1
|
Hickey MJ, Sudhakar V. Looking below the surface: using intravital imaging to decipher inflammatory renal disease and renal cell injury. Am J Physiol Renal Physiol 2025; 328:F418-F430. [PMID: 39918796 DOI: 10.1152/ajprenal.00321.2024] [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: 11/08/2024] [Revised: 11/22/2024] [Accepted: 02/02/2025] [Indexed: 03/04/2025] Open
Abstract
Renal function can be perturbed by a range of stimuli that cause cellular injury and inflammation in the kidney. These injurious and inflammatory processes are typically dynamic and progressive, involving the actions of highly migratory cells such as leukocytes and cellular responses that occur over time spans ranging from seconds to weeks. Understanding these dynamic responses has entailed the use of imaging technologies that allow visualization and capture of events over different time spans, ideally in intact organs in live, experimental animals. The technique that allows this is intravital imaging. Intravital imaging, particularly multiphoton intravital microscopy, has been crucial to the investigation of dynamic physiological and pathophysiological processes in the kidney for many years, driving key developments in our understanding of renal (patho)physiology. This includes the mechanisms of ultrafiltrate generation, the response to acute kidney injury, and how inflammatory leukocytes are recruited to and cause injury in the kidney. This review describes the key studies that have applied intravital imaging to the investigation of models of inflammatory renal disease. The responses examined include those restricted to the glomerulus and the effects of acute kidney injury on the tubulointerstitium. Future innovations and directions in this field of research are also discussed.
Collapse
Affiliation(s)
- Michael J Hickey
- Centre for Inflammatory Diseases, Department of Medicine, Monash Medical Centre, Monash University, Clayton, Victoria, Australia
| | - Vaishnavi Sudhakar
- Centre for Inflammatory Diseases, Department of Medicine, Monash Medical Centre, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
2
|
Hackl A, Weber LT. The Ca 2+-actin-cytoskeleton axis in podocytes is an important, non-immunologic target of immunosuppressive therapy in proteinuric kidney diseases. Pediatr Nephrol 2025:10.1007/s00467-025-06670-z. [PMID: 39856247 DOI: 10.1007/s00467-025-06670-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 12/19/2024] [Accepted: 12/19/2024] [Indexed: 01/27/2025]
Abstract
The integrity of the filtration barrier of the kidney relies on the proper composition of podocyte interdigitating foot processes. Their architecture is supported by a complex actin-cytoskeleton. Following podocyte stress or injury, podocytes encounter structural changes, including rearrangement of the actin network and subsequent effacement of the foot processes. Immunosuppressive drugs, which are currently used as treatment in proteinuric kidney diseases, have been shown to exert not only immune-mediated effects. This review will focus on the direct effects of glucocorticoids, cyclosporine A, tacrolimus, mycophenolate mofetil, and rituximab on podocytes by regulation of Ca2+ ion channels and consecutive downstream signaling which prevent cytoskeletal rearrangements and ultimately proteinuria. In addition, the efficacy of these drugs in genetic nephrotic syndrome will be discussed.
Collapse
Affiliation(s)
- Agnes Hackl
- Department of Pediatrics, University of Cologne, Faculty of Medicine and University Hospital Cologne, Kerpener Street 62, 50937, Cologne, Germany.
| | - Lutz T Weber
- Department of Pediatrics, University of Cologne, Faculty of Medicine and University Hospital Cologne, Kerpener Street 62, 50937, Cologne, Germany
| |
Collapse
|
3
|
Figurek A, Jankovic N, Hall AM. Quantitative Intravital Calcium Imaging in Mouse Kidney. Methods Mol Biol 2025; 2861:187-193. [PMID: 39395106 DOI: 10.1007/978-1-0716-4164-4_14] [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] [Indexed: 10/14/2024]
Abstract
Intracellular calcium is an important regulator of solute transport in renal epithelial cells, and disordered calcium signaling may underlie the pathogenesis of certain kidney diseases. Intravital multiphoton imaging of the kidney in transgenic mice expressing highly sensitive fluorescent reporters allows detailed study of calcium signals within different specialized segments of the renal tubule and how these are integrated with other cellular processes. Moreover, changes in activity can be observed in real time in response to physiological interventions or disease-causing insults. In this chapter, we will provide a detailed protocol for performing this powerful research technique.
Collapse
Affiliation(s)
- Andreja Figurek
- Institute of Anatomy, University of Zurich, Zürich, Switzerland
| | - Nevena Jankovic
- Institute of Anatomy, University of Zurich, Zürich, Switzerland
| | - Andrew M Hall
- Institute of Anatomy, University of Zurich, Zürich, Switzerland.
- Department of Nephrology, University Hospital Zurich, Zürich, Switzerland.
- Zurich Kidney Center, University of Zurich, Zürich, Switzerland.
| |
Collapse
|
4
|
Martinez MM, Walsh JR, Kamocka MM, Lee H, Dunn KW. Longitudinal intravital microscopy of the mouse kidney: inflammatory responses to abdominal imaging windows. Am J Physiol Renal Physiol 2024; 327:F845-F868. [PMID: 39323386 PMCID: PMC11563595 DOI: 10.1152/ajprenal.00071.2024] [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: 03/01/2024] [Revised: 09/10/2024] [Accepted: 09/10/2024] [Indexed: 09/27/2024] Open
Abstract
Intravital microscopy enables direct observation of cell biology and physiology at subcellular resolution in real time in living animals. Implanted windows extend the scope of intravital microscopy to processes extending for weeks or even months, such as disease progression or tumor development. However, a question that must be addressed in such studies is whether the imaging window, like any foreign body, triggers an inflammatory response, and whether that response alters the biological process under investigation. To directly evaluate this question, we conducted large-scale intravital microscopy of the kidney of LysM-EGFP mice over time after implantation of abdominal imaging windows. These studies demonstrate that windows stimulated a variety of changes consistent with a foreign body response. Within a few days of implantation, leukocytes were recruited to the window and the region between the window and kidney where, over the next 16 days, they increased in number in an expanding volume that developed a new vascular network. These changes were accompanied by a dramatic increase in glomerular albumin permeability within 2-5 days of implantation. Similar results were obtained from mice implanted with windows coated with poly(l-lysine)-graft-polyethylene glycol (PLL-g-PEG), but not from immune-deficient mice. These studies demonstrate the importance of evaluating whether implanted windows induce an inflammatory response, and whether that response impacts the processes under evaluation in longitudinal intravital microscopy studies.NEW & NOTEWORTHY Intravital microscopy studies of LysM-EGFP mice demonstrate that abdominal imaging windows placed over the kidney stimulated a variety of changes consistent with a foreign body response. Within a day of implantation, leukocytes were recruited to the window where, over the next 16 days, they increased in number in an expanding volume that developed a new vascular network. These changes were accompanied by a dramatic increase in glomerular permeability to albumin.
Collapse
Affiliation(s)
- Michelle M Martinez
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Julia R Walsh
- Weldon School of Biomedical Engineering, Center for Implantable Devices, Purdue University, West Lafayette, Indiana, United States
| | - Malgorzata M Kamocka
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Hyowon Lee
- Weldon School of Biomedical Engineering, Center for Implantable Devices, Purdue University, West Lafayette, Indiana, United States
| | - Kenneth W Dunn
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States
- Weldon School of Biomedical Engineering, Center for Implantable Devices, Purdue University, West Lafayette, Indiana, United States
| |
Collapse
|
5
|
Gyarmati G, Shroff UN, Izuhara A, Deepak S, Komers R, Bedard PW, Peti-Peterdi J. Sparsentan improves glomerular hemodynamics, cell functions, and tissue repair in a mouse model of FSGS. JCI Insight 2024; 9:e177775. [PMID: 39226116 PMCID: PMC11466195 DOI: 10.1172/jci.insight.177775] [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: 11/20/2023] [Accepted: 08/28/2024] [Indexed: 09/05/2024] Open
Abstract
Dual endothelin-1 (ET-1) and angiotensin II (AngII) receptor antagonism with sparsentan has strong antiproteinuric actions via multiple potential mechanisms that are more pronounced, or additive, compared with current standard of care using angiotensin receptor blockers (ARBs). Considering the many actions of ET-1 and AngII on multiple cell types, this study aimed to determine glomeruloprotective mechanisms of sparsentan compared to the ARB losartan by direct visualization of its effects in the intact kidney in focal segmental glomerulosclerosis (FSGS) using intravital multiphoton microscopy. In both healthy and FSGS models, sparsentan treatment increased afferent/efferent arteriole diameters; increased or preserved blood flow and single-nephron glomerular filtration rate; attenuated acute ET-1 and AngII-induced increases in podocyte calcium; reduced proteinuria; preserved podocyte number; increased both endothelial and renin lineage cells and clones in vasculature, glomeruli, and tubules; restored glomerular endothelial glycocalyx; and attenuated mitochondrial stress and immune cell homing. These effects were either not observed or of smaller magnitude with losartan. The pleiotropic nephroprotective effects of sparsentan included improved hemodynamics, podocyte and endothelial cell functions, and tissue repair. Compared with losartan, sparsentan was more effective in the sustained preservation of kidney structure and function, which underscores the importance of the ET-1 component in FSGS pathogenesis and therapy.
Collapse
Affiliation(s)
- Georgina Gyarmati
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Urvi Nikhil Shroff
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Audrey Izuhara
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Sachin Deepak
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Radko Komers
- Travere Therapeutics, San Diego, California, USA
| | | | - Janos Peti-Peterdi
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
6
|
Ma R, Tao Y, Wade ML, Mallet RT. Non-voltage-gated Ca 2+ channel signaling in glomerular cells in kidney health and disease. Am J Physiol Renal Physiol 2024; 327:F249-F264. [PMID: 38867675 PMCID: PMC11460346 DOI: 10.1152/ajprenal.00130.2024] [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: 04/29/2024] [Revised: 06/10/2024] [Accepted: 06/10/2024] [Indexed: 06/14/2024] Open
Abstract
Positioned at the head of the nephron, the renal corpuscle generates a plasma ultrafiltrate to initiate urine formation. Three major cell types within the renal corpuscle, the glomerular mesangial cells, podocytes, and glomerular capillary endothelial cells, communicate via endocrine- and paracrine-signaling mechanisms to maintain the structure and function of the glomerular capillary network and filtration barrier. Ca2+ signaling mediated by several distinct plasma membrane Ca2+ channels impacts the functions of all three cell types. The past two decades have witnessed pivotal advances in understanding of non-voltage-gated Ca2+ channel function and regulation in the renal corpuscle in health and renal disease. This review summarizes the current knowledge of the physiological and pathological impact of non-voltage-gated Ca2+ channel signaling in mesangial cells, podocytes and glomerular capillary endothelium. The main focus is on transient receptor potential and store-operated Ca2+ channels, but ionotropic N-methyl-d-aspartate receptors and purinergic receptors also are discussed. This update of Ca2+ channel functions and their cellular signaling cascades in the renal corpuscle is intended to inform the development of therapeutic strategies targeting these channels to treat kidney diseases, particularly diabetic nephropathy.
Collapse
Affiliation(s)
- Rong Ma
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Yu Tao
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Michael L Wade
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Robert T Mallet
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| |
Collapse
|
7
|
Wiesner E, Binz-Lotter J, Hackl A, Unnersjö-Jess D, Rutkowski N, Benzing T, Hackl MJ. Correlative multiphoton-STED microscopy of podocyte calcium levels and slit diaphragm ultrastructure in the renal glomerulus. Sci Rep 2024; 14:13019. [PMID: 38844492 PMCID: PMC11156906 DOI: 10.1038/s41598-024-63507-9] [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: 02/23/2024] [Accepted: 05/29/2024] [Indexed: 06/09/2024] Open
Abstract
In recent years functional multiphoton (MP) imaging of vital mouse tissues and stimulation emission depletion (STED) imaging of optically cleared tissues allowed new insights into kidney biology. Here, we present a novel workflow where MP imaging of calcium signals can be combined with super-resolved STED imaging for morphological analysis of the slit diaphragm (SD) within the same glomerulus. Mice expressing the calcium indicator GCaMP3 in podocytes served as healthy controls or were challenged with two different doses of nephrotoxic serum (NTS). NTS induced glomerular damage in a dose dependent manner measured by shortening of SD length. In acute kidney slices (AKS) intracellular calcium levels increased upon disease but showed a high variation between glomeruli. We could not find a clear correlation between intracellular calcium levels and SD length in the same glomerulus. Remarkably, analysis of the SD morphology of glomeruli selected during MP calcium imaging revealed a higher percentage of completely disrupted SD architecture than estimated by STED imaging alone. Our novel co-imaging protocol is applicable to a broad range of research questions. It can be used with different tissues and is compatible with diverse reporters and target proteins.
Collapse
Affiliation(s)
- Eva Wiesner
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Julia Binz-Lotter
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Agnes Hackl
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- Department of Pediatrics, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - David Unnersjö-Jess
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Nelli Rutkowski
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Thomas Benzing
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Matthias J Hackl
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.
| |
Collapse
|
8
|
Bohovyk R, Khedr S, Levchenko V, Stefanenko M, Semenikhina M, Kravtsova O, Isaeva E, Geurts AM, Klemens CA, Palygin O, Staruschenko A. Protease-Activated Receptor 1-Mediated Damage of Podocytes in Diabetic Nephropathy. Diabetes 2023; 72:1795-1808. [PMID: 37722138 PMCID: PMC10658073 DOI: 10.2337/db23-0032] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 09/04/2023] [Indexed: 09/20/2023]
Abstract
There is clinical evidence that increased urinary serine proteases are associated with the disease severity in the setting of diabetic nephropathy (DN). Elevation of serine proteases may mediate [Ca2+]i dynamics in podocytes through the protease-activated receptors (PARs) pathway, including associated activation of nonspecific cation channels. Cultured human podocytes and freshly isolated glomeruli were used for fluorescence and immunohistochemistry stainings, calcium imaging, Western blot analysis, scanning ion conductance microscopy, and patch clamp analysis. Goto-Kakizaki, Wistar, type 2 DN (T2DN), and a novel PAR1 knockout on T2DN rat background rats were used to test the importance of PAR1-mediated signaling in DN settings. We found that PAR1 activation increases [Ca2+]i via TRPC6 channels. Both human cultured podocytes exposed to high glucose and podocytes from freshly isolated glomeruli of T2DN rats had increased PAR1-mediated [Ca2+]i compared with controls. Imaging experiments revealed that PAR1 activation plays a role in podocyte morphological changes. T2DN rats exhibited a significantly higher response to thrombin and urokinase. Moreover, the plasma concentration of thrombin in T2DN rats was significantly elevated compared with Wistar rats. T2DNPar1-/- rats were embryonically lethal. T2DNPar1+/- rats had a significant decrease in glomerular damage associated with DN lesions. Overall, these data provide evidence that, during the development of DN, elevated levels of serine proteases promote an excessive [Ca2+]i influx in podocytes through PAR1-TRPC6 signaling, ultimately leading to podocyte apoptosis, the development of albuminuria, and glomeruli damage. ARTICLE HIGHLIGHTS Increased urinary serine proteases are associated with diabetic nephropathy. During the development of diabetic nephropathy in type 2 diabetes, the elevation of serine proteases could overstimulate protease-activated receptor 1 (PAR1). PAR1 signaling is involved in the development of DN via TRPC6-mediated intracellular calcium signaling. This study provides fundamental knowledge that can be used to develop efficient therapeutic approaches targeting serine proteases or corresponding PAR pathways to prevent or slow the progression of diabetes-associated kidney diseases.
Collapse
Affiliation(s)
- Ruslan Bohovyk
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL
| | - Sherif Khedr
- Department of Physiology, Faculty of Medicine, Ain-Shams University, Cairo, Egypt
| | - Vladislav Levchenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL
| | - Mariia Stefanenko
- Department of Medicine, Division of Nephrology, Medical University of South Carolina, Charleston, SC
| | - Marharyta Semenikhina
- Department of Medicine, Division of Nephrology, Medical University of South Carolina, Charleston, SC
| | - Olha Kravtsova
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL
| | - Elena Isaeva
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI
| | - Aron M. Geurts
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI
| | - Christine A. Klemens
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL
- Hypertension and Kidney Research Center, University of South Florida, Tampa, FL
| | - Oleg Palygin
- Department of Medicine, Division of Nephrology, Medical University of South Carolina, Charleston, SC
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC
| | - Alexander Staruschenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL
- Hypertension and Kidney Research Center, University of South Florida, Tampa, FL
- James A. Haley Veterans’ Hospital, Tampa, FL
| |
Collapse
|
9
|
Hackl A, Nüsken E, Voggel J, Abo Zed SED, Binz-Lotter J, Unnersjö-Jess D, Müller C, Fink G, Bohl K, Wiesner E, Diefenhardt P, Dafinger C, Chen H, Wohlfarth M, Müller RU, Hackl MJ, Schermer B, Nüsken KD, Weber LT. The effect of mycophenolate mofetil on podocytes in nephrotoxic serum nephritis. Sci Rep 2023; 13:14167. [PMID: 37644089 PMCID: PMC10465485 DOI: 10.1038/s41598-023-41222-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 08/23/2023] [Indexed: 08/31/2023] Open
Abstract
Mycophenolate mofetil (MMF) is applied in proteinuric kidney diseases, but the exact mechanism of its effect on podocytes is still unknown. Our previous in vitro experiments suggested that MMF can ameliorate podocyte damage via restoration of the Ca2+-actin cytoskeleton axis. The goal of this study was to characterize podocyte biology during MMF treatment in nephrotoxic serum (NTS) nephritis (NTN). NTN was induced in three-week old wild-type mice. On day 3, half of the mice were treated with MMF (100 mg/kgBW/d p.o.) for one week. On day 10, we performed proteomic analysis of glomeruli as well as super-resolution imaging of the slit diaphragm. For multiphoton imaging of Ca2+ concentration ([Ca2+]i), the experimental design was repeated in mice expressing podocyte-specific Ca2+ sensor. MMF ameliorated the proteinuria and crescent formation induced by NTS. We identified significant changes in the abundance of proteins involved in Ca2+ signaling and actin cytoskeleton regulation, which was further confirmed by direct [Ca2+]i imaging in podocytes showing decreased Ca2+ levels after MMF treatment. This was associated with a tendency to restoration of podocyte foot process structure. Here, we provide evidence that MPA has a substantial direct effect on podocytes. MMF contributes to improvement of [Ca2+]i and amelioration of the disorganized actin cytoskeleton in podocytes. These data extend the knowledge of direct effects of immunosuppressants on podocytes that may contribute to a more effective treatment of proteinuric glomerulopathies with the least possible side effects.
Collapse
Affiliation(s)
- A Hackl
- Department of Pediatrics, Faculty of Medicine, University Hospital Cologne, University of Cologne, Kerpener Street 62, 50937, Cologne, Germany.
- CECAD, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany.
| | - E Nüsken
- Department of Pediatrics, Faculty of Medicine, University Hospital Cologne, University of Cologne, Kerpener Street 62, 50937, Cologne, Germany
| | - J Voggel
- Department of Pediatrics, Faculty of Medicine, University Hospital Cologne, University of Cologne, Kerpener Street 62, 50937, Cologne, Germany
| | - S E D Abo Zed
- Department of Pediatrics, Faculty of Medicine, University Hospital Cologne, University of Cologne, Kerpener Street 62, 50937, Cologne, Germany
- CECAD, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - J Binz-Lotter
- CECAD, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
- Department 2 of Internal Medicine, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - D Unnersjö-Jess
- CECAD, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
- Department 2 of Internal Medicine, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - C Müller
- Department of Therapeutic Drug Monitoring, Pharmacology at the Laboratory Centre, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - G Fink
- Department of Pediatrics, Faculty of Medicine, University Hospital Cologne, University of Cologne, Kerpener Street 62, 50937, Cologne, Germany
| | - K Bohl
- CECAD, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
- Department 2 of Internal Medicine, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - E Wiesner
- CECAD, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
- Department 2 of Internal Medicine, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - P Diefenhardt
- CECAD, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
- Department 2 of Internal Medicine, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - C Dafinger
- Department of Pediatrics, Faculty of Medicine, University Hospital Cologne, University of Cologne, Kerpener Street 62, 50937, Cologne, Germany
- CECAD, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - H Chen
- CECAD, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
- Department 2 of Internal Medicine, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - M Wohlfarth
- Department of Pediatrics, Faculty of Medicine, University Hospital Cologne, University of Cologne, Kerpener Street 62, 50937, Cologne, Germany
| | - R-U Müller
- CECAD, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
- Department 2 of Internal Medicine, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Rare Kidney Diseases Cologne, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - M J Hackl
- CECAD, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
- Department 2 of Internal Medicine, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - B Schermer
- CECAD, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
- Department 2 of Internal Medicine, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - K-D Nüsken
- Department of Pediatrics, Faculty of Medicine, University Hospital Cologne, University of Cologne, Kerpener Street 62, 50937, Cologne, Germany
| | - L T Weber
- Department of Pediatrics, Faculty of Medicine, University Hospital Cologne, University of Cologne, Kerpener Street 62, 50937, Cologne, Germany
- Center for Rare Kidney Diseases Cologne, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
10
|
Bordoni L, Kristensen AM, Sardella D, Kidmose H, Pohl L, Krag SRP, Schiessl IM. Longitudinal tracking of acute kidney injury reveals injury propagation along the nephron. Nat Commun 2023; 14:4407. [PMID: 37479698 PMCID: PMC10362041 DOI: 10.1038/s41467-023-40037-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 07/10/2023] [Indexed: 07/23/2023] Open
Abstract
Acute kidney injury (AKI) is an important risk factor for chronic kidney disease (CKD), but the underlying mechanisms of failed tubule repair and AKI-CKD transition are incompletely understood. In this study, we aimed for dynamic tracking of tubule injury and remodeling to understand if focal injury upon AKI may spread over time. Here, we present a model of AKI, in which we rendered only half of the kidney ischemic. Using serial intravital 2-photon microscopy and genetic identification of cycling cells, we tracked dynamic tissue remodeling in post- and non-ischemic kidney regions simultaneously and over 3 weeks. Spatial and temporal analysis of cycling cells relative to initial necrotic cell death demonstrated pronounced injury propagation and expansion into non-necrotic tissue regions, which predicted tubule atrophy with epithelial VCAM1 expression. In summary, our longitudinal analyses of tubule injury, remodeling, and fate provide important insights into AKI pathology.
Collapse
Affiliation(s)
- Luca Bordoni
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- GliaLab and Letten Centre, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | | | - Donato Sardella
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Hanne Kidmose
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Layla Pohl
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | | |
Collapse
|
11
|
Bagang N, Gupta K, Singh G, Kanuri SH, Mehan S. Protease-activated receptors in kidney diseases: A comprehensive review of pathological roles, therapeutic outcomes and challenges. Chem Biol Interact 2023; 377:110470. [PMID: 37011708 DOI: 10.1016/j.cbi.2023.110470] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/21/2023] [Accepted: 03/31/2023] [Indexed: 04/03/2023]
Abstract
Studies have demonstrated that protease-activated receptors (PARs) with four subtypes (PAR1-4) are mainly expressed in the renal epithelial, endothelial, and podocyte cells. Some endogenous and urinary proteases, namely thrombin, trypsin, urokinase, and kallikrein released during diseased conditions, are responsible for activating different subtypes of PARs. Each PAR receptor subtype is involved in kidney disease of distinct aetiology. PAR1 and PAR2 have shown differential therapeutic outcomes in rodent models of type-1 and type-2 diabetic kidney diseases due to the distinct etiological basis of each disease type, however such findings need to be confirmed in other diabetic renal injury models. PAR1 and PAR2 blockers have been observed to abolish drug-induced nephrotoxicity in rodents by suppressing tubular inflammation and fibrosis and preventing mitochondrial dysfunction. Notably, PAR2 inhibition improved autophagy and prevented fibrosis, inflammation, and remodeling in the urethral obstruction model. Only the PAR1/4 subtypes have emerged as a therapeutic target for treating experimentally induced nephrotic syndrome, where their respective antibodies attenuated the podocyte apoptosis induced upon thrombin activation. Strikingly PAR2 and PAR4 subtypes involvement has been tested in sepsis-induced acute kidney injury (AKI) and renal ischemia-reperfusion injury models. Thus, more studies are required to delineate the role of other subtypes in the sepsis-AKI model. Evidence suggests that PARs regulate oxidative, inflammatory stress, immune cell activation, fibrosis, autophagic flux, and apoptosis during kidney diseases.
Collapse
|
12
|
Staruschenko A, Ma R, Palygin O, Dryer SE. Ion channels and channelopathies in glomeruli. Physiol Rev 2023; 103:787-854. [PMID: 36007181 PMCID: PMC9662803 DOI: 10.1152/physrev.00013.2022] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 08/15/2022] [Accepted: 08/21/2022] [Indexed: 11/22/2022] Open
Abstract
An essential step in renal function entails the formation of an ultrafiltrate that is delivered to the renal tubules for subsequent processing. This process, known as glomerular filtration, is controlled by intrinsic regulatory systems and by paracrine, neuronal, and endocrine signals that converge onto glomerular cells. In addition, the characteristics of glomerular fluid flow, such as the glomerular filtration rate and the glomerular filtration fraction, play an important role in determining blood flow to the rest of the kidney. Consequently, disease processes that initially affect glomeruli are the most likely to lead to end-stage kidney failure. The cells that comprise the glomerular filter, especially podocytes and mesangial cells, express many different types of ion channels that regulate intrinsic aspects of cell function and cellular responses to the local environment, such as changes in glomerular capillary pressure. Dysregulation of glomerular ion channels, such as changes in TRPC6, can lead to devastating glomerular diseases, and a number of channels, including TRPC6, TRPC5, and various ionotropic receptors, are promising targets for drug development. This review discusses glomerular structure and glomerular disease processes. It also describes the types of plasma membrane ion channels that have been identified in glomerular cells, the physiological and pathophysiological contexts in which they operate, and the pathways by which they are regulated and dysregulated. The contributions of these channels to glomerular disease processes, such as focal segmental glomerulosclerosis (FSGS) and diabetic nephropathy, as well as the development of drugs that target these channels are also discussed.
Collapse
Affiliation(s)
- Alexander Staruschenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida
- Hypertension and Kidney Research Center, University of South Florida, Tampa, Florida
- James A. Haley Veterans Hospital, Tampa, Florida
| | - Rong Ma
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - Oleg Palygin
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Stuart E Dryer
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
- Department of Biomedical Sciences, Tilman J. Fertitta Family College of Medicine, University of Houston, Houston, Texas
| |
Collapse
|
13
|
Sivakumar S, Miellet S, Clarke C, Hartley PS. Insect nephrocyte function is regulated by a store operated calcium entry mechanism controlling endocytosis and Amnionless turnover. JOURNAL OF INSECT PHYSIOLOGY 2022; 143:104453. [PMID: 36341969 DOI: 10.1016/j.jinsphys.2022.104453] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/22/2022] [Accepted: 10/21/2022] [Indexed: 05/26/2023]
Abstract
Insect nephrocytes are ultrafiltration cells that remove circulating proteins and exogenous toxins from the haemolymph. Experimental disruption of nephrocyte development or function leads to systemic impairment of insect physiology as evidenced by cardiomyopathy, chronic activation of immune signalling and shortening of lifespan. The genetic and structural basis of the nephrocyte's ultrafiltration mechanism is conserved between arthropods and mammals, making them an attractive model for studying human renal function and systemic clearance mechanisms in general. Although dynamic changes to intracellular calcium are fundamental to the function of many cell types, there are currently no studies of intracellular calcium signalling in nephrocytes. In this work we aimed to characterise calcium signalling in the pericardial nephrocytes of Drosophila melanogaster. To achieve this, a genetically encoded calcium reporter (GCaMP6) was expressed in nephrocytes to monitor intracellular calcium both in vivo within larvae and in vitro within dissected adults. Larval nephrocytes exhibited stochastically timed calcium waves. A calcium signal could be initiated in preparations of adult nephrocytes and abolished by EGTA, or the store operated calcium entry (SOCE) blocker 2-APB, as well as RNAi mediated knockdown of the SOCE genes Stim and Orai. Neither the presence of calcium-free buffer nor EGTA affected the binding of the endocytic cargo albumin to nephrocytes but they did impair the subsequent accumulation of albumin within nephrocytes. Pre-treatment with EGTA, calcium-free buffer or 2-APB led to significantly reduced albumin binding. Knock-down of Stim and Orai was non-lethal, caused an increase to nephrocyte size and reduced albumin binding, reduced the abundance of the endocytic cargo receptor Amnionless and disrupted the localisation of Dumbfounded at the filtration slit diaphragm. These data indicate that pericardial nephrocytes exhibit stochastically timed calcium waves in vivo and that SOCE mediates the localisation of the endocytic co-receptor Amnionless. Identifying the signals both up and downstream of SOCE may highlight mechanisms relevant to the renal and excretory functions of a broad range of species, including humans.
Collapse
Affiliation(s)
- Shruthi Sivakumar
- Department of Life and Environmental Sciences, Faculty of Science and Technology, Bournemouth University, Dorset BH12 5BB, UK
| | - Sara Miellet
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia; Molecular Horizons, University of Wollongong, NSW, Australia
| | - Charlotte Clarke
- Department of Life and Environmental Sciences, Faculty of Science and Technology, Bournemouth University, Dorset BH12 5BB, UK
| | - Paul S Hartley
- Department of Life and Environmental Sciences, Faculty of Science and Technology, Bournemouth University, Dorset BH12 5BB, UK.
| |
Collapse
|
14
|
Arndt P, Sradnick J, Kroeger H, Holtzhausen S, Kessel F, Gerlach M, Todorov V, Hugo C. A quantitative 3D intravital look at the juxtaglomerular renin-cell-niche reveals an individual intra/extraglomerular feedback system. Front Physiol 2022; 13:980787. [PMID: 36237522 PMCID: PMC9550881 DOI: 10.3389/fphys.2022.980787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 09/01/2022] [Indexed: 11/13/2022] Open
Abstract
The juxtaglomerular niche occupied by renin cells (RCN) plays an important role in glomerular repair but the precise temporal and spatial interrelations remain unclear. This study proposes the hypothesis of a local intra-extraglomerular regenerative feedback system and establishes a new quantifiable system for RCN responses in individual glomeruli in vivo. A strictly intraglomerular two-photon laser-induced injury model was established. Labeled renin cells (RC) in transgenic renin reporter mice were fate-traced in healthy and injured glomeruli over several days by intravital microscopy and quantified via new three-dimensional image processing algorithms based on ray tracing. RC in healthy glomeruli demonstrated dynamic extraglomerular protrusions. Upon intraglomerular injury the corresponding RCN first increased in volume and then increased in area of dynamic migration up to threefold compared to their RCN. RC started migration reaching the site of injury within 3 hours and acquired a mesangial cell phenotype without losing physical RCN-contact. During intraglomerular repair only the corresponding RCN responded via stimulated neogenesis, a process of de novo differentiation of RC to replenish the RCN. Repeated continuous intravital microscopy provides a state-of-the-art tool to prove and further study the local intraglomerular RCN repair feedback system in individual glomeruli in vivo in a quantifiable manner.
Collapse
Affiliation(s)
- Patrick Arndt
- Experimental Nephrology, Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Jan Sradnick
- Experimental Nephrology, Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Hannah Kroeger
- Experimental Nephrology, Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Stefan Holtzhausen
- Institute of Machine Elements and Machine Design, Chair of Virtual Product Development, Dresden University of Technology, Dresden, Germany
| | - Friederike Kessel
- Experimental Nephrology, Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Michael Gerlach
- Core Facility Cellular Imaging, Experimental Center, Faculty of Medicine Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Vladimir Todorov
- Experimental Nephrology, Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Christian Hugo
- Experimental Nephrology, Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
- *Correspondence: Christian Hugo,
| |
Collapse
|
15
|
The Calcium-Sensing Receptor Stabilizes Podocyte Function in Proteinuric Humans and Mice. Kidney Int 2022; 101:1186-1199. [DOI: 10.1016/j.kint.2022.01.036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/15/2021] [Accepted: 01/21/2022] [Indexed: 12/30/2022]
|
16
|
GYARMATI GEORGINA, TOMA ILDIKÓ, IZUHARA AUDREY, BURFORD JAMESL, SHROFF URVINIKHIL, PAPADOURI STELLA, DEEPAK SACHIN, PETI-PETERDI JÁNOS. The role of TRPC6 calcium channels and P2 purinergic receptors in podocyte mechanical and metabolic sensing. Physiol Int 2021; 109:2021.00205. [PMID: 34978536 PMCID: PMC9200898 DOI: 10.1556/2060.2021.00205] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 11/22/2021] [Indexed: 12/17/2022]
Abstract
Podocyte calcium (Ca2+) signaling plays important roles in the (patho)physiology of the glomerular filtration barrier. Overactivation of podocyte transient receptor potential canonical (TRPC) channels including TRPC6 and purinergic signaling via P2 receptors that are known mechanosensors can increase podocyte intracellular Ca2+ levels ([Ca2+]i) and cause cell injury, proteinuria and glomerular disease including in diabetes. However, important mechanistic details of the trigger and activation of these pathways in vivo in the intact glomerular environment are lacking. Here we show direct visual evidence that podocytes can sense mechanical overload (increased glomerular capillary pressure) and metabolic alterations (increased plasma glucose) via TRPC6 and purinergic receptors including P2Y2. Multiphoton microscopy of podocyte [Ca2+]i was performed in vivo using wild-type and TRPC6 or P2Y2 knockout (KO) mice expressing the calcium reporter GCaMP3/5 only in podocytes and in vitro using freshly dissected microperfused glomeruli. Single-nephron intra-glomerular capillary pressure elevations induced by obstructing the efferent arteriole lumen with laser-induced microthrombus in vivo and by a micropipette in vitro triggered >2-fold increases in podocyte [Ca2+]i. These responses were blocked in TRPC6 and P2Y2 KO mice. Acute elevations of plasma glucose caused >4-fold increases in podocyte [Ca2+]i that were abolished by pharmacological inhibition of TRPC6 or P2 receptors using SAR7334 or suramin treatment, respectively. This study established the role of Ca2+ signaling via TRPC6 channels and P2 receptors in mechanical and metabolic sensing of podocytes in vivo, which are promising therapeutic targets in conditions with high intra-glomerular capillary pressure and plasma glucose, such as diabetic and hypertensive nephropathy.
Collapse
Affiliation(s)
- GEORGINA GYARMATI
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA 90033, USA
| | - ILDIKÓ TOMA
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA 90033, USA
| | - AUDREY IZUHARA
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA 90033, USA
| | - JAMES L. BURFORD
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA 90033, USA
| | - URVI NIKHIL SHROFF
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA 90033, USA
| | - STELLA PAPADOURI
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA 90033, USA
| | - SACHIN DEEPAK
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA 90033, USA
| | - JÁNOS PETI-PETERDI
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
17
|
Szrejder M, Rogacka D, Piwkowska A. Purinergic P2 receptors: Involvement and therapeutic implications in diabetes-related glomerular injury. Arch Biochem Biophys 2021; 714:109078. [PMID: 34742673 DOI: 10.1016/j.abb.2021.109078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/15/2021] [Accepted: 10/30/2021] [Indexed: 02/08/2023]
Abstract
The purinergic activation of P2 receptors initiates a powerful and rapid signaling cascade that contributes to the regulation of an array of physiological and pathophysiological processes in many organs, including the kidney. P2 receptors are broadly distributed in both epithelial and vascular renal cells. Disturbances of purinergic signaling can lead to impairments in renal function. A growing body of evidence indicates changes in P2 receptor expression and nucleotide metabolism in chronic renal injury and inflammatory diseases. Increasing attention has focused on purinergic P2X7 receptors, which are not normally expressed in healthy kidney tissue but are highly expressed at sites of tissue damage and inflammation. Under hyperglycemic conditions, several mechanisms that are linked to purinergic signaling and involve nucleotide release and degradation are disrupted, resulting in the accumulation of adenosine 5'-triphosphate in the bloodstream in diabetes. Dysfunction of the purinergic system might be associated with serious vascular complications in diabetes, including diabetic nephropathy. This review summarizes our current knowledge of the role of P2 receptors in diabetes-related glomerular injury and its implications for new therapeutics for diabetic nephropathy.
Collapse
Affiliation(s)
- Maria Szrejder
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Gdańsk, Poland.
| | - Dorota Rogacka
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Gdańsk, Poland; Department of Molecular Biotechnology, University of Gdańsk, Faculty of Chemistry, Gdańsk, Poland
| | - Agnieszka Piwkowska
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Gdańsk, Poland; Department of Molecular Biotechnology, University of Gdańsk, Faculty of Chemistry, Gdańsk, Poland
| |
Collapse
|
18
|
Gyarmati G, Jacob CO, Peti-Peterdi J. New Endothelial Mechanisms in Glomerular (Patho)biology and Proteinuria Development Captured by Intravital Multiphoton Imaging. Front Med (Lausanne) 2021; 8:765356. [PMID: 34722598 PMCID: PMC8548465 DOI: 10.3389/fmed.2021.765356] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 09/17/2021] [Indexed: 11/23/2022] Open
Abstract
In the past two decades, intravital imaging using multiphoton microscopy has provided numerous new visual and mechanistic insights into glomerular biology and disease processes including the function of glomerular endothelial cells (GEnC), podocytes, and the development of proteinuria. Although glomerular endothelial injury is known to precede podocyte damage in several renal diseases, the primary role of GEnCs in proteinuria development received much less attention compared to the vast field of podocyte pathobiology. Consequently, our knowledge of GEnC mechanisms in glomerular diseases is still emerging. This review highlights new visual clues on molecular and cellular mechanisms of GEnCs and their crosstalk with podocytes and immune cells that were acquired recently by the application of multiphoton imaging of the intact glomerular microenvironment in various proteinuric disease models. New mechanisms of glomerular tissue remodeling and regeneration are discussed based on results of tracking the fate and function of individual GEnCs using serial intravital multiphoton imaging over several days and weeks. The three main topics of this review include (i) the role of endothelial injury and microthrombi in podocyte detachment and albumin leakage via hemodynamic and mechanical forces, (ii) the alterations of the endothelial surface layer (glycocalyx) and its interactions with circulating immune cells in lupus nephritis, and (iii) the structural and functional remodeling and regeneration of GEnCs in hypertension, diabetes, and other experimental injury conditions. By the comprehensive visual portrayal of GEnCs and the many other contributing glomerular cell types, this review emphasizes the complexity of pathogenic mechanisms that result in proteinuria development.
Collapse
Affiliation(s)
- Georgina Gyarmati
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, United States
| | - Chaim O Jacob
- Division of Rheumatology and Immunology, Department of Medicine, University of Southern California, Los Angeles, CA, United States
| | - János Peti-Peterdi
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
19
|
Liu W, Shi L, Wan Q, Wu Y, Huang D, Ou J, Liu Q, Guan X, Yang Y, Zhang X, Gao J. Huangqi Guizhi Wuwu Decoction attenuates Podocyte cytoskeletal protein damage in IgA nephropathy rats by regulating AT1R/Nephrin/c-Abl pathway. Biomed Pharmacother 2021; 142:111907. [PMID: 34339916 DOI: 10.1016/j.biopha.2021.111907] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 07/02/2021] [Accepted: 07/06/2021] [Indexed: 01/17/2023] Open
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Huangqi Guizhi Wuwu Decoction(HQGZWWD) is a Traditional Chinese Medicine formula from Synopsis of Golden Chamber used to treat blood arthralgia. According to the principle that the same treatment can be used for different diseases, HQGZWWD has proven effective for IgA nephropathy (IgAN) associated with spleen and kidney yang deficiency. AIM OF THE STUDY In this study, we investigated the mechanism by which HQGZWWD alleviates proteinuria and protects renal function in rats with IgAN by regulating the AT1R/Nephrin/c-Abl pathway. METHODS Rats were randomly divided into six groups: control, IgAN model, IgAN model treated with low-dose HQGZWWD, IgAN model treated with medium-dose HQGZWWD, IgAN model treated with high-dose HQGZWWD, and IgAN model treated with valsartan. IgAN was induced using bovine γ-globulin. We evaluated the mediating effects of HQGZWWD on podocyte cytoskeletal proteins, the AT1R/Nephrin/c-Abl pathway, upstream tumor necrosis factor-α (TNF-α), and TNF-α receptor-1 (TNFR1). RESULTS The IgAN rats displayed proteinuria, IgA deposition in the mesangial region, and podocyte cytoskeletal protein damage. The expression of TNF-α, TNFR1, AT1R, and c-Abl was increased in the IgAN rat kidney, whereas the expression of nephrin, podocin, ACTN4, and phosphorylated nephrin (p-nephrin) was reduced. HQGZWWD treatment significantly alleviated podocyte cytoskeletal protein damage in the IgAN rats, upregulated the expression of nephrin, podocin, and ACTN4, and the colocalized expression of F-actin and nephrin. This study demonstrates that HQGZWWD attenuates podocyte cytoskeletal protein damage by regulating the AT1R-nephrin- c-Abl pathway, upregulating the expression of p-nephrin, and downregulating the expression of AT1R and c-Abl. CONCLUSIONS These results indicate that HQGZWWD attenuates podocyte cytoskeletal protein damage in IgAN rats by regulating the AT1R/Nephrin/c-Abl pathway, providing a potential therapeutic approach for IgAN.
Collapse
MESH Headings
- Actinin/genetics
- Actinin/metabolism
- Actins/metabolism
- Animals
- Cytoskeletal Proteins/metabolism
- Disease Models, Animal
- Down-Regulation/drug effects
- Drugs, Chinese Herbal/chemistry
- Drugs, Chinese Herbal/pharmacology
- Drugs, Chinese Herbal/therapeutic use
- Glomerulonephritis, IGA/drug therapy
- Glomerulonephritis, IGA/metabolism
- Glomerulonephritis, IGA/pathology
- Glomerulonephritis, IGA/physiopathology
- Immunoglobulin A/metabolism
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Male
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Podocytes/drug effects
- Protective Agents/chemistry
- Protective Agents/pharmacology
- Protective Agents/therapeutic use
- Proteinuria/metabolism
- Proto-Oncogene Proteins c-abl/metabolism
- Rats, Sprague-Dawley
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/metabolism
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Signal Transduction/drug effects
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/metabolism
- Rats
Collapse
Affiliation(s)
- Weiwei Liu
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine; TCM Institute of Kidney Disease of Shanghai University of Traditional Chinese Medicine; Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education; Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, No. 528 Zhangheng Road, Shanghai 201203, China
| | - Liqiang Shi
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine; TCM Institute of Kidney Disease of Shanghai University of Traditional Chinese Medicine; Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education; Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, No. 528 Zhangheng Road, Shanghai 201203, China
| | - Qiang Wan
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine; TCM Institute of Kidney Disease of Shanghai University of Traditional Chinese Medicine; Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education; Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, No. 528 Zhangheng Road, Shanghai 201203, China
| | - Yansheng Wu
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine; TCM Institute of Kidney Disease of Shanghai University of Traditional Chinese Medicine; Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education; Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, No. 528 Zhangheng Road, Shanghai 201203, China
| | - Di Huang
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine; TCM Institute of Kidney Disease of Shanghai University of Traditional Chinese Medicine; Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education; Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, No. 528 Zhangheng Road, Shanghai 201203, China
| | - Jiaoying Ou
- Department of Internal Medicine, Shanghai TCM-Integrated Hospital, Affiliated to Shanghai University of Traditional Chinese Medicine, No. 184 Road Baoding, Shanghai 200082, China
| | - Qiuyu Liu
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine; TCM Institute of Kidney Disease of Shanghai University of Traditional Chinese Medicine; Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education; Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, No. 528 Zhangheng Road, Shanghai 201203, China
| | - Xin Guan
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine; TCM Institute of Kidney Disease of Shanghai University of Traditional Chinese Medicine; Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education; Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, No. 528 Zhangheng Road, Shanghai 201203, China
| | - Yuzhu Yang
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine; TCM Institute of Kidney Disease of Shanghai University of Traditional Chinese Medicine; Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education; Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, No. 528 Zhangheng Road, Shanghai 201203, China
| | - Xiaotian Zhang
- Preventive treatment of disease center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No. 528 Zhangheng Road, Shanghai 201203, China
| | - Jiandong Gao
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine; TCM Institute of Kidney Disease of Shanghai University of Traditional Chinese Medicine; Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education; Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, No. 528 Zhangheng Road, Shanghai 201203, China.
| |
Collapse
|
20
|
Djenoune L, Tomar R, Dorison A, Ghobrial I, Schenk H, Hegermann J, Beverly-Staggs L, Hidalgo-Gonzalez A, Little MH, Drummond IA. Autonomous Calcium Signaling in Human and Zebrafish Podocytes Controls Kidney Filtration Barrier Morphogenesis. J Am Soc Nephrol 2021; 32:1697-1712. [PMID: 33911000 PMCID: PMC8425667 DOI: 10.1681/asn.2020101525] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 02/12/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Podocytes are critical to maintaining the glomerular filtration barrier, and mutations in nephrotic syndrome genes are known to affect podocyte calcium signaling. However, the role of calcium signaling during podocyte development remains unknown. METHODS We undertook live imaging of calcium signaling in developing podocytes, using zebrafish larvae and human kidney organoids. To evaluate calcium signaling during development and in response to channel blockers and genetic defects, the calcium biosensor GCaMP6s was expressed in zebrafish podocytes. We used electron microscopy to evaluate filtration barrier formation in zebrafish, and Fluo-4 to detect calcium signals in differentiating podocytes in human kidney organoids. RESULTS Immature zebrafish podocytes (2.5 days postfertilization) generated calcium transients that correlated with interactions with forming glomerular capillaries. Calcium transients persisted until 4 days postfertilization, and were absent after glomerular barrier formation was complete. We detected similar calcium transients in maturing human organoid glomeruli, suggesting a conserved mechanism. In both models, inhibitors of SERCA or IP3 receptor calcium-release channels blocked calcium transients in podocytes, whereas lanthanum was ineffective, indicating the calcium source is from intracellular podocyte endoplasmic-reticulum stores. Calcium transients were not affected by blocking heartbeat or by blocking development of endothelium or endoderm, and they persisted in isolated glomeruli, suggesting podocyte-autonomous calcium release. Inhibition of expression of phospholipase C-γ1, but not nephrin or phospholipase C-ε1, led to significantly decreased calcium activity. Finally, blocking calcium release affected glomerular shape and podocyte foot process formation, supporting the critical role of calcium signaling in glomerular morphogenesis. CONCLUSIONS These findings establish podocyte cell-autonomous calcium signaling as a prominent and evolutionarily conserved feature of podocyte differentiation and demonstrate its requirement for podocyte foot process formation.
Collapse
Affiliation(s)
- Lydia Djenoune
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Charlestown, Massachusetts
| | - Ritu Tomar
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Charlestown, Massachusetts
| | - Aude Dorison
- Murdoch Children’s Research Institute, The Royal Children’s Hospital, Parkville, Victoria, Australia
| | - Irene Ghobrial
- Murdoch Children’s Research Institute, The Royal Children’s Hospital, Parkville, Victoria, Australia
| | - Heiko Schenk
- Department of Medicine/Nephrology, Hannover Medical School, Hannover, Germany,Research Core Unit Electron Microscopy, Hannover Medical School, Hannover, Germany
| | - Jan Hegermann
- Research Core Unit Electron Microscopy, Hannover Medical School, Hannover, Germany
| | - Lynne Beverly-Staggs
- Davis Center for Regenerative Biology and Aging, Mount Desert Island Biological Laboratory, Bar Harbor, Maine
| | | | - Melissa H. Little
- Murdoch Children’s Research Institute, The Royal Children’s Hospital, Parkville, Victoria, Australia,Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia,Department of Anatomy and Neuroscience, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Victoria, Australia
| | - Iain A. Drummond
- Davis Center for Regenerative Biology and Aging, Mount Desert Island Biological Laboratory, Bar Harbor, Maine
| |
Collapse
|
21
|
Palygin O, Klemens CA, Isaeva E, Levchenko V, Spires DR, Dissanayake LV, Nikolaienko O, Ilatovskaya DV, Staruschenko A. Characterization of purinergic receptor 2 signaling in podocytes from diabetic kidneys. iScience 2021; 24:102528. [PMID: 34142040 PMCID: PMC8188476 DOI: 10.1016/j.isci.2021.102528] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 04/12/2021] [Accepted: 05/08/2021] [Indexed: 02/08/2023] Open
Abstract
Growing evidence suggests that renal purinergic signaling undergoes significant remodeling during pathophysiological conditions such as diabetes. This study examined the renal P2 receptor profile and ATP-mediated calcium response from podocytes in glomeruli from kidneys with type 1 or type 2 diabetic kidney disease (DKD), using type 2 diabetic nephropathy (T2DN) rats and streptozotocin-injected Dahl salt-sensitive (type 1 diabetes) rats. A dramatic increase in the ATP-mediated intracellular calcium flux in podocytes was observed in both models. Pharmacological inhibition established that P2X4 and P2X7 are the major receptors contributing to the augmented ATP-mediated intracellular calcium signaling in diabetic podocytes. The transition in purinergic receptor composition from metabotropic to ionotropic may disrupt intracellular calcium homeostasis in podocytes resulting in their dysfunction and potentially further aggravating DKD progression. Diabetic podocytes have sustained intracellular Ca2+ signaling in response to ATP Podocyte purinergic receptor signaling is predominantly ionotropic in diabetes Both type 1 and 2 diabetic podocytes have similar purinergic receptor remodeling
Collapse
Affiliation(s)
- Oleg Palygin
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Christine A Klemens
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Elena Isaeva
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Vladislav Levchenko
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Denisha R Spires
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Lashodya V Dissanayake
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Oksana Nikolaienko
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Daria V Ilatovskaya
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA.,Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Alexander Staruschenko
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA.,Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, WI, USA
| |
Collapse
|
22
|
Sachs M, Wetzel S, Reichelt J, Sachs W, Schebsdat L, Zielinski S, Seipold L, Heintz L, Müller SA, Kretz O, Lindenmeyer M, Wiech T, Huber TB, Lüllmann-Rauch R, Lichtenthaler SF, Saftig P, Meyer-Schwesinger C. ADAM10-Mediated Ectodomain Shedding Is an Essential Driver of Podocyte Damage. J Am Soc Nephrol 2021; 32:1389-1408. [PMID: 33785583 PMCID: PMC8259650 DOI: 10.1681/asn.2020081213] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 02/08/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Podocytes embrace the glomerular capillaries with foot processes, which are interconnected by a specialized adherens junction to ultimately form the filtration barrier. Altered adhesion and loss are common features of podocyte injury, which could be mediated by shedding of cell-adhesion molecules through the regulated activity of cell surface-expressed proteases. A Disintegrin and Metalloproteinase 10 (ADAM10) is such a protease known to mediate ectodomain shedding of adhesion molecules, among others. Here we evaluate the involvement of ADAM10 in the process of antibody-induced podocyte injury. METHODS Membrane proteomics, immunoblotting, high-resolution microscopy, and immunogold electron microscopy were used to analyze human and murine podocyte ADAM10 expression in health and kidney injury. The functionality of ADAM10 ectodomain shedding for podocyte development and injury was analyzed, in vitro and in vivo, in the anti-podocyte nephritis (APN) model in podocyte-specific, ADAM10-deficient mice. RESULTS ADAM10 is selectively localized at foot processes of murine podocytes and its expression is dispensable for podocyte development. Podocyte ADAM10 expression is induced in the setting of antibody-mediated injury in humans and mice. Podocyte ADAM10 deficiency attenuates the clinical course of APN and preserves the morphologic integrity of podocytes, despite subepithelial immune-deposit formation. Functionally, ADAM10-related ectodomain shedding results in cleavage of the cell-adhesion proteins N- and P-cadherin, thus decreasing their injury-related surface levels. This favors podocyte loss and the activation of downstream signaling events through the Wnt signaling pathway in an ADAM10-dependent manner. CONCLUSIONS ADAM10-mediated ectodomain shedding of injury-related cadherins drives podocyte injury.
Collapse
Affiliation(s)
- Marlies Sachs
- Institute of Cellular and Integrative Physiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sebastian Wetzel
- Institute of Biochemistry, Christian-Albrechts University Kiel, Kiel, Germany
| | - Julia Reichelt
- Institute of Cellular and Integrative Physiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Wiebke Sachs
- Institute of Cellular and Integrative Physiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lisa Schebsdat
- Institute of Cellular and Integrative Physiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stephanie Zielinski
- Institute of Cellular and Integrative Physiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lisa Seipold
- Institute of Biochemistry, Christian-Albrechts University Kiel, Kiel, Germany
| | - Lukas Heintz
- Institute of Cellular and Integrative Physiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stephan A. Müller
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Neuroproteomics, School of Medicine, University Hospital rechts der Isar, Technical University of Munich, Munich, Germany
| | - Oliver Kretz
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maja Lindenmeyer
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thorsten Wiech
- Nephropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias B. Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Stefan F. Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Neuroproteomics, School of Medicine, University Hospital rechts der Isar, Technical University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Paul Saftig
- Institute of Biochemistry, Christian-Albrechts University Kiel, Kiel, Germany
| | - Catherine Meyer-Schwesinger
- Institute of Cellular and Integrative Physiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
23
|
Desposito D, Schiessl IM, Gyarmati G, Riquier-Brison A, Izuhara AK, Kadoya H, Der B, Shroff UN, Hong YK, Peti-Peterdi J. Serial intravital imaging captures dynamic and functional endothelial remodeling with single-cell resolution. JCI Insight 2021; 6:123392. [PMID: 33848265 PMCID: PMC8262275 DOI: 10.1172/jci.insight.123392] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 04/12/2021] [Indexed: 01/01/2023] Open
Abstract
Endothelial cells are important in the maintenance of healthy blood vessels and in the development of vascular diseases. However, the origin and dynamics of endothelial precursors and remodeling at the single-cell level have been difficult to study in vivo owing to technical limitations. Therefore, we aimed to develop a direct visual approach to track the fate and function of single endothelial cells over several days and weeks in the same vascular bed in vivo using multiphoton microscopy (MPM) of transgenic Cdh5-Confetti mice and the kidney glomerulus as a model. Individual cells of the vascular endothelial lineage were identified and tracked owing to their unique color combination, based on the random expression of cyan/green/yellow/red fluorescent proteins. Experimental hypertension, hyperglycemia, and laser-induced endothelial cell ablation rapidly increased the number of new glomerular endothelial cells that appeared in clusters of the same color, suggesting clonal cell remodeling by local precursors at the vascular pole. Furthermore, intravital MPM allowed the detection of distinct structural and functional alterations of proliferating endothelial cells. No circulating Cdh5-Confetti+ cells were found in the renal cortex. Moreover, the heart, lung, and kidneys showed more significant clonal endothelial cell expansion compared with the brain, pancreas, liver, and spleen. In summary, we have demonstrated that serial MPM of Cdh5-Confetti mice in vivo is a powerful technical advance to study endothelial remodeling and repair in the kidney and other organs under physiological and disease conditions.
Collapse
Affiliation(s)
- Dorinne Desposito
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, and
| | - Ina Maria Schiessl
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, and
| | - Georgina Gyarmati
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, and
| | - Anne Riquier-Brison
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, and
| | - Audrey K Izuhara
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, and
| | - Hiroyuki Kadoya
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, and
| | - Balint Der
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, and
| | - Urvi Nikhil Shroff
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, and
| | - Young-Kwon Hong
- Department of Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Janos Peti-Peterdi
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, and
| |
Collapse
|
24
|
Martins JR, Haenni D, Bugarski M, Polesel M, Schuh C, Hall AM. Intravital kidney microscopy: entering a new era. Kidney Int 2021; 100:527-535. [PMID: 34015315 DOI: 10.1016/j.kint.2021.02.042] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/01/2021] [Accepted: 02/16/2021] [Indexed: 02/07/2023]
Abstract
The development of intravital imaging with multiphoton microscopy has had a major impact on kidney research. It provides the unique opportunity to visualize dynamic behavior of cells and organelles in their native environment and to relate this to the complex 3-dimensional structure of the organ. Moreover, changes in cell/organelle function can be followed in real time in response to physiological interventions or disease-causing insults. However, realizing the enormous potential of this exciting approach has necessitated overcoming several substantial practical hurdles. In this article, we outline the nature of these challenges and how a variety of technical advances have provided effective solutions. In particular, improvements in laser/microscope technology, fluorescent probes, transgenic animals, and abdominal windows are collectively making previously opaque processes visible. Meanwhile, the rise of machine learning-based image analysis is facilitating the rapid generation of large amounts of quantitative data, amenable to deeper statistical interrogation. Taken together, the increased capabilities of multiphoton imaging are opening up huge new possibilities to study structure-function relationships in the kidney in unprecedented detail. In addition, they are yielding important new insights into cellular mechanisms of tissue damage, repair, and adaptive remodeling during disease states. Thus, intravital microscopy is truly entering an exciting new era in translational kidney research.
Collapse
Affiliation(s)
- Joana R Martins
- Center for Microscopy and Image Analysis, University of Zurich, Zurich, Switzerland
| | - Dominik Haenni
- Center for Microscopy and Image Analysis, University of Zurich, Zurich, Switzerland; Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Milica Bugarski
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | | | - Claus Schuh
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Andrew M Hall
- Institute of Anatomy, University of Zurich, Zurich, Switzerland; Department of Nephrology, University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
25
|
Abstract
AbstractAcute kidney injury (AKI) is a common clinical symptom, which is mainly manifested by elevated serum creatinine and blood urea nitrogen levels. When AKI is not repaired in time, the patient is prone to develop chronic kidney disease (CKD). The kidney is composed of more than 30 different cells, and its structure is complex. It is extremely challenging to understand the lineage relationships and cell fate of these cells in the process of kidney injury and regeneration. Since the 20th century, lineage tracing technology has provided an important mean for studying organ development, tissue damage repair, and the differentiation and fate of single cells. However, traditional lineage tracing methods rely on sacrificing animals to make tissue slices and then take snapshots with conventional imaging tools to obtain interesting information. This method cannot achieve dynamic and continuous monitoring of cell actions on living animals. As a kind of intravital microscopy (IVM), two-photon microscopy (TPM) has successfully solved the above problems. Because TPM has the ability to penetrate deep tissues and can achieve imaging at the single cell level, lineage tracing technology with TPM is gradually becoming popular. In this review, we provided the key technical elements of lineage tracing, and how to use intravital imaging technology to visualize and quantify the fate of renal cells.
Collapse
|
26
|
Astragaloside IV inhibits palmitic acid-induced apoptosis through regulation of calcium homeostasis in mice podocytes. Mol Biol Rep 2021; 48:1453-1464. [PMID: 33606151 PMCID: PMC7925475 DOI: 10.1007/s11033-021-06204-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 01/28/2021] [Indexed: 11/04/2022]
Abstract
Loss of podocytes is a hallmark of diabetic nephropathy, and a growing body of evidence indicates that podocytes are susceptible to palmitic acid (PA). We have previously shown that AS-IV inhibited PA-induced podocyte apoptosis by activating sarcoendoplasmic reticulum Ca2+ ATPase (SERCA), which indicate calcium regulation may involve in the process. Immunofluorescence staining, Western blot and flow cytometry were used to measure the protective efficacy of AS-IV to ameliorate PA-induced ER stress and podocyte apoptosis. Meanwhile, AS-IV inhibited cytochrome c release, decreased mitochondrial membrane potential, accompany with the depletion of endoplasmic reticulum Ca2+ and elevation of cytosolic and mitochondrial Ca2+. Sequestration of cytosolic calcium with BAPTA-AM limited the response of podocyte apoptosis, while during the process the effect of AS-IV was also restrained. In contrast, elevation of cytosolic calcium with calcium ionophore ionomycin was depressed by AS-IV addition. Furthermore, inhibiting TRPC6 expression with SKF96365 or TRPC6 siRNA counteracted the beneficial effect of AS-IV. Our study provides further evidence to conclude the inhibitory effect of AS-IV to podocyte apoptosis is Ca2+-dependent. And the efficacy correlates with inhibiting TRPC6-mediated Ca2+ influx, and then cellular Ca2+ disturbance was coordinated.
Collapse
|
27
|
Tian X, Inoue K, Zhang Y, Wang Y, Sperati CJ, Pedigo CE, Zhao T, Yan M, Groener M, Moledina DG, Ebenezer K, Li W, Zhang Z, Liebermann DA, Greene L, Greer P, Parikh CR, Ishibe S. Inhibiting calpain 1 and 2 in cyclin G associated kinase-knockout mice mitigates podocyte injury. JCI Insight 2020; 5:142740. [PMID: 33208557 PMCID: PMC7710277 DOI: 10.1172/jci.insight.142740] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/08/2020] [Indexed: 12/27/2022] Open
Abstract
Evidence for reduced expression of cyclin G associated kinase (GAK) in glomeruli of patients with chronic kidney disease was observed in the Nephroseq human database, and GAK was found to be associated with the decline in kidney function. To examine the role of GAK, a protein that functions to uncoat clathrin during endocytosis, we generated podocyte-specific Gak-knockout mice (Gak-KO), which developed progressive proteinuria and kidney failure with global glomerulosclerosis. We isolated glomeruli from the mice carrying the mutation to perform messenger RNA profiling and unearthed evidence for dysregulated podocyte calpain protease activity as an important contributor to progressive podocyte damage. Treatment with calpain inhibitor III specifically inhibited calpain-1/-2 activities, mitigated the degree of proteinuria and glomerulosclerosis, and led to a striking increase in survival in the Gak-KO mice. Podocyte-specific deletion of Capns1, essential for calpain-1 and calpain-2 activities, also improved proteinuria and glomerulosclerosis in Gak-KO mice. Increased podocyte calpain activity-mediated proteolysis of IκBα resulted in increased NF-κB p65-induced expression of growth arrest and DNA-damage-inducible 45 beta in the Gak-KO mice. Our results suggest that loss of podocyte-associated Gak induces glomerular injury secondary to calcium dysregulation and aberrant calpain activation, which when inhibited, can provide a protective role.
Collapse
MESH Headings
- Animals
- Calpain/antagonists & inhibitors
- Diabetic Nephropathies/etiology
- Diabetic Nephropathies/metabolism
- Diabetic Nephropathies/pathology
- Diabetic Nephropathies/therapy
- Female
- Glomerulosclerosis, Focal Segmental/etiology
- Glomerulosclerosis, Focal Segmental/metabolism
- Glomerulosclerosis, Focal Segmental/pathology
- Glomerulosclerosis, Focal Segmental/therapy
- Humans
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Podocytes/metabolism
- Podocytes/pathology
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Protein Serine-Threonine Kinases/physiology
- Renal Insufficiency, Chronic/etiology
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Renal Insufficiency, Chronic/therapy
Collapse
Affiliation(s)
- 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
| | - Yan Zhang
- State Key Laboratory of Organ Failure Research, Southern Medical University, Nanfang Hospital, Guangzhou, China
- Center for Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Ying Wang
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - C. John Sperati
- Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Christopher E. Pedigo
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Tingting Zhao
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Meihua Yan
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Marwin Groener
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Dennis G. Moledina
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Karen Ebenezer
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Wei Li
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Zhenhai Zhang
- State Key Laboratory of Organ Failure Research, Southern Medical University, Nanfang Hospital, Guangzhou, China
- Center for Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Dan A. Liebermann
- Fels Institute of Cancer Research and Molecular Biology and Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania USA
| | - Lois Greene
- Laboratory of Cell Biology, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Peter Greer
- Queen’s Cancer Research Institute, Kingston, Ontario, Canada
| | - Chirag R. Parikh
- Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Shuta Ishibe
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
28
|
Extracellular ATP modulates podocyte function through P2Y purinergic receptors and pleiotropic effects on AMPK and cAMP/PKA signaling pathways. Arch Biochem Biophys 2020; 695:108649. [PMID: 33122160 DOI: 10.1016/j.abb.2020.108649] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 09/25/2020] [Accepted: 10/20/2020] [Indexed: 02/06/2023]
Abstract
Podocytes and their foot processes interlinked by slit diaphragms, constitute a continuous outermost layer of the glomerular capillary and seem to be crucial for maintaining the integrity of the glomerular filtration barrier. Purinergic signaling is involved in a wide range of physiological processes in the renal system, including regulating glomerular filtration. We evaluated the role of nucleotide receptors in cultured rat podocytes using non-selective P2 receptor agonists and agonists specific for the P2Y1, P2Y2, and P2Y4 receptors. The results showed that extracellular ATP evokes cAMP-dependent pathways through P2 receptors and influences remodeling of the podocyte cytoskeleton and podocyte permeability to albumin via coupling with RhoA signaling. Our findings highlight the relevance of the P2Y4 receptor in protein kinase A-mediated signal transduction to the actin cytoskeleton. We observed increased cAMP concentration and decreased RhoA activity after treatment with a P2Y4 agonist. Moreover, protein kinase A inhibitors reversed P2Y4-induced changes in RhoA activity and intracellular F-actin staining. P2Y4 stimulation resulted in enhanced AMPK phosphorylation and reduced reactive oxygen species generation. Our findings identify P2Y-PKA-RhoA signaling as the regulatory mechanism of the podocyte contractile apparatus and glomerular filtration. We describe a protection mechanism for the glomerular barrier linked to reduced oxidative stress and reestablished energy balance.
Collapse
|
29
|
Ranjit S, Lanzanò L, Libby AE, Gratton E, Levi M. Advances in fluorescence microscopy techniques to study kidney function. Nat Rev Nephrol 2020; 17:128-144. [PMID: 32948857 DOI: 10.1038/s41581-020-00337-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/30/2020] [Indexed: 02/07/2023]
Abstract
Fluorescence microscopy, in particular immunofluorescence microscopy, has been used extensively for the assessment of kidney function and pathology for both research and diagnostic purposes. The development of confocal microscopy in the 1950s enabled imaging of live cells and intravital imaging of the kidney; however, confocal microscopy is limited by its maximal spatial resolution and depth. More recent advances in fluorescence microscopy techniques have enabled increasingly detailed assessment of kidney structure and provided extraordinary insights into kidney function. For example, nanoscale precise imaging by rapid beam oscillation (nSPIRO) is a super-resolution microscopy technique that was originally developed for functional imaging of kidney microvilli and enables detection of dynamic physiological events in the kidney. A variety of techniques such as fluorescence recovery after photobleaching (FRAP), fluorescence correlation spectroscopy (FCS) and Förster resonance energy transfer (FRET) enable assessment of interaction between proteins. The emergence of other super-resolution techniques, including super-resolution stimulated emission depletion (STED), photoactivated localization microscopy (PALM), stochastic optical reconstruction microscopy (STORM) and structured illumination microscopy (SIM), has enabled functional imaging of cellular and subcellular organelles at ≤50 nm resolution. The deep imaging via emission recovery (DIVER) detector allows deep, label-free and high-sensitivity imaging of second harmonics, enabling assessment of processes such as fibrosis, whereas fluorescence lifetime imaging microscopy (FLIM) enables assessment of metabolic processes.
Collapse
Affiliation(s)
- Suman Ranjit
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, USA. .,Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, CA, USA.
| | - Luca Lanzanò
- Nanoscopy and NIC@IIT, Istituto Italiano di Tecnologia, Genoa, Italy.,Department of Physics and Astronomy "Ettore Majorana", University of Catania, Catania, Italy
| | - Andrew E Libby
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, USA
| | - Enrico Gratton
- Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, CA, USA.
| | - Moshe Levi
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, USA.
| |
Collapse
|
30
|
Struk T, Nair V, Eichinger F, Kretzler M, Wedlich-Söldner R, Bayraktar S, Pavenstädt H. Transcriptome analysis of primary podocytes reveals novel calcium regulated regulatory networks. FASEB J 2020; 34:14490-14506. [PMID: 32931033 DOI: 10.1096/fj.201902493rr] [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: 10/05/2019] [Revised: 08/11/2020] [Accepted: 08/14/2020] [Indexed: 11/11/2022]
Abstract
Podocytes are pivotal in establishing the selective permeability of the glomerular filtration barrier. Recently, we showed that an increase of the intracellular calcium ion concentration [Ca2+ ] causes a rapid and transient actin reset (CaAR) measurable through live imaging microscopy using lifeact-mCherry as an actin dye in different cell types including the podocyte. This and other studies show the critical role [Ca2+ ] and the actin cytoskeleton play in podocyte homeostasis. To further investigate the role of [Ca2+ ] and the actin cytoskeleton in podocytes, we used a double fluorescent reporter mouse model to establish a primary podocyte culture system. We treated these podocytes temporarily with a Calcium Ionophore and facultatively with Latrunculin A, an inhibitor of actin polymerization. Unbiased genome wide transcriptional analysis identified a transcriptional response in podocytes to elevated [Ca2+ ] levels, affecting mRNA levels of PDGF-BB, RICTOR, and MIR17HG as mediators of Ca2+ -signaling. Comparison of the ex vivo transcriptional response from the primary podocyte culture with glomerular transcripts across a wide spectrum of CKD disease confirmed co-regulation of transcript sets, establishing the disease relevance of the model system. Our findings demonstrate novel [Ca2+ ] regulated gene networks in podocytes deepening our understanding of podocyte biology and disease.
Collapse
Affiliation(s)
- Thaddäus Struk
- Department of Medicine, University of Münster, Münster, Germany
| | - Viji Nair
- Michigan Kidney Translational Medical Core, University of Michigan, Ann Arbor, MI, USA
| | - Felix Eichinger
- Michigan Kidney Translational Medical Core, University of Michigan, Ann Arbor, MI, USA
| | - Matthias Kretzler
- Michigan Kidney Translational Medical Core, University of Michigan, Ann Arbor, MI, USA.,Internal Medicine, Department of Nephrology, University of Michigan, Ann Arbor, MI, USA
| | | | - Samet Bayraktar
- Department of Medicine, University of Münster, Münster, Germany
| | | |
Collapse
|
31
|
Martins JR, Haenni D, Bugarski M, Figurek A, Hall AM. Quantitative intravital Ca2+ imaging maps single cell behavior to kidney tubular structure. Am J Physiol Renal Physiol 2020; 319:F245-F255. [DOI: 10.1152/ajprenal.00052.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Ca2+ is an important second messenger that translates extracellular stimuli into intracellular responses. Although there has been significant progress in understanding Ca2+ dynamics in organs such as the brain, the nature of Ca2+ signals in the kidney is still poorly understood. Here, we show that by using a genetically expressed highly sensitive reporter (GCaMP6s), it is possible to perform imaging of Ca2+ signals at high resolution in the mouse kidney in vivo. Moreover, by applying machine learning-based automated analysis using a Ca2+-independent signal, quantitative data can be extracted in an unbiased manner. By projecting the resulting data onto the structure of the kidney, we show that different tubular segments display highly distinct spatiotemporal patterns of Ca2+ signals. Furthermore, we provide evidence that Ca2+ activity in the proximal tubule decreases with increasing distance from the glomerulus. Finally, we demonstrate that substantial changes in intracellular Ca2+ can be detected in proximal tubules in a cisplatin model of acute kidney injury, which can be linked to alterations in cell structure and transport function. In summary, we describe a powerful new tool to investigate how single cell behavior is integrated with whole organ structure and function and how it is altered in disease states relevant to humans.
Collapse
Affiliation(s)
| | - Dominik Haenni
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
- Center for Microscopy and Image Analysis, University of Zurich, Zurich, Switzerland
| | - Milica Bugarski
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Andreja Figurek
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Andrew M. Hall
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
- Department of Nephrology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
32
|
Hu C, Lakshmipathi J, Stuart D, Peti-Peterdi J, Gyarmati G, Hao CM, Hansell P, Kohan DE. Renomedullary Interstitial Cell Endothelin A Receptors Regulate BP and Renal Function. J Am Soc Nephrol 2020; 31:1555-1568. [PMID: 32487560 DOI: 10.1681/asn.2020020232] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 04/06/2020] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND The physiologic role of renomedullary interstitial cells, which are uniquely and abundantly found in the renal inner medulla, is largely unknown. Endothelin A receptors regulate multiple aspects of renomedullary interstitial cell function in vitro. METHODS To assess the effect of targeting renomedullary interstitial cell endothelin A receptors in vivo, we generated a mouse knockout model with inducible disruption of renomedullary interstitial cell endothelin A receptors at 3 months of age. RESULTS BP and renal function were similar between endothelin A receptor knockout and control mice during normal and reduced sodium or water intake. In contrast, on a high-salt diet, compared with control mice, the knockout mice had reduced BP; increased urinary sodium, potassium, water, and endothelin-1 excretion; increased urinary nitrite/nitrate excretion associated with increased noncollecting duct nitric oxide synthase-1 expression; increased PGE2 excretion associated with increased collecting duct cyclooxygenase-1 expression; and reduced inner medullary epithelial sodium channel expression. Water-loaded endothelin A receptor knockout mice, compared with control mice, had markedly enhanced urine volume and reduced urine osmolality associated with increased urinary endothelin-1 and PGE2 excretion, increased cyclooxygenase-2 protein expression, and decreased inner medullary aquaporin-2 protein content. No evidence of endothelin-1-induced renomedullary interstitial cell contraction was observed. CONCLUSIONS Disruption of renomedullary interstitial cell endothelin A receptors reduces BP and increases salt and water excretion associated with enhanced production of intrinsic renal natriuretic and diuretic factors. These studies indicate that renomedullary interstitial cells can modulate BP and renal function under physiologic conditions.
Collapse
Affiliation(s)
- Chunyan Hu
- Division of Nephrology, University of Utah Health Center, Salt Lake City, Utah
| | | | - Deborah Stuart
- Division of Nephrology, University of Utah Health Center, Salt Lake City, Utah
| | - Janos Peti-Peterdi
- Departments of Physiology and Neuroscience and Medicine, University of Southern California, Los Angeles, California
| | - Georgina Gyarmati
- Departments of Physiology and Neuroscience and Medicine, University of Southern California, Los Angeles, California
| | - Chuan-Ming Hao
- Division of Nephrology, Huashan Hospital, Fudan University, Shanghai, China
| | - Peter Hansell
- Department of Medical Cell Biology, Section of Integrative Physiology, Uppsala University Biomedical Center, Uppsala, Sweden
| | - Donald E Kohan
- Division of Nephrology, University of Utah Health Center, Salt Lake City, Utah
| |
Collapse
|
33
|
Binz-Lotter J, Jüngst C, Rinschen MM, Koehler S, Zentis P, Schauss A, Schermer B, Benzing T, Hackl MJ. Injured Podocytes Are Sensitized to Angiotensin II-Induced Calcium Signaling. J Am Soc Nephrol 2020; 31:532-542. [PMID: 31924670 DOI: 10.1681/asn.2019020109] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 12/01/2019] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Inhibition of angiotensin II (AngII) signaling, a therapeutic mainstay of glomerular kidney diseases, is thought to act primarily through regulating glomerular blood flow and reducing filtration pressure. Although extravascular actions of AngII have been suggested, a direct effect of AngII on podocytes has not been demonstrated in vivo. METHODS To study the effects of AngII on podocyte calcium levels in vivo, we used intravital microscopy of the kidney in mice expressing the calcium indicator protein GCaMP3. RESULTS In healthy animals, podocytes displayed limited responsiveness to AngII stimulation. In contrast, in animals subjected to either adriamycin-induced acute chemical injury or genetic deletion of the podocin-encoding gene Nphs2, the consequent podocyte damage and proteinuria rendered the cells responsive to AngII and resulted in AngII-induced calcium transients in significantly more podocytes. The angiotensin type 1 receptor blocker losartan could fully inhibit this response. Also, responsiveness to AngII was at least partly mediated through the transient receptor potential channel 6, which has been implicated in podocyte calcium handling. Interestingly, loss of a single Nphs2 allele also increased podocytes' responsiveness to AngII signaling. This direct effect of AngII on injured podocytes results in increased calcium transients, which can further aggravate the underlying kidney disease. CONCLUSIONS Our discovery that podocytes become sensitized to AngII-induced calcium signaling upon injury might explain results from large, randomized, controlled trials in which improved renal outcomes occur only in the subgroup of patients with proteinuria, indicating podocyte damage. Our findings also emphasize the need to treat every patient with a glomerular disease with either an angiotensin-converting enzyme inhibitor or an angiotensin type 1 receptor blocker.
Collapse
Affiliation(s)
- Julia Binz-Lotter
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; and
| | - Christian Jüngst
- Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Markus M Rinschen
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; and
| | - Sybille Koehler
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; and
| | - Peter Zentis
- Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Astrid Schauss
- Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Bernhard Schermer
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; and.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Thomas Benzing
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; and.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Matthias J Hackl
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; and
| |
Collapse
|
34
|
Inoue K, Tian X, Velazquez H, Soda K, Wang Z, Pedigo CE, Wang Y, Cross E, Groener M, Shin JW, Li W, Hassan H, Yamamoto K, Mundel P, Ishibe S. Inhibition of Endocytosis of Clathrin-Mediated Angiotensin II Receptor Type 1 in Podocytes Augments Glomerular Injury. J Am Soc Nephrol 2019; 30:2307-2320. [PMID: 31511362 PMCID: PMC6900791 DOI: 10.1681/asn.2019010053] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 08/04/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Inhibition of the renin-angiotensin system remains a cornerstone in reducing proteinuria and progression of kidney failure, effects believed to be the result of reduction in BP and glomerular hyperfiltration. However, studies have yielded conflicting results on whether podocyte-specific angiotensin II (AngII) signaling directly induces podocyte injury. Previous research has found that after AngII stimulation, β-arrestin-bound angiotensin II receptor type 1 (AT1R) is internalized in a clathrin- and dynamin-dependent manner, and that Dynamin1 and Dynamin2 double-knockout mice exhibit impaired clathrin-mediated endocytosis. METHODS We used podocyte-specific Dyn double-knockout mice to examine AngII-stimulated AT1R internalization and signaling in primary podocytes and controls. We also examined the in vivo effect of AngII in these double-knockout mice through renin-angiotensin system blockers and through deletion of Agtr1a (which encodes the predominant AT1R isoform expressed in kidney, AT1aR). We tested calcium influx, Rac1 activation, and lamellipodial extension in control and primary podocytes of Dnm double-knockout mice treated with AngII. RESULTS We confirmed augmented AngII-stimulated AT1R signaling in primary Dnm double-knockout podocytes resulting from arrest of clathrin-coated pit turnover. Genetic ablation of podocyte Agtr1a in Dnm double-knockout mice demonstrated improved albuminuria and kidney function compared with the double-knockout mice. Isolation of podocytes from Dnm double-knockout mice revealed abnormal membrane dynamics, with increased Rac1 activation and lamellipodial extension, which was attenuated in Dnm double-knockout podocytes lacking AT1aR. CONCLUSIONS Our results indicate that inhibiting aberrant podocyte-associated AT1aR signaling pathways has a protective effect in maintaining the integrity of the glomerular filtration barrier.
Collapse
Affiliation(s)
- Kazunori Inoue
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Xuefei Tian
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Heino Velazquez
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Keita Soda
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Zhen Wang
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Christopher E Pedigo
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Ying Wang
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Elizabeth Cross
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Marwin Groener
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Jee-Won Shin
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Wei Li
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Hossam Hassan
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Koichi Yamamoto
- Department of Geriatric Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan; and
| | - Peter Mundel
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Shuta Ishibe
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut;
| |
Collapse
|
35
|
Vallon V, Unwin R, Inscho EW, Leipziger J, Kishore BK. Extracellular Nucleotides and P2 Receptors in Renal Function. Physiol Rev 2019; 100:211-269. [PMID: 31437091 DOI: 10.1152/physrev.00038.2018] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The understanding of the nucleotide/P2 receptor system in the regulation of renal hemodynamics and transport function has grown exponentially over the last 20 yr. This review attempts to integrate the available data while also identifying areas of missing information. First, the determinants of nucleotide concentrations in the interstitial and tubular fluids of the kidney are described, including mechanisms of cellular release of nucleotides and their extracellular breakdown. Then the renal cell membrane expression of P2X and P2Y receptors is discussed in the context of their effects on renal vascular and tubular functions. Attention is paid to effects on the cortical vasculature and intraglomerular structures, autoregulation of renal blood flow, tubuloglomerular feedback, and the control of medullary blood flow. The role of the nucleotide/P2 receptor system in the autocrine/paracrine regulation of sodium and fluid transport in the tubular and collecting duct system is outlined together with its role in integrative sodium and fluid homeostasis and blood pressure control. The final section summarizes the rapidly growing evidence indicating a prominent role of the extracellular nucleotide/P2 receptor system in the pathophysiology of the kidney and aims to identify potential therapeutic opportunities, including hypertension, lithium-induced nephropathy, polycystic kidney disease, and kidney inflammation. We are only beginning to unravel the distinct physiological and pathophysiological influences of the extracellular nucleotide/P2 receptor system and the associated therapeutic perspectives.
Collapse
Affiliation(s)
- Volker Vallon
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| | - Robert Unwin
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| | - Edward W Inscho
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| | - Jens Leipziger
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| | - Bellamkonda K Kishore
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| |
Collapse
|
36
|
Felizardo RJF, de Almeida DC, Pereira RL, Watanabe IKM, Doimo NTS, Ribeiro WR, Cenedeze MA, Hiyane MI, Amano MT, Braga TT, Ferreira CM, Parmigiani RB, Andrade-Oliveira V, Volpini RA, Vinolo MAR, Mariño E, Robert R, Mackay CR, Camara NOS. Gut microbial metabolite butyrate protects against proteinuric kidney disease through epigenetic- and GPR109a-mediated mechanisms. FASEB J 2019; 33:11894-11908. [PMID: 31366236 DOI: 10.1096/fj.201901080r] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Butyrate is a short-chain fatty acid derived from the metabolism of indigestible carbohydrates by the gut microbiota. Butyrate contributes to gut homeostasis, but it may also control inflammatory responses and host physiology in other tissues. Butyrate inhibits histone deacetylases, thereby affecting gene transcription, and also signals through the metabolite-sensing G protein receptor (GPR)109a. We produced an mAb to mouse GPR109a and found high expression on podocytes in the kidney. Wild-type and Gpr109a-/- mice were induced to develop nephropathy by a single injection of Adriamycin and treated with sodium butyrate or high butyrate-releasing high-amylose maize starch diet. Butyrate improved proteinuria by preserving podocyte at glomerular basement membrane and attenuated glomerulosclerosis and tissue inflammation. This protective phenotype was associated with increased podocyte-related proteins and a normalized pattern of acetylation and methylation at promoter sites of genes essential for podocyte function. We found that GPR109a is expressed by podocytes, and the use of Gpr109a-/- mice showed that the protective effects of butyrate depended on GPR109a expression. A prebiotic diet that releases high amounts of butyrate also proved highly effective for protection against kidney disease. Butyrate and GPR109a play a role in the pathogenesis of kidney disease and provide one of the important molecular connections between diet, the gut microbiota, and kidney disease.-Felizardo, R. J. F., de Almeida, D. C., Pereira, R. L., Watanabe, I. K. M., Doimo, N. T. S., Ribeiro, W. R., Cenedeze, M. A., Hiyane, M. I., Amano, M. T., Braga, T. T., Ferreira, C. M., Parmigiani, R. B., Andrade-Oliveira, V., Volpini, R. A., Vinolo, M. A. R., Mariño, E., Robert, R., Mackay, C. R., Camara, N. O. S. Gut microbial metabolite butyrate protects against proteinuric kidney disease through epigenetic- and GPR109a-mediated mechanisms.
Collapse
Affiliation(s)
- Raphael J F Felizardo
- Nephrology Division, Department of Medicine, Universidade Federal de São Paulo, São Paulo, Brazil.,Department of Immunology, Institute of Biomedical Sciences IV, Universidade de São Paulo, São Paulo, Brazil.,Department of Biochemistry and Molecular Biology, Biodiscovery Institute, Monash University, Clayton, Victoria, Australia
| | - Danilo C de Almeida
- Nephrology Division, Department of Medicine, Universidade Federal de São Paulo, São Paulo, Brazil.,Department of Immunology, Institute of Biomedical Sciences IV, Universidade de São Paulo, São Paulo, Brazil
| | - Rafael L Pereira
- Department of Physiology, Universidade Federal do Paraná, Curitiba, Brazil
| | - Ingrid K M Watanabe
- Nephrology Division, Department of Medicine, Universidade Federal de São Paulo, São Paulo, Brazil.,Department of Immunology, Institute of Biomedical Sciences IV, Universidade de São Paulo, São Paulo, Brazil
| | - Nayara T S Doimo
- Center for Molecular Oncology, Hospital Sírio-Libanês, São Paulo, Brazil
| | - Willian R Ribeiro
- Department of Pharmaceutics Sciences, Institute of Environmental Chemistry and Pharmaceutical Sciences, Universidade Federal de São Paulo, Diadema, Brazil
| | - Marcos A Cenedeze
- Nephrology Division, Department of Medicine, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Meire I Hiyane
- Department of Immunology, Institute of Biomedical Sciences IV, Universidade de São Paulo, São Paulo, Brazil
| | - Mariane T Amano
- Department of Immunology, Institute of Biomedical Sciences IV, Universidade de São Paulo, São Paulo, Brazil.,Center for Molecular Oncology, Hospital Sírio-Libanês, São Paulo, Brazil
| | - Tárcio T Braga
- Department of Immunology, Institute of Biomedical Sciences IV, Universidade de São Paulo, São Paulo, Brazil
| | - Caroline M Ferreira
- Department of Pharmaceutics Sciences, Institute of Environmental Chemistry and Pharmaceutical Sciences, Universidade Federal de São Paulo, Diadema, Brazil
| | | | - Vinicius Andrade-Oliveira
- Department of Immunology, Institute of Biomedical Sciences IV, Universidade de São Paulo, São Paulo, Brazil
| | - Rildo A Volpini
- Laboratório de Investigação Médica 12 (LIM12), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Marco Aurélio R Vinolo
- Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, Universidade Estadual de Campinas, Campinas, Brazil
| | - Eliana Mariño
- Department of Biochemistry and Molecular Biology, Biodiscovery Institute, Monash University, Clayton, Victoria, Australia
| | - Remy Robert
- Department of Biochemistry and Molecular Biology, Biodiscovery Institute, Monash University, Clayton, Victoria, Australia
| | - Charles R Mackay
- Department of Biochemistry and Molecular Biology, Biodiscovery Institute, Monash University, Clayton, Victoria, Australia
| | - Niels O S Camara
- Nephrology Division, Department of Medicine, Universidade Federal de São Paulo, São Paulo, Brazil.,Department of Immunology, Institute of Biomedical Sciences IV, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
37
|
Zhong C, Schleifenbaum J. Genetically Encoded Calcium Indicators: A New Tool in Renal Hypertension Research. Front Med (Lausanne) 2019; 6:128. [PMID: 31263699 PMCID: PMC6585435 DOI: 10.3389/fmed.2019.00128] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 05/23/2019] [Indexed: 11/13/2022] Open
Abstract
Hypertension is ranked as the third cause of disability-adjusted life-years. The percentage of the population suffering from hypertension will continue to increase over the next years. Renovascular disease is one of the most common causes of secondary hypertension. Vascular changes seen in hypertension are partially based on dysfunctional calcium signaling. This signaling can be studied using calcium indicators (loading dyes and genetically encoded calcium indicators; GECIs). Most progress in development has been seen in GECIs, which are used in an increasing number of publications concerning calcium signaling in vasculature and the kidney. The use of transgenic mouse models expressing GECIs will facilitate new possibilities to study dysfunctional calcium signaling in a cell type-specific manner, thus helping to identify more specific targets for treatment of (renal) hypertension.
Collapse
Affiliation(s)
- Cheng Zhong
- Institute of Vegetative Physiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Johanna Schleifenbaum
- Institute of Vegetative Physiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
38
|
Shroff UN, Schiessl IM, Gyarmati G, Riquier-Brison A, Peti-Peterdi J. Novel fluorescence techniques to quantitate renal cell biology. Methods Cell Biol 2019; 154:85-107. [PMID: 31493823 PMCID: PMC6748388 DOI: 10.1016/bs.mcb.2019.04.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Fluorescence microscopy techniques are powerful tools to study tissue dynamics, cellular function and biology both in vivo and in vitro. These tools allow for functional assessment and quantification along with qualitative analysis, thus providing a comprehensive understanding of various cellular processes under normal physiological and disease conditions. The main focus of this chapter is the recently developed method of serial intravital multiphoton microscopy that has helped shed light on the dynamic alterations of the spatial distribution and fate of single renal cells or cell populations and their migration patterns in the same tissue region over several days in response to various stimuli within the living kidney. This technique is very useful for studying in vivo the molecular and cellular mechanisms of tissue remodeling and repair after injury. In addition, complementary in vitro imaging tools are also described and discussed, like tissue clearing techniques and protein synthesis measurement in tissues in situ that provide an in depth assessment of changes at the cellular level. Thus, these novel fluorescence techniques can be effectively leveraged for different tissue types, experimental conditions as well as disease models to improve our understanding of renal cell biology.
Collapse
Affiliation(s)
- Urvi Nikhil Shroff
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Ina Maria Schiessl
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Georgina Gyarmati
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Anne Riquier-Brison
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Janos Peti-Peterdi
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.
| |
Collapse
|
39
|
Gyarmati G, Kadoya H, Moon JY, Burford JL, Ahmadi N, Gill IS, Hong YK, Dér B, Peti-Peterdi J. Advances in Renal Cell Imaging. Semin Nephrol 2019; 38:52-62. [PMID: 29291762 DOI: 10.1016/j.semnephrol.2017.09.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A great variety of cell imaging technologies are used routinely every day for the investigation of kidney cell types in applications ranging from basic science research to drug development and pharmacology, clinical nephrology, and pathology. Quantitative visualization of the identity, density, and fate of both resident and nonresident cells in the kidney, and imaging-based analysis of their altered function, (patho)biology, metabolism, and signaling in disease conditions, can help to better define pathomechanism-based disease subgroups, identify critical cells and structures that play a role in the pathogenesis, critically needed biomarkers of disease progression, and cell and molecular pathways as targets for novel therapies. Overall, renal cell imaging has great potential for improving the precision of diagnostic and treatment paradigms for individual acute kidney injury or chronic kidney disease patients or patient populations. This review highlights and provides examples for some of the recently developed renal cell optical imaging approaches, mainly intravital multiphoton fluorescence microscopy, and the new knowledge they provide for our better understanding of renal pathologies.
Collapse
Affiliation(s)
- Georgina Gyarmati
- Department of Physiology and Neuroscience, Department of Medicine, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Hiroyuki Kadoya
- Department of Physiology and Neuroscience, Department of Medicine, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA; Department of Nephrology and Hypertension, Kawasaki Medical School, Kurashiki, Japan
| | - Ju-Young Moon
- Department of Physiology and Neuroscience, Department of Medicine, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA; Division of Nephrology, Department of Internal Medicine, Kyung Hee University, College of Medicine, Seoul, Korea
| | - James L Burford
- Department of Physiology and Neuroscience, Department of Medicine, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Nariman Ahmadi
- Institute of Urology, Catherine & Joseph Aresty Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Inderbir S Gill
- Institute of Urology, Catherine & Joseph Aresty Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Young-Kwon Hong
- Department of Surgery and Biochemistry and Molecular Biology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Bálint Dér
- Department of Physiology and Neuroscience, Department of Medicine, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - János Peti-Peterdi
- Department of Physiology and Neuroscience, Department of Medicine, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA.
| |
Collapse
|
40
|
Schiessl IM, Fremter K, Burford JL, Castrop H, Peti-Peterdi J. Long-Term Cell Fate Tracking of Individual Renal Cells Using Serial Intravital Microscopy. Methods Mol Biol 2019; 2150:25-44. [PMID: 31087287 DOI: 10.1007/7651_2019_232] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Intravital multiphoton microscopy of the kidney is a powerful technique to study alterations in tissue morphology and function simultaneously in the living animal and represents a dynamic and developing research tool in the field. Recent technological advances include serial intravital multiphoton microscopy of the same kidney regions over several weeks and combined with ex vivo histology for cellular biomarker expression of the same cells, which had been subject to serial imaging before. Thus, serial intravital multiphoton microscopy followed by ex vivo histology provides unique tools to perform long-term cell fate tracing of the same renal cells during physiological and pathophysiological conditions, thereby allowing the detection of structural changes of the same renal cells over time. Examples include renal cell migration and proliferation while linking these events to local functional alterations and eventually to the expression of distinct cellular biomarkers. Here, we provide a detailed step-by-step protocol to facilitate serial intravital multiphoton microscopy for long-term in vivo tracking of renal cells and subsequent ex vivo histology for immunohistological staining of the same cells in the fixed tissue.
Collapse
Affiliation(s)
- Ina Maria Schiessl
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Katharina Fremter
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - James L Burford
- Department of Ophthalmology, University of Southern California, Los Angeles, CA, USA
| | - Hayo Castrop
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Janos Peti-Peterdi
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
41
|
Rennert L, Zschiedrich S, Sandner L, Hartleben B, Cicko S, Ayata CK, Meyer C, Zech A, Zeiser R, Huber TB, Idzko M, Grahammer F. P2Y2R Signaling Is Involved in the Onset of Glomerulonephritis. Front Immunol 2018; 9:1589. [PMID: 30061884 PMCID: PMC6054981 DOI: 10.3389/fimmu.2018.01589] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 06/27/2018] [Indexed: 12/19/2022] Open
Abstract
Endogenously released adenosine-5’-triphosphate (ATP) is a key regulator of physiological function and inflammatory responses in the kidney. Genetic or pharmacological inhibition of purinergic receptors has been linked to attenuation of inflammatory disorders and hence constitutes promising new avenues for halting and reverting inflammatory renal diseases. However, the involvement of purinergic receptors in glomerulonephritis (GN) has only been incompletely mapped. Here, we demonstrate that induction of GN in an experimental antibody-mediated GN model results in a significant increase of urinary ATP-levels and an upregulation of P2Y2R expression in resident kidney cells as well as infiltrating leukocytes pointing toward a possible role of the ATP/P2Y2R-axis in glomerular disease initiation. In agreement, decreasing extracellular ATP-levels or inhibition of P2R during induction of antibody-mediated GN leads to a reduction in all cardinal features of GN such as proteinuria, glomerulosclerosis, and renal failure. The specific involvement of P2Y2R could be further substantiated by demonstrating the protective effect of the lack of P2Y2R in antibody-mediated GN. To systematically differentiate between the function of P2Y2R on resident renal cells versus infiltrating leukocytes, we performed bone marrow-chimera experiments revealing that P2Y2R on hematopoietic cells is the main driver of the ATP/P2Y2R-mediated disease progression in antibody-mediated GN. Thus, these data unravel an important pro-inflammatory role for P2Y2R in the pathogenesis of GN.
Collapse
Affiliation(s)
- Laura Rennert
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stefan Zschiedrich
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Lukas Sandner
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Pneumology, University Medical Center Freiburg, Freiburg, Germany
| | - Björn Hartleben
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sanja Cicko
- Department of Pneumology, University Medical Center Freiburg, Freiburg, Germany
| | - Cemil Korcan Ayata
- Department of Pneumology, University Medical Center Freiburg, Freiburg, Germany
| | - Charlotte Meyer
- Department of Pneumology, University Medical Center Freiburg, Freiburg, Germany
| | - Andreas Zech
- Department of Pneumology, University Medical Center Freiburg, Freiburg, Germany
| | - Robert Zeiser
- Department of Hematology, Oncology and Stem Cell Transplantation, University Medical Center Freiburg, Freiburg, Germany
| | - Tobias 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, Albert-Ludwigs-University, Freiburg, Germany
| | - Marco Idzko
- Department of Pneumology, University Medical Center Freiburg, Freiburg, Germany.,Division of Pulmonology, Department of Medicine II, Medical University Vienna, Vienna, Austria
| | - Florian 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
| |
Collapse
|
42
|
Siegerist F, Endlich K, Endlich N. Novel Microscopic Techniques for Podocyte Research. Front Endocrinol (Lausanne) 2018; 9:379. [PMID: 30050501 PMCID: PMC6050355 DOI: 10.3389/fendo.2018.00379] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 06/22/2018] [Indexed: 01/16/2023] Open
Abstract
Together with endothelial cells and the glomerular basement membrane, podocytes form the size-specific filtration barrier of the glomerulus with their interdigitating foot processes. Since glomerulopathies are associated with so-called foot process effacement-a severe change of well-formed foot processes into flat and broadened processes-visualization of the three-dimensional podocyte morphology is a crucial part for diagnosis of nephrotic diseases. However, interdigitating podocyte foot processes are too narrow to be resolved by classic light microscopy due to Ernst Abbe's law making electron microscopy necessary. Although three dimensional electron microscopy approaches like serial block face and focused ion beam scanning electron microscopy and electron tomography allow volumetric reconstruction of podocytes, these techniques are very time-consuming and too specialized for routine use or screening purposes. During the last few years, different super-resolution microscopic techniques were developed to overcome the optical resolution limit enabling new insights into podocyte morphology. Super-resolution microscopy approaches like three dimensional structured illumination microscopy (3D-SIM), stimulated emission depletion microscopy (STED) and localization microscopy [stochastic optical reconstruction microscopy (STORM), photoactivated localization microscopy (PALM)] reach resolutions down to 80-20 nm and can be used to image and further quantify podocyte foot process morphology. Furthermore, in vivo imaging of podocytes is essential to study the behavior of these cells in situ. Therefore, multiphoton laser microscopy was a breakthrough for in vivo studies of podocytes in transgenic animal models like rodents and zebrafish larvae because it allows imaging structures up to several hundred micrometer in depth within the tissue. Additionally, along with multiphoton microscopy, lightsheet microscopy is currently used to visualize larger tissue volumes and therefore image complete glomeruli in their native tissue context. Alongside plain visualization of cellular structures, atomic force microscopy has been used to study the change of mechanical properties of podocytes in diseased states which has been shown to be a culprit in podocyte maintenance. This review discusses recent advances in the field of microscopic imaging and demonstrates their currently used and other possible applications for podocyte research.
Collapse
Affiliation(s)
| | | | - Nicole Endlich
- Institute for Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
43
|
Riquier-Brison ADM, Sipos A, Prókai Á, Vargas SL, Toma L, Meer EJ, Villanueva KG, Chen JCM, Gyarmati G, Yih C, Tang E, Nadim B, Pendekanti S, Garrelds IM, Nguyen G, Danser AHJ, Peti-Peterdi J. The macula densa prorenin receptor is essential in renin release and blood pressure control. Am J Physiol Renal Physiol 2018; 315:F521-F534. [PMID: 29667908 DOI: 10.1152/ajprenal.00029.2018] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The prorenin receptor (PRR) was originally proposed to be a member of the renin-angiotensin system (RAS); however, recent work questioned their association. The present paper describes a functional link between the PRR and RAS in the renal juxtaglomerular apparatus (JGA), a classic anatomical site of the RAS. PRR expression was found in the sensory cells of the JGA, the macula densa (MD), and immunohistochemistry-localized PRR to the MD basolateral cell membrane in mouse, rat, and human kidneys. MD cell PRR activation led to MAP kinase ERK1/2 signaling and stimulation of PGE2 release, the classic pathway of MD-mediated renin release. Exogenous renin or prorenin added to the in vitro microperfused JGA-induced acute renin release, which was inhibited by removing the MD or by the administration of a PRR decoy peptide. To test the function of MD PRR in vivo, we established a new mouse model with inducible conditional knockout (cKO) of the PRR in MD cells based on neural nitric oxide synthase-driven Cre-lox recombination. Deletion of the MD PRR significantly reduced blood pressure and plasma renin. Challenging the RAS by low-salt diet + captopril treatment caused further significant reductions in blood pressure, renal renin, cyclooxygenase-2, and microsomal PGE synthase expression in cKO vs. wild-type mice. These results suggest that the MD PRR is essential in a novel JGA short-loop feedback mechanism, which is integrated within the classic MD mechanism to control renin synthesis and release and to maintain blood pressure.
Collapse
Affiliation(s)
- Anne D M Riquier-Brison
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California , Los Angeles, California
| | - Arnold Sipos
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California , Los Angeles, California
| | - Ágnes Prókai
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California , Los Angeles, California
| | - Sarah L Vargas
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California , Los Angeles, California
| | - Lldikó Toma
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California , Los Angeles, California
| | - Elliott J Meer
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California , Los Angeles, California
| | - Karie G Villanueva
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California , Los Angeles, California
| | - Jennifer C M Chen
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California , Los Angeles, California
| | - Georgina Gyarmati
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California , Los Angeles, California
| | - Christopher Yih
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California , Los Angeles, California
| | - Elaine Tang
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California , Los Angeles, California
| | - Bahram Nadim
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California , Los Angeles, California
| | - Sujith Pendekanti
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California , Los Angeles, California
| | - Ingrid M Garrelds
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, Rotterdam , The Netherlands
| | - Genevieve Nguyen
- Centre for Interdisciplinary Research in Biology, UMR INSERM U1050, Collège de France, Paris , France
| | - A H Jan Danser
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, Rotterdam , The Netherlands
| | - János Peti-Peterdi
- Departments of Physiology and Neuroscience, and Medicine, Zilkha Neurogenetic Institute, University of Southern California , Los Angeles, California
| |
Collapse
|
44
|
Schiessl IM, Grill A, Fremter K, Steppan D, Hellmuth MK, Castrop H. Renal Interstitial Platelet-Derived Growth Factor Receptor- β Cells Support Proximal Tubular Regeneration. J Am Soc Nephrol 2018; 29:1383-1396. [PMID: 29444905 DOI: 10.1681/asn.2017101069] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Accepted: 01/16/2018] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND The kidney is considered to be a structurally stable organ with limited baseline cellular turnover. Nevertheless, single cells must be constantly replaced to conserve the functional integrity of the organ. PDGF chain B (PDGF-BB) signaling through fibroblast PDGF receptor-β (PDGFRβ) contributes to interstitial-epithelial cell communication and facilitates regenerative functions in several organs. However, the potential role of interstitial cells in renal tubular regeneration has not been examined. METHODS In mice with fluorescent protein expression in renal tubular cells and PDGFRβ-positive interstitial cells, we ablated single tubular cells by high laser exposure. We then used serial intravital multiphoton microscopy with subsequent three-dimensional reconstruction and ex vivo histology to evaluate the cellular and molecular processes involved in tubular regeneration. RESULTS Single-tubular cell ablation caused the migration and division of dedifferentiated tubular epithelial cells that preceded tubular regeneration. Moreover, tubular cell ablation caused immediate calcium responses in adjacent PDGFRβ-positive interstitial cells and the rapid migration thereof toward the injury. These PDGFRβ-positive cells enclosed the injured epithelium before the onset of tubular cell dedifferentiation, and the later withdrawal of these PDGFRβ-positive cells correlated with signs of tubular cell redifferentiation. Intraperitoneal administration of trapidil to block PDGFRβ impeded PDGFRβ-positive cell migration to the tubular injury site and compromised the recovery of tubular function. CONCLUSIONS Ablated tubular cells are exclusively replaced by resident tubular cell proliferation in a process dependent on PDGFRβ-mediated communication between the renal interstitium and the tubular system.
Collapse
Affiliation(s)
- Ina Maria Schiessl
- Institute of Physiology, University of Regensburg, Regensburg, Germany; and .,Department of Physiology and Biophysics, University of Southern California, Los Angeles, California
| | - Alexandra Grill
- Institute of Physiology, University of Regensburg, Regensburg, Germany; and
| | - Katharina Fremter
- Institute of Physiology, University of Regensburg, Regensburg, Germany; and
| | - Dominik Steppan
- Institute of Physiology, University of Regensburg, Regensburg, Germany; and
| | | | - Hayo Castrop
- Institute of Physiology, University of Regensburg, Regensburg, Germany; and
| |
Collapse
|
45
|
Ma Y, Yang Q, Zhong Z, Liang W, Zhang L, Yang Y, Ding G. Role of c-Abl and nephrin in podocyte cytoskeletal remodeling induced by angiotensin II. Cell Death Dis 2018; 9:185. [PMID: 29416010 PMCID: PMC5833834 DOI: 10.1038/s41419-017-0225-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 12/05/2017] [Accepted: 12/06/2017] [Indexed: 12/18/2022]
Abstract
Our previous study showed that angiotensin II (Ang II) exposure diminished the interaction between nephrin and c-Abl, then c-Abl mediated SHIP2-Akt pathway in the process of podocyte injury in vivo and vitro. However, the relationship between nephrin and c-Abl was unknown. Recently, various studies showed that nephrin was required for cytoskeletal remodeling in glomerular podocytes. But its specific mechanisms remain incompletely understood. As a nonreceptor tyrosine kinase involved in cytoskeletal regulation, c-Abl may be a candidate of signaling proteins interacting with Src homology 2/3 (SH2/SH3) domains of nephrin. Therefore, it is proposed that c-Abl contributes to nephrin-dependent cytoskeletal remodeling of podocytes. Herein, we observed that nephrin-c-Abl colocalization were suppressed in glomeruli of patients with proteinuria. Next, CD16/7-nephrin and c-Abl vectors were constructed to investigate the nephrin-c-Abl signaling pathway in podocyte actin-cytoskeletal remodeling. The disorganized cytoskeleton stimulated by cytochalasin D in COS7 cells was dramatically restored by co-transfection with phosphorylated CD16/7-nephrin and c-Abl full-length constructs. Further, co-immunoprecipitation showed that phosphorylated CD16/7-nephrin interacted with wild-type c-Abl, but not with SH2/SH3-defective c-Abl. These findings suggest that phosphorylated nephrin is able to recruit c-Abl in a SH2/SH3-dependent manner and detached c-Abl from dephosphorylated nephrin contributes to cytoskeletal remodeling in podocytes.
Collapse
Affiliation(s)
- Yiqiong Ma
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Qian Yang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zhentong Zhong
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Wei Liang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Lu Zhang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yingjie Yang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Guohua Ding
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
46
|
Dunn KW, Sutton TA, Sandoval RM. Live-Animal Imaging of Renal Function by Multiphoton Microscopy. ACTA ACUST UNITED AC 2018; 83:12.9.1-12.9.25. [PMID: 29345326 DOI: 10.1002/cpcy.32] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Intravital microscopy, microscopy of living animals, is a powerful research technique that combines the resolution and sensitivity found in microscopic studies of cultured cells with the relevance and systemic influences of cells in the context of the intact animal. The power of intravital microscopy has recently been extended with the development of multiphoton fluorescence microscopy systems capable of collecting optical sections from deep within the kidney at subcellular resolution, supporting high-resolution characterizations of the structure and function of glomeruli, tubules, and vasculature in the living kidney. Fluorescent probes are administered to an anesthetized, surgically prepared animal, followed by image acquisition for up to 3 hr. Images are transferred via a high-speed network to specialized computer systems for digital image analysis. This general approach can be used with different combinations of fluorescent probes to evaluate processes such as glomerular permeability, proximal tubule endocytosis, microvascular flow, vascular permeability, mitochondrial function, and cellular apoptosis/necrosis. © 2018 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Kenneth W Dunn
- Indiana University School of Medicine, Indianapolis, Indiana
| | | | | |
Collapse
|
47
|
Abstract
Kidney cell death plays a key role in the progression of life-threatening renal diseases, such as acute kidney injury and chronic kidney disease. Injured and dying epithelial and endothelial cells take part in complex communication with the innate immune system, which drives the progression of cell death and the decrease in renal function. To improve our understanding of kidney cell death dynamics and its impact on renal disease, a study approach is needed that facilitates the visualization of renal function and morphology in real time. Intravital multiphoton microscopy of the kidney has been used for more than a decade and made substantial contributions to our understanding of kidney physiology and pathophysiology. It is a unique tool that relates renal structure and function in a time- and spatial-dependent manner. Basic renal function, such as microvascular blood flow regulation and glomerular filtration, can be determined in real time and homeostatic alterations, which are linked inevitably to cell death and can be depicted down to the subcellular level. This review provides an overview of the available techniques to study kidney dysfunction and inflammation in terms of cell death in vivo, and addresses how this novel approach can be used to improve our understanding of cell death dynamics in renal disease.
Collapse
|
48
|
Guan Y, Nakano D, Zhang Y, Li L, Liu W, Nishida M, Kuwabara T, Morishita A, Hitomi H, Mori K, Mukoyama M, Masaki T, Hirano K, Nishiyama A. A protease-activated receptor-1 antagonist protects against podocyte injury in a mouse model of nephropathy. J Pharmacol Sci 2017; 135:S1347-8613(17)30128-7. [PMID: 29110957 DOI: 10.1016/j.jphs.2017.09.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 08/13/2017] [Accepted: 08/15/2017] [Indexed: 12/19/2022] Open
Abstract
The kidney expresses protease-activated receptor-1 (PAR-1). PAR-1 is known as a thrombin receptor, but its role in kidney injury is not well understood. In this study, we examined the contribution of PAR-1 to kidney glomerular injury and the effects of its inhibition on development of nephropathy. Mice were divided into 3 groups: control, doxorubicin + vehicle (15 mg/kg doxorubicin and saline) and doxorubicin + Q94 (doxorubicin at 15 mg/kg and the PAR-1 antagonist Q94 at 5 mg/kg/d) groups. Where indicated, doxorubicin was administered intravenously and PAR-1 antagonist or saline vehicle by subcutaneous osmotic mini-pump. PAR-1 expression was increased in glomeruli of mice treated with doxorubicin. Q94 treatment significantly suppressed the increased albuminuria in these nephropathic mice. Pathological analysis showed that Q94 treatment significantly attenuated periodic acid-Schiff and desmin staining, indicators of podocyte injury, and also decreased glomerular levels of podocin and nephrin. Furthermore, thrombin increased intracellular calcium levels in podocytes. This increase was suppressed by Q94 and Rox4560, a transient receptor potential cation channel (TRPC)3/6 antagonist. In addition, both Q94 and Rox4560 suppressed the doxorubicin-induced increase in activities of caspase-9 and caspase-3 in podocytes. These data suggested that PAR-1 contributes to development of podocyte and glomerular injury and that PAR-1 antagonists have therapeutic potential.
Collapse
Affiliation(s)
- Yu Guan
- Department of Pharmacology, Kagawa University, Kagawa, Japan
| | - Daisuke Nakano
- Department of Pharmacology, Kagawa University, Kagawa, Japan.
| | - Yifan Zhang
- Department of Pharmacology, Kagawa University, Kagawa, Japan
| | - Lei Li
- Department of Pharmacology, Kagawa University, Kagawa, Japan
| | - Wenhua Liu
- Department of Cardiovascular Physiology, Kagawa University, Kagawa, Japan
| | - Motohiro Nishida
- Division of Cardiocirculatory Signaling, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), National Institutes of Natural Sciences, Okazaki, Aichi, Japan
| | - Takashige Kuwabara
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Asahiro Morishita
- Department of Gastroenterology & Neurology, Kagawa University, Kagawa, Japan
| | - Hirofumi Hitomi
- Department of Pharmacology, Kagawa University, Kagawa, Japan
| | - Kiyoshi Mori
- Department of Nephrology and Kidney Research, Shizuoka General Hospital, Shizuoka, Japan
| | - Masashi Mukoyama
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Tsutomu Masaki
- Department of Gastroenterology & Neurology, Kagawa University, Kagawa, Japan
| | - Katsuya Hirano
- Department of Cardiovascular Physiology, Kagawa University, Kagawa, Japan
| | - Akira Nishiyama
- Department of Pharmacology, Kagawa University, Kagawa, Japan
| |
Collapse
|
49
|
Kavvadas P, Abed A, Poulain C, Authier F, Labéjof LP, Calmont A, Afieri C, Prakoura N, Dussaule JC, Chatziantoniou C, Chadjichristos CE. Decreased Expression of Connexin 43 Blunts the Progression of Experimental GN. J Am Soc Nephrol 2017; 28:2915-2930. [PMID: 28667079 DOI: 10.1681/asn.2016111211] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 05/05/2017] [Indexed: 11/03/2022] Open
Abstract
GN refers to a variety of renal pathologies that often progress to ESRD, but the molecular mechanisms underlying this progression remain incompletely characterized. Here, we determined whether dysregulated expression of the gap junction protein connexin 43, which has been observed in the progression of renal disease, contributes to GN progression. Immunostaining revealed de novo expression of connexin 43 in damaged glomeruli in patients with glomerular diseases as well as in mice after induction of experimental GN. Notably, 2 weeks after the induction of GN with nephrotoxic serum, mice with a heterozygous deletion of the connexin 43 gene (connexin 43+/-) had proteinuria, BUN, and serum creatinine levels significantly lower than those of wild-type animals. Additionally, the connexin 43+/- mice showed less crescent formation, tubular dilation, monocyte infiltration, and interstitial renal fibrosis. Treatment of cultured podocytes with connexin 43-specific blocking peptides attenuated TGF-β-induced cytoskeletal and morphologic changes and apoptosis as did treatment with the purinergic blocker suramin. Finally, therapeutic treatment of GN mice with connexin 43-specific antisense oligodeoxynucleotide improved functional and structural renal parameters. These findings suggest that crosstalk between connexin 43 and purinergic signaling contributes to podocyte damage in GN. Given that this protein is highly induced in individuals with glomerular diseases, connexin 43 may be a novel target for therapeutic treatment of GN.
Collapse
Affiliation(s)
- Panagiotis Kavvadas
- National Institute for Health and Medical Research Unité Mixte de Recherche-S1155, Batiment Recherche, Tenon Hospital, Paris, France
| | - Ahmed Abed
- National Institute for Health and Medical Research Unité Mixte de Recherche-S1155, Batiment Recherche, Tenon Hospital, Paris, France.,Sorbonne Universites, University Pierre et Marie Curie University Paris 6, Paris, France
| | - Coralie Poulain
- National Institute for Health and Medical Research Unité Mixte de Recherche-S1155, Batiment Recherche, Tenon Hospital, Paris, France.,University René Descartes, Paris, France.,University Denis Diderot, Paris, France
| | - Florence Authier
- National Institute for Health and Medical Research Unité Mixte de Recherche-S1155, Batiment Recherche, Tenon Hospital, Paris, France
| | - Lise-Paule Labéjof
- National Institute for Health and Medical Research Unité Mixte de Recherche-S1155, Batiment Recherche, Tenon Hospital, Paris, France.,Universidade Estadual de Santa Cruz, Ilhéus, Bahia, Brazil
| | - Amelie Calmont
- National Institute for Health and Medical Research Unité Mixte de Recherche-S1155, Batiment Recherche, Tenon Hospital, Paris, France
| | - Carlo Afieri
- National Institute for Health and Medical Research Unité Mixte de Recherche-S1155, Batiment Recherche, Tenon Hospital, Paris, France.,Unit of Nephrology Dialysis and Kidney Transplantation, Fondazione Istituto Di Ricovero e Cura a Carattere Scientifico Ca Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy; and
| | - Niki Prakoura
- National Institute for Health and Medical Research Unité Mixte de Recherche-S1155, Batiment Recherche, Tenon Hospital, Paris, France
| | - Jean-Claude Dussaule
- National Institute for Health and Medical Research Unité Mixte de Recherche-S1155, Batiment Recherche, Tenon Hospital, Paris, France.,Sorbonne Universites, University Pierre et Marie Curie University Paris 6, Paris, France.,Department of Physiology, Saint Antoine Hospital, Paris, France
| | - Christos Chatziantoniou
- National Institute for Health and Medical Research Unité Mixte de Recherche-S1155, Batiment Recherche, Tenon Hospital, Paris, France.,Sorbonne Universites, University Pierre et Marie Curie University Paris 6, Paris, France
| | - Christos E Chadjichristos
- National Institute for Health and Medical Research Unité Mixte de Recherche-S1155, Batiment Recherche, Tenon Hospital, Paris, France; .,Sorbonne Universites, University Pierre et Marie Curie University Paris 6, Paris, France
| |
Collapse
|
50
|
Combined use of electron microscopy and intravital imaging captures morphological and functional features of podocyte detachment. Pflugers Arch 2017; 469:965-974. [PMID: 28664407 DOI: 10.1007/s00424-017-2020-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 06/13/2017] [Accepted: 06/14/2017] [Indexed: 12/12/2022]
Abstract
The development of podocyte injury and albuminuria in various glomerular pathologies is still incompletely understood due to technical limitations in studying the glomerular filtration barrier (GFB) in real-time. We aimed to directly visualize the early morphological and functional changes of the GFB during the development of focal segmental glomerulosclerosis (FSGS) using a combination of transmission electron microscopy (TEM) and in vivo multiphoton microscopy (MPM) in the rat puromycin aminonucleoside (PAN) model. We hypothesized that this combined TEM + MPM experimental approach would provide a major technical improvement that would benefit our mechanistic understanding of podocyte detachment. Male Sprague-Dawley (for TEM) or Munich-Wistar-Frömter (for MPM) rats were given a single dose of 100-150 mg/kg body weight PAN i.p. and were either sacrificed and the kidneys processed for TEM or surgically instrumented for in vivo MPM imaging at various times 2-14 days after PAN administration. Both techniques demonstrated hypertrophy and cystic dilatations of the subpodocyte space that developed as early as 2-3 days after PAN. Adhesions of the visceral epithelium to the parietal Bowman's capsule (synechiae) appeared at days 8-10. TEM provided unmatched resolution of podocyte foot process remodeling, while MPM revealed the rapid dynamics of pseudocyst filling, emptying, and rupture, as well as endothelial and podocyte injury, misdirected filtration, and podocyte shedding. Due to the complementary advantages of TEM and MPM, this combined approach can provide an unusally comprehensive and dynamic portrayal of the alterations in podocyte morphology and function during FSGS development. The results advance our understanding of the role and importance of the various cell types, hemodynamics, and mechanical forces in the development of glomerular pathology.
Collapse
|