Wilms' tumor suppressor) and present in lower abundance in renal tubule cells. Fluorescence in situ hybridization detected APOL1 mRNA in glomeruli (podocytes and endothelial cells) and tubules, consistent with endogenous synthesis in these cell types. When these analyses were extended to renal-derived cell lines, quantitative RT-PCR did not detect APOL1 mRNA in human mesangial cells; however, abundant levels of APOL1 mRNA were observed in proximal tubule cells and glomerular endothelial cells, with lower expression in podocytes. Western blot analysis revealed corresponding levels of APOL1 protein in these cell lines. To explain the apparent discrepancy between the marked abundance of APOL1 protein in kidney podocytes observed in cryosections versus the lesser abundance in podocyte cell lines, we explored APOL1 cellular uptake. APOL1 protein was taken up readily by human podocytes in vitro but was not taken up efficiently by mesangial cells, glomerular endothelial cells, or proximal tubule cells. We hypothesize that the higher levels of APOL1 protein in human cryosectioned podocytes may reflect both endogenous protein synthesis and APOL1 uptake from the circulation or glomerular filtrate.
Collapse
Affiliation(s)
| | | | | | | | - Peter A Antinozzi
- Biochemistry, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | | | - Moin A Saleem
- Children's Renal Unit, Bristol Royal Hospital for Children, University of Bristol, Bristol, United Kingdom
| | - Simon C Satchell
- Learning and Research Southmead Hospital Bristol, University of Bristol, Bristol, United Kingdom
| | - Bernhard Banas
- Internal Medicine II-Nephrology/Transplantation, University Medical Center, Regensburg, Germany
| | - Peter W Mathieson
- Children's Renal Unit, Bristol Royal Hospital for Children, University of Bristol, Bristol, United Kingdom
| | - Matthias Kretzler
- Department of Internal Medicine-Nephrology, University of Michigan at Ann Arbor Medical School, Ann Arbor, Michigan
| | | | | | - Snezana Petrovic
- Internal Medicine-Nephrology, Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | | | - Martin R Pollak
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts; and
| | - Michael D Ross
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - John S Parks
- Pathology-Lipid Sciences, and Biochemistry, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | | |
Collapse
|
26
|
Jennings P, Aschauer L, Wilmes A, Gstraunthaler G. Renal Cell Culture. METHODS IN PHARMACOLOGY AND TOXICOLOGY 2014. [DOI: 10.1007/978-1-4939-0521-8_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
27
|
Yang B, Hodgkinson AD, Shaw NA, Millward BA, Demaine AG. Protective effect of statin therapy on connective tissue growth factor induction by diabetes in vivo and high glucose in vitro. Growth Factors 2013; 31:199-208. [PMID: 24192280 DOI: 10.3109/08977194.2013.852189] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Transcriptional activity of connective tissue growth factor (CTGF) promoter in transfected HEK293 cells was determined by luciferase assays. Secreted CTGF in cultured human mesangial cells was measured by enzyme-linked immunosorbent assay (ELISA). CTGF in urine and plasma was also measured in 405 subjects with/without type 2 diabetes. Our results showed that high glucose significantly increased transcription of the promoter in the transfected cells by more than 2.5-folds (p < 0.0005). CTGF secretion was induced by high glucose in the cells (p < 0.0005). These increases were inhibited by simvastatin. Urine CTGF was positively associated with plasma CTGF in both type 2 diabetes (p = 0.0005) and controls (p = 0.01). Urine CTGF levels in patients with macroalbuminuria were significantly higher than patients without macroalbuminuria (p < 0.05). In conclusion, our in vitro study suggests that statin may have a renal-protective effect through the inhibition of CTGF expression. Urine CTGF may be a good marker for the prediction of diabetic nephropathy.
Collapse
Affiliation(s)
- Bingmei Yang
- Molecular Medicine, Institute of Translational & Stratified Medicine, Plymouth University Schools of Medicine & Dentistry , United Kingdom
| | | | | | | | | |
Collapse
|
28
|
Hale LJ, Welsh GI, Perks CM, Hurcombe JA, Moore S, Hers I, Saleem MA, Mathieson PW, Murphy AJ, Jeansson M, Holly JM, Hardouin SN, Coward RJ. Insulin-like growth factor-II is produced by, signals to and is an important survival factor for the mature podocyte in man and mouse. J Pathol 2013; 230:95-106. [PMID: 23299523 DOI: 10.1002/path.4165] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Revised: 12/03/2012] [Accepted: 12/26/2012] [Indexed: 01/10/2023]
Abstract
Podocytes are crucial for preventing the passage of albumin into the urine and, when lost, are associated with the development of albuminuria, renal failure and cardiovascular disease. Podocytes have limited capacity to regenerate, therefore pro-survival mechanisms are critically important. Insulin-like growth factor-II (IGF-II) is a potent survival and growth factor; however, its major function is thought to be in prenatal development, when circulating levels are high. IGF-II has only previously been reported to continue to be expressed in discrete regions of the brain into adulthood in rodents, with systemic levels being undetectable. Using conditionally immortalized human and ex vivo adult mouse cells of the glomerulus, we demonstrated the podocyte to be the major glomerular source and target of IGF-II; it signals to this cell via the IGF-I receptor via the PI3 kinase and MAPK pathways. Functionally, a reduction in IGF signalling causes podocyte cell death in vitro and glomerular disease in vivo in an aged IGF-II transgenic mouse that produces approximately 60% of IGF-II due to a lack of the P2 promoter of this gene. Collectively, this work reveals the fundamental importance of IGF-II in the mature podocyte for glomerular health across mammalian species.
Collapse
Affiliation(s)
- L J Hale
- Academic and Children's Renal Unit, University of Bristol, Learning and Research, Southmead Hospital, Bristol, BS10 5NB, UK
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Holme A, Hurcombe JA, Straatman-Iwanowska A, Inward CI, Gissen P, Coward RJ. Glomerular involvement in the arthrogryposis, renal dysfunction and cholestasis syndrome. Clin Kidney J 2013; 6:183-8. [PMID: 26019847 PMCID: PMC4432437 DOI: 10.1093/ckj/sfs182] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 12/12/2012] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Arthrogryposis, renal dysfunction and cholestasis (ARC) syndrome is a multisystem autosomal-recessive disorder caused by defects in the VPS33B and VIPAR genes, involved in localization of apical membrane proteins. Affected children usually die by 1 year of age, often secondary to infective complications. The classic renal manifestation previously described in ARC syndrome is proximal-tubular dysfunction. The aim of this study is to gain further insight into the renal manifestations of this syndrome. METHODS Clinical review of three cases of ARC syndrome presenting to a tertiary centre. Together with measurement of VPS33B and VIPAR protein expression in the human glomerulus. RESULTS The cases demonstrated severe failure to thrive and in addition to commonly described features profound proteinuria and albuminuria, together with hypoalbuminaemia, suggesting glomerular involvement of this syndrome. Western blotting of conditionally immortalized human glomerular cells and ex vivo immunofluorescent analysis of the human glomerulus revealed that VPS33B and VIPAR were highly expressed in glomerular endothelium, and podocytes, but not in the mesangium. CONCLUSIONS ARC syndrome affects the glomerulus as well as the proximal tubule in the kidney. Our molecular studies suggest that both cell types that constitute the glomerular filtration barrier are affected in this condition, providing an explanation for the albuminuria that we have observed in our cases.
Collapse
Affiliation(s)
- Amelia Holme
- Department of Child and Adolescent Health , University of Bristol , Bristol , UK ; Department of Paediatric Nephrology , Bristol Royal Hospital for Children , Bristol , UK
| | | | | | - Carol I Inward
- Department of Paediatric Nephrology , Bristol Royal Hospital for Children , Bristol , UK
| | - Paul Gissen
- MRC Laboratory for Molecular Cell Biology , University College London , London , UK ; Department of Paediatric Metabolic Medicine , Great Ormond Street Hospital , London , UK
| | - Richard J Coward
- Department of Paediatric Nephrology , Bristol Royal Hospital for Children , Bristol , UK ; Academic Renal Unit , University of Bristol , Bristol , UK
| |
Collapse
|
30
|
Abstract
Podocytes (glomerular epithelial cells) lie on the urinary aspect of the glomerular capillary and play a key role in the selective filter that underlies kidney function. They are injured in various forms of renal disease: the extents of this injury and its reversibility have major implications for treatment and prognosis. Until recently, podocytes were difficult to study in vitro because of a previous lack of techniques for obtaining differentiated cells in quantities adequate for research. In recent years, this problem has been solved for rodent and human podocytes and there has been an explosion of research using cultured cells. These authors have led the development and characterization of human podocyte cell lines and in this article describe the methods that have allowed them to do this.
Collapse
Affiliation(s)
- Lan Ni
- Academic Renal Unit, University of Bristol, Southmead Hospital, Bristol, UK
| | | | | |
Collapse
|