1
|
Mann N, Sun H, Majmundar AJ. Mechanisms of podocyte injury in genetic kidney disease. Pediatr Nephrol 2024:10.1007/s00467-024-06551-x. [PMID: 39485497 DOI: 10.1007/s00467-024-06551-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 11/03/2024]
Abstract
Glomerular diseases are a leading cause of chronic kidney disease worldwide. Both acquired and hereditary glomerulopathies frequently share a common final disease mechanism: disruption of the glomerular filtration barrier, podocyte injury, and ultimately podocyte death and detachment. Over 70 monogenic causes of proteinuric kidney disease have been identified, and most of these genes are highly expressed in podocytes, regulating key processes such as maintenance of the slit diaphragm, regulation of actin cytoskeleton remodeling, and modulation of downstream transcriptional pathways. Collectively, these are increasingly being referred to as hereditary "podocytopathies," in which podocyte injury is the central feature driving proteinuria and kidney dysfunction. In this review, we provide an overview of the monogenic podocytopathies and discuss the molecular mechanisms by which single-gene defects lead to podocyte injury and ultimately glomerulosclerosis. We review how advances in genomic technology and a better understanding of the cell biological basis of disease have led to the development of more targeted and personalized therapeutic strategies, including an overview of small molecule and gene therapy approaches.
Collapse
Affiliation(s)
- Nina Mann
- Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| | - Hua Sun
- Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Amar J Majmundar
- Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
2
|
Saito A, Kise R, Inoue A. Generation of Comprehensive GPCR-Transducer-Deficient Cell Lines to Dissect the Complexity of GPCR Signaling. Pharmacol Rev 2024; 76:599-619. [PMID: 38719480 DOI: 10.1124/pharmrev.124.001186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/02/2024] [Accepted: 05/06/2024] [Indexed: 06/16/2024] Open
Abstract
G-protein-coupled receptors (GPCRs) compose the largest family of transmembrane receptors and are targets of approximately one-third of Food and Drug Administration-approved drugs owing to their involvement in almost all physiologic processes. GPCR signaling occurs through the activation of heterotrimeric G-protein complexes and β-arrestins, both of which serve as transducers, resulting in distinct cellular responses. Despite seeming simple at first glance, accumulating evidence indicates that activation of either transducer is not a straightforward process as a stimulation of a single molecule has the potential to activate multiple signaling branches. The complexity of GPCR signaling arises from the aspects of G-protein-coupling selectivity, biased signaling, interpathway crosstalk, and variable molecular modifications generating these diverse signaling patterns. Numerous questions relative to these aspects of signaling remained unanswered until the recent development of CRISPR genome-editing technology. Such genome editing technology presents opportunities to chronically eliminate the expression of G-protein subunits, β-arrestins, G-protein-coupled receptor kinases (GRKs), and many other signaling nodes in the GPCR pathways at one's convenience. Here, we review the practicality of using CRISPR-derived knockout (KO) cells in the experimental contexts of unraveling the molecular details of GPCR signaling mechanisms. To mention a few, KO cells have revealed the contribution of β-arrestins in ERK activation, Gα protein selectivity, GRK-based regulation of GPCRs, and many more, hence validating its broad applicability in GPCR studies. SIGNIFICANCE STATEMENT: This review emphasizes the practical application of G-protein-coupled receptor (GPCR) transducer knockout (KO) cells in dissecting the intricate regulatory mechanisms of the GPCR signaling network. Currently available cell lines, along with accumulating KO cell lines in diverse cell types, offer valuable resources for systematically elucidating GPCR signaling regulation. Given the association of GPCR signaling with numerous diseases, uncovering the system-based signaling map is crucial for advancing the development of novel drugs targeting specific diseases.
Collapse
Affiliation(s)
- Ayaki Saito
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Ryoji Kise
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| |
Collapse
|
3
|
Ran L, Li W, Zhang H, Lin J, Zhu L, Long H, Xiang L, Chen L, Li Q, Hu Y, Gong M, Xiao B, Zhao H. Identification of Plasma hsa_circ_0001230 and hsa_circ_0023879 as Potential Novel Biomarkers for Focal Segmental Glomerulosclerosis and circRNA-miRNA-mRNA Network Analysis. Kidney Blood Press Res 2024; 49:310-325. [PMID: 38648755 DOI: 10.1159/000538825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 03/03/2024] [Indexed: 04/25/2024] Open
Abstract
INTRODUCTION Focal segmental glomerulosclerosis (FSGS) is a common glomerulopathy with an unclear mechanism. The demand for FSGS clinical diagnostic biomarkers has not yet been met. Circular RNA (circRNA) is a novel non-coding RNA with multiple functions, but its diagnostic value for FSGS remains unexplored. This study aimed to identify circRNAs that could aid in early clinical diagnosis and to investigate their mechanisms in podocyte injury. METHODS The signature of plasma circRNAs for FSGS was identified by circRNA microarray. The existence of circRNAs was confirmed by quantitative real-time polymerase chain reaction (qRT-PCR), RNase R assay, and DNA sequencing. Plasma levels of circRNAs were evaluated by qRT-PCR. The diagnostic value was appraised by the receiver operating characteristic curve. The circRNA-miRNA-mRNA network was built with Cytoscape 7.3.2. Statistically significant differences were calculated by the Mann-Whitney U test. RESULTS A total of 493 circRNAs (165 upregulated, 328 downregulated) were differentially expressed in the plasma of FSGS patients (n = 3) and normal controls (n = 3). Eight candidate circRNAs were demonstrated to be circular and stable transcripts. Among them, hsa_circ_0001230 and hsa_circ_0023879 were significantly upregulated in FSGS patients (n = 29) compared to normal controls (n = 51). The areas under the curve value of hsa_circ_0001230 and hsa_circ_0023879 were 0.668 and 0.753, respectively, while that of the two-circRNA panel was 0.763. The RNA pull-down analysis revealed that hsa_circ_0001230 and hsa_circ_0023879 could sponge hsa-miR-106a. Additionally, hsa_circ_0001230 and hsa_circ_0023879 positively regulated hsa-miR-106a target genes phosphatase and tensin homolog (PTEN) and Bcl-2-like protein 11 (BCL2L11) in podocytes. CONCLUSION hsa_circ_0001230 and hsa_circ_0023879 are novel blood biomarkers for FSGS. They may regulate podocyte apoptosis by competitively binding to hsa-miR-106a.
Collapse
Affiliation(s)
- Lingyu Ran
- Department of Kidney, Southwest Hospital, Army Medical University, Chongqing, China,
| | - Wei Li
- Department of Pharmacy, Chongqing University Cancer Hospital, Chongqing, China
| | - Huhai Zhang
- Department of Kidney, Southwest Hospital, Army Medical University, Chongqing, China
| | - Jie Lin
- Department of Disease Control and Prevention, The 904th Hospital of Joint Logistic Support Force of the PLA, Wuxi, China
| | - Longyin Zhu
- Department of Kidney, Southwest Hospital, Army Medical University, Chongqing, China
| | - Huanping Long
- Department of Kidney, Southwest Hospital, Army Medical University, Chongqing, China
| | - Lunli Xiang
- Department of Kidney, Southwest Hospital, Army Medical University, Chongqing, China
| | - Liping Chen
- Department of Kidney, Southwest Hospital, Army Medical University, Chongqing, China
| | - Qixuan Li
- Department of Kidney, Southwest Hospital, Army Medical University, Chongqing, China
| | - Yuhan Hu
- Department of Clinical Lab, Southwest Hospital, Army Medical University, Chongqing, China
| | - Min Gong
- College of Traditional Chinese Medicine, Chongqing Three Gorges Medical College, Chongqing, China
| | - Bin Xiao
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Hongwen Zhao
- Department of Kidney, Southwest Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
4
|
Lu P, Zhang M, Chen Z, Xu Q, Liu M, Zhao F, Liu X, Wang X. Salvianolic Acid B Inhibits Myocardial Fibrosis during Diabetic Cardiomyopathy via Suppressing TRPC6 and TGF‐ β/Smad3 Pathway. J Food Biochem 2024; 2024. [DOI: 10.1155/2024/5525825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 07/18/2024] [Indexed: 01/05/2025]
Abstract
Salvianolic acid B (Sal B), the main water‐soluble polyphenolic constituent of Danshen, is noted for its anti‐inflammatory, antioxidant, and antiapoptotic properties, particularly in cardiovascular protection. However, the mechanisms by which Sal B affects myocardial fibrosis require further investigation. In vivo, we established a diabetic mouse model using a high‐fat diet and intraperitoneal streptozotocin (STZ) administration. Mice were then treated with Sal B, the transient receptor potential channel 6 (TRPC6) inducers, or their combination. Upregulation of TRPC6 worsened myocardial pathology, leading to cardiac hypertrophy and collagen fiber deposition. In vitro, transforming growth factor (TGF)‐β1 induced transdifferentiation of cardiac fibroblasts into myofibroblasts, creating a myofibroblast cell model. Sal B, TRPC6 inducers, or their combination were administered. TRPC6 upregulation increased procollagen type I C‐terminal propeptide (PICP) and procollagen type III N‐terminal propeptide (PIIINP) secretion, promoting myofibroblast proliferation and migration. Our study indicates that TRPC6 expression is upregulated in myocardial fibrosis, enhancing TGF‐β/Smad3 signaling and promoting collagen I (COL‐1) synthesis. Sal B inhibited abnormal TRPC6 expression and TGF‐β/Smad3 activation, mitigating these effects. Thus, Sal B alleviates myocardial fibrosis in diabetes by modulating TRPC6 expression and TGF‐β/Smad3 signaling pathway.
Collapse
|
5
|
Llancalahuen FM, Vallejos A, Aravena D, Prado Y, Gatica S, Otero C, Simon F. α1-Adrenergic Stimulation Increases Platelet Adhesion to Endothelial Cells Mediated by TRPC6. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1408:65-82. [PMID: 37093422 DOI: 10.1007/978-3-031-26163-3_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Stimulation of a1-adrenergic nervous system is increased during systemic inflammation and other pathological conditions with the consequent adrenergic receptors (ARs) activation. It has been reported that a1-stimulation contributes to coagulation since a1-AR blockers inhibit coagulation and its organic consequences. Also, coagulation induced by a1-AR stimulation can be greatly decreased using a1-AR blockers. In health, endothelial cells (ECs) perform anticoagulant actions at cellular and molecular level. However, during inflammation, ECs turn dysfunctional promoting a procoagulant state. Endothelium-dependent coagulation progresses at cellular and molecular levels, promoting endothelial acquisition of procoagulant properties to potentiate coagulation by means of prothrombotic and antifibrinolytic proteins expression increase in ECs releasing them to circulation, the thrombus formation is strengthened. Calcium signaling is a main feature of coagulation. Inhibition of ion channels involved in Ca2+ entry severely decreases coagulation. The transient receptor potential canonical 6 (TRPC6) is a non-selective Ca2+-permeable ion channel. TRPC6 activity is induced by diacylglycerol, suggesting that is regulated by a1-ARs. Furthermore, a1-ARs stimulation elicits a TRPC-like current in rat mesenteric artery smooth muscle and mesangial cells. However, whether TRPC6 could promote an ECs-mediated platelet adhesion induced by a1-adrenergic stimulation is currently not known. Therefore, the aim of this study was to examine if the TRPC6 calcium channel mediates platelet adhesion induced by a1-adrenergic stimulation. Our results suggest that platelet adhesion to ECs is enhanced by the a1-adrenergic stimulation evoked by phenylephrine mediated by TRPC6 activity. We conclude that TRPC6 is a molecular determinant in platelet adhesion to ECs with implications in systemic inflammatory diseases treatment.
Collapse
Affiliation(s)
- Felipe M Llancalahuen
- Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Alejando Vallejos
- Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Diego Aravena
- Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Yolanda Prado
- Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Sebastian Gatica
- Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Carolina Otero
- Escuela de Química y Farmacia, Facultad de Medicina, Universidad Andres Bello, Santiago, Chile
| | - Felipe Simon
- Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile.
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile.
- Millennium Nucleus of Ion Channel-Associated Diseases, Santiago, Chile.
| |
Collapse
|
6
|
Hermann C, Treder A, Näher M, Geiseler R, Gudermann T, Mederos Y Schnitzler M, Storch U. The normalized slope conductance as a tool for quantitative analysis of current-voltage relations. Biophys J 2022; 121:1435-1448. [PMID: 35300969 PMCID: PMC9072577 DOI: 10.1016/j.bpj.2022.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/02/2021] [Accepted: 03/11/2022] [Indexed: 11/29/2022] Open
Abstract
The patch-clamp method which was rewarded with the Nobel Prize in 1991 is a well-established and indispensable method to study ion channels in living cells and to biophysically characterize non-voltage-gated ion channels that comprise about 70% of all ion channels in the human genome. To investigate the biophysical properties of non-voltage-gated ion channels, whole-cell measurements with application of continuous voltage-ramps are routinely conducted to obtain current-voltage (IV) relationships. However, adequate tools for detailed and quantitative analysis of IV curves are still missing. We use the example of the transient receptor potential classical (TRPC) channel family to elucidate whether the normalized slope conductance (NSC) is an appropriate tool for a reliable discrimination of the IV curves of diverse TRPC channels that differ in their individual curve progression. We provide a robust calculation method for the NSC, and by applying this method we find that TRPC channel activators and modulators can evoke different NSC progressions independent from their expression levels which is pointing to distinguishable active channel states. TRPC6 mutations of patients suffering from focal segmental glomerulosclerosis (FSGS) resulted in distinct NSC progressions suggesting that the NSC is suitable to investigate structure-function relations and might help unravel the unknown pathomechanisms leading to FSGS. Altogether, the NCS represents an effective algorithm for extended biophysical characterization of non-voltage-gated ion channels.
Collapse
Affiliation(s)
- Christian Hermann
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig Maximilian University of Munich, 80336 Munich, Germany
| | - Aaron Treder
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig Maximilian University of Munich, 80336 Munich, Germany
| | - Marius Näher
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig Maximilian University of Munich, 80336 Munich, Germany
| | - Roman Geiseler
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig Maximilian University of Munich, 80336 Munich, Germany
| | - Thomas Gudermann
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig Maximilian University of Munich, 80336 Munich, Germany;; Comprehensive Pneumology Center Munich (CPC-M), German Center for Lung Research, Munich, Germany
| | - Michael Mederos Y Schnitzler
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig Maximilian University of Munich, 80336 Munich, Germany;; DZHK (German Centre for Cardiovascular Research), Munich Heart Alliance, Munich, Germany.
| | - Ursula Storch
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig Maximilian University of Munich, 80336 Munich, Germany;; Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian University of Munich, 80336 Munich, Germany.
| |
Collapse
|
7
|
Guo P, Tai Y, Wang M, Sun H, Zhang L, Wei W, Xiang YK, Wang Q. Gα 12 and Gα 13: Versatility in Physiology and Pathology. Front Cell Dev Biol 2022; 10:809425. [PMID: 35237598 PMCID: PMC8883321 DOI: 10.3389/fcell.2022.809425] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 01/17/2022] [Indexed: 01/14/2023] Open
Abstract
G protein-coupled receptors (GPCRs), as the largest family of receptors in the human body, are involved in the pathological mechanisms of many diseases. Heterotrimeric G proteins represent the main molecular switch and receive cell surface signals from activated GPCRs. Growing evidence suggests that Gα12 subfamily (Gα12/13)-mediated signaling plays a crucial role in cellular function and various pathological processes. The current research on the physiological and pathological function of Gα12/13 is constantly expanding, Changes in the expression levels of Gα12/13 have been found in a wide range of human diseases. However, the mechanistic research on Gα12/13 is scattered. This review briefly describes the structural sequences of the Gα12/13 isoforms and introduces the coupling of GPCRs and non-GPCRs to Gα12/13. The effects of Gα12/13 on RhoA and other signaling pathways and their roles in cell proliferation, migration, and immune cell function, are discussed. Finally, we focus on the pathological impacts of Gα12/13 in cancer, inflammation, metabolic diseases, fibrotic diseases, and circulatory disorders are brought to focus.
Collapse
Affiliation(s)
- Paipai Guo
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Yu Tai
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Manman Wang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Hanfei Sun
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Lingling Zhang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Wei Wei
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Yang K Xiang
- Department of Pharmacology, University of California, Davis, Davis, CA, United States.,VA Northern California Health Care System, Mather, CA, United States
| | - Qingtong Wang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| |
Collapse
|
8
|
Ding Y, Diao Z, Cui H, Yang A, Liu W, Jiang L. Bioinformatics analysis reveals the roles of cytoskeleton protein transgelin in occurrence and development of proteinuria. Transl Pediatr 2021; 10:2250-2268. [PMID: 34733666 PMCID: PMC8506063 DOI: 10.21037/tp-21-83] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 07/15/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Proteinuria is a sensitive hallmark for progressive renal dysfunction. Transgelin (TAGLN) has been demonstrated to participate in etiology of proteinuria and dynamics of podocyte foot process; however, the mechanism of TAGLN involvement in proteinuria is unknown. The present study aimed to explore the roles of TAGLN in the development of proteinuria. METHODS Differentially expressed genes (DEGs) were detected from microarray expression profiling datasets from Gene Expression Omnibus, and analyzed by the short time series expression miner to cluster the DEGs in proteinuria progression. Kyoto Encyclopedia of Genes and Genomes pathway analysis was used to determine the top 20 enriched pathways, and construct a gene interaction network. RESULTS In total, 2,409 DEGs for nephropathy and 10,612 DEGs for podocyte foot process and proteinuria were detected. Additionally, 76 common DEGs (25 upregulated and 41 downregulated) between nephropathy and podocyte foot process were primarily involved in innate immunity, positive regulation of transcription-DNA-templated, immunity and negative regulation of cell proliferation, enriched in cytokine-cytokine receptor interaction signaling pathway, Ras signaling pathway, axon guidance, tumor necrosis factor (TNF) signaling pathway and apoptosis. CONCLUSIONS We discovered a TAGLN-mediated regulatory network involved in proteinuria progression. These findings provide novel insight to understand the molecular mechanisms underlying the pathogenesis of proteinuria.
Collapse
Affiliation(s)
- Yingxue Ding
- Pediatric Department, Beijing Friendship Hospital, Capital University of Medical Sciences, Beijing, China
| | - Zongli Diao
- Nephrology Department, Beijing Friendship Hospital, Capital University of Medical Sciences, Beijing, China
| | - Hong Cui
- Pediatric Department, Beijing Friendship Hospital, Capital University of Medical Sciences, Beijing, China
| | - Aijun Yang
- Pediatric Department, Beijing Friendship Hospital, Capital University of Medical Sciences, Beijing, China
| | - Wenhu Liu
- Nephrology Department, Beijing Friendship Hospital, Capital University of Medical Sciences, Beijing, China
| | - Lina Jiang
- Pediatric Department, Beijing Friendship Hospital, Capital University of Medical Sciences, Beijing, China
| |
Collapse
|
9
|
Weber J, Rajan S, Schremmer C, Chao YK, Krasteva-Christ G, Kannler M, Yildirim AÖ, Brosien M, Schredelseker J, Weissmann N, Grimm C, Gudermann T, Dietrich A. TRPV4 channels are essential for alveolar epithelial barrier function as protection from lung edema. JCI Insight 2020; 5:134464. [PMID: 32931478 PMCID: PMC7605532 DOI: 10.1172/jci.insight.134464] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 09/09/2020] [Indexed: 12/25/2022] Open
Abstract
Ischemia/reperfusion-induced edema (IRE), one of the most significant causes of mortality after lung transplantation, can be mimicked ex vivo in isolated perfused mouse lungs (IPL). Transient receptor potential vanilloid 4 (TRPV4) is a nonselective cation channel studied in endothelium; however, its role in the lung epithelium remains elusive. Here, we show enhanced IRE in TRPV4-deficient (TRPV4–/–) IPL compared with that of WT controls, indicating a protective role of TRPV4 in maintenance of the alveolar epithelial barrier. By immunohistochemistry, mRNA profiling, and electrophysiological characterization, we detected TRPV4 in bronchial epithelium, alveolar epithelial type I (ATI), and alveolar epithelial type II (ATII) cells. Genetic ablation of TRPV4 resulted in reduced expression of the water-conducting aquaporin-5 (AQP-5) channel in ATI cells. Migration of TRPV4–/– ATI cells was reduced, and cell barrier function was impaired. Analysis of isolated primary TRPV4–/– ATII cells revealed a reduced expression of surfactant protein C, and the TRPV4 activator GSK1016790A induced increases in current densities only in WT ATII cells. Moreover, TRPV4–/– lungs of adult mice developed significantly larger mean chord lengths and altered lung function compared with WT lungs. Therefore, our data illustrate essential functions of TRPV4 channels in alveolar epithelial cells and in protection from edema formation. TRPV4, a non-selective cation channel, is essential for alveolar epithelial function and protects from ischemia-reperfusion-induced lung edema.
Collapse
Affiliation(s)
- Jonas Weber
- Walther Straub Institute of Pharmacology and Toxicology, a member of the German Center for Lung Research (DZL), Ludwig Maximilian University of Munich, Munich Germany
| | - Suhasini Rajan
- Walther Straub Institute of Pharmacology and Toxicology, a member of the German Center for Lung Research (DZL), Ludwig Maximilian University of Munich, Munich Germany
| | - Christian Schremmer
- Walther Straub Institute of Pharmacology and Toxicology, a member of the German Center for Lung Research (DZL), Ludwig Maximilian University of Munich, Munich Germany
| | - Yu-Kai Chao
- Walther Straub Institute of Pharmacology and Toxicology, a member of the German Center for Lung Research (DZL), Ludwig Maximilian University of Munich, Munich Germany
| | - Gabriela Krasteva-Christ
- Institute of Anatomy and Cell Biology, School of Medicine, Saarland University, Homburg, Germany
| | - Martina Kannler
- Walther Straub Institute of Pharmacology and Toxicology, a member of the German Center for Lung Research (DZL), Ludwig Maximilian University of Munich, Munich Germany
| | - Ali Önder Yildirim
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, a member of the DZL, Helmholtz Center Munich, German Research Center for Environmental Health, Munich, Germany
| | - Monika Brosien
- Justus Liebig University Giessen, Cardio-Pulmonary Institute, University of Giessen and Marburg Lung Center, a member of the DZL, Giessen, Germany
| | - Johann Schredelseker
- Walther Straub Institute of Pharmacology and Toxicology, a member of the German Center for Lung Research (DZL), Ludwig Maximilian University of Munich, Munich Germany
| | - Norbert Weissmann
- Justus Liebig University Giessen, Cardio-Pulmonary Institute, University of Giessen and Marburg Lung Center, a member of the DZL, Giessen, Germany
| | - Christian Grimm
- Walther Straub Institute of Pharmacology and Toxicology, a member of the German Center for Lung Research (DZL), Ludwig Maximilian University of Munich, Munich Germany
| | - Thomas Gudermann
- Walther Straub Institute of Pharmacology and Toxicology, a member of the German Center for Lung Research (DZL), Ludwig Maximilian University of Munich, Munich Germany
| | - Alexander Dietrich
- Walther Straub Institute of Pharmacology and Toxicology, a member of the German Center for Lung Research (DZL), Ludwig Maximilian University of Munich, Munich Germany
| |
Collapse
|
10
|
Bendiks L, Geiger F, Gudermann T, Feske S, Dietrich A. Store-operated Ca 2+ entry in primary murine lung fibroblasts is independent of classical transient receptor potential (TRPC) channels and contributes to cell migration. Sci Rep 2020; 10:6812. [PMID: 32321939 PMCID: PMC7176639 DOI: 10.1038/s41598-020-63677-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 03/16/2020] [Indexed: 12/14/2022] Open
Abstract
Stromal interaction molecules (STIM1, 2) are acting as sensors for Ca2+ in intracellular stores and activate Orai channels at the plasma membrane for store-operated Ca2+ entry (SOCE), while classical transient receptor potential (TRPC) channel mediate receptor-operated Ca2+ entry (ROCE). Several reports, however, indicate a role for TRPC in SOCE in certain cell types. Here, we analyzed Ca2+ influx and cell function in TRPC1/6-deficient (TRPC1/6-/-) and STIM1/2- deficient (STIM1/2ΔpmLF) primary murine lung fibroblasts (pmLF). As expected, SOCE was decreased in STIM1/2- deficient pmLF and ROCE was decreased in TRPC1/6-/- pmLF compared to control cells. By contrast, SOCE was not significantly different in TRPC1/6-/- pmLF and ROCE was similar in STIM1/2-deficient pmLF compared to Wt cells. Most interestingly, cell proliferation, migration and nuclear localization of nuclear factor of activated T-cells (NFATc1 and c3) were decreased after ablation of STIM1/2 proteins in pmLF. In conclusion, TRPC1/6 channels are not involved in SOCE and STIM1/2 deficiency resulted in decreased cell proliferation and migration in pmLF.
Collapse
Affiliation(s)
- Larissa Bendiks
- Walther Straub Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL), Medical Faculty, LMU-Munich, Munich, Germany
| | - Fabienne Geiger
- Walther Straub Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL), Medical Faculty, LMU-Munich, Munich, Germany
| | - Thomas Gudermann
- Walther Straub Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL), Medical Faculty, LMU-Munich, Munich, Germany
| | - Stefan Feske
- Department of Pathology, New York University School of Medicine, New York, NY, 10016, USA
| | - Alexander Dietrich
- Walther Straub Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL), Medical Faculty, LMU-Munich, Munich, Germany.
| |
Collapse
|
11
|
Yu S, Choi WI, Choi YJ, Kim HY, Hildebrandt F, Gee HY. PLCE1 regulates the migration, proliferation, and differentiation of podocytes. Exp Mol Med 2020; 52:594-603. [PMID: 32238860 PMCID: PMC7210307 DOI: 10.1038/s12276-020-0410-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 02/07/2020] [Accepted: 02/12/2020] [Indexed: 12/31/2022] Open
Abstract
PLCE1 encodes phospholipase C epsilon, and its mutations cause recessive nephrotic syndrome. However, the mechanisms by which PLCE1 mutations result in defects associated with glomerular function are not clear. To address this, we investigated the function of PLCE1 in podocytes called glomerular epithelial cells, where the pathogenesis of nephrotic syndrome converges. PLCE1 colocalized with Rho GTPases in glomeruli. Further, it interacted with Rho GTPases through the pleckstrin homology domain and Ras GTP-binding domains 1/2. Knockdown or knockout of PLCE1 in podocytes resulted in decreased levels of GTP-bound Rac1 and Cdc42, but not those of RhoA, and caused a reduction in cell migration. PLCE1 interacted with NCK2 but not with NCK1. Similar to the PLCE1 knockout, NCK2 knockout resulted in decreased podocyte migration. Knockout of PLCE1 reduced the EGF-induced activation of ERK and cell proliferation in podocytes, whereas knockout of NCK2 did not affect proliferation. Further, the knockout of PLCE1 also resulted in decreased expression of podocyte markers, including NEPH1, NPHS1, WT1, and SYNPO, upon differentiation, but the knockout of NCK2 did not affect the expression of these markers. Therefore, our findings demonstrate that PLCE1 regulates Rho GTPase activity and cell migration through interacting with NCK2 and that PLCE1 also plays a role in the proliferation and differentiation of podocytes, regardless of the presence of NCK2. A genetic mutation associated with kidney disease impairs the maturation and migration of cells that filter waste products out of the blood. Tiny tendrils from kidney cells called podocytes establish a tight meshwork that keeps blood proteins in circulation while allowing unwanted contaminants to pass through. Mutations in the PLCE1 gene disrupt this filter, leading to a disorder called nephrotic syndrome Researchers led by Heon Yung Gee at Yonsei University College of Medicine, Seoul, South Korea, have uncovered mechanisms underlying this malfunction. Working with cultured podocytes, they showed that loss of PLCE1 impairs cell migration, potentially undermining their ability to form a meshwork. The researchers also found that the protein encoded by PLCE1 interacts with other molecules that promote cell division and maturation, revealing another mechanism by which mutations could contribute to loss of podocyte function.
Collapse
Affiliation(s)
- Seyoung Yu
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Won-Il Choi
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Yo Jun Choi
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Hye-Youn Kim
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Friedhelm Hildebrandt
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Heon Yung Gee
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, 03722, Korea.
| |
Collapse
|
12
|
Atchison DK, O'Connor CL, Menon R, Otto EA, Ganesh SK, Wiggins RC, Smrcka AV, Bitzer M. Hypertension induces glomerulosclerosis in phospholipase C-ε1 deficiency. Am J Physiol Renal Physiol 2020; 318:F1177-F1187. [PMID: 32223311 DOI: 10.1152/ajprenal.00541.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Loss-of-function mutations in phospholipase C-ε1 (PLCE1) have been detected in patients with nephrotic syndrome, but other family members with the same mutation were asymptomatic, suggesting additional stressor are required to cause the full phenotype. Consistent with these observations, we determined that global Plce1-deficient mice have histologically normal glomeruli and no albuminuria at baseline. Angiotensin II (ANG II) is known to induce glomerular damage in genetically susceptible individuals. Therefore, we tested whether ANG II enhances glomerular damage in Plce1-deficient mice. ANG II increased blood pressure equally in Plce1-deficient and wild-type littermates. Additionally, it led to 20-fold increased albuminuria and significantly more sclerotic glomeruli in Plce1-deficient mice compared with wild-type littermates. Furthermore, Plce1-deficient mice demonstrated diffuse mesangial expansion, podocyte loss, and focal podocyte foot process effacement. To determine whether these effects are mediated by hypertension and hyperfiltration, rather than directly through ANG II, we raised blood pressure to a similar level using DOCA + salt + uninephrectomy and norepinephrine. This caused a fivefold increase in albuminuria in Plce1-deficient mice and a significant increase in the number of sclerotic glomeruli. Consistent with previous findings in mice, we detected strong PLCE1 transcript expression in podocytes using single cell sequencing of human kidney tissue. In hemagglutinin-tagged Plce1 transgenic mice, Plce1 was detected in podocytes and also in glomerular arterioles using immunohistochemistry. Our data demonstrate that Plce1 deficiency in mice predisposes to glomerular damage secondary to hypertensive insults.
Collapse
Affiliation(s)
- Douglas K Atchison
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Christopher L O'Connor
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Rajasree Menon
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Edgar A Otto
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Santhi K Ganesh
- Cardiovascular Division, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Roger C Wiggins
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Alan V Smrcka
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan
| | - Markus Bitzer
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
13
|
Strazza M, Adam K, Smrcka AV, Lerrer S, Mor A. PLCε1 suppresses tumor growth by regulating murine T cell mobilization. Clin Exp Immunol 2019; 200:53-60. [PMID: 31867717 DOI: 10.1111/cei.13409] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2019] [Indexed: 01/01/2023] Open
Abstract
Phospholipase C epsilon 1 (PLCε1) is a unique member of the phospholipase family, in that it also functions as a guanine nucleotide exchange factor (GEF) for the small GTPase Rap1. It is this function as a Rap1 GEF that gives PLCε1 an essential role in chemokine-mediated T cell adhesion. We have utilized a syngeneic tumor model, MC38 cells in C57BL/6 mice, and observed that tumors grow larger and more quickly in the absence of PLCε1. Single-cell analysis revealed an increased CD4+ /CD8+ ratio in the spleens, lymph nodes and tumors of PLCε1 knock-out tumor-bearing mice. T cells isolated from PLCε1 knock-out mice were less activated by multiple phenotypical parameters than those from wild-type mice. We additionally noted a decrease in expression of the chemokine receptors C-X-C chemokine receptor type 4 (CXCR4) and C-C motif chemokine receptor 4 (CCR4) on CD4+ T cells from the spleens, lymph nodes and tumors of PLCε1 knock-out mice compared to wild-type mice, and diminished migration of PLCε1-depleted CD3+ T cells towards stromal cell-derived factor (SDF)-1α. Based on these results, we conclude that PLCε1 is a potential regulator of tumor-infiltrating lymphocytes, functioning, at least in part, at the level of T cell trafficking and recruitment.
Collapse
Affiliation(s)
- M Strazza
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
| | - K Adam
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
| | - A V Smrcka
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - S Lerrer
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
| | - A Mor
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
14
|
Fiedler S, Storch U, Erdogmus S, Gudermann T, Mederos Y Schnitzler M, Dietrich A. Small Fluorescein Arsenical Hairpin-Based Förster Resonance Energy Transfer Analysis Reveals Changes in Amino- to Carboxyl-Terminal Interactions upon OAG Activation of Classical Transient Receptor Potential 6. Mol Pharmacol 2019; 96:90-98. [PMID: 31171574 DOI: 10.1124/mol.119.115949] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 05/02/2019] [Indexed: 01/19/2023] Open
Abstract
Although the overall structure of many classical transient receptor potential proteins (TRPC), including human and murine TRPC6, were recently resolved by cryoelectron microscopy analysis, structural changes during channel activation by 1-oleoyl-1-acetyl-sn-glycerol (OAG), the membrane-permeable analog of diacylglycerol, were not defined. Moreover, data on carboxyl- and amino-terminal interactions were not provided, as the amino-terminal regions of murine and human TRPC6 were not resolved. Therefore, we employed a Förster resonance energy transfer (FRET) approach using a small fluorescein arsenical hairpin (FlAsH) targeted to a short tetracysteine sequence at the unresolved amino-terminus and cerulean, a cyan fluorescent protein, as a tag at the carboxyl-terminus of the murine TRPC6 protein. After OAG as well as GSK-1702934A activation, FRET efficiency was simultaneously and significantly reduced, indicating a decreased interaction between the amino to carboxyl termini in the functional tagged murine TRPC6 tetramer (TRPC6 WT) heterologously expressed in human embryonic kidney 293T cells. There was a significant reduction in the FRET signal obtained from analysis of murine TRPC6 FRET constructs with homologous amino-terminal mutations (M131T, G108S) that had been identified in human patients with inherited focal segmental glomerulosclerosis, a condition that can lead to end-stage renal disease. A novel, designed loss-of-function TRPC6 mutation (N109A) in the amino-terminus in close proximity to the carboxyl-terminus produced similar FRET ratios. SIGNIFICANCE STATEMENT: Our data show for the first time that FlAsH-tagging of ion channels is a promising tool to study conformational changes after channel opening and may significantly advance the analysis of ion channel activation as well as their mutants involved in channelopathies.
Collapse
Affiliation(s)
- Susanne Fiedler
- Walther-Straub-Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL) (S.F., U.S., S.E., T.G., M.M.S., A.D.) and Institute for Cardiovascular Prevention (IPEK) (U.S.), Ludwig-Maximilians-Universitaet Muenchen, Munich, Germany; German Centre for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany (T.G., M.M.S.); and Comprehensive Pneumology Center Munich (CPC-M), Munich, Germany (T.G., A.D.)
| | - Ursula Storch
- Walther-Straub-Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL) (S.F., U.S., S.E., T.G., M.M.S., A.D.) and Institute for Cardiovascular Prevention (IPEK) (U.S.), Ludwig-Maximilians-Universitaet Muenchen, Munich, Germany; German Centre for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany (T.G., M.M.S.); and Comprehensive Pneumology Center Munich (CPC-M), Munich, Germany (T.G., A.D.)
| | - Serap Erdogmus
- Walther-Straub-Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL) (S.F., U.S., S.E., T.G., M.M.S., A.D.) and Institute for Cardiovascular Prevention (IPEK) (U.S.), Ludwig-Maximilians-Universitaet Muenchen, Munich, Germany; German Centre for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany (T.G., M.M.S.); and Comprehensive Pneumology Center Munich (CPC-M), Munich, Germany (T.G., A.D.)
| | - Thomas Gudermann
- Walther-Straub-Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL) (S.F., U.S., S.E., T.G., M.M.S., A.D.) and Institute for Cardiovascular Prevention (IPEK) (U.S.), Ludwig-Maximilians-Universitaet Muenchen, Munich, Germany; German Centre for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany (T.G., M.M.S.); and Comprehensive Pneumology Center Munich (CPC-M), Munich, Germany (T.G., A.D.)
| | - Michael Mederos Y Schnitzler
- Walther-Straub-Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL) (S.F., U.S., S.E., T.G., M.M.S., A.D.) and Institute for Cardiovascular Prevention (IPEK) (U.S.), Ludwig-Maximilians-Universitaet Muenchen, Munich, Germany; German Centre for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany (T.G., M.M.S.); and Comprehensive Pneumology Center Munich (CPC-M), Munich, Germany (T.G., A.D.)
| | - Alexander Dietrich
- Walther-Straub-Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL) (S.F., U.S., S.E., T.G., M.M.S., A.D.) and Institute for Cardiovascular Prevention (IPEK) (U.S.), Ludwig-Maximilians-Universitaet Muenchen, Munich, Germany; German Centre for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany (T.G., M.M.S.); and Comprehensive Pneumology Center Munich (CPC-M), Munich, Germany (T.G., A.D.)
| |
Collapse
|
15
|
Dryer SE, Roshanravan H, Kim EY. TRPC channels: Regulation, dysregulation and contributions to chronic kidney disease. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1041-1066. [PMID: 30953689 DOI: 10.1016/j.bbadis.2019.04.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 12/20/2018] [Accepted: 01/06/2019] [Indexed: 12/13/2022]
Abstract
Mutations in the gene encoding canonical transient receptor potential-6 (TRPC6) channels result in severe nephrotic syndromes that typically lead to end-stage renal disease. Many but not all of these mutations result in a gain in the function of the resulting channel protein. Since those observations were first made, substantial work has supported the hypothesis that TRPC6 channels can also contribute to progression of acquired (non-genetic) glomerular diseases, including primary and secondary FSGS, glomerulosclerosis during autoimmune glomerulonephritis, and possibly in type-1 diabetes. Their regulation has been extensively studied, especially in podocytes, but also in mesangial cells and other cell types present in the kidney. More recent evidence has implicated TRPC6 in renal fibrosis and tubulointerstitial disease caused by urinary obstruction. Consequently TRPC6 is being extensively investigated as a target for drug discovery. Other TRPC family members are present in kidney. TRPC6 can form a functional heteromultimer with TRPC3, and it has been suggested that TRPC5 may also play a role in glomerular disease progression, although the evidence on this is contradictory. Here we review literature on the expression and regulation of TRPC6, TRPC3 and TRPC5 in various cell types of the vertebrate kidney, the evidence that these channels are dysregulated in disease models, and research showing that knock-out or pharmacological inhibition of these channels can reduce the severity of kidney disease. We also summarize several areas that remain controversial, and some of the large gaps of knowledge concerning the fundamental role of these proteins in regulation of renal function.
Collapse
Affiliation(s)
- Stuart E Dryer
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA; Department of Internal Medicine, Division of Nephrology, Baylor College of Medicine, Houston, TX, USA.
| | - Hila Roshanravan
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Eun Young Kim
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| |
Collapse
|
16
|
Cholesterol-Dependent Gating Effects on Ion Channels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1115:167-190. [PMID: 30649760 DOI: 10.1007/978-3-030-04278-3_8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Biomembranes separate a live cell from its environment and keep it in an off-equilibrium, steady state. They contain both phospholipids and nonphospholipids, depending on whether there are phosphate groups in the headgroup regions. Cholesterol (CHOL) is one type of nonphospholipids, and one of the most abundant lipid molecules in humans. Its content in plasma membranes and intracellular membranes varies and is tightly regulated. Voltage-gated ion channels are universally present in every cell and are fairly diversified in the eukaryotic domain of life. Our lipid-dependent gating hypothesis postulates that the controlled switch of the voltage-sensor domains (VSDs) in a voltage-gated potassium (Kv) channel between the "down" and the "up" state (gating) is sensitive to the ratio of phospholipids:nonphospholipids in the annular layer around the channel. High CHOL content is found to exert strong inhibitory effects on Kv channels. Such effects have been observed in in vitro membranes, cultured cells, and animal models for cholesterol metabolic defects. Thermodynamic analysis of the CHOL-dependent gating suggests that the inhibitory effects of CHOL result from collective interactions between annular CHOL molecules and the channel, which appear to be a more generic principle behind the CHOL effects on other ion channels and transporters. We will review the recent progress in the CHOL-dependent gating of voltage-gated ion channels, discuss the current technical limitations, and then expand briefly the learned principles to other ion channels that are known to be sensitive to the CHOL-channel interactions.
Collapse
|
17
|
Krauszman A, Mak TW, Szaszi K, Kuebler WM. Role of phosphatase and tensin homolog in hypoxic pulmonary vasoconstriction. Cardiovasc Res 2018; 113:869-878. [PMID: 28430879 DOI: 10.1093/cvr/cvx076] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 04/16/2017] [Indexed: 01/26/2023] Open
Abstract
Aims Hypoxic pulmonary vasoconstriction (HPV) redistributes blood flow from poorly ventilated to better aerated areas in the lung, thereby optimizing ventilation-perfusion ratio (V/Q). Pulmonary artery smooth muscle cell (PASMC) contraction in response to hypoxia is triggered by Ca2+ influx via transient receptor potential canonical 6 (TRPC6) cation channels that have translocated to caveolae in the plasma membrane. Since phosphatase and tensin homolog (PTEN) was suggested to regulate TRPC6 in endothelial cells, we aimed to define its role in the hypoxic response of PASMCs and as a putative mediator of HPV. Methods and results In isolated perfused mouse lungs, smooth muscle specific PTEN deficiency attenuated pulmonary vasoconstriction in response to hypoxia but not to angiotensin II (Ang II). Analogously, siRNA-mediated knock down of PTEN in human PASMC inhibited the hypoxia-induced increase in cytosolic Ca2+ concentration ([Ca2+]i). Co-immunoprecipitation and proximity ligation assays revealed increased interaction of PTEN with TRPC6 in human PASMC and murine lungs in response to hypoxia. In hypoxic PASMC, both PTEN and TRPC6 translocated to caveolae, and this response was blocked by pharmacological inhibition of Rho-associated protein kinase (ROCK) which in parallel prevented PTEN-TRPC6 interaction, hypoxia-induced [Ca2+]i increase, and HPV in PASMC and murine lungs, respectively. Conclusion Our data indicate a novel interplay between ROCK and [Ca2+]i signalling in HPV via PTEN, in that ROCK mediates interaction of PTEN and TRPC6 which then conjointly translocate to caveolae allowing for Ca2+ influx into and subsequent contraction of PASMC.
Collapse
Affiliation(s)
- Adrienn Krauszman
- Department of Surgery, The Keenan Research Centre for Biomedical Science at St. Michael's Hospital, 209 Victoria Street, M5B 1T8, Toronto, ON, Canada.,Department of Physiology, Institute of Physiology, Charité - Universitaetsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Tak W Mak
- Departments of Medical Biophysics and Immunology, Campbell Family Institute for Breast Cancer Research, Princess Margaret Cancer Centre, University Health Network, 620 University Ave, M5G 2C1, Toronto, ON, Canada
| | - Katalin Szaszi
- Department of Surgery, The Keenan Research Centre for Biomedical Science at St. Michael's Hospital, 209 Victoria Street, M5B 1T8, Toronto, ON, Canada.,Department of Surgery
| | - Wolfgang M Kuebler
- Department of Surgery, The Keenan Research Centre for Biomedical Science at St. Michael's Hospital, 209 Victoria Street, M5B 1T8, Toronto, ON, Canada.,Department of Physiology, Institute of Physiology, Charité - Universitaetsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany.,Department of Surgery.,Department of Physiology, University of Toronto, 27 King's College Circle, M5S 1A1, Toronto, ON, Canada
| |
Collapse
|
18
|
TRPA1 channels: expression in non-neuronal murine lung tissues and dispensability for hyperoxia-induced alveolar epithelial hyperplasia. Pflugers Arch 2018; 470:1231-1241. [PMID: 29754249 DOI: 10.1007/s00424-018-2148-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 03/22/2018] [Accepted: 04/27/2018] [Indexed: 01/31/2023]
Abstract
Transient receptor potential A1 (TRPA1) channels were originally characterized in neuronal tissues but also identified in lung epithelium by staining with fluorescently coupled TRPA1 antibodies. Its exact function in non-neuronal tissues, however, is elusive. TRPA1 is activated in vitro by hypoxia and hyperoxia and is therefore a promising TRP candidate for sensing hyperoxia in pulmonary epithelial cells and for inducing alveolar epithelial hyperplasia. Here, we isolated tracheal, bronchial, and alveolar epithelial cells and show low but detectable TRPA1 mRNA levels in all these cells as well as TRPA1 protein by Western blotting in alveolar type II (AT II) cells. We quantified changes in intracellular Ca2+ ([Ca2+]i) levels induced by application of hyperoxic solutions in primary tracheal epithelial, bronchial epithelial, and AT II cells isolated from wild-type (WT) and TRPA1-deficient (TRPA1-/-) mouse lungs. In all cell types, we detected hyperoxia-induced rises in [Ca2+]i levels, which were not significantly different in TRPA1-deficient cells compared to WT cells. We also tested TRPA1 function in a mouse model for hyperoxia-induced alveolar epithelial hyperplasia. A characteristic significant increase in thickening of alveolar tissues was detected in mouse lungs after exposure to hyperoxia, but not in normoxic WT and TRPA1-/- controls. Quantification of changes in lung morphology in hyperoxic WT and TRPA1-/- mice, however, again revealed no significant changes. Therefore, TRPA1 expression does neither appear to be a key player for hyperoxia-induced changes in [Ca2+]i levels in primary lung epithelial cells, nor being essential for the development of hyperoxia-induced alveolar epithelial hyperplasia.
Collapse
|
19
|
Urban N, Neuser S, Hentschel A, Köhling S, Rademann J, Schaefer M. Pharmacological inhibition of focal segmental glomerulosclerosis-related, gain of function mutants of TRPC6 channels by semi-synthetic derivatives of larixol. Br J Pharmacol 2017; 174:4099-4122. [PMID: 28800680 DOI: 10.1111/bph.13977] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 07/24/2017] [Accepted: 07/31/2017] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND PURPOSE Gain of function mutations in TRPC6 channels can cause autosomal dominant forms of focal segmental glomerulosclerosis (FSGS). Validated inhibitors of TRPC6 channels that are biologically active on FSGS-related TRPC6 mutants are eagerly sought. EXPERIMENTAL APPROACH We synthesized new TRPC6-inhibiting modulators from larixol, a resiniferous constituent of Larix decidua, and tested the potency and selectivity in cell lines stably expressing various TRPC channel isoforms. Channel activation was followed by Ca2+ influx analyses and electrophysiological recordings. The most promising compound larixyl carbamate (LC) was tested on native TRPC6 channels and TRPC6 constructs carrying FSGS-related point mutations. KEY RESULTS LC exhibited an about 30-fold preference for TRPC6 over TRPC3 channels and a fivefold preference for TRPC6 over TRPC7 channels. Six FSGS-related TRPC6 mutants, including the highly active M132T and R175Q variants, were strongly inhibited by 1 μM LC. Surprisingly, no TRPC6-related Ca2+ signals were detectable in primary murine podocytes, or in acutely isolated glomeruli. in these preparations. Quantitative PCR revealed a 20-fold to 50-fold lower abundance of TRPC6 transcripts in rat or mouse podocytes, compared with pulmonary artery smooth muscle cells from the same species. Accordingly, electrophysiological recordings demonstrated that DAG-induced currents in murine podocytes are very small, but sensitive to LC. CONCLUSIONS AND IMPLICATIONS In spite of their low abundance in native podocytes, native TRPC6 channels are targetable using larixol-derived TRPC6 inhibitors. As observed with wild-type TRPC6 channels, FSGS-related TRPC6 mutants were sensitive to the newly developed inhibitors, paving the way for experimental therapies.
Collapse
Affiliation(s)
- Nicole Urban
- Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, Universität Leipzig, Leipzig, Germany
| | - Sonja Neuser
- Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, Universität Leipzig, Leipzig, Germany
| | - Anika Hentschel
- Institut für Pharmazie, Freie Universität Berlin, Berlin, Germany
| | | | - Jörg Rademann
- Institut für Pharmazie, Freie Universität Berlin, Berlin, Germany
| | - Michael Schaefer
- Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, Universität Leipzig, Leipzig, Germany
| |
Collapse
|
20
|
PLCε1 regulates SDF-1α-induced lymphocyte adhesion and migration to sites of inflammation. Proc Natl Acad Sci U S A 2017; 114:2693-2698. [PMID: 28213494 DOI: 10.1073/pnas.1612900114] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Regulation of integrins is critical for lymphocyte adhesion to endothelium and migration throughout the body. Inside-out signaling to integrins is mediated by the small GTPase Ras-proximate-1 (Rap1). Using an RNA-mediated interference screen, we identified phospholipase Cε 1 (PLCε1) as a crucial regulator of stromal cell-derived factor 1 alpha (SDF-1α)-induced Rap1 activation. We have shown that SDF-1α-induced activation of Rap1 is transient in comparison with the sustained level following cross-linking of the antigen receptor. We identified that PLCε1 was necessary for SDF-1α-induced adhesion using shear stress, cell morphology alterations, and crawling on intercellular adhesion molecule 1 (ICAM-1)-expressing cells. Structure-function experiments to separate the dual-enzymatic function of PLCε1 uncover necessary contributions of the CDC25, Pleckstrin homology, and Ras-associating domains, but not phospholipase activity, to this pathway. In the mouse model of delayed type hypersensitivity, we have shown an essential role for PLCε1 in T-cell migration to inflamed skin, but not for cytokine secretion and proliferation in regional lymph nodes. Our results reveal a signaling pathway where SDF-1α induces T-cell adhesion through activation of PLCε1, suggesting that PLCε1 is a specific potential target in treating conditions involving migration of T cells to inflamed organs.
Collapse
|
21
|
Zhai S, Liu C, Zhang L, Zhu J, Guo J, Zhang J, Chen Z, Zhou W, Chang T, Xu S, Qi Y, Zhuang T, Yu N, Wang W, Wang H, Yu S, Li X. PLCE1 Promotes Esophageal Cancer Cell Progression by Maintaining the Transcriptional Activity of Snail. Neoplasia 2017; 19:154-164. [PMID: 28147304 PMCID: PMC5279705 DOI: 10.1016/j.neo.2016.12.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 12/07/2016] [Accepted: 12/12/2016] [Indexed: 02/07/2023] Open
Abstract
Esophageal cancer is among the most deadly malignant diseases. However, the genetic factors contributing to its occurrence are poorly understood. Multiple studies with large clinic-based cohorts revealed that variations of the phospholipase C epsilon (PLCE1) gene were associated with esophageal cancer susceptibility. However, the causative role of PLCE1 in esophageal cancer is not clear. We inactivated the functional alleles of PLCE1 by CRISPR/Cas9 genome editing technology. The resultant PLCE1 inactivated cells were analyzed both in vitro and in vivo. Our results showed that loss of PLCE1 dramatically decreased the invasion and proliferation capacity of esophageal carcinoma cells in vitro. Moreover, such PLCE1 inactivated tumor grafts exhibited significantly decreased tumor size in mice. We found that PLCE1 was required to maintain protein level of snail a key transcription factor responsible for invasion. Our further transcriptomic data revealed that deficient cells were significantly decreased in expression of genes enriched as targets of Snail. Strikingly, recovery of Snail protein at least partially rescued the invasion and proliferation capacity in PLCE1 inactivated cells. In ESCC clinical specimens, PLCE1 was correlated with tumor stage (P<.0001). Interestingly, PLCE1 expression was positively correlated Snail by immunohistochemistry in such specimens (P<.0001). Therefore, our functional experiments showed the essential roles of PLCE1 in esophageal carcinoma cells and provided evidences that targeting PLCE1 and its downstream molecules could be effective therapies for esophageal cancer.
Collapse
Affiliation(s)
- Shicong Zhai
- Center for Cancer Research, Xinxiang Medical University, Xinxiang, Henan, China; Department of Gastroenterology, the Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Cui Liu
- School of Nursing, Xinxiang Medical University, Henan, China
| | - Lichen Zhang
- School of Laboratory Medicine, Xinxiang Medical University
| | - Jian Zhu
- School of Laboratory Medicine, Xinxiang Medical University; Research Center for Immunology, Xinxiang Medical University, Henan, China
| | - Jiqiang Guo
- Research Center for Immunology, Xinxiang Medical University, Henan, China
| | - Jinghang Zhang
- Department of Pathology, the First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, China
| | - Zhijun Chen
- Department of Thoracic Surgery, the First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, China
| | - Wenping Zhou
- Lymphoma Institute, Zhengzhou, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Tingmin Chang
- Department of Gastroenterology, the First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, China
| | - Siguang Xu
- Institute of Lung and Molecular Therapy, Xinxiang Medical University, China
| | - Yijun Qi
- Key Laboratory of Cellular and Molecular Immunology, College of Medicine, Henan University, Kaifeng, Henan, China
| | - Ting Zhuang
- School of Laboratory Medicine, Xinxiang Medical University; Research Center for Immunology, Xinxiang Medical University, Henan, China
| | - Na Yu
- Department of Gastroenterology, the Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Weilong Wang
- Department of Gastroenterology, the Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Hui Wang
- Ontario Cancer Institute, Campbell Family Institute for Breast Cancer Research, University of Toronto, Toronto, Canada
| | - Sifan Yu
- Research Center for Immunology, Xinxiang Medical University, Henan, China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education) Department of Renal cancer and Melanoma, Peking University School of Oncology, Beijing Cancer Hospital and Institute, Beijing, China
| | - Xiumin Li
- Center for Cancer Research, Xinxiang Medical University, Xinxiang, Henan, China; Department of Gastroenterology, the Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China.
| |
Collapse
|
22
|
Roshanravan H, Kim EY, Dryer SE. 20-Hydroxyeicosatetraenoic Acid (20-HETE) Modulates Canonical Transient Receptor Potential-6 (TRPC6) Channels in Podocytes. Front Physiol 2016; 7:351. [PMID: 27630573 PMCID: PMC5005377 DOI: 10.3389/fphys.2016.00351] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 08/02/2016] [Indexed: 01/08/2023] Open
Abstract
The arachidonic acid metabolite 20-hydroxyeicosatetraenoic acid (20-HETE) regulates renal function, including changes in glomerular function evoked during tubuloglomerular feedback (TGF). This study describes the cellular actions of 20-HETE on cultured podocytes, assessed by whole-cell recordings from cultured podocytes combined with pharmacological and cell-biological manipulations of cells. Bath superfusion of 20-HETE activates cationic currents that are blocked by the pan-TRP blocker SKF-96365 and by 50 μM La3+, and which are attenuated after siRNA knockdown of TRPC6 subunits. Similar currents are evoked by a membrane-permeable analog of diacylgycerol (OAG), but OAG does not occlude responses to maximally-activating concentrations of 20-HETE (20 μM). Exposure to 20-HETE also increased steady-state surface abundance of TRPC6 subunits in podocytes as assessed by cell-surface biotinylation assays, and increased cytosolic concentrations of reactive oxygen species (ROS). TRPC6 activation by 20-HETE was eliminated in cells pretreated with TEMPOL, a membrane-permeable superoxide dismutase mimic. Activation of TRPC6 by 20-HETE was also blocked when whole-cell recording pipettes contained GDP-βS, indicating a role for either small or heterotrimeric G proteins in the transduction cascade. Responses to 20-HETE were eliminated by siRNA knockdown of podocin, a protein that organizes NADPH oxidase complexes with TRPC6 subunits in this cell type. In summary, modulation of ionic channels in podocytes may contribute to glomerular actions of 20-HETE.
Collapse
Affiliation(s)
- Hila Roshanravan
- Department of Biology and Biochemistry, University of Houston Houston, TX, USA
| | - Eun Y Kim
- Department of Biology and Biochemistry, University of Houston Houston, TX, USA
| | - Stuart E Dryer
- Department of Biology and Biochemistry, University of HoustonHouston, TX, USA; Division of Nephrology, Baylor College of MedicineHouston, TX, USA
| |
Collapse
|
23
|
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: 5.3] [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
|
24
|
Forst AL, Olteanu VS, Mollet G, Wlodkowski T, Schaefer F, Dietrich A, Reiser J, Gudermann T, Mederos y Schnitzler M, Storch U. Podocyte Purinergic P2X4 Channels Are Mechanotransducers That Mediate Cytoskeletal Disorganization. J Am Soc Nephrol 2015; 27:848-62. [PMID: 26160898 DOI: 10.1681/asn.2014111144] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 06/10/2015] [Indexed: 11/03/2022] Open
Abstract
Podocytes are specialized, highly differentiated epithelial cells in the kidney glomerulus that are exposed to glomerular capillary pressure and possible increases in mechanical load. The proteins sensing mechanical forces in podocytes are unconfirmed, but the classic transient receptor potential channel 6 (TRPC6) interacting with the MEC-2 homolog podocin may form a mechanosensitive ion channel complex in podocytes. Here, we observed that podocytes respond to mechanical stimulation with increased intracellular calcium concentrations and increased inward cation currents. However, TRPC6-deficient podocytes responded in a manner similar to that of control podocytes, and mechanically induced currents were unaffected by genetic inactivation of TRPC1/3/6 or administration of the broad-range TRPC blocker SKF-96365. Instead, mechanically induced currents were significantly decreased by the specific P2X purinoceptor 4 (P2X4) blocker 5-BDBD. Moreover, mechanical P2X4 channel activation depended on cholesterol and podocin and was inhibited by stabilization of the actin cytoskeleton. Because P2X4 channels are not intrinsically mechanosensitive, we investigated whether podocytes release ATP upon mechanical stimulation using a fluorometric approach. Indeed, mechanically induced ATP release from podocytes was observed. Furthermore, 5-BDBD attenuated mechanically induced reorganization of the actin cytoskeleton. Altogether, our findings reveal a TRPC channel-independent role of P2X4 channels as mechanotransducers in podocytes.
Collapse
Affiliation(s)
- Anna-Lena Forst
- Walther-Straub-Institute of Pharmacology and Toxicology, University of Munich, Munich, Germany
| | - Vlad Sorin Olteanu
- Walther-Straub-Institute of Pharmacology and Toxicology, University of Munich, Munich, Germany
| | - Géraldine Mollet
- INSERM U1163, Laboratory of Hereditary Kidney Diseases, Imagine Institute, Paris, France
| | - Tanja Wlodkowski
- Division of Pediatric Nephrology, Heidelberg University Center for Pediatrics and Adolescent Medicine, Heidelberg, Germany
| | - Franz Schaefer
- Division of Pediatric Nephrology, Heidelberg University Center for Pediatrics and Adolescent Medicine, Heidelberg, Germany
| | - Alexander Dietrich
- Walther-Straub-Institute of Pharmacology and Toxicology, University of Munich, Munich, Germany
| | - Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, Illinois; and
| | - Thomas Gudermann
- Walther-Straub-Institute of Pharmacology and Toxicology, University of Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), Munich Heart Alliance, Munich, Germany
| | - Michael Mederos y Schnitzler
- Walther-Straub-Institute of Pharmacology and Toxicology, University of Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), Munich Heart Alliance, Munich, Germany
| | - Ursula Storch
- Walther-Straub-Institute of Pharmacology and Toxicology, University of Munich, Munich, Germany
| |
Collapse
|