1
|
Tanzi A, Buono L, Grange C, Iampietro C, Brossa A, Arcolino FO, Arigoni M, Calogero R, Perin L, Deaglio S, Levtchenko E, Peruzzi L, Bussolati B. Urine-derived podocytes from steroid resistant nephrotic syndrome patients as a model for renal-progenitor derived extracellular vesicles effect and drug screening. J Transl Med 2024; 22:762. [PMID: 39143486 PMCID: PMC11323595 DOI: 10.1186/s12967-024-05575-z] [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: 02/14/2024] [Accepted: 08/04/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND Personalized disease models are crucial for evaluating how diseased cells respond to treatments, especially in case of innovative biological therapeutics. Extracellular vesicles (EVs), nanosized vesicles released by cells for intercellular communication, have gained therapeutic interest due to their ability to reprogram target cells. We here utilized urinary podocytes obtained from children affected by steroid-resistant nephrotic syndrome with characterized genetic mutations as a model to test the therapeutic potential of EVs derived from kidney progenitor cells (nKPCs). METHODS EVs were isolated from nKPCs derived from the urine of a preterm neonate. Three lines of urinary podocytes obtained from nephrotic patients' urine and a line of Alport syndrome patient podocytes were characterized and used to assess albumin permeability in response to nKPC-EVs or various drugs. RNA sequencing was conducted to identify commonly modulated pathways after nKPC-EV treatment. siRNA transfection was used to demonstrate the involvement of SUMO1 and SENP2 in the modulation of permeability. RESULTS Treatment with the nKPC-EVs significantly reduced permeability across all the steroid-resistant patients-derived and Alport syndrome-derived podocytes. At variance, podocytes appeared unresponsive to standard pharmacological treatments, with the exception of one line, in alignment with the patient's clinical response at 48 months. By RNA sequencing, only two genes were commonly upregulated in nKPC-EV-treated genetically altered podocytes: small ubiquitin-related modifier 1 (SUMO1) and Sentrin-specific protease 2 (SENP2). SUMO1 and SENP2 downregulation increased podocyte permeability confirming the role of the SUMOylation pathway. CONCLUSIONS nKPCs emerge as a promising non-invasive source of EVs with potential therapeutic effects on podocytes with genetic dysfunction, through modulation of SUMOylation, an important pathway for the stability of podocyte slit diaphragm proteins. Our findings also suggest the feasibility of developing a non-invasive in vitro model for screening regenerative compounds on patient-derived podocytes.
Collapse
Affiliation(s)
- Adele Tanzi
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, Turin, 10125, Italy
| | - Lola Buono
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, Turin, 10125, Italy
| | - Cristina Grange
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Corinne Iampietro
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, Turin, 10125, Italy
| | - Alessia Brossa
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, Turin, 10125, Italy
| | - Fanny Oliveira Arcolino
- Department of Pediatric Nephrology, Emma Children's Hospital, Amsterdam UMC, Amsterdam, The Netherlands
- Emma Centrum of Personalized Medicine, Emma Children's Hospital, Amsterdam UMC, Amsterdam, The Netherlands
| | - Maddalena Arigoni
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, Turin, 10125, Italy
| | - Raffaele Calogero
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, Turin, 10125, Italy
| | - Laura Perin
- Department of Urology, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Silvia Deaglio
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Elena Levtchenko
- Department of Pediatric Nephrology, Emma Children's Hospital, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Development and Regeneration, Cluster Woman and Child, Laboratory of Pediatric Nephrology, KU Leuven, Leuven, Belgium
| | - Licia Peruzzi
- Pediatric Nephrology, ERKNet Center, Regina Margherita Children's Hospital, AOU Città della, Salute e della Scienza di Torino, Turin, Italy
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, Turin, 10125, Italy.
| |
Collapse
|
2
|
Ugodnikov A, Persson H, Simmons CA. Bridging barriers: advances and challenges in modeling biological barriers and measuring barrier integrity in organ-on-chip systems. LAB ON A CHIP 2024; 24:3199-3225. [PMID: 38689569 DOI: 10.1039/d3lc01027a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
Biological barriers such as the blood-brain barrier, skin, and intestinal mucosal barrier play key roles in homeostasis, disease physiology, and drug delivery - as such, it is important to create representative in vitro models to improve understanding of barrier biology and serve as tools for therapeutic development. Microfluidic cell culture and organ-on-a-chip (OOC) systems enable barrier modelling with greater physiological fidelity than conventional platforms by mimicking key environmental aspects such as fluid shear, accurate microscale dimensions, mechanical cues, extracellular matrix, and geometrically defined co-culture. As the prevalence of barrier-on-chip models increases, so does the importance of tools that can accurately assess barrier integrity and function without disturbing the carefully engineered microenvironment. In this review, we first provide a background on biological barriers and the physiological features that are emulated through in vitro barrier models. Then, we outline molecular permeability and electrical sensing barrier integrity assessment methods, and the related challenges specific to barrier-on-chip implementation. Finally, we discuss future directions in the field, as well important priorities to consider such as fabrication costs, standardization, and bridging gaps between disciplines and stakeholders.
Collapse
Affiliation(s)
- Alisa Ugodnikov
- Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada.
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Henrik Persson
- Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada.
| | - Craig A Simmons
- Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada.
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
- Department of Mechanical & Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada
| |
Collapse
|
3
|
LeBleu VS, Kanasaki K, Lovisa S, Alge JL, Kim J, Chen Y, Teng Y, Gerami-Naini B, Sugimoto H, Kato N, Revuelta I, Grau N, Sleeman JP, Taduri G, Kizu A, Rafii S, Hochedlinger K, Quaggin SE, Kalluri R. Genetic reprogramming with stem cells regenerates glomerular epithelial podocytes in Alport syndrome. Life Sci Alliance 2024; 7:e202402664. [PMID: 38561223 PMCID: PMC10985218 DOI: 10.26508/lsa.202402664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 04/04/2024] Open
Abstract
Glomerular filtration relies on the type IV collagen (ColIV) network of the glomerular basement membrane, namely, in the triple helical molecules containing the α3, α4, and α5 chains of ColIV. Loss of function mutations in the genes encoding these chains (Col4a3, Col4a4, and Col4a5) is associated with the loss of renal function observed in Alport syndrome (AS). Precise understanding of the cellular basis for the patho-mechanism remains unknown and a specific therapy for this disease does not currently exist. Here, we generated a novel allele for the conditional deletion of Col4a3 in different glomerular cell types in mice. We found that podocytes specifically generate α3 chains in the developing glomerular basement membrane, and that its absence is sufficient to impair glomerular filtration as seen in AS. Next, we show that horizontal gene transfer, enhanced by TGFβ1 and using allogenic bone marrow-derived mesenchymal stem cells and induced pluripotent stem cells, rescues Col4a3 expression and revive kidney function in Col4a3-deficient AS mice. Our proof-of-concept study supports that horizontal gene transfer such as cell fusion enables cell-based therapy in Alport syndrome.
Collapse
Affiliation(s)
- Valerie S LeBleu
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Division of Matrix Biology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Northwestern University Feinberg School of Medicine and Kellogg School of Management, Chicago, IL, USA
- https://ror.org/02pttbw34 Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Keizo Kanasaki
- Division of Matrix Biology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Sara Lovisa
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joseph L Alge
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- https://ror.org/02pttbw34 Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Jiha Kim
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yang Chen
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yingqi Teng
- Division of Matrix Biology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Behzad Gerami-Naini
- Division of Matrix Biology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Hikaru Sugimoto
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Division of Matrix Biology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Noritoshi Kato
- Division of Matrix Biology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Ignacio Revuelta
- Division of Matrix Biology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Nicole Grau
- Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Jonathan P Sleeman
- Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
- Karlsruhe Institute of Technology (IBCS-BIP), Karlsruhe, Germany
| | - Gangadhar Taduri
- Division of Matrix Biology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Akane Kizu
- Division of Matrix Biology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Shahin Rafii
- Department of Genetic Medicine and Ansary Stem Cell Institute, Weill Cornell Medical College, New York, NY, USA
| | - Konrad Hochedlinger
- Massachusetts General Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Boston, MA, USA
| | - Susan E Quaggin
- Northwestern University Feinberg School of Medicine & Feinberg Cardiovascular and Renal Research Institute, Chicago, IL, USA
| | - Raghu Kalluri
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Division of Matrix Biology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Boston, MA, USA
- Harvard-MIT Division of Health Sciences and Technology, Boston, MA, USA
- Department of Bioengineering, Rice University, Houston, TX, USA
- https://ror.org/02pttbw34 Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
4
|
Tanzi A, Buono L, Grange C, Iampietro C, Brossa A, Arcolino FO, Arigoni M, Calogero R, Perin L, Deaglio S, Levtchenko E, Peruzzi L, Bussolati B. Urine-derived podocytes from steroid resistant nephrotic syndrome patients as a model for renal-progenitor derived extracellular vesicles effect and drug screening. RESEARCH SQUARE 2024:rs.3.rs-3959549. [PMID: 38464119 PMCID: PMC10925474 DOI: 10.21203/rs.3.rs-3959549/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Background Personalized disease models are crucial for assessing the specific response of diseased cells to drugs, particularly novel biological therapeutics. Extracellular vesicles (EVs), nanosized vesicles released by cells for intercellular communication, have gained therapeutic interest due to their ability to reprogram target cells. We here utilized urinary podocytes obtained from children affected by steroid-resistant nephrotic syndrome with characterized genetic mutations as a model to test the therapeutic potential of EVs derived from kidney progenitor cells. Methods EVs were isolated from kidney progenitor cells (nKPCs) derived from the urine of a preterm neonate. Three lines of urinary podocytes obtained from nephrotic patients' urine and a line of Alport patient podocytes were characterized and used to assess albumin permeability in response to various drugs or to nKPC-EVs. RNA sequencing was conducted to identify commonly modulated pathways. Results Podocytes appeared unresponsive to pharmacological treatments, except for a podocyte line demonstrating responsiveness, in alignment with the patient's clinical response at 48 months. At variance, treatment with the nKPC-EVs was able to significantly reduce permeability in all the steroid-resistant patients-derived podocytes as well as in the line of Alport-derived podocytes. RNA sequencing of nKPC-EV-treated podocytes revealed the common upregulation of two genes (small ubiquitin-related modifier 1 (SUMO1) and Sentrin-specific protease 2 (SENP2)) involved in the SUMOylation pathway, a process recently demonstrated to play a role in slit diaphragm stabilization. Gene ontology analysis on podocyte expression profile highlighted cell-to-cell adhesion as the primary upregulated biological activity in treated podocytes. Conclusions nKPCs emerge as a promising non-invasive source of EVs with potential therapeutic effects on podocyte dysfunction. Furthermore, our findings suggest the possibility of establishing a non-invasive in vitro model for screening regenerative compounds on patient-derived podocytes.
Collapse
Affiliation(s)
- Adele Tanzi
- University of Turin: Universita degli Studi di Torino
| | - Lola Buono
- University of Turin: Universita degli Studi di Torino
| | | | | | | | | | | | | | | | | | | | - Licia Peruzzi
- Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino: Azienda Ospedaliero Universitaria Citta della Salute e della Scienza di Torino
| | | |
Collapse
|
5
|
Proteomics and Phosphoproteomics of Circulating Extracellular Vesicles Provide New Insights into Diabetes Pathobiology. Int J Mol Sci 2022; 23:ijms23105779. [PMID: 35628588 PMCID: PMC9147902 DOI: 10.3390/ijms23105779] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/14/2022] [Accepted: 05/18/2022] [Indexed: 02/04/2023] Open
Abstract
The purpose of this study was to define the proteomic and phosphoproteomic landscape of circulating extracellular vesicles (EVs) in people with normal glucose tolerance (NGT), prediabetes (PDM), and diabetes (T2DM). Archived serum samples from 30 human subjects (n = 10 per group, ORIGINS study, NCT02226640) were used. EVs were isolated using EVtrap®. Mass spectrometry-based methods were used to detect the global EV proteome and phosphoproteome. Differentially expressed features, correlation, enriched pathways, and enriched tissue-specific protein sets were identified using custom R scripts. Phosphosite-centric analyses were conducted using directPA and PhosR software packages. A total of 2372 unique EV proteins and 716 unique EV phosphoproteins were identified among all samples. Unsupervised clustering of the differentially expressed (fold change ≥ 2, p < 0.05, FDR < 0.05) proteins and, particularly, phosphoproteins showed excellent discrimination among the three groups. CDK1 and PKCδ appear to drive key upstream phosphorylation events that define the phosphoproteomic signatures of PDM and T2DM. Circulating EVs from people with diabetes carry increased levels of specific phosphorylated kinases (i.e., AKT1, GSK3B, LYN, MAP2K2, MYLK, and PRKCD) and could potentially distribute activated kinases systemically. Among characteristic changes in the PDM and T2DM EVs, “integrin switching” appeared to be a central feature. Proteins involved in oxidative phosphorylation (OXPHOS), known to be reduced in various tissues in diabetes, were significantly increased in EVs from PDM and T2DM, which suggests that an abnormally elevated EV-mediated secretion of OXPHOS components may underlie the development of diabetes. A highly enriched signature of liver-specific markers among the downregulated EV proteins and phosphoproteins in both PDM and T2DM groups was also detected. This suggests that an alteration in liver EV composition and/or secretion may occur early in prediabetes. This study identified EV proteomic and phosphoproteomic signatures in people with prediabetes and T2DM and provides novel insight into the pathobiology of diabetes.
Collapse
|
6
|
Bejoy J, Qian ES, Woodard LE. Accelerated protocol for the differentiation of podocytes from human pluripotent stem cells. STAR Protoc 2021; 2:100898. [PMID: 34746862 PMCID: PMC8551929 DOI: 10.1016/j.xpro.2021.100898] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Several kidney diseases including congenital nephrotic syndrome, Alport syndrome, and diabetic nephropathy are linked to podocyte dysfunction. Human podocytopathies may be modeled in either primary or immortalized podocyte cell lines. Human induced pluripotent stem cell (hiPSC)-derived podocytes are a source of human podocytes, but the existing protocols have variable efficiency and expensive media components. We developed an accelerated, feeder-free protocol for deriving functional, mature podocytes from hiPSCs in only 12 days, saving time and money compared with other approaches.
Collapse
Affiliation(s)
- Julie Bejoy
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Eddie Spencer Qian
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Lauren Elizabeth Woodard
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Veterans Affairs, Nashville, TN 37212, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
7
|
Greiten JK, Kliewe F, Schnarre A, Artelt N, Schröder S, Rogge H, Amann K, Daniel C, Lindenmeyer MT, Cohen CD, Endlich K, Endlich N. The role of filamins in mechanically stressed podocytes. FASEB J 2021; 35:e21560. [PMID: 33860543 DOI: 10.1096/fj.202001179rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 02/26/2021] [Accepted: 03/15/2021] [Indexed: 11/11/2022]
Abstract
Glomerular hypertension induces mechanical load to podocytes, often resulting in podocyte detachment and the development of glomerulosclerosis. Although it is well known that podocytes are mechanosensitive, the mechanosensors and mechanotransducers are still unknown. Since filamin A, an actin-binding protein, is already described to be a mechanosensor and mechanotransducer, we hypothesized that filamins could be important for the outside-in signaling as well as the actin cytoskeleton of podocytes under mechanical stress. In this study, we demonstrate that filamin A is the main isoform of the filamin family that is expressed in cultured podocytes. Together with filamin B, filamin A was significantly up-regulated during mechanical stretch (3 days, 0.5 Hz, and 5% extension). To study the role of filamin A in cultured podocytes under mechanical stress, filamin A was knocked down (Flna KD) by specific siRNA. Additionally, we established a filamin A knockout podocyte cell line (Flna KO) by CRISPR/Cas9. Knockdown and knockout of filamin A influenced the expression of synaptopodin, a podocyte-specific protein, focal adhesions as well as the morphology of the actin cytoskeleton. Moreover, the cell motility of Flna KO podocytes was significantly increased. Since the knockout of filamin A has had no effect on cell adhesion of podocytes during mechanical stress, we simultaneously knocked down the expression of filamin A and B. Thereby, we observed a significant loss of podocytes during mechanical stress indicating a compensatory mechanism. Analyzing hypertensive mice kidneys as well as biopsies of patients suffering from diabetic nephropathy, we found an up-regulation of filamin A in podocytes in contrast to the control. In summary, filamin A and B mediate matrix-actin cytoskeleton interactions which are essential for the adaptation of cultured podocyte to mechanical stress.
Collapse
Affiliation(s)
- Jonas K Greiten
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Felix Kliewe
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Annabel Schnarre
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Nadine Artelt
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Sindy Schröder
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Henrik Rogge
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Kerstin Amann
- Department of Nephropathology, Friedrich-Alexander University (FAU) Erlangen-Nuremberg, Erlangen, Germany
| | - Christoph Daniel
- Department of Nephropathology, Friedrich-Alexander University (FAU) Erlangen-Nuremberg, Erlangen, Germany
| | - Maja T Lindenmeyer
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Clemens D Cohen
- Nephrological Center, Medical Clinic and Policlinic IV, University of Munich, Munich, Germany
| | - Karlhans Endlich
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Nicole Endlich
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
8
|
Lay AC, Hale LJ, Stowell-Connolly H, Pope RJP, Nair V, Ju W, Marquez E, Rollason R, Hurcombe JA, Hayes B, Roberts T, Gillam L, Allington J, Nelson RG, Kretzler M, Holly JMP, Perks CM, McArdle CA, Welsh GI, Coward RJM. IGFBP-1 expression is reduced in human type 2 diabetic glomeruli and modulates β1-integrin/FAK signalling in human podocytes. Diabetologia 2021; 64:1690-1702. [PMID: 33758952 PMCID: PMC8187213 DOI: 10.1007/s00125-021-05427-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 01/14/2021] [Indexed: 12/25/2022]
Abstract
AIMS/HYPOTHESIS Podocyte loss or injury is one of the earliest features observed in the pathogenesis of diabetic kidney disease (DKD), which is the leading cause of end-stage renal failure worldwide. Dysfunction in the IGF axis, including in IGF binding proteins (IGFBPs), is associated with DKD, particularly in the early stages of disease progression. The aim of this study was to investigate the potential roles of IGFBPs in the development of type 2 DKD, focusing on podocytes. METHODS IGFBP expression was analysed in the Pima DKD cohort, alongside data from the Nephroseq database, and in ex vivo human glomeruli. Conditionally immortalised human podocytes and glomerular endothelial cells were studied in vitro, where IGFBP-1 expression was analysed using quantitative PCR and ELISAs. Cell responses to IGFBPs were investigated using migration, cell survival and adhesion assays; electrical cell-substrate impedance sensing; western blotting; and high-content automated imaging. RESULTS Data from the Pima DKD cohort and from the Nephroseq database demonstrated a significant reduction in glomerular IGFBP-1 in the early stages of human type 2 DKD. In the glomerulus, IGFBP-1 was predominantly expressed in podocytes and controlled by phosphoinositide 3-kinase (PI3K)-forkhead box O1 (FoxO1) activity. In vitro, IGFBP-1 signalled to podocytes via β1-integrins, resulting in increased phosphorylation of focal-adhesion kinase (FAK), increasing podocyte motility, adhesion, electrical resistance across the adhesive cell layer and cell viability. CONCLUSIONS/INTERPRETATION This work identifies a novel role for IGFBP-1 in the regulation of podocyte function and that the glomerular expression of IGFBP-1 is reduced in the early stages of type 2 DKD, via reduced FoxO1 activity. Thus, we hypothesise that strategies to maintain glomerular IGFBP-1 levels may be beneficial in maintaining podocyte function early in DKD.
Collapse
Affiliation(s)
- Abigail C Lay
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | - Lorna J Hale
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | | | - Robert J P Pope
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Viji Nair
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Wenjun Ju
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Eva Marquez
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | - Ruth Rollason
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | - Jenny A Hurcombe
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | - Bryony Hayes
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | - Timothy Roberts
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | - Lawrence Gillam
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | - Jonathan Allington
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | - Robert G Nelson
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, USA
| | - Matthias Kretzler
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Jeff M P Holly
- IGFs and Metabolic Endocrinology Group, Bristol Medical School, University of Bristol, Bristol, UK
| | - Claire M Perks
- IGFs and Metabolic Endocrinology Group, Bristol Medical School, University of Bristol, Bristol, UK
| | - Craig A McArdle
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Gavin I Welsh
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK
| | - Richard J M Coward
- Bristol Renal, Bristol Medical School, University of Bristol, Bristol, UK.
| |
Collapse
|
9
|
Tomas NM, Mortensen SA, Wilmanns M, Huber TB. Across scales: novel insights into kidney health and disease by structural biology. Kidney Int 2021; 100:281-288. [PMID: 33940110 DOI: 10.1016/j.kint.2021.03.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/16/2021] [Accepted: 03/25/2021] [Indexed: 11/25/2022]
Abstract
Over the past decades, structural biology methods such as X-ray crystallography and cryo-electron microscopy have been increasingly used to study protein functions, molecular interactions, physiological processes, and disease mechanisms. This review outlines a selection of structural biology methods, highlights recent examples of how structural analyses have contributed to a more profound understanding of the machinery of life, and gives a perspective on how these methods can be applied to investigate functions of kidney molecules and pathogenic mechanisms of renal diseases.
Collapse
Affiliation(s)
- Nicola M Tomas
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Simon A Mortensen
- European Molecular Biology Laboratory, Hamburg Unit, Hamburg, Germany
| | - Matthias Wilmanns
- European Molecular Biology Laboratory, Hamburg Unit, Hamburg, Germany; University Hamburg Clinical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias B Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
10
|
Zheng JM, Wang SS, Tian X, Che DJ. Sustained activation of C3aR in a human podocyte line impairs the morphological maturation of the cells. Mol Med Rep 2020; 22:5326-5338. [PMID: 33174024 PMCID: PMC7646996 DOI: 10.3892/mmr.2020.11626] [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: 03/30/2020] [Accepted: 09/02/2020] [Indexed: 12/03/2022] Open
Abstract
The C3a receptor (C3aR) has been reported to be involved in various physiological and pathological processes, including the regulation of cellular structure development. Expression of C3aR has been reported in podocytes; however, data concerning the role of C3aR in podocyte morphology is scarce. The aim of the present study was to examine the effect of C3aR activation on the architectural development of podocytes. An immortal human podocyte line (HPC) was transfected with a C3a expression lentivirus vector or recombinant C3a. SB290157 was used to block the activation of C3aR. The expression of C3a in HPC cells was analyzed by reverse transcription-quantitative PCR (RT-qPCR) and ELISAs. Phase contrast and fluorescence microscopy were used to observe the morphology of the podocytes. The adhesive ability of HPC cells was analyzed using an attachment assay. RT-qPCR, cyto-immunofluorescence and western blotting were used to determine the expression levels of the adhesion-associated genes. The expression levels of carboxypeptidases in HPC cells was also detected by RT-qPCR. Compared with the untransfected and control virus-transfected HPC cells, the C3a-overexpressing cells (HPC-C3a) failed to expand their cell bodies and develop an arborized appearance in the process of maturation, which the control cells exhibited. In addition, HPC-C3a cells presented with decreased adhesive capacity, altered focal adhesion (FA) plaques and decreased expression of FA-associated genes. These effects were blocked by a C3aR antagonist; however, the addition of purified C3a could not completely mimic the effects of C3a overexpression. Furthermore, HPC cells expressed carboxypeptidases, which have been reported to be able to inactivate C3a. In summary, the results demonstrated that sustained C3aR activation impaired the morphological maturation of HPC cells, which may be associated with the altered expression of FA-associated genes and impaired FA. Since chronic complement activation has been reported in renal diseases, which indicate sustained C3aR activation in renal cells, including podocytes and podocyte progenitors, the possible role of C3aR in the dysregulation of podocyte architecture and podocyte regeneration requires further research.
Collapse
Affiliation(s)
- Jing-Min Zheng
- Department of Nephrology, Taizhou Hospital, Wenzhou Medical University, Linhai, Zhejiang 317000, P.R. China
| | - Sha-Sha Wang
- Department of Nephrology, Taizhou Hospital, Wenzhou Medical University, Linhai, Zhejiang 317000, P.R. China
| | - Xiong Tian
- Department of Nephrology, Taizhou Hospital, Wenzhou Medical University, Linhai, Zhejiang 317000, P.R. China
| | - De-Jun Che
- Department of Nephrology, Taizhou Hospital, Wenzhou Medical University, Linhai, Zhejiang 317000, P.R. China
| |
Collapse
|
11
|
Saoud R, Jaffa MA, Habib A, Zhao J, Al Hariri M, Zhu R, Hasan A, Ziyadeh FN, Kobeissy F, Mechref Y, Jaffa AA. Modulation of proteomic and inflammatory signals by Bradykinin in podocytes. J Adv Res 2020; 24:409-422. [PMID: 32518694 PMCID: PMC7270529 DOI: 10.1016/j.jare.2020.05.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 05/16/2020] [Accepted: 05/18/2020] [Indexed: 01/19/2023] Open
Abstract
Podocyte damage is one of the hallmarks of diabetic nephropathy leading to proteinuria and kidney damage. The underlying mechanisms of podocyte injury are not well defined. Bradykinin (BK) was shown to contribute to diabetic kidney disease. Here, we evaluated the temporal changes in proteome profile and inflammatory signals of podocytes in response to BK (10−7M). Protein profile was evaluated by liquid chromatography mass Spectrometry (LC-MS/MS) analysis. Proteome profile analysis of podocytes treated with BK (10−7M) for 3 and 6 h, revealed 61 proteins that were differentially altered compared to unstimulated control podocytes. Pathway enrichment analysis suggested inhibition of cell death pathways, engagement of cytoskeletal elements and activation of inflammatory pathways. One of the inflammatory proteins that was identified to be induced by BK treatment is Prostaglandin (PG) H Synthase-2 (Cyclooxygenase-2, COX-2). In addition, BK significantly induced the production and release of PGE2 and this effect was inhibited by both COX-2 and MEK Kinase inhibitors, demonstrating that the production of PGE2 by BK is mediated via COX-2 and MAPK-dependent mechanisms. These findings provide a global understanding of the effector modulated proteome in response to BK and also reveal BK as an important modulator of inflammation and a potential player in podocyte injury.
Collapse
Affiliation(s)
- Richard Saoud
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Lebanon
| | - Miran A Jaffa
- Epidemiology and Population Health Department, Faculty of Health Sciences, American University of Beirut, Lebanon
| | - Aida Habib
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Lebanon.,INSERM-U1149, Centre de Recherche sur l'Inflammation, Sorbonne Paris Cité, Laboratoire d'Excellence Inflamex, Faculté de Médecine, Site Xavier Bichat, Université de Paris, France
| | - Jingfu Zhao
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX 79409, United States
| | - Moustafa Al Hariri
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Lebanon
| | - Rui Zhu
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX 79409, United States
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, Qatar University, Qatar
| | - Fuad N Ziyadeh
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Lebanon.,Department of Internal Medicine, Faculty of Medicine, American University of Beirut, Lebanon
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Lebanon
| | - Yehia Mechref
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX 79409, United States
| | - Ayad A Jaffa
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Lebanon
| |
Collapse
|
12
|
Uchio-Yamada K, Yasuda K, Monobe Y, Akagi KI, Suzuki O, Manabe N. Tensin2 is important for podocyte-glomerular basement membrane interaction and integrity of the glomerular filtration barrier. Am J Physiol Renal Physiol 2020; 318:F1520-F1530. [PMID: 32390516 DOI: 10.1152/ajprenal.00055.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Tensin2 (Tns2), an integrin-linked protein, is enriched in podocytes within the glomerulus. Previous studies have revealed that Tns2-deficient mice exhibit defects of the glomerular basement membrane (GBM) soon after birth in a strain-dependent manner. However, the mechanisms for the onset of defects caused by Tns2 deficiency remains unidentified. Here, we aimed to determine the role of Tns2 using newborn Tns2-deficient mice and murine primary podocytes. Ultrastructural analysis revealed that developing glomeruli during postnatal nephrogenesis exhibited abnormal GBM processing due to ectopic laminin-α2 accumulation followed by GBM thickening. In addition, analysis of primary podocytes revealed that Tns2 deficiency led to impaired podocyte-GBM interaction and massive expression of laminin-α2 in podocytes. Our study suggests that weakened podocyte-GBM interaction due to Tns2 deficiency causes increased mechanical stress on podocytes by continuous daily filtration after birth, resulting in stressed podocytes ectopically producing laminin-α2, which interrupts GBM processing. We conclude that Tns2 plays important roles in the podocyte-GBM interaction and maintenance of the glomerular filtration barrier.
Collapse
Affiliation(s)
- Kozue Uchio-Yamada
- Laboratory of Animal Models for Human Diseases, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
| | - Keiko Yasuda
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yoko Monobe
- Section of Laboratory Equipment, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
| | - Ken-Ichi Akagi
- Section of Laboratory Equipment, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
| | - Osamu Suzuki
- Laboratory of Animal Models for Human Diseases, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
| | - Noboru Manabe
- Department of Human Sciences, Osaka International University, Moriguchi, Osaka, Japan
| |
Collapse
|
13
|
Kanasaki K. N-acetyl-seryl-aspartyl-lysyl-proline is a valuable endogenous antifibrotic peptide for kidney fibrosis in diabetes: An update and translational aspects. J Diabetes Investig 2020; 11:516-526. [PMID: 31997585 PMCID: PMC7232267 DOI: 10.1111/jdi.13219] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 01/23/2020] [Accepted: 01/24/2020] [Indexed: 02/06/2023] Open
Abstract
N-acetyl-seryl-aspartyl-lysyl-proline (AcSDKP) is an endogenous peptide that has been confirmed to show excellent organ-protective effects. Even though originally discovered as a modulator of hemotopoietic stem cells, during the recent two decades, AcSDKP has been recognized as valuable antifibrotic peptide. The antifibrotic mechanism of AcSDKP is not yet clear; we have established that AcSDKP could target endothelial-mesenchymal transition program through the induction of the endothelial fibroblast growth factor receptor signaling pathway. Also, recent reports suggested the clinical significance of AcSDKP. The aim of this review was to update recent advances of the mechanistic action of AcSDKP and discuss translational research aspects.
Collapse
Affiliation(s)
- Keizo Kanasaki
- Internal Medicine 1Faculty of MedicineShimane UniversityIzumoJapan
- Department of Diabetology and EndocrinologyKanazawa Medical UniversityUchinadaJapan
- Division of Anticipatory Molecular Food Science and TechnologyKanazawa Medical UniversityUchinadaJapan
| |
Collapse
|
14
|
Plosa EJ, Benjamin JT, Sucre JM, Gulleman PM, Gleaves LA, Han W, Kook S, Polosukhin VV, Haake SM, Guttentag SH, Young LR, Pozzi A, Blackwell TS, Zent R. β1 Integrin regulates adult lung alveolar epithelial cell inflammation. JCI Insight 2020; 5:129259. [PMID: 31873073 PMCID: PMC7098727 DOI: 10.1172/jci.insight.129259] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 12/12/2019] [Indexed: 01/04/2023] Open
Abstract
Integrins, the extracellular matrix receptors that facilitate cell adhesion and migration, are necessary for organ morphogenesis; however, their role in maintaining adult tissue homeostasis is poorly understood. To define the functional importance of β1 integrin in adult mouse lung, we deleted it after completion of development in type 2 alveolar epithelial cells (AECs). Aged β1 integrin-deficient mice exhibited chronic obstructive pulmonary disease-like (COPD-like) pathology characterized by emphysema, lymphoid aggregates, and increased macrophage infiltration. These histopathological abnormalities were preceded by β1 integrin-deficient AEC dysfunction such as excessive ROS production and upregulation of NF-κB-dependent chemokines, including CCL2. Genetic deletion of the CCL2 receptor, Ccr2, in mice with β1 integrin-deficient type 2 AECs impaired recruitment of monocyte-derived macrophages and resulted in accelerated inflammation and severe premature emphysematous destruction. The lungs exhibited reduced AEC efferocytosis and excessive numbers of inflamed type 2 AECs, demonstrating the requirement for recruited monocytes/macrophages in limiting lung injury and remodeling in the setting of a chronically inflamed epithelium. These studies support a critical role for β1 integrin in alveolar homeostasis in the adult lung.
Collapse
Affiliation(s)
| | | | | | | | - Linda A. Gleaves
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and
| | - Wei Han
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and
| | | | - Vasiliy V. Polosukhin
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and
| | - Scott M. Haake
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Nashville Veterans Affairs Medical Center, Nashville, Tennessee, USA
| | | | - Lisa R. Young
- Division of Pulmonary Medicine, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Ambra Pozzi
- Nashville Veterans Affairs Medical Center, Nashville, Tennessee, USA.,Division of Nephrology and Hypertension, Department of Medicine,,Department of Molecular Physiology and Biophysics, and
| | - Timothy S. Blackwell
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and,Nashville Veterans Affairs Medical Center, Nashville, Tennessee, USA.,Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Roy Zent
- Nashville Veterans Affairs Medical Center, Nashville, Tennessee, USA.,Division of Nephrology and Hypertension, Department of Medicine,,Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
15
|
Chen CA, Chang JM, Yang YL, Chang EE, Chen HC. Macrophage migration inhibitory factor regulates integrin-β1 and cyclin D1 expression via ERK pathway in podocytes. Biomed Pharmacother 2020; 124:109892. [PMID: 31986415 DOI: 10.1016/j.biopha.2020.109892] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/02/2020] [Accepted: 01/10/2020] [Indexed: 11/29/2022] Open
Abstract
AIMS Macrophage migration inhibitory factor (MIF) is found to increase in proliferative glomerulonephritis. MIF binds to the MIF receptor (CD74) that activates MAP kinase (ERK and p38). Integrins and cyclinD1 regulate cell proliferation, differentiation and adhesion. This study evaluates whether MIF can regulate integrin-β1/cyclin D1 expression and cell adhesion of podocytes. MAIN METHODS Expression of integrin-β1 mRNA/protein and cyclin D1 mRNA under stimulation of MIF was evaluated by real-time PCR and Western blotting. MIF receptor (CD74) and MAP kinase under MIF treatment were examined to determine which pathway regulated integrin-β1 and cyclin D1 expression. Cell adhesion was evaluated under MIF treatment and/or anti-integrin-β1 antibody by cell adhesion assay. KEY FINDINGS Protein levels of integrin-β1 were up-regulated under MIF treatment in a dosage-dependent manner. CD74 protein levels were not changed after MIF treatment. Integrin-β1 and cyclin D1 mRNA levels were up-regulated after MIF 100 ng/ml treatment. ERK inhibitor U0126 reduced MIF-induced the increase in integrin-β1 mRNA and protein expression following MIF stimulation. However, p38 inhibitor SB 203580 did not inhibit MIF-induced increase in integrin-β1 mRNA and protein expression following MIF stimulation. MIF-induced increase in cyclin D1 mRNA level also was inhibited only by U0126 following MIF stimulation. Podocyte adhesion was increased after MIF treatment, but, anti-integrin-β1 antibody decreased MIF-enhanced podocyte adhesion. SIGNIFICANCE MIF increases integrin-β1 and cyclin D1 expression through the ERK pathway in podocytes, and the up-regulated expression of integrin-β1 increases podocyte adhesion. These results provide further understanding for the role of MIF in developing proliferative glomerulonephritis.
Collapse
Affiliation(s)
- Chien-An Chen
- Department of Nephrology, Tainan Sinlau Hospital, Tainan, 701, Taiwan; Department of Health Care Administration, Chang Jung Christian University, Tainan, 711, Taiwan.
| | - Jer-Ming Chang
- Department of Nephrology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Yu-Lin Yang
- Graduate Institute of Medical Laboratory Science and Biotechnology, Chung Hwa University of Medical Technology, Tainan, 703, Taiwan
| | - Eddy-Essen Chang
- Department of Nephrology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Hung-Chun Chen
- Department of Nephrology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| |
Collapse
|
16
|
Huang M, Zhu S, Huang H, He J, Tsuji K, Jin WW, Xie D, Ham O, Capen DE, Lu W, Păunescu TG, Yang B, Lu HAJ. Integrin-Linked Kinase Deficiency in Collecting Duct Principal Cell Promotes Necroptosis of Principal Cell and Contributes to Kidney Inflammation and Fibrosis. J Am Soc Nephrol 2019; 30:2073-2090. [PMID: 31653783 PMCID: PMC6830785 DOI: 10.1681/asn.2018111162] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 07/15/2019] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Necroptosis is a newly discovered cell death pathway that plays a critical role in AKI. The involvement of integrin-linked kinase (ILK) in necroptosis has not been studied. METHODS We performed experiments in mice with an Ilk deletion in collecting duct (CD) principal cells (PCs), and cultured tubular epithelial cells treated with an ILK inhibitor or ILK siRNA knockdown. RESULTS Ilk deletion in CD PCs resulted in acute tubular injury and early mortality in mice. Progressive interstitial fibrosis and inflammation associated with the activation of the canonical TGF-β signaling cascade were detected in the kidneys of the mice lacking ILK in the CD PCs. In contrast to the minimal apoptosis detected in the animals' injured CDs, widespread necroptosis was present in ILK-deficient PCs, characterized by cell swelling, deformed mitochondria, and rupture of plasma membrane. In addition, ILK deficiency resulted in increased expression and activation of necroptotic proteins MLKL and RIPK3, and membrane translocation of MLKL in CD PCs. ILK inhibition and siRNA knockdown reduced cell survival in cultured tubular cells, concomitant with increased membrane accumulation of MLKL and/or phospho-MLKL. Administration of a necroptosis inhibitor, necrostatin-1, blocked cell death in vitro and significantly attenuated inflammation, interstitial fibrosis, and renal failure in ILK-deficient mice. CONCLUSIONS The study demonstrates the critical involvement of ILK in necroptosis through modulation of the RIPK3 and MLKL pathway and highlights the contribution of CD PC injury to the development of inflammation and interstitial fibrosis of the kidney.
Collapse
Affiliation(s)
- Ming Huang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Shuai Zhu
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Huihui Huang
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jinzhao He
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Kenji Tsuji
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - William W Jin
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Dongping Xie
- Department of Physiology, Tongji University School of Medicine, Shanghai, China; and
| | - Onju Ham
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Diane E Capen
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Weining Lu
- Renal Section, Departments of Medicine, and Pathology & Laboratory Medicine, Boston University Medical Center, Boston, Massachusetts
| | - Teodor G Păunescu
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Baoxue Yang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China;
| | - Hua A Jenny Lu
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts;
| |
Collapse
|
17
|
Molecular stratification of idiopathic nephrotic syndrome. Nat Rev Nephrol 2019; 15:750-765. [DOI: 10.1038/s41581-019-0217-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2019] [Indexed: 01/03/2023]
|
18
|
Niculovic KM, Blume L, Wedekind H, Kats E, Albers I, Groos S, Abeln M, Schmitz J, Beuke E, Bräsen JH, Melk A, Schiffer M, Weinhold B, Münster-Kühnel AK. Podocyte-Specific Sialylation-Deficient Mice Serve as a Model for Human FSGS. J Am Soc Nephrol 2019; 30:1021-1035. [PMID: 31040189 DOI: 10.1681/asn.2018090951] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 02/26/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND The etiology of steroid-resistant nephrotic syndrome, which manifests as FSGS, is not completely understood. Aberrant glycosylation is an often underestimated factor for pathologic processes, and structural changes in the glomerular endothelial glycocalyx have been correlated with models of nephrotic syndrome. Glycans are frequently capped by sialic acid (Sia), and sialylation's crucial role for kidney function is well known. Human podocytes are highly sialylated; however, sialylation's role in podocyte homeostasis remains unclear. METHODS We generated a podocyte-specific sialylation-deficient mouse model (PCmas-/- ) by targeting CMP-Sia synthetase, and used histologic and ultrastructural analysis to decipher the phenotype. We applied CRISPR/Cas9 technology to generate immortalized sialylation-deficient podocytes (asialo-podocytes) for functional studies. RESULTS Progressive loss of sialylation in PCmas-/- mice resulted in onset of proteinuria around postnatal day 28, accompanied by foot process effacement and loss of slit diaphragms. Podocyte injury led to severe glomerular defects, including expanded capillary lumen, mesangial hypercellularity, synechiae formation, and podocyte loss. In vivo, loss of sialylation resulted in mislocalization of slit diaphragm components, whereas podocalyxin localization was preserved. In vitro, asialo-podocytes were viable, able to proliferate and differentiate, but showed impaired adhesion to collagen IV. CONCLUSIONS Loss of cell-surface sialylation in mice resulted in disturbance of podocyte homeostasis and FSGS development. Impaired podocyte adhesion to the glomerular basement membrane most likely contributed to disease development. Our data support the notion that loss of sialylation might be part of the complex process causing FSGS. Sialylation, such as through a Sia supplementation therapy, might provide a new therapeutic strategy to cure or delay FSGS and potentially other glomerulopathies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Esther Beuke
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Hannover, Germany; and
| | - Jan H Bräsen
- Nephropathology Unit, Institute of Pathology, and
| | - Anette Melk
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Hannover, Germany; and
| | - Mario Schiffer
- Department of Nephrology and Hypertension, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | | | | |
Collapse
|
19
|
Musah S, Dimitrakakis N, Camacho DM, Church GM, Ingber DE. Directed differentiation of human induced pluripotent stem cells into mature kidney podocytes and establishment of a Glomerulus Chip. Nat Protoc 2019; 13:1662-1685. [PMID: 29995874 DOI: 10.1038/s41596-018-0007-8] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Protocols have been established to direct the differentiation of human induced pluripotent stem (iPS) cells into nephron progenitor cells and organoids containing many types of kidney cells, but it has been difficult to direct the differentiation of iPS cells to form specific types of mature human kidney cells with high yield. Here, we describe a detailed protocol for the directed differentiation of human iPS cells into mature, post-mitotic kidney glomerular podocytes with high (>90%) efficiency within 26 d and under chemically defined conditions, without genetic manipulations or subpopulation selection. We also describe how these iPS cell-derived podocytes may be induced to form within a microfluidic organ-on-a-chip (Organ Chip) culture device to build a human kidney Glomerulus Chip that mimics the structure and function of the kidney glomerular capillary wall in vitro within 35 d (starting with undifferentiated iPS cells). The podocyte differentiation protocol requires skills for culturing iPS cells, and the development of a Glomerulus Chip requires some experience with building and operating microfluidic cell culture systems. This method could be useful for applications in nephrotoxicity screening, therapeutic development, and regenerative medicine, as well as mechanistic study of kidney development and disease.
Collapse
Affiliation(s)
- Samira Musah
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA.,Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Nikolaos Dimitrakakis
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - Diogo M Camacho
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - George M Church
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA.,Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA. .,Vascular Biology Program, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA. .,Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, MA, USA.
| |
Collapse
|
20
|
Dipeptidyl peptidase-4 inhibition and renoprotection: the role of antifibrotic effects. Curr Opin Nephrol Hypertens 2018; 26:56-66. [PMID: 27820706 DOI: 10.1097/mnh.0000000000000291] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW This article analyzes the potential beneficial effects of dipeptidyl peptidase (DPP)-4 inhibitors on renal diseases. RECENT FINDINGS The pathological significance of DPP-4, either dependent or independent on catalytic activities, on renal diseases has been reported in preclinical studies. With regard to this, we have shown that damaged endothelial cells are converted to a mesenchymal cell phenotype, which is associated with the induction of DPP-4 in endothelial cells. The endothelial mesenchymal transition may contribute to kidney fibrosis; indeed, the antifibrotic effects of DPP-4 inhibitors have been reported elsewhere. However, even though such potential benefits of DPP-4 inhibitors on renal diseases were shown in preclinical studies, clinical trials have not yet revealed significant benefits in renal hard outcomes of DPP-4 inhibitors. SUMMARY To completely understand the beneficial effects of DPP-4 inhibitors, both the following studies are required: first, preclinical studies that analyze deeper molecular mechanisms of DPP-4 inhibition, and, second, clinical studies that investigate whether such potential beneficial effects of DPP-4 inhibitors are relevant to the patients in the clinic.
Collapse
|
21
|
Yang R, Hong H, Wang M, Ma Z. Correlation Between Single-Nucleotide Polymorphisms Within miR-30a and Related Target Genes and Risk or Prognosis of Nephrotic Syndrome. DNA Cell Biol 2018; 37:233-243. [PMID: 29356585 DOI: 10.1089/dna.2017.4024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
This study was aimed to figure out the association of single-nucleotide polymorphisms (SNPs) within miR-30a and its downstream molecules (i.e., Notch1, Snail1, p53, CD73, and TET1) with susceptibility to and prognosis of nephrotic syndrome (NS). In the aggregate, 265 patients and 281 healthy controls were gathered, and related laboratory indicators were examined. The miR-30a, Notch1, Snail1, TET1, p53, and CD73 expressions were also evaluated by quantitative real-time polymerase chain reaction (qRT-PCR), immunohistochemistry, or enzyme-linked immunosorbent assay. Besides, the SNPs were genotyped by RT-PCR with aid of ABI-PRISM™ 377 DNA sequencing instrument. As a result, the NS patients were correlated with remarkably higher 24-h protein excretion, random urine protein/creatinine (UPCR), and serum creatinine, along with lower estimated glomerular filtration rate and serum albumin, when compared with normal subjects (p < 0.05). Furthermore, significant correlations were present between miR-30a expression and the expressions of Notch1 (rs = -0.350), p53 (rs = -0.339), CD73 (rs = -0.300), TET1 (rs = -0.249), and Snail1 (rs = -0.829) (all p < 0.05). The SNPs of miR-30a [i.e., rs2222722 (C>T)], Notch1 [i.e., rs3124599 (G>A), rs3124591 (C>T), and rs139994842 (G>A)], Snail1 [i.e., rs6020178 (T>C)], p53 [i.e., rs1042522 (C>G)], and CD73 [i.e., rs9444348 (G>A) and rs4431401 (T>C)] were significantly correlated with both differed NS risk and altered hormone sensitivity to NS (all p < 0.05). Moreover, haplotype AC of CD73 and haplotype ATG of Notch1 were the helpful factors against NS (p < 0.05), yet haplotype GT of CD73 functioned oppositely (p < 0.05). The haplotype AT of CD73 was beneficial to the NS patients for that the carriers could be treated with hormones without severe complications (p < 0.05). Conclusively, the SNPs situated within miR-30a and its downstream molecules (i.e., Notch1, Snail1, p53, CD73, and TET1) could become the promising biomarkers for both NS diagnosis and prediction of NS prognosis.
Collapse
Affiliation(s)
- Ruiheng Yang
- Department of Nephrology, Liaocheng People's Hospital , Liaocheng, China
| | - Hong Hong
- Department of Nephrology, Liaocheng People's Hospital , Liaocheng, China
| | - Mengjun Wang
- Department of Nephrology, Liaocheng People's Hospital , Liaocheng, China
| | - Zhongchao Ma
- Department of Nephrology, Liaocheng People's Hospital , Liaocheng, China
| |
Collapse
|
22
|
Lausecker F, Tian X, Inoue K, Wang Z, Pedigo CE, Hassan H, Liu C, Zimmer M, Jinno S, Huckle AL, Hamidi H, Ross RS, Zent R, Ballestrem C, Lennon R, Ishibe S. Vinculin is required to maintain glomerular barrier integrity. Kidney Int 2017; 93:643-655. [PMID: 29241625 PMCID: PMC5846847 DOI: 10.1016/j.kint.2017.09.021] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 08/23/2017] [Accepted: 09/14/2017] [Indexed: 01/08/2023]
Abstract
Cell-matrix interactions and podocyte intercellular junctions are key for maintaining the glomerular filtration barrier. Vinculin, a cytoplasmic protein, couples actin filaments to integrin-mediated cell-matrix adhesions and to cadherin-based intercellular junctions. Here, we examined the role of vinculin in podocytes by the generation of a podocyte-specific knockout mouse. Mice lacking podocyte vinculin had increased albuminuria and foot process effacement following injury in vivo. Analysis of primary podocytes isolated from the mutant mice revealed defects in cell protrusions, altered focal adhesion size and signaling, as well as impaired cell migration. Furthermore, we found a marked mislocalization of the intercellular junction protein zonula occludens-1. In kidney sections from patients with focal segmental glomerulosclerosis, minimal change disease and membranous nephropathy, we observed dramatic differences in the expression levels and localization of vinculin. Thus, our results suggest that vinculin is necessary to maintain the integrity of the glomerular filtration barrier by modulating podocyte foot processes and stabilizing intercellular junctions.
Collapse
Affiliation(s)
- Franziska Lausecker
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Xuefei Tian
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Kazunori Inoue
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Zhen Wang
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Christopher E Pedigo
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Hossam Hassan
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Chang Liu
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Margaret Zimmer
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Stephanie Jinno
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Abby L Huckle
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Hellyeh Hamidi
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Robert S Ross
- Department of Medicine/Cardiology, University of California, San Diego, School of Medicine, La Jolla, California, USA and Veterans Affairs San Diego Healthcare System, San Diego, California, USA
| | - Roy Zent
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Veterans Affairs Hospital, Nashville, Tennessee, USA
| | - Christoph Ballestrem
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| | - Rachel Lennon
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK; School of Biology, Faculty of Biology, Medicine and Health, University of Medicine, Manchester Academic Health Science Centre, UK.
| | - Shuta Ishibe
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.
| |
Collapse
|
23
|
Rogg M, Yasuda-Yamahara M, Abed A, Dinse P, Helmstädter M, Conzelmann AC, Frimmel J, Sellung D, Biniossek ML, Kretz O, Grahammer F, Schilling O, Huber TB, Schell C. The WD40-domain containing protein CORO2B is specifically enriched in glomerular podocytes and regulates the ventral actin cytoskeleton. Sci Rep 2017; 7:15910. [PMID: 29162887 PMCID: PMC5698439 DOI: 10.1038/s41598-017-15844-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 10/31/2017] [Indexed: 11/13/2022] Open
Abstract
Podocytes are highly specialized epithelial cells essentially required to establish and maintain the kidney filtration barrier. Due to their complex cellular architecture these cells rely on an elaborated cytoskeletal apparatus providing plasticity as well as adaptive adhesion properties to withstand significant physical filtration forces. However, our knowledge about podocyte specific components of the cytoskeletal machinery is still incomplete. Employing cross-analysis of various quantitative omics-data sets we identify the WD40-domain containing protein CORO2B as a podocyte enriched protein. Furthermore, we demonstrate the distinct localization pattern of CORO2B to the ventral actin cytoskeleton serving as a physical linkage module to cell-matrix adhesion sites. Analysis of a novel Coro2b knockout mouse revealed that CORO2B modulates stress response of podocytes in an experimental nephropathy model. Using quantitative focal adhesome proteomics we identify the recruitment of CFL1 via CORO2B to focal adhesions as an underlying mechanism. Thus, we describe CORO2B as a novel podocyte enriched protein influencing cytoskeletal plasticity and stress adaptation.
Collapse
Affiliation(s)
- M Rogg
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - M Yasuda-Yamahara
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Medicine, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - A Abed
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - P Dinse
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - M Helmstädter
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - A C Conzelmann
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - J Frimmel
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - D Sellung
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - M L Biniossek
- Institute of Molecular Medicine and Cell Research, University of Freiburg, Freiburg, Germany
| | - O Kretz
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Institute of Anatomy and Cell Biology, Dept. for Neuroanatomy, Medical Faculty, Albert-Ludwigs-University Freiburg, Freiburg, Germany.,III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - F Grahammer
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - O Schilling
- Institute of Molecular Medicine and Cell Research, University of Freiburg, Freiburg, Germany.,BIOSS Center for Biological Signalling Studies and Center for Systems Biology (ZBSA), Albert-Ludwigs-University, Freiburg, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - T B Huber
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany. .,III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany. .,BIOSS Center for Biological Signalling Studies and Center for Systems Biology (ZBSA), Albert-Ludwigs-University, Freiburg, Germany.
| | - C Schell
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Institute for Surgical Pathology, Medical Center Freiburg, Freiburg, Germany.,Berta-Ottenstein Programme, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
24
|
Abstract
Podocytes exhibit a unique cytoskeletal architecture that is fundamentally linked to their function in maintaining the kidney filtration barrier. The cytoskeleton regulates podocyte shape, structure, stability, slit diaphragm insertion, adhesion, plasticity, and dynamic response to environmental stimuli. Genetic mutations demonstrate that even slight impairment of the podocyte cytoskeletal apparatus results in proteinuria and glomerular disease. Moreover, mechanisms underpinning all acquired glomerular pathologies converge on disruption of the cytoskeleton, suggesting that this subcellular structure could be targeted for therapeutic purposes. This review summarizes our current understanding of the function of the cytoskeleton in podocytes and the associated implications for pathophysiology.
Collapse
Affiliation(s)
- Christoph Schell
- Institute of Surgical Pathology and.,Department of Medicine IV, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Tobias B Huber
- Department of Medicine IV, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Germany; .,III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; and.,BIOSS Centre for Biological Signalling Studies and Center for Biological Systems Analysis (ZBSA), Albert-Ludwigs-University, Freiburg, Germany
| |
Collapse
|
25
|
Michgehl U, Pavenstädt H, Vollenbröker B. Cross talk between the Crumbs complex and Hippo signaling in renal epithelial cells. Pflugers Arch 2017; 469:917-926. [PMID: 28612137 DOI: 10.1007/s00424-017-2004-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 05/18/2017] [Accepted: 05/19/2017] [Indexed: 12/12/2022]
Abstract
Cell polarity has a crucial role in organizing cells into tissues and in mediating transport processes and cell-cell communication. Especially the cells of the nephron require apicobasal polarity to establish and maintain their barrier function. The Crumbs complex including the integral membrane protein Crumbs, as well as Pals1 and Patj, is essential for the establishment of apicobasal polarity. The interactions of the core proteins and the interplay with other processes have been characterized in various epithelial cell lines in detail. Notably, Crb2 and Crb3 are expressed within the kidney and play an important role in the proper function of podocytes and tubules, respectively. The interaction of polarity proteins and components of the Hippo pathway-an evolutionarily highly conserved kinase cascade regulating cell proliferation, organ size, and tissue regeneration-has been discovered recently. Here, we discuss potential molecular and physiological links between the Crumbs complex and the Hippo pathway in renal cells.
Collapse
Affiliation(s)
- U Michgehl
- Internal Medicine D, University Hospital of Muenster, Albert-Schweitzer-Campus 1, A14, D-48149, Muenster, Germany.
| | - H Pavenstädt
- Internal Medicine D, University Hospital of Muenster, Albert-Schweitzer-Campus 1, A14, D-48149, Muenster, Germany
| | - B Vollenbröker
- Internal Medicine D, University Hospital of Muenster, Albert-Schweitzer-Campus 1, A14, D-48149, Muenster, Germany
| |
Collapse
|
26
|
The FERM protein EPB41L5 regulates actomyosin contractility and focal adhesion formation to maintain the kidney filtration barrier. Proc Natl Acad Sci U S A 2017; 114:E4621-E4630. [PMID: 28536193 DOI: 10.1073/pnas.1617004114] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Podocytes form the outer part of the glomerular filter, where they have to withstand enormous transcapillary filtration forces driving glomerular filtration. Detachment of podocytes from the glomerular basement membrane precedes most glomerular diseases. However, little is known about the regulation of podocyte adhesion in vivo. Thus, we systematically screened for podocyte-specific focal adhesome (FA) components, using genetic reporter models in combination with iTRAQ-based mass spectrometry. This approach led to the identification of FERM domain protein EPB41L5 as a highly enriched podocyte-specific FA component in vivo. Genetic deletion of Epb41l5 resulted in severe proteinuria, detachment of podocytes, and development of focal segmental glomerulosclerosis. Remarkably, by binding and recruiting the RhoGEF ARGHEF18 to the leading edge, EPB41L5 directly controls actomyosin contractility and subsequent maturation of focal adhesions, cell spreading, and migration. Furthermore, EPB41L5 controls matrix-dependent outside-in signaling by regulating the focal adhesome composition. Thus, by linking extracellular matrix sensing and signaling, focal adhesion maturation, and actomyosin activation EPB41L5 ensures the mechanical stability required for podocytes at the kidney filtration barrier. Finally, a diminution of EPB41L5-dependent signaling programs appears to be a common theme of podocyte disease, and therefore offers unexpected interventional therapeutic strategies to prevent podocyte loss and kidney disease progression.
Collapse
|
27
|
Mature induced-pluripotent-stem-cell-derived human podocytes reconstitute kidney glomerular-capillary-wall function on a chip. Nat Biomed Eng 2017; 1. [PMID: 29038743 PMCID: PMC5639718 DOI: 10.1038/s41551-017-0069] [Citation(s) in RCA: 320] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
An in vitro model of the human kidney glomerulus — the major site of blood filtration — could facilitate drug discovery and illuminate kidney-disease mechanisms. Microfluidic organ-on-a-chip technology has been used to model the human proximal tubule, yet a kidney-glomerulus-on-a-chip has not been possible because of the lack of functional human podocytes — the cells that regulate selective permeability in the glomerulus. Here, we demonstrate an efficient (> 90%) and chemically defined method for directing the differentiation of human induced pluripotent stem (hiPS) cells into podocytes that express markers of the mature phenotype (nephrin+, WT1+, podocin+, Pax2−) and that exhibit primary and secondary foot processes. We also show that the hiPS-cell-derived podocytes produce glomerular basement-membrane collagen and recapitulate the natural tissue/tissue interface of the glomerulus, as well as the differential clearance of albumin and inulin, when co-cultured with human glomerular endothelial cells in an organ-on-a-chip microfluidic device. The glomerulus-on-a-chip also mimics adriamycin-induced albuminuria and podocyte injury. This in vitro model of human glomerular function with mature human podocytes may facilitate drug development and personalized-medicine applications.
Collapse
|
28
|
Reiser J, Lee HW, Gupta V, Altintas MM. A High-Content Screening Technology for Quantitatively Studying Podocyte Dynamics. Adv Chronic Kidney Dis 2017; 24:183-188. [PMID: 28501082 DOI: 10.1053/j.ackd.2017.04.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Podocytes form the visceral layer of a kidney glomerulus and express a characteristic octopus-like cellular architecture specialized for the ultrafiltration of blood. The cytoskeletal dynamics and structural elasticity of podocytes rely on the self-organization of highly interconnected actin bundles, and the maintenance of these features is important for the intact glomerular filtration. Development of more differentiated podocytes in culture has dramatically increased our understanding of the molecular mechanisms regulating podocyte actin dynamics. Podocytes are damaged in a variety of kidney diseases, and therapies targeting podocytes are being investigated with increasing efforts. Association between podocyte damage and disease severity-or between podocyte recovery and the performance of therapeutic molecules-have been the venues of research for years. In this perspective, more standardized high--content screening has emerged as a powerful tool for visualization and analysis of podocyte morphology. This high-throughput fluorescence microscopy technique is based on an automated image analysis with simultaneous detection of various phenotypes (multiplexing) across multiple phenotypic parameters (multiparametric). Here, we review the principles of high-content screening technology and summarize efforts to carry out small compound screen using podocytes.
Collapse
|
29
|
Kidney development and perspectives for organ engineering. Cell Tissue Res 2017; 369:171-183. [DOI: 10.1007/s00441-017-2616-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 03/21/2017] [Indexed: 12/17/2022]
|
30
|
Viquez OM, Yazlovitskaya EM, Tu T, Mernaugh G, Secades P, McKee KK, Georges-Labouesse E, De Arcangelis A, Quaranta V, Yurchenco P, Gewin LC, Sonnenberg A, Pozzi A, Zent R. Integrin alpha6 maintains the structural integrity of the kidney collecting system. Matrix Biol 2016; 57-58:244-257. [PMID: 28043890 DOI: 10.1016/j.matbio.2016.12.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 12/10/2016] [Indexed: 10/20/2022]
Abstract
Laminins are a major constituent of the basement membranes of the kidney collecting system. Integrins, transmembrane receptors formed by non-covalently bound α and β subunits, serve as laminin receptors, but their role in development and homeostasis of the kidney collecting system is poorly defined. Integrin α3β1, one of the major laminin receptors, plays a minor role in kidney collecting system development, while the role of α6 containing integrins (α6β1 and α6β4), the other major laminin receptors, is unknown. Patients with mutations in α6 containing integrins not only develop epidermolysis bullosa, but also have abnormalities in the kidney collecting system. In this study, we show that selectively deleting the α6 or β4 integrin subunits at the initiation of ureteric bud development in mice does not affect morphogenesis. However, the collecting system becomes dilated and dysmorphic as the mice age. The collecting system in both null genotypes was also highly susceptible to unilateral ureteric obstruction injury with evidence of excessive tubule dilatation and epithelial cell apoptosis. Mechanistically, integrin α6-null collecting duct cells are unable to withstand high mechanical force when adhered to laminin. Thus, we conclude that α6 integrins are important for maintaining the integrity of the kidney collecting system by enhancing tight adhesion of the epithelial cells to the basement membrane. These data give a mechanistic explanation for the association between kidney collecting system abnormalities in patients and epidermolysis bullosa.
Collapse
Affiliation(s)
- Olga M Viquez
- Division of Nephrology and Hypertension and Vanderbilt Center for Kidney Disease, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Eugenia M Yazlovitskaya
- Division of Nephrology and Hypertension and Vanderbilt Center for Kidney Disease, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Tianxiang Tu
- Division of Nephrology and Hypertension and Vanderbilt Center for Kidney Disease, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Glenda Mernaugh
- Division of Nephrology and Hypertension and Vanderbilt Center for Kidney Disease, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Pablo Secades
- Division of Cell Biology, Netherlands Cancer Institute, 1066 CX Amsterdam, Netherlands
| | - Karen K McKee
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | - Elizabeth Georges-Labouesse
- Department of Development and Stem Cells, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, Illkirch, Inserm, U964, Illkirch, CNRS, UMR 7104, Illkirch, Université de Strasbourg, Strasbourg, France
| | - Adele De Arcangelis
- Department of Development and Stem Cells, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, Illkirch, Inserm, U964, Illkirch, CNRS, UMR 7104, Illkirch, Université de Strasbourg, Strasbourg, France
| | - Vito Quaranta
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Peter Yurchenco
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | - Leslie C Gewin
- Division of Nephrology and Hypertension and Vanderbilt Center for Kidney Disease, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Veterans Affairs Hospital, Nashville, TN 37232, USA
| | - Arnoud Sonnenberg
- Division of Cell Biology, Netherlands Cancer Institute, 1066 CX Amsterdam, Netherlands
| | - Ambra Pozzi
- Division of Nephrology and Hypertension and Vanderbilt Center for Kidney Disease, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Veterans Affairs Hospital, Nashville, TN 37232, USA
| | - Roy Zent
- Division of Nephrology and Hypertension and Vanderbilt Center for Kidney Disease, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Veterans Affairs Hospital, Nashville, TN 37232, USA.
| |
Collapse
|
31
|
Marshall CB. Rethinking glomerular basement membrane thickening in diabetic nephropathy: adaptive or pathogenic? Am J Physiol Renal Physiol 2016; 311:F831-F843. [PMID: 27582102 DOI: 10.1152/ajprenal.00313.2016] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 08/21/2016] [Indexed: 12/12/2022] Open
Abstract
Diabetic nephropathy (DN) is the leading cause of chronic kidney disease in the United States and is a major cause of cardiovascular disease and death. DN develops insidiously over a span of years before clinical manifestations, including microalbuminuria and declining glomerular filtration rate (GFR), are evident. During the clinically silent period, structural lesions develop, including glomerular basement membrane (GBM) thickening, mesangial expansion, and glomerulosclerosis. Once microalbuminuria is clinically apparent, structural lesions are often considerably advanced, and GFR decline may then proceed rapidly toward end-stage kidney disease. Given the current lack of sensitive biomarkers for detecting early DN, a shift in focus toward examining the cellular and molecular basis for the earliest structural change in DN, i.e., GBM thickening, may be warranted. Observed within one to two years following the onset of diabetes, GBM thickening precedes clinically evident albuminuria. In the mature glomerulus, the podocyte is likely key in modifying the GBM, synthesizing and assembling matrix components, both in physiological and pathological states. Podocytes also secrete matrix metalloproteinases, crucial mediators in extracellular matrix turnover. Studies have shown that the critical podocyte-GBM interface is disrupted in the diabetic milieu. Just as healthy podocytes are essential for maintaining the normal GBM structure and function, injured podocytes likely have a fundamental role in upsetting the balance between the GBM's synthetic and degradative pathways. This article will explore the biological significance of GBM thickening in DN by reviewing what is known about the GBM's formation, its maintenance during health, and its disruption in DN.
Collapse
Affiliation(s)
- Caroline B Marshall
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and Department of Veterans Affairs Medical Center, Birmingham, Alabama
| |
Collapse
|
32
|
Abstract
Genetic studies of hereditary forms of nephrotic syndrome have identified several proteins that are involved in regulating the permselective properties of the glomerular filtration system. Further extensive research has elucidated the complex molecular basis of the glomerular filtration barrier and clearly established the pivotal role of podocytes in the pathophysiology of glomerular diseases. Podocyte architecture is centred on focal adhesions and slit diaphragms - multiprotein signalling hubs that regulate cell morphology and function. A highly interconnected actin cytoskeleton enables podocytes to adapt in order to accommodate environmental changes and maintain an intact glomerular filtration barrier. Actin-based endocytosis has now emerged as a regulator of podocyte integrity, providing an impetus for understanding the precise mechanisms that underlie the steady-state control of focal adhesion and slit diaphragm components. This Review outlines the role of actin dynamics and endocytosis in podocyte biology, and discusses how molecular heterogeneity in glomerular disorders could be exploited to deliver more rational therapeutic interventions, paving the way for targeted medicine in nephrology.
Collapse
|
33
|
Wu Y, Xu JX, El-Jouni W, Lu T, Li S, Wang Q, Tran M, Yu W, Wu M, Barrera IE, Bonventre JV, Zhou J, Denker BM, Kong T. Gα12 is required for renal cystogenesis induced by Pkd1 inactivation. J Cell Sci 2016; 129:3675-3684. [PMID: 27505895 DOI: 10.1242/jcs.190496] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 07/21/2016] [Indexed: 01/09/2023] Open
Abstract
Mutation of PKD1, encoding the protein polycystin-1 (PC1), is the main cause of autosomal dominant polycystic kidney disease (ADPKD). The signaling pathways downstream of PC1 in ADPKD are still not fully understood. Here, we provide genetic evidence for the necessity of Gα12 (encoded by Gna12, hereafter Gα12) for renal cystogenesis induced by Pkd1 knockout. There was no phenotype in mice with deletion of Gα12 (Gα12-/-). Polyinosine-polycytosine (pI:pC)-induced deletion of Pkd1 (Mx1Cre+Pkd1f/fGα12+/+) in 1-week-old mice resulted in multiple kidney cysts by 9 weeks, but the mice with double knockout of Pkd1 and Gα12 (Mx1Cre+Pkd1f/fGα12-/-) had no structural and functional abnormalities in the kidneys. These mice could survive more than one year without kidney abnormalities except multiple hepatic cysts in some mice, which indicates that the effect of Gα12 on cystogenesis is kidney specific. Furthermore, Pkd1 knockout promoted Gα12 activation, which subsequently decreased cell-matrix and cell-cell adhesion by affecting the function of focal adhesion and E-cadherin, respectively. Our results demonstrate that Gα12 is required for the development of kidney cysts induced by Pkd1 mutation in mouse ADPKD.
Collapse
Affiliation(s)
- Yong Wu
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jen X Xu
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Wassim El-Jouni
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Tzongshi Lu
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Suyan Li
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Qingyi Wang
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Mei Tran
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA Renal Division, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Wanfeng Yu
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Maoqing Wu
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ivan E Barrera
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Joseph V Bonventre
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jing Zhou
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Bradley M Denker
- Renal Division, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Tianqing Kong
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
34
|
Kanasaki K. The pathological significance of dipeptidyl peptidase-4 in endothelial cell homeostasis and kidney fibrosis. Diabetol Int 2016; 7:212-220. [PMID: 30603266 DOI: 10.1007/s13340-016-0281-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 07/22/2016] [Indexed: 02/06/2023]
Abstract
Endothelial dysfunction and tubulointerstitial fibrosis are characteristics of diabetic kidneys. Recent evidence has suggested that the diabetic kidney is associated with dipeptidyl peptidase (DPP)-4 overexpression in endothelial cells. Several insults can induce endothelial cells to alter their phenotype into a mesenchymal-like phenotype via endothelial-mesenchymal transition (EndMT), which plays pivotal roles in tissue fibrosis. We have recently revealed the fibrogenic role of DPP-4 through the induction of EndMT in diabetic kidneys. This review mainly focuses on the biological and pathological significance of DPP-4 overexpression in endothelial cells through the mechanisms of endothelial homeostasis defects, EndMT, and kidney fibrosis.
Collapse
Affiliation(s)
- Keizo Kanasaki
- 1Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Ishikawa 920-0293 Japan.,2Division of Anticipatory Molecular Food Science and Technology, Kanazawa Medical University, Uchinada, Ishikawa 920-0293 Japan
| |
Collapse
|
35
|
Tian X, Ishibe S. Targeting the podocyte cytoskeleton: from pathogenesis to therapy in proteinuric kidney disease. Nephrol Dial Transplant 2016; 31:1577-83. [PMID: 26968197 DOI: 10.1093/ndt/gfw021] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 01/24/2016] [Indexed: 01/11/2023] Open
Abstract
Glomerular injury often incites a progression to chronic kidney disease, which affects millions of patients worldwide. Despite our current understanding of this disease's pathogenesis, there is still a lack of therapy available to curtail its progression. However, exciting new data strongly suggest the podocyte-an actin-rich, terminally differentiated epithelial cell that lines the outside of the glomerular filtration barrier-as a therapeutic target. The importance of podocytes in the pathogenesis of human nephrotic syndrome is best characterized by identification of genetic mutations, many of which regulate the actin cytoskeleton. The intricate regulation of the podocyte actin cytoskeleton is fundamental in preserving an intact glomerular filtration barrier, and this knowledge has inspired new research targeting actin-regulating proteins in these cells. This review will shed light on recent findings, which have furthered our understanding of the molecular mechanisms regulating podocyte actin dynamics, as well as discoveries that have therapeutic implications in the treatment of proteinuric kidney disease.
Collapse
Affiliation(s)
- Xuefei Tian
- Section of Nephrology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Shuta Ishibe
- Section of Nephrology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
36
|
Verma R, Venkatareddy M, Kalinowski A, Patel SR, Garg P. Integrin Ligation Results in Nephrin Tyrosine Phosphorylation In Vitro. PLoS One 2016; 11:e0148906. [PMID: 26848974 PMCID: PMC4743922 DOI: 10.1371/journal.pone.0148906] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 01/24/2016] [Indexed: 11/19/2022] Open
Abstract
Nephrin is expressed at the basolateral aspect of podocytes and is an important signaling protein at the glomerular slit diaphragm. In vitro studies have demonstrated that Nephrin phosphorylation-dependent signaling is able to assemble a protein complex that is able to polymerize actin. However, proximal signaling events that result in nephrin tyrosine phosphorylation are not well understood. Nephrin deletion in mice and human nephrin mutations result in developmental failure of the podocyte intercellular junction resutling in proteinuria. This has been presumed to be due to a failure to respond to an external polarized cue in the absence of nephrin or a failure to transduce an outside-in signal in patients with nephrin mutations. The nephrin extracellular domain binds to itself or neph1 across the foot process intercellular junction. Nephrin is tyrosine phosphorylation-silent in healthy glomeruli when presumably the nephrin extracellular domain is in an engaged state. These observations raise the possibility of an alternate proximal signaling mechanism that might be responsible for nephrin tyrosine phosphorylation. Here we present data showing that integrin engagement at the basal aspect of cultured podocytes results in nephrin tyrosine phosphorylation. This is abrogated by incubating podocytes with an antibody that prevents integrin β1 ligation and activation in response to binding to extracellular matrix. Furthermore, nephrin tyrosine phosphorylation was observed in podocytes expressing a membrane-targeted nephrin construct that lacks the extracellular domain. We propose, integrin-activation based signaling might be responsible for nephrin phosphorylation rather than engagment of the nephrin extracellular domain by a ligand.
Collapse
Affiliation(s)
- Rakesh Verma
- Division of Nephroloigy, Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Madhusudan Venkatareddy
- Division of Nephroloigy, Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Anne Kalinowski
- Division of Nephroloigy, Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Sanjeevkumar R. Patel
- Division of Nephroloigy, Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Puneet Garg
- Division of Nephroloigy, Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
37
|
Abstract
Podocytes are highly specialized cells of the kidney glomerulus that wrap around capillaries and that neighbor cells of the Bowman’s capsule. When it comes to glomerular filtration, podocytes play an active role in preventing plasma proteins from entering the urinary ultrafiltrate by providing a barrier comprising filtration slits between foot processes, which in aggregate represent a dynamic network of cellular extensions. Foot processes interdigitate with foot processes from adjacent podocytes and form a network of narrow and rather uniform gaps. The fenestrated endothelial cells retain blood cells but permit passage of small solutes and an overlying basement membrane less permeable to macromolecules, in particular to albumin. The cytoskeletal dynamics and structural plasticity of podocytes as well as the signaling between each of these distinct layers are essential for an efficient glomerular filtration and thus for proper renal function. The genetic or acquired impairment of podocytes may lead to foot process effacement (podocyte fusion or retraction), a morphological hallmark of proteinuric renal diseases. Here, we briefly discuss aspects of a contemporary view of podocytes in glomerular filtration, the patterns of structural changes in podocytes associated with common glomerular diseases, and the current state of basic and clinical research.
Collapse
Affiliation(s)
- Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Mehmet M Altintas
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
38
|
Koeners MP, Wesseling S, Sánchez M, Braam B, Joles JA. Perinatal Inhibition of NF-KappaB Has Long-Term Antihypertensive and Renoprotective Effects in Fawn-Hooded Hypertensive Rats. Am J Hypertens 2016; 29:123-31. [PMID: 25958302 DOI: 10.1093/ajh/hpv065] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 04/02/2015] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Inhibition of transcription factor nuclear factor-kappa B (NFκB) is beneficial in various models of hypertension and renal disease. We hypothesized first that NFκB inhibition during renal development ameliorates hereditary hypertensive renal disease and next whether this was mediated via suppression of peroxisome proliferator-activated receptor (PPAR)γ coactivator 1α (PGC-1α). METHODS AND RESULTS Prior to the development of renal injury in fawn-hooded hypertensive (FHH) rats, a model of hypertension, glomerular hyperfiltration, and progressive renal injury, NFkB activity, measured by nuclear protein expression of NFkB subunit p65, was enhanced twofold in 2-day-old male and female FHH kidneys as compared to normotensive Wistar-Kyoto (WKY) rats (P < 0.05). Treating FHH dams with pyrrolidine di thio carbamate (PDTC), an NFκB inhibitor, from 2 weeks before birth to 4 weeks after birth diminished NFkB activity in 2-day-FHH offspring to 2-day-WKY levels (P < 0.01). Perinatal PDTC reduced systolic blood pressure from 20 weeks onwards by on average 25 mm Hg (P < 0.001) and ameliorated proteinuria (P < 0.05) and glomerulosclerosis (P < 0.05). In kidneys of 2-day-, 2-week-, and adult offspring of PDTC-treated FHH dams, PGC-1α was induced on average by 67% (quantitative polymerase chain reaction (qPCR)) suggesting that suppression of this factor by NFkB could be involved in renal damage. Follow-up experiments with perinatal pioglitazone (Pio), a PPARγ agonist, failed to confer persistent antihypertensive or renoprotective effects. CONCLUSIONS Perinatal inhibition of enhanced active renal NFκB in 2-day FHH had persistent antihypertensive and renoprotective effects. However, this was not the case for PPARγ stimulation. NFkB stimulation is therefore involved in renal damage in the FHH model of proteinuric renal disease by pathways other than via PPARγ.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Antihypertensive Agents/therapeutic use
- Blood Pressure/drug effects
- Disease Models, Animal
- Female
- Gene Expression Regulation, Developmental
- Hypertension, Renal/complications
- Hypertension, Renal/drug therapy
- Hypertension, Renal/genetics
- Hypertension, Renal/metabolism
- Hypertension, Renal/physiopathology
- Hypertension, Renal/prevention & control
- Male
- NF-kappa B/antagonists & inhibitors
- NF-kappa B/biosynthesis
- NF-kappa B/genetics
- Nephritis/genetics
- Nephritis/physiopathology
- Nephritis/prevention & control
- RNA/genetics
- Rats
- Rats, Inbred WKY
- Renal Circulation
Collapse
Affiliation(s)
- Maarten P Koeners
- Department of Nephrology and Hypertension, University Medical Center, Utrecht, Netherlands; School of Physiology and Pharmacology, University of Bristol, Bristol, UK
| | - Sebastiaan Wesseling
- Department of Nephrology and Hypertension, University Medical Center, Utrecht, Netherlands; Division of Toxicology, Wageningen University, Wageningen, The Netherlands
| | - Manuel Sánchez
- Department of Nephrology and Hypertension, University Medical Center, Utrecht, Netherlands; Department of Pharmacology, School of Pharmacy, University of Granada, Spain
| | - Branko Braam
- Division of Nephrology & Immunology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada; Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
| | - Jaap A Joles
- Department of Nephrology and Hypertension, University Medical Center, Utrecht, Netherlands;
| |
Collapse
|
39
|
Song Y, Qin X, Wang H, Miao R, Zhang Y, Miao C, Wang Z. Effects of integrin α5β1 on the proliferation and migration of human aortic vascular smooth muscle cells. Mol Med Rep 2015; 13:1147-55. [PMID: 26648324 PMCID: PMC4732837 DOI: 10.3892/mmr.2015.4649] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 04/30/2015] [Indexed: 01/13/2023] Open
Abstract
Integrin (ITG) α5β1 is a dominant fibronectin receptor that is abundantly expressed on the surface of vascular smooth muscle cells (VSMCs). However, the association between integrin α5β1 and the proliferation and migration of VSMCs has yet to be elucidated. The aim of the present study was to characterize the roles of ITGα5 and ITGβ1 in the proliferation and migration of VSMCs, and to determine the effects of ITGα5β1 on integrin-linked kinase (ILK) and focal adhesion kinase (FAK) mRNA expression. Lentiviral expression vectors as well as RNA interference vectors of ITGα5 and ITGβ1 were successfully constructed and transfected into VSMCs to obtain ITGα5‑ and ITGβ1‑overexpressing or -silenced cells, respectively. Cell cycle distribution, proliferation and migration were analyzed in the transfected VSMCs in order to clarify the roles of ITGβ1 and ITGα5 in the proliferation and migration of VSMCs. ITGβ1 was markedly associated with the proliferation and migration of VSMCs, and FAK was shown to be involved in the signaling pathways of ITGβ1. ITGα5 did not exert any effects on VSMCs. The results of the present study may provide a possible therapeutic target for the prevention and treatment of early vascular disease associated with VSMCs.
Collapse
Affiliation(s)
- Yan Song
- Department of Vascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Xiaoyu Qin
- Department of Vascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Hanjie Wang
- Department of Vascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Renying Miao
- Department of Vascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yonggan Zhang
- Department of Vascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Chaofeng Miao
- Department of Vascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Zifan Wang
- Department of Vascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
40
|
Ye M, Zheng J, Chen X, Chen X, Wu X, Lin X, Liu Y. Prednisone inhibits the focal adhesion kinase/receptor activator of NF-κB ligand/mitogen-activated protein kinase signaling pathway in rats with adriamycin-induced nephropathy. Mol Med Rep 2015; 12:7471-8. [PMID: 26459042 DOI: 10.3892/mmr.2015.4370] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 08/27/2015] [Indexed: 11/05/2022] Open
Abstract
The aim of the present study was to investigate the mechanisms underlying the effects of prednisone on adriamycin-induced nephritic rat kidney damage via the focal adhesion kinase (FAK)/receptor activator of nuclear factor-κB ligand (RANKL)/mitogen‑activated protein kinase (MAPK) signaling pathway. An adriamycin‑induced nephritic rat model was established to investigate these mechanisms. A total of 30 healthy male Sprague‑Dawley rats were randomly assigned to the normal, model or prednisone group. Samples of urine were collected over the course of 24 h at days 7, 14, and 28, and renal cortex tissue samples were harvested at days 14, and 28 following nephritic rat model establishment. The total urinary protein content was measured by biuret colorimetry. Pathological changes in the kidney tissue samples were observed using an electron microscope. The mRNA expressions levels of FAK, RANKL, p38, extracellular signal‑regulated kinase (ERK), c‑Jun N‑terminal kinase (JNK), and nephrin were then quantified by reverse transcription‑quantitative polymerase chain reaction. In addition, the protein expressions levels of FAK, RANKL, p38, ERK, JNK, phosphorylated (p)‑FAK, p‑ERK, and p‑JNK were quantified by western blotting. As compared with the normal group, the protein expression levels of FAK, RANKL, p-FAK, p38 and p-ERK in the model group were increased. In the prednisone group, the protein expression levels of p-ERK decreased, as compared with the normal group. In the prednisone group, the urinary protein levels, the protein expression levels of FAK, RANKL, p38, p-FAK, p-p38 and the mRNA expression levels of FAK, p38, RANKL, ERK, JNK decreased, as compared with the model group. In the prednisone group, the mRNA and protein expression levels of nephrin and the serum expression levels of RANKL increased, the serum expression levels of osteoprotegerin (OPG) were decreased, as compared with the model group. No significant changes in the protein expression levels of JNK were observed among the groups. These results suggested that prednisone is able to protect podocytes from apoptosis, and reduce urinary protein levels by inhibiting the FAK/RANKL/MAPK signaling pathway in kidney tissue samples. Serum prednisone may induce osteoporosis via the OPG/RANK/RANKL signaling pathway.
Collapse
Affiliation(s)
- Minyuan Ye
- Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Jing Zheng
- Department of Nephropathy, The People's Hospital of Fujian Province, Fuzhou, Fujian 350004, P.R. China
| | - Xiaoying Chen
- Department of Nephropathy, The People's Hospital of Fujian Province, Fuzhou, Fujian 350004, P.R. China
| | - Xuelan Chen
- Department of Nephropathy, The People's Hospital of Fujian Province, Fuzhou, Fujian 350004, P.R. China
| | - Xinhong Wu
- Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Xiuqin Lin
- Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Yafang Liu
- Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| |
Collapse
|
41
|
LI ZHUO, ZHANG LI, SHI WEI, CHEN YUANHAN, ZHANG HONG, LIU SHUANGXIN, LIANG XINLING, LING TING, YU CHUNPING, HUANG ZHONGSHUN, TAN XIAOFAN, ZHAO XINCHEN, YE ZHIMING, ZHANG BIN, WANG WENJIAN, LI RUIZHAO, MA JIANCHAO. Spironolactone inhibits podocyte motility via decreasing integrin β1 and increasing integrin β3 in podocytes under high-glucose conditions. Mol Med Rep 2015; 12:6849-54. [PMID: 26352002 DOI: 10.3892/mmr.2015.4295] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 07/29/2015] [Indexed: 01/19/2023] Open
|
42
|
Chen Z, Migeon T, Verpont MC, Zaidan M, Sado Y, Kerjaschki D, Ronco P, Plaisier E. HANAC Syndrome Col4a1 Mutation Causes Neonate Glomerular Hyperpermeability and Adult Glomerulocystic Kidney Disease. J Am Soc Nephrol 2015; 27:1042-54. [PMID: 26260163 DOI: 10.1681/asn.2014121217] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 06/16/2015] [Indexed: 12/21/2022] Open
Abstract
Hereditary angiopathy, nephropathy, aneurysms, and muscle cramps (HANAC) syndrome is an autosomal dominant syndrome caused by mutations in COL4A1 that encodes the α1 chain of collagen IV, a major component of basement membranes. Patients present with cerebral small vessel disease, retinal tortuosity, muscle cramps, and kidney disease consisting of multiple renal cysts, chronic kidney failure, and sometimes hematuria. Mutations producing HANAC syndrome localize within the integrin binding site containing CB3[IV] fragment of the COL4A1 protein. To investigate the pathophysiology of HANAC syndrome, we generated mice harboring the Col4a1 p.Gly498Val mutation identified in a family with the syndrome. Col4a1 G498V mutation resulted in delayed glomerulogenesis and podocyte differentiation without reduction of nephron number, causing albuminuria and hematuria in newborns. The glomerular defects resolved within the first month, but glomerular cysts developed in 3-month-old mutant mice. Abnormal structure of Bowman's capsule was associated with metalloproteinase induction and activation of the glomerular parietal epithelial cells that abnormally expressed CD44,α-SMA, ILK, and DDR1. Inflammatory infiltrates were observed around glomeruli and arterioles. Homozygous Col4a1 G498V mutant mice additionally showed dysmorphic papillae and urinary concentration defects. These results reveal a developmental role for the α1α1α2 collagen IV molecule in the embryonic glomerular basement membrane, affecting podocyte differentiation. The observed association between molecular alteration of the collagenous network in Bowman's capsule of the mature kidney and activation of parietal epithelial cells, matrix remodeling, and inflammation may account for glomerular cyst development and CKD in patients with COL4A1-related disorders.
Collapse
Affiliation(s)
- Zhiyong Chen
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche (UMR)S 1155, Paris, France
| | - Tiffany Migeon
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche (UMR)S 1155, Paris, France; Sorbonne University, Université Pierre et Marie Curie, Paris 06, UMR_S 1155, Paris, France
| | - Marie-Christine Verpont
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche (UMR)S 1155, Paris, France; Sorbonne University, Université Pierre et Marie Curie, Paris 06, UMR_S 1155, Paris, France
| | - Mohamad Zaidan
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche (UMR)S 1155, Paris, France
| | - Yoshikazu Sado
- Division of Immunology, Shigei Medical Research Institute, Okayama, Japan
| | - Dontscho Kerjaschki
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria; and
| | - Pierre Ronco
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche (UMR)S 1155, Paris, France; Sorbonne University, Université Pierre et Marie Curie, Paris 06, UMR_S 1155, Paris, France; Assistance Publique-Hôpitaux de Paris, Department of Nephrology and Dialysis, Tenon Hospital, Paris, France
| | - Emmanuelle Plaisier
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche (UMR)S 1155, Paris, France; Sorbonne University, Université Pierre et Marie Curie, Paris 06, UMR_S 1155, Paris, France; Assistance Publique-Hôpitaux de Paris, Department of Nephrology and Dialysis, Tenon Hospital, Paris, France
| |
Collapse
|
43
|
Inoue K, Ishibe S. Podocyte endocytosis in the regulation of the glomerular filtration barrier. Am J Physiol Renal Physiol 2015; 309:F398-405. [PMID: 26084928 DOI: 10.1152/ajprenal.00136.2015] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 06/15/2015] [Indexed: 02/06/2023] Open
Abstract
Severe defects in the glomerular filtration barrier result in nephrotic syndrome, which is characterized by massive proteinuria. The podocyte, a specialized epithelial cell with interdigitating foot processes separated by a slit diaphragm, plays a vital role in regulating the passage of proteins from the capillary lumen to Bowman's space. Recent findings suggest a critical role for endocytosis in podocyte biology as highlighted by genetic mouse models of disease and human genetic mutations that result in the loss of the integrity of the glomerular filtration barrier. In vitro podocyte studies have also unraveled a plethora of constituents that are differentially internalized to maintain homeostasis. These observations provide a framework and impetus for understanding the precise regulation of podocyte endocytic machinery in both health and disease.
Collapse
Affiliation(s)
- Kazunori Inoue
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Shuta Ishibe
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
44
|
Kuroda Y, Ohashi I, Naruto T, Ida K, Enomoto Y, Saito T, Nagai JI, Wada T, Kurosawa K. Delineation of the KIAA2022 mutation phenotype: two patients with X-linked intellectual disability and distinctive features. Am J Med Genet A 2015; 167:1349-53. [PMID: 25900396 DOI: 10.1002/ajmg.a.37002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 01/19/2015] [Indexed: 11/06/2022]
Abstract
Next-generation sequencing has enabled the screening for a causative mutation in X-linked intellectual disability (XLID). We identified KIAA2022 mutations in two unrelated male patients by targeted sequencing. We selected 13 Japanese male patients with severe intellectual disability (ID), including four sibling patients and nine sporadic patients. Two of thirteen had a KIAA2022 mutation. Patient 1 was a 3-year-old boy. He had severe ID with autistic behavior and hypotonia. Patient 2 was a 5-year-old boy. He also had severe ID with autistic behavior, hypotonia, central hypothyroidism, and steroid-dependent nephrotic syndrome. Both patients revealed consistent distinctive features, including upswept hair, narrow forehead, downslanting eyebrows, wide palpebral fissures, long nose, hypoplastic alae nasi, open mouth, and large ears. De novo KIAA2022 mutations (p.Q705X in Patient 1, p.R322X in Patient 2) were detected by targeted sequencing and confirmed by Sanger sequencing. KIAA2022 mutations and alterations have been reported in only four families with nonsyndromic ID and epilepsy. KIAA2022 is highly expressed in the fetal and adult brain and plays a crucial role in neuronal development. These additional patients support the evidence that KIAA2022 is a causative gene for XLID.
Collapse
Affiliation(s)
- Yukiko Kuroda
- Division of Medical Genetics, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Ikuko Ohashi
- Division of Medical Genetics, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Takuya Naruto
- Division of Medical Genetics, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Kazumi Ida
- Division of Medical Genetics, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Yumi Enomoto
- Division of Medical Genetics, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Toshiyuki Saito
- Department of Clinical Laboratory, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Jun-Ichi Nagai
- Department of Clinical Laboratory, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Takahito Wada
- Division of Pediatric Neurology, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Kenji Kurosawa
- Division of Medical Genetics, Kanagawa Children's Medical Center, Yokohama, Japan
| |
Collapse
|
45
|
Lee HW, Khan SQ, Faridi MH, Wei C, Tardi NJ, Altintas MM, Elshabrawy HA, Mangos S, Quick KL, Sever S, Reiser J, Gupta V. A Podocyte-Based Automated Screening Assay Identifies Protective Small Molecules. J Am Soc Nephrol 2015; 26:2741-52. [PMID: 25858967 DOI: 10.1681/asn.2014090859] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 01/08/2015] [Indexed: 12/30/2022] Open
Abstract
Podocyte injury and loss mark an early step in the pathogenesis of various glomerular diseases, making these cells excellent targets for therapeutics. However, cell-based high-throughput screening assays for the rational development of podocyte-directed therapeutics are currently lacking. Here, we describe a novel high-content screening-based phenotypic assay that analyzes thousands of podocytes per assay condition in 96-well plates to quantitatively measure dose-dependent changes in multiple cellular features. Our assay consistently produced a Z' value >0.44, making it suitable for compound screening. On screening with >2100 pharmacologically active agents, we identified 24 small molecules that protected podocytes against injury in vitro (1% hit rate). Among the identified hits, we confirmed an β1-integrin agonist, pyrintegrin, as a podocyte-protective agent. Treatment with pyrintegrin prevented damage-induced decreases in F-actin stress fibers, focal adhesions, and active β1-integrin levels in cultured cells. In vivo, administration of pyrintegrin protected mice from LPS-induced podocyte foot process effacement and proteinuria. Analysis of the murine glomeruli showed that LPS administration reduced the levels of active β1 integrin in the podocytes, which was prevented by cotreatment with pyrintegrin. In rats, pyrintegrin reduced peak proteinuria caused by puromycin aminonucleoside-induced nephropathy. Our findings identify pyrintegrin as a potential therapeutic candidate and show the use of podocyte-based screening assays for identifying novel therapeutics for proteinuric kidney diseases.
Collapse
Affiliation(s)
- Ha Won Lee
- Department of Medicine, Rush University Medical Center, Chicago, Illinois
| | - Samia Q Khan
- Department of Medicine, Rush University Medical Center, Chicago, Illinois
| | - Mohd Hafeez Faridi
- Department of Medicine, Rush University Medical Center, Chicago, Illinois
| | - Changli Wei
- Department of Medicine, Rush University Medical Center, Chicago, Illinois
| | - Nicholas J Tardi
- Department of Medicine, Rush University Medical Center, Chicago, Illinois
| | - Mehmet M Altintas
- Department of Medicine, Rush University Medical Center, Chicago, Illinois
| | - Hatem A Elshabrawy
- Department of Medicine, Rush University Medical Center, Chicago, Illinois
| | - Steve Mangos
- Department of Medicine, Rush University Medical Center, Chicago, Illinois
| | - Kevin L Quick
- Cellular Imaging and Analysis, PerkinElmer Life Sciences, Waltham, Massachusetts; and
| | - Sanja Sever
- Department of Medicine, Harvard Medical School, Division of Nephrology, Massachusetts General Hospital, Charlestown, Massachusetts
| | - Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, Illinois;
| | - Vineet Gupta
- Department of Medicine, Rush University Medical Center, Chicago, Illinois;
| |
Collapse
|
46
|
Interactions of DPP-4 and integrin β1 influences endothelial-to-mesenchymal transition. Kidney Int 2015; 88:479-89. [PMID: 25830763 DOI: 10.1038/ki.2015.103] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 02/03/2015] [Accepted: 02/12/2015] [Indexed: 02/07/2023]
Abstract
Integrin β1 and dipeptidyl peptidase (DPP)-4 play roles in endothelial cell biology. Vascular endothelial growth factor (VEGF)-A inhibits endothelial-to-mesenchymal transition (EndMT) through VEGF-R2, but through VEGF-R1 promotes EndMT by reducing the bioavailability of VEGF-A. Here we tested whether DPP-4-integrin β1 interactions have a role in EndMT in the renal fibrosis of diabetic nephropathy. In streptozotocin-induced fibrotic kidneys in diabetic CD-1 mice, levels of endothelial DPP-4, integrin β1, and phospho-integrin β1 were all higher and associated with plasma cystatin C elevation. The DPP-4 inhibitor linagliptin ameliorated kidney fibrosis, reduced plasma cystatin C levels, and suppressed endothelial levels of DPP-4, integrin β1, and phospho-integrin β1. In cultured endothelial cells, DPP-4 and integrin β1 physically interacted. Suppression of DPP-4 by siRNA was associated with suppression of integrin β1 and vice versa. Knockdown of either integrin β1 or DPP-4 resulted in the silencing of TGF-β2-induced TGF-β receptor heterodimer formation, smad3 phosphorylation, and EndMT. DPP-4 negatively regulated endothelial viability signaling by VEGF-R2 suppression and VEGF-R1 induction in endothelial cells. Thus, DPP-4 and integrin β1 interactions regulate key endothelial cell signal transduction in both physiological and pathological conditions including EndMT. Hence, inhibiting DPP-4 may be a therapeutic target for treating kidney fibrosis in diabetes.
Collapse
|
47
|
Borza CM, Chen X, Zent R, Pozzi A. Cell Receptor-Basement Membrane Interactions in Health and Disease: A Kidney-Centric View. CURRENT TOPICS IN MEMBRANES 2015; 76:231-53. [PMID: 26610916 PMCID: PMC4913201 DOI: 10.1016/bs.ctm.2015.07.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cell-extracellular matrix (ECM) interactions are essential for tissue development, homeostasis, and response to injury. Basement membranes (BMs) are specialized ECMs that separate epithelial or endothelial cells from stromal components and interact with cells via cellular receptors, including integrins and discoidin domain receptors. Disruption of cell-BM interactions due to either injury or genetic defects in either the ECM components or cellular receptors often lead to irreversible tissue injury and loss of organ function. Animal models that lack specific BM components or receptors either globally or in selective tissues have been used to help with our understanding of the molecular mechanisms whereby cell-BM interactions regulate organ function in physiological and pathological conditions. We review recently published works on animal models that explore how cell-BM interactions regulate kidney homeostasis in both health and disease.
Collapse
Affiliation(s)
- Corina M. Borza
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, 37232
- Vanderbilt Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, TN, 37232
| | - Xiwu Chen
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, 37232
| | - Roy Zent
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, 37232
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN, 37232
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN, 37232
- Vanderbilt Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, TN, 37232
- Department of Medicine, Veterans Administration Hospital, Nashville, TN, 37232
| | - Ambra Pozzi
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, 37232
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN, 37232
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN, 37232
- Vanderbilt Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, TN, 37232
- Department of Medicine, Veterans Administration Hospital, Nashville, TN, 37232
| |
Collapse
|
48
|
Ichii O, Otsuka-Kanazawa S, Nakamura T, Ueno M, Kon Y, Chen W, Rosenberg AZ, Kopp JB. Podocyte injury caused by indoxyl sulfate, a uremic toxin and aryl-hydrocarbon receptor ligand. PLoS One 2014; 9:e108448. [PMID: 25244654 PMCID: PMC4171541 DOI: 10.1371/journal.pone.0108448] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 08/21/2014] [Indexed: 02/07/2023] Open
Abstract
Indoxyl sulfate is a uremic toxin and a ligand of the aryl-hydrocarbon receptor (AhR), a transcriptional regulator. Elevated serum indoxyl sulfate levels may contribute to progressive kidney disease and associated vascular disease. We asked whether indoxyl sulfate injures podocytes in vivo and in vitro. Mice exposed to indoxyl sulfate for 8 w exhibited prominent tubulointerstitial lesions with vascular damage. Indoxyl sulfate-exposed mice with microalbuminuria showed ischemic changes, while more severely affected mice showed increased mesangial matrix, segmental solidification, and mesangiolysis. In normal mouse kidneys, AhR was predominantly localized to the podocyte nuclei. In mice exposed to indoxyl sulfate for 2 h, isolated glomeruli manifested increased Cyp1a1 expression, indicating AhR activation. After 8 w of indoxyl sulfate, podocytes showed foot process effacement, cytoplasmic vacuoles, and a focal granular and wrinkled pattern of podocin and synaptopodin expression. Furthermore, vimentin and AhR expression in the glomerulus was increased in the indoxyl sulfate-exposed glomeruli compared to controls. Glomerular expression of characteristic podocyte mRNAs was decreased, including Actn4, Cd2ap, Myh9, Nphs1, Nphs2, Podxl, Synpo, and Wt1. In vitro, immortalized-mouse podocytes exhibited AhR nuclear translocation beginning 30 min after 1 mM indoxyl sulfate exposure, and there was increased phospho-Rac1/Cdc42 at 2 h. After exposure to indoxyl sulfate for 24 h, mouse podocytes exhibited a pro-inflammatory phenotype, perturbed actin cytoskeleton, decreased expression of podocyte-specific genes, and decreased cell viability. In immortalized human podocytes, indoxyl sulfate treatment caused cell injury, decreased mRNA expression of podocyte-specific proteins, as well as integrins, collagens, cytoskeletal proteins, and bone morphogenetic proteins, and increased cytokine and chemokine expression. We propose that basal levels of AhR activity regulate podocyte function under normal conditions, and that increased activation of podocyte AhR by indoxyl sulfate contributes to progressive glomerular injury.
Collapse
Affiliation(s)
- Osamu Ichii
- Laboratory of Anatomy, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Hokkaido, Japan
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| | - Saori Otsuka-Kanazawa
- Laboratory of Anatomy, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Hokkaido, Japan
| | - Teppei Nakamura
- Laboratory of Anatomy, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Hokkaido, Japan
- Section of Biological Safety Research, Chitose Laboratory, Japan Food Research Laboratories, Chitose, Japan
| | - Masaaki Ueno
- Section of Biological Safety Research, Chitose Laboratory, Japan Food Research Laboratories, Chitose, Japan
| | - Yasuhiro Kon
- Laboratory of Anatomy, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Hokkaido, Japan
| | - Weiping Chen
- Microarray Core Facility, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Avi Z. Rosenberg
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jeffrey B. Kopp
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
49
|
Sasaki H, Sasaki N, Nishino T, Nagasaki KI, Kitamura H, Torigoe D, Agui T. Quantitative trait Loci for resistance to the congenital nephropathy in tensin 2-deficient mice. PLoS One 2014; 9:e99602. [PMID: 24967628 PMCID: PMC4072594 DOI: 10.1371/journal.pone.0099602] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 05/16/2014] [Indexed: 11/18/2022] Open
Abstract
The ICR-derived glomerulonephritis (ICGN) mouse is a chronic kidney disease (CKD) model that is characterized histologically by glomerulosclerosis, vascular sclerosis and tubulointerstitial fibrosis, and clinically by proteinuria and anemia, which are common symptoms and pathological changes associated with a variety of kidney diseases. Previously, we performed a quantitative trait locus (QTL) analysis to identify the causative genes for proteinuria in ICGN mice, and found a deletion mutation of the tensin 2 gene (Tns2nph, MGI no: 2447990). Interestingly, the congenic strain carrying the Tns2nph mutation on a C57BL/6J (B6) genetic background exhibited milder phenotypes than did ICGN mice, indicating the presence of several modifier genes controlling the disease phenotype. In this study, to identify the modifier/resistant loci for CKD progression in Tns2-deficient mice, we performed QTL analysis using backcross progenies from susceptible ICGN and resistant B6 mice. We identified a significant locus on chromosome (Chr) 2 (LOD = 5.36; 31 cM) and two suggestive loci on Chrs 10 (LOD = 2.27; 64 cM) and X (LOD = 2.65; 67 cM) with linkage to the severity of tubulointerstitial injury. One significant locus on Chr 13 (LOD = 3.49; approximately 14 cM) and one suggestive locus on Chr 2 (LOD = 2.41; 51 cM) were identified as QTLs for the severity of glomerulosclerosis. Suggestive locus in BUN was also detected in the same Chr 2 region (LOD = 2.34; 51 cM). A locus on Chr 2 (36 cM) was significantly linked with HGB (LOD = 4.47) and HCT (LOD = 3.58). Four novel epistatic loci controlling either HGB or tubulointerstitial injury were discovered. Further genetic analysis should lead to identification of CKD modifier gene(s), aiding early diagnosis and providing novel approaches to the discovery of drugs for the treatment and possible prevention of kidney disease.
Collapse
Affiliation(s)
- Hayato Sasaki
- Laboratory of Laboratory Animal Science and Medicine, Department of Disease Control, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Nobuya Sasaki
- Laboratory of Laboratory Animal Science and Medicine, Faculty of Veterinary Medicine, Kitasato University, Towada, Japan
- * E-mail:
| | - Tomohiro Nishino
- Laboratory of Laboratory Animal Science and Medicine, Department of Disease Control, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Ken-ichi Nagasaki
- Section of Biological Safety Research, Chitose Laboratory, Japan Food Research Laboratories, Chitose, Japan
| | - Hiroshi Kitamura
- Department of Veterinary Physiology, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan
| | - Daisuke Torigoe
- Laboratory of Laboratory Animal Science and Medicine, Department of Disease Control, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Takashi Agui
- Laboratory of Laboratory Animal Science and Medicine, Department of Disease Control, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
50
|
Schlondorff J. How many Achilles' heels does a podocyte have? An update on podocyte biology: Table 1. Nephrol Dial Transplant 2014; 30:1091-7. [DOI: 10.1093/ndt/gfu214] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 05/07/2014] [Indexed: 11/15/2022] Open
|