1
|
Hall AM. Protein handling in kidney tubules. Nat Rev Nephrol 2025; 21:241-252. [PMID: 39762367 DOI: 10.1038/s41581-024-00914-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2024] [Indexed: 03/23/2025]
Abstract
The kidney proximal tubule reabsorbs and degrades filtered plasma proteins to reclaim valuable nutrients and maintain body homeostasis. Defects in this process result in proteinuria, one of the most frequently used biomarkers of kidney disease. Filtered proteins enter proximal tubules via receptor-mediated endocytosis and are processed within a highly developed apical endo-lysosomal system (ELS). Proteinuria is a strong risk factor for chronic kidney disease progression and genetic disorders of the ELS cause hereditary kidney diseases, so deepening understanding of how the proximal tubule handles proteins is crucial for translational nephrology. Moreover, the ELS is both an entry point for nephrotoxins that induce tubular damage and a target for novel therapies to prevent it. Cutting-edge research techniques, such as functional intravital imaging and computational modelling, are shedding light on spatial and integrative aspects of renal tubular protein processing in vivo, how these are altered under pathological conditions and the consequences for other tubular functions. These insights have potentially important implications for understanding the origins of systemic complications arising in proteinuric states, and might lead to the development of new ways of monitoring and treating kidney diseases.
Collapse
Affiliation(s)
- Andrew M Hall
- Institute of Anatomy, University of Zurich, Zurich, Switzerland.
- Zurich Kidney Center, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
2
|
Wagner CA. Beyond SGLT2: proximal tubule transporters as potential drug targets for chronic kidney disease. Nephrol Dial Transplant 2025; 40:i18-i28. [PMID: 39907544 PMCID: PMC11795650 DOI: 10.1093/ndt/gfae211] [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: 05/02/2024] [Indexed: 02/06/2025] Open
Abstract
The kidneys produce daily about 180 liters of urine but only about 2 liters are excreted. The proximal tubule plays an important role in reabsorbing the majority of filtered urine and many metabolites such as sugars, amino acids, salts or phosphate that are contained in this large volume. Reabsorption of these important metabolites is mediated by a diverse group of highly specialized transport proteins. Another group of transport proteins in the proximal tubule is responsible for the active secretion of metabolic waste products or toxins and drugs into urine. All these transporters have in common that they are directly linked to kidney metabolism and indirectly to whole-body metabolism and functions. In recent years, it has become evident that modulation of these transporters may influence the onset, progression and consequences of kidney disease. This review summarizes recent developments in this field and discusses some examples of drugs already in clinical use or in development. The examples include inhibitors of sugar transporters (SGLT2 inhibitors) that are successfully used in patients with kidney disease, diabetes or heart failure. Likewise, indirect inhibitors (acetazolamide) of an transporter absorbing sodium in exchange for protons (NHE3) are used mostly in patients with heart failure or for prevention of high altitude disease, while direct inhibitors show promise in preclinical studies to reduce damage in episodes of acute kidney disease or high blood pressure. Modulators of transporters mediating the excretion of urate have been used in patients with gout and are also discussed to prevent kidney disease. Novel drugs in development target transporters for phosphate, amino acids, or toxin and drug excretion and may be helpful for specific conditions associated with kidney disease. The advantages and challenges associated with these (novel) drugs targeting proximal tubule transport are discussed. ABSTRACT The proximal tubule is responsible for reabsorbing about 60% of filtered solutes and water and is critical for the secretion of metabolic waste products, drugs and toxins. A large number of highly specialized ion channels and transport proteins belonging to the SLC and ABC transporter families are involved. Their activity is directly or indirectly linked to ATP consumption and requires large quantities of energy and oxygen supply. Moreover, the activity of these transporters is often coupled to the movement of Na+ ions thus influencing also salt and water balance, as well as transport and regulatory processes in downstream segments. Because of their relevance for systemic ion balance, for renal metabolism or for affecting regulatory processes, proximal tubule transporters are attractive targets for existing drug and for novel strategies to reduce kidney disease progression or to alleviate the consequences of decreased kidney function. In this review, the relevance of some major proximal tubule transport systems as drug targets in individuals with chronic kidney disease (CKD) is discussed. Inhibitors of the sodium-glucose cotransporter 2, SGLT2, are now part of standard therapy in patients with CKD and/or heart failure. Also, indirect inhibition of Na+/H+-exchangers by carbonic anhydrase inhibitors and uricosuric drugs have been used for decades. Inhibition of phosphate and amino acid transporters have recently been proposed as novel principles to remove excess phosphate or to protect the proximal tubule metabolically, respectively. In addition, organic cation and anion transporters involved in drug and toxin excretion may serve as targets of new drugs. The advantages and challenges associated with (novel) drugs targeting proximal tubule transport are discussed.
Collapse
Affiliation(s)
- Carsten A Wagner
- University of Zurich – Institute of Physiology, Zurich, Switzerland
| |
Collapse
|
3
|
Hoenig MP, Brooks CR, Hoorn EJ, Hall AM. Biology of the proximal tubule in body homeostasis and kidney disease. Nephrol Dial Transplant 2025; 40:234-243. [PMID: 39066502 PMCID: PMC11852287 DOI: 10.1093/ndt/gfae177] [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: 02/29/2024] [Indexed: 07/28/2024] Open
Abstract
The proximal tubule (PT) is known as the workhorse of the kidney, for both the range and magnitude of the functions that it performs. It is not only responsible for reabsorbing most solutes and proteins filtered by glomeruli, but also for secreting non-filtered substances including drugs and uremic toxins. The PT therefore plays a pivotal role in kidney physiology and body homeostasis. Moreover, it is the major site of damage in acute kidney injury and nephrotoxicity. In this review, we will provide an introduction to the cell biology of the PT and explore how it is adapted to the execution of a myriad of different functions and how these can differ between males and females. We will then discuss how the PT regulates phosphate, glucose and acid-base balance, and the consequences of alterations in PT function for bone and cardiovascular health. Finally, we explore why the PT is vulnerable to ischemic and toxic insults, and how acute injury in the PT can lead to maladaptive repair, chronic damage and kidney fibrosis. In summary, we will demonstrate that knowledge of the basic cell biology of the PT is critical for understanding kidney disease phenotypes and their associated systemic complications, and for developing new therapeutic strategies to prevent these.
Collapse
Affiliation(s)
- Melanie P Hoenig
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Craig R Brooks
- Department of Medicine at Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ewout J Hoorn
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Andrew M Hall
- Institute of Anatomy, University of Zurich, Switzerland. Zurich Kidney Center, University of Zurich, Zürich, Switzerland
| |
Collapse
|
4
|
Ayasse N, Berg P, Sørensen MV, Svendsen SL, Weinstein AM, Leipziger J. Revisiting voltage-coupled H + secretion in the collecting duct. Am J Physiol Renal Physiol 2024; 327:F931-F945. [PMID: 39323388 PMCID: PMC11918339 DOI: 10.1152/ajprenal.00023.2024] [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: 01/18/2024] [Revised: 09/17/2024] [Accepted: 09/17/2024] [Indexed: 09/27/2024] Open
Abstract
Experimental studies have shown that V-type ATPase-driven H+ secretion is dependent on transepithelial voltage. On this basis, the "voltage hypothesis" of urinary acidification by the collecting duct was derived. Accordingly, it has been supposed that the lumen-negative potential created by the reabsorption of Na+ via the epithelial Na+ channel (ENaC) enhances electrogenic H+ secretion via V-type H+-ATPase. This concept continues to be widely used to explain acid/base disorders. Importantly, however, a solid proof of principle for the voltage hypothesis in physiologically relevant situations has not been reached. Rather, it has been challenged by recent in vivo functional studies. In this review, we outline the arguments and experimental observations explaining why voltage-coupled H+ secretion in the collecting duct often appears poorly applicable for rationalizing changes in H+ secretion as a function of more or less ENaC function in the collecting duct.
Collapse
Affiliation(s)
- Niklas Ayasse
- Vth Department of Medicine, University Hospital Mannheim, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany
| | - Peder Berg
- Department of Biomedicine, Physiology, Aarhus University, Aarhus, Denmark
| | - Mads V Sørensen
- Department of Biomedicine, Physiology, Aarhus University, Aarhus, Denmark
| | - Samuel L Svendsen
- Department of Biomedicine, Physiology, Aarhus University, Aarhus, Denmark
| | - Alan M Weinstein
- Department of Physiology and Biophysics, Weill Medical College of Cornell University, New York, New York, United States
| | - Jens Leipziger
- Department of Biomedicine, Physiology, Aarhus University, Aarhus, Denmark
| |
Collapse
|
5
|
Faivre A, Bugarski M, Rinaldi A, Sakhi IB, Verissimo T, Legouis D, Rutkowski JM, Correia S, Kaminska M, Dalga D, Malpetti D, Cippa PE, de Seigneux S, Hall AM. Spatiotemporal Landscape of Kidney Tubular Responses to Glomerular Proteinuria. J Am Soc Nephrol 2024; 35:854-869. [PMID: 38652545 PMCID: PMC11230716 DOI: 10.1681/asn.0000000000000357] [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: 03/16/2023] [Accepted: 04/12/2024] [Indexed: 04/25/2024] Open
Abstract
Key Points Glomerular proteinuria induces large-scale changes in gene expression along the nephron. Increased protein uptake in the proximal tubule results in axial remodeling and injury. Increased protein delivery to the distal tubule causes dedifferentiation of the epithelium. Background Large increases in glomerular protein filtration induce major changes in body homeostasis and are associated with a higher risk of kidney functional decline and cardiovascular disease. We investigated how elevated protein exposure modifies the landscape of tubular function along the entire nephron, to understand the cellular changes that mediate these important clinical phenomena. Methods We conducted single-nucleus RNA sequencing, functional intravital imaging, and antibody staining to spatially map transport processes along the mouse kidney tubule. We then delineated how these were altered in a transgenic mouse model of inducible glomerular proteinuria (POD-ATTAC) at 7 and 28 days. Results Glomerular proteinuria activated large-scale and pleiotropic changes in gene expression in all major nephron sections. Extension of protein uptake from early (S1) to later (S2) parts of the proximal tubule initially triggered dramatic expansion of a hybrid S1/2 population, followed by injury and failed repair, with the cumulative effect of loss of canonical S2 functions. Proteinuria also induced acute injury in S3. Meanwhile, overflow of luminal proteins to the distal tubule caused transcriptional convergence between specialized regions and generalized dedifferentiation. Conclusions Proteinuria modulated cell signaling in tubular epithelia and caused distinct patterns of remodeling and injury in a segment-specific manner. Podcast This article contains a podcast at https://dts.podtrac.com/redirect.mp3/www.asn-online.org/media/podcast/JASN/2024_05_01_ASN0000000000000357.mp3
Collapse
Affiliation(s)
- Anna Faivre
- Department of Medicine and Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Milica Bugarski
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Anna Rinaldi
- Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Division of Nephrology, Department of Medicine, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Imene B. Sakhi
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Thomas Verissimo
- Department of Medicine and Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - David Legouis
- Department of Medicine and Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
- Division of Intensive Care, Department of Acute Medicine, University Hospital of Geneva, Geneva, Switzerland
| | | | - Sara Correia
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Monika Kaminska
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Delal Dalga
- Department of Medicine and Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Daniele Malpetti
- Istituto Dalle Molle di Studi sull'Intelligenza Artificiale (IDSIA), USI/SUPSI, Lugano, Switzerland
| | - Pietro E. Cippa
- Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Division of Nephrology, Department of Medicine, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Sophie de Seigneux
- Department of Medicine and Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
- Department of Medicine, Service of Nephrology, Geneva University Hospitals, Geneva, Switzerland
| | - Andrew M. Hall
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
- Department of Nephrology, University Hospital Zurich, Zurich, Switzerland
- Zurich Kidney Center, Zurich, Switzerland
| |
Collapse
|
6
|
Hoogstraten CA, Hoenderop JG, de Baaij JHF. Mitochondrial Dysfunction in Kidney Tubulopathies. Annu Rev Physiol 2024; 86:379-403. [PMID: 38012047 DOI: 10.1146/annurev-physiol-042222-025000] [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: 11/29/2023]
Abstract
Mitochondria play a key role in kidney physiology and pathology. They produce ATP to fuel energy-demanding water and solute reabsorption processes along the nephron. Moreover, mitochondria contribute to cellular health by the regulation of autophagy, (oxidative) stress responses, and apoptosis. Mitochondrial abundance is particularly high in cortical segments, including proximal and distal convoluted tubules. Dysfunction of the mitochondria has been described for tubulopathies such as Fanconi, Gitelman, and Bartter-like syndromes and renal tubular acidosis. In addition, mitochondrial cytopathies often affect renal (tubular) tissues, such as in Kearns-Sayre and Leigh syndromes. Nevertheless, the mechanisms by which mitochondrial dysfunction results in renal tubular diseases are only scarcely being explored. This review provides an overview of mitochondrial dysfunction in the development and progression of kidney tubulopathies. Furthermore, it emphasizes the need for further mechanistic investigations to identify links between mitochondrial function and renal electrolyte reabsorption.
Collapse
Affiliation(s)
- Charlotte A Hoogstraten
- Department of Medical Biosciences, Radboud University Medical Center, Nijmegen, The Netherlands;
| | - Joost G Hoenderop
- Department of Medical Biosciences, Radboud University Medical Center, Nijmegen, The Netherlands;
| | - Jeroen H F de Baaij
- Department of Medical Biosciences, Radboud University Medical Center, Nijmegen, The Netherlands;
| |
Collapse
|
7
|
Hsiao HY, Yen TH, Wu FY, Cheng CM, Liu JW, Fan YT, Huang JJ, Nien CY. Delivery and Transcriptome Assessment of an In Vitro Three-Dimensional Proximal Tubule Model Established by Human Kidney 2 Cells in Clinical Gelatin Sponges. Int J Mol Sci 2023; 24:15547. [PMID: 37958530 PMCID: PMC10650118 DOI: 10.3390/ijms242115547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/10/2023] [Accepted: 10/14/2023] [Indexed: 11/15/2023] Open
Abstract
The high prevalence of kidney diseases and the low identification rate of drug nephrotoxicity in preclinical studies reinforce the need for representative yet feasible renal models. Although in vitro cell-based models utilizing renal proximal tubules are widely used for kidney research, many proximal tubule cell (PTC) lines have been indicated to be less sensitive to nephrotoxins, mainly due to altered expression of transporters under a two-dimensional culture (2D) environment. Here, we selected HK-2 cells to establish a simplified three-dimensional (3D) model using gelatin sponges as scaffolds. In addition to cell viability and morphology, we conducted a comprehensive transcriptome comparison and correlation analysis of 2D and 3D cultured HK-2 cells to native human PTCs. Our 3D model displayed stable and long-term growth with a tubule-like morphology and demonstrated a more comparable gene expression profile to native human PTCs compared to the 2D model. Many missing or low expressions of major genes involved in PTC transport and metabolic processes were restored, which is crucial for successful nephrotoxicity prediction. Consequently, we established a cost-effective yet more representative model for in vivo PTC studies and presented a comprehensive transcriptome analysis for the systematic characterization of PTC lines.
Collapse
Affiliation(s)
- Hui-Yi Hsiao
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
- Center for Tissue Engineering, Linkuo Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan;
| | - Tzung-Hai Yen
- Department of Nephrology, Clinical Poison Center, Linkuo Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan;
- Department of Nephrology, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Fang-Yu Wu
- Department of Life Science, National Central University, Taoyuan 32001, Taiwan; (F.-Y.W.); (Y.-T.F.)
| | - Chao-Min Cheng
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 300193, Taiwan;
| | - Jia-Wei Liu
- Center for Tissue Engineering, Linkuo Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan;
| | - Yu-Ting Fan
- Department of Life Science, National Central University, Taoyuan 32001, Taiwan; (F.-Y.W.); (Y.-T.F.)
| | - Jung-Ju Huang
- Division of Reconstructive Microsurgery, Department of Plastic and Reconstructive Surgery, Linkou Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan;
| | - Chung-Yi Nien
- Department of Life Science, National Central University, Taoyuan 32001, Taiwan; (F.-Y.W.); (Y.-T.F.)
| |
Collapse
|
8
|
Rega LR, Janssens V, Graversen JH, Moestrup SK, Cairoli S, Goffredo BM, Nevo N, Courtoy GE, Jouret F, Antignac C, Emma F, Pierreux CE, Courtoy PJ. Dietary supplementation of cystinotic mice by lysine inhibits the megalin pathway and decreases kidney cystine content. Sci Rep 2023; 13:17276. [PMID: 37828038 PMCID: PMC10570359 DOI: 10.1038/s41598-023-43105-x] [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: 04/07/2023] [Accepted: 09/20/2023] [Indexed: 10/14/2023] Open
Abstract
Megalin/LRP2 is a major receptor supporting apical endocytosis in kidney proximal tubular cells. We have previously reported that kidney-specific perinatal ablation of the megalin gene in cystinotic mice, a model of nephropathic cystinosis, essentially blocks renal cystine accumulation and partially preserves kidney tissue integrity. Here, we examined whether inhibition of the megalin pathway in adult cystinotic mice by dietary supplementation (5x-fold vs control regular diet) with the dibasic amino-acids (dAAs), lysine or arginine, both of which are used to treat patients with other rare metabolic disorders, could also decrease renal cystine accumulation and protect cystinotic kidneys. Using surface plasmon resonance, we first showed that both dAAs compete for protein ligand binding to immobilized megalin in a concentration-dependent manner, with identical inhibition curves by L- and D-stereoisomers. In cystinotic mice, 2-month diets with 5x-L-lysine and 5x-L-arginine were overall well tolerated, while 5x-D-lysine induced strong polyuria but no weight loss. All diets induced a marked increase of dAA urinary excretion, most prominent under 5x-D-lysine, without sign of kidney insufficiency. Renal cystine accumulation was slowed down approx. twofold by L-dAAs, and totally suppressed by D-lysine. We conclude that prolonged dietary manipulation of the megalin pathway in kidneys is feasible, tolerable and can be effective in vivo.
Collapse
Affiliation(s)
- L R Rega
- Nephrology Research Unit, Translational Pediatrics and Clinical Genetics Research Area, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - V Janssens
- Cell Biology Unit, de Duve Institute and Louvain University Medical School, Brussels, Belgium
| | - J H Graversen
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - S K Moestrup
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - S Cairoli
- Division of Metabolic Diseases and Drug Biology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - B M Goffredo
- Division of Metabolic Diseases and Drug Biology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - N Nevo
- Laboratoire des Maladies Rénales Héréditaires, Inserm UMR 1163, Institut Imagine, Université Paris Cité, Paris, France
| | - G E Courtoy
- Imaging Platform (2IP), Institut de Recherche Expérimentale et Clinique, Louvain University Medical School, Brussels, Belgium
| | - F Jouret
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA), Cardiovascular Sciences, University of Liège, Liège, Belgium
| | - C Antignac
- Laboratoire des Maladies Rénales Héréditaires, Inserm UMR 1163, Institut Imagine, Université Paris Cité, Paris, France
| | - F Emma
- Nephrology Research Unit, Translational Pediatrics and Clinical Genetics Research Area, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - C E Pierreux
- Cell Biology Unit, de Duve Institute and Louvain University Medical School, Brussels, Belgium.
| | - P J Courtoy
- Cell Biology Unit, de Duve Institute and Louvain University Medical School, Brussels, Belgium.
| |
Collapse
|
9
|
Wagner CA, Unwin R, Lopez-Garcia SC, Kleta R, Bockenhauer D, Walsh S. The pathophysiology of distal renal tubular acidosis. Nat Rev Nephrol 2023; 19:384-400. [PMID: 37016093 DOI: 10.1038/s41581-023-00699-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2023] [Indexed: 04/06/2023]
Abstract
The kidneys have a central role in the control of acid-base homeostasis owing to bicarbonate reabsorption and production of ammonia and ammonium in the proximal tubule and active acid secretion along the collecting duct. Impaired acid excretion by the collecting duct system causes distal renal tubular acidosis (dRTA), which is characterized by the failure to acidify urine below pH 5.5. This defect originates from reduced function of acid-secretory type A intercalated cells. Inherited forms of dRTA are caused by variants in SLC4A1, ATP6V1B1, ATP6V0A4, FOXI1, WDR72 and probably in other genes that are yet to be discovered. Inheritance of dRTA follows autosomal-dominant and -recessive patterns. Acquired forms of dRTA are caused by various types of autoimmune diseases or adverse effects of some drugs. Incomplete dRTA is frequently found in patients with and without kidney stone disease. These patients fail to appropriately acidify their urine when challenged, suggesting that incomplete dRTA may represent an intermediate state in the spectrum of the ability to excrete acids. Unrecognized or insufficiently treated dRTA can cause rickets and failure to thrive in children, osteomalacia in adults, nephrolithiasis and nephrocalcinosis. Electrolyte disorders are also often present and poorly controlled dRTA can increase the risk of developing chronic kidney disease.
Collapse
Affiliation(s)
- Carsten A Wagner
- Institute of Physiology, University of Zurich, Zurich, Switzerland.
- Department of Renal Medicine, Royal Free Hospital, University College London, London, UK.
| | - Robert Unwin
- Department of Renal Medicine, Royal Free Hospital, University College London, London, UK
| | - Sergio C Lopez-Garcia
- Department of Renal Medicine, Royal Free Hospital, University College London, London, UK
- Department of Paediatric Nephrology, Great Ormond Street Hospital for Children, NHS Foundation Trust, London, UK
| | - Robert Kleta
- Department of Renal Medicine, Royal Free Hospital, University College London, London, UK
| | - Detlef Bockenhauer
- Department of Renal Medicine, Royal Free Hospital, University College London, London, UK
- Department of Paediatric Nephrology, Great Ormond Street Hospital for Children, NHS Foundation Trust, London, UK
| | - Stephen Walsh
- Department of Renal Medicine, Royal Free Hospital, University College London, London, UK
| |
Collapse
|
10
|
Goto S, Hosojima M, Kabasawa H, Saito A. The endocytosis receptor megalin: From bench to bedside. Int J Biochem Cell Biol 2023; 157:106393. [PMID: 36863658 DOI: 10.1016/j.biocel.2023.106393] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/27/2023] [Indexed: 03/04/2023]
Abstract
The large (∼600 kDa) endocytosis receptor megalin/low-density lipoprotein receptor-related protein 2 is highly expressed at the apical membrane of proximal tubular epithelial cells (PTECs). Megalin plays an important role in the endocytosis of various ligands via interactions with intracellular adaptor proteins, which mediate the trafficking of megalin in PTECs. Megalin mediates the retrieval of essential substances, including carrier-bound vitamins and elements, and impairment of the endocytic process may result in the loss of those substances. In addition, megalin reabsorbs nephrotoxic substances such as antimicrobial (colistin, vancomycin, and gentamicin) or anticancer (cisplatin) drugs and advanced glycation end product-modified or fatty acid-containing albumin. The megalin-mediated uptake of these nephrotoxic ligands causes metabolic overload in PTECs and leads to kidney injury. Blockade or suppression of the megalin-mediated endocytosis of nephrotoxic substances may represent a novel therapeutic strategy for drug-induced nephrotoxicity or metabolic kidney disease. Megalin reabsorbs urinary biomarker proteins such as albumin, α1-microglobulin, β2-microglobulin, and liver-type fatty acid-binding protein; thus, the above-mentioned megalin-targeted therapy may have an effect on the urinary excretion of these biomarkers. We have previously established a sandwich enzyme-linked immunosorbent assay to measure the ectodomain (A-megalin) and full-length (C-megalin) forms of urinary megalin using monoclonal antibodies against the amino- and carboxyl-terminals of megalin, respectively, and reported their clinical usefulness. In addition, there have been reports of patients with novel pathological anti-brush border autoantibodies targeting megalin in the kidney. Even with these breakthroughs in the characterization of megalin, a large number of issues remain to be addressed in future research.
Collapse
Affiliation(s)
- Sawako Goto
- Departments of Applied Molecular Medicine, Japan
| | - Michihiro Hosojima
- Departments of Clinical Nutrition Science, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata City, Niigata, Japan
| | - Hideyuki Kabasawa
- Departments of Clinical Nutrition Science, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata City, Niigata, Japan
| | | |
Collapse
|
11
|
Santos R, Bürgi M, Mateos JM, Luciani A, Loffing J. Too bright for 2 dimensions: recent progress in advanced 3-dimensional microscopy of the kidney. Kidney Int 2022; 102:1238-1246. [PMID: 35963448 DOI: 10.1016/j.kint.2022.06.031] [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: 03/19/2022] [Revised: 06/19/2022] [Accepted: 06/24/2022] [Indexed: 01/12/2023]
Abstract
The kidney is a structurally and functionally complex organ responsible for the control of water, ion, and other solute homeostasis. Moreover, the kidneys excrete metabolic waste products and produce hormones, such as renin and erythropoietin. The functional unit of the kidney is the nephron, which is composed by a serial arrangement of a filter unit called the renal corpuscle and several tubular segments that modulate the filtered fluid by reabsorption and secretion. Within each kidney, thousands of nephrons are closely intermingled and surrounded by an intricate network of blood vessels and various interstitial cell types, including fibroblasts and immune cells. This complex tissue architecture is essential for proper kidney function. In fact, kidney disease is often reflected or even caused by a derangement of the histologic structures. Frequently, kidney histology is studied using microscopic analysis of 2-dimensional tissue sections, which, however, misses important 3-dimensional spatial information. Reconstruction of serial sections tries to overcome this limitation, but is technically challenging, time-consuming, and often inherently linked to sectioning artifacts. In recent years, advances in tissue preparation (e.g., optical clearing) and new light- and electron-microscopic methods have provided novel avenues for 3-dimensional kidney imaging. Combined with novel machine-learning algorithms, these approaches offer unprecedented options for large-scale and automated analysis of kidney structure and function. This review provides a brief overview of these emerging imaging technologies and presents key examples of how these approaches are already used to study the normal and the diseased kidney.
Collapse
Affiliation(s)
- Rui Santos
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Max Bürgi
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - José María Mateos
- Centre for Microscopy and Image Analysis, University of Zurich, Zurich, Switzerland
| | - Alessandro Luciani
- Institute of Physiology, University of Zurich, Zurich, Switzerland; National Centre of Competence in Research "Kidney.CH," University of Zurich, Zurich, Switzerland
| | - Johannes Loffing
- Institute of Anatomy, University of Zurich, Zurich, Switzerland; National Centre of Competence in Research "Kidney.CH," University of Zurich, Zurich, Switzerland.
| |
Collapse
|
12
|
Rinschen MM, Harder JL, Carter-Timofte ME, Zanon Rodriguez L, Mirabelli C, Demir F, Kurmasheva N, Ramakrishnan SK, Kunke M, Tan Y, Billing A, Dahlke E, Larionov AA, Bechtel-Walz W, Aukschun U, Grabbe M, Nielsen R, Christensen EI, Kretzler M, Huber TB, Wobus CE, Olagnier D, Siuzdak G, Grahammer F, Theilig F. VPS34-dependent control of apical membrane function of proximal tubule cells and nutrient recovery by the kidney. Sci Signal 2022; 15:eabo7940. [PMID: 36445937 PMCID: PMC10350314 DOI: 10.1126/scisignal.abo7940] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
The lipid kinase VPS34 orchestrates autophagy, endocytosis, and metabolism and is implicated in cancer and metabolic disease. The proximal tubule in the kidney is a key metabolic organ that controls reabsorption of nutrients such as fatty acids, amino acids, sugars, and proteins. Here, by combining metabolomics, proteomics, and phosphoproteomics analyses with functional and superresolution imaging assays of mice with an inducible deficiency in proximal tubular cells, we revealed that VPS34 controlled the metabolome of the proximal tubule. In addition to inhibiting pinocytosis and autophagy, VPS34 depletion induced membrane exocytosis and reduced the abundance of the retromer complex necessary for proper membrane recycling and lipid retention, leading to a loss of fuel and biomass. Integration of omics data into a kidney cell metabolomic model demonstrated that VPS34 deficiency increased β-oxidation, reduced gluconeogenesis, and enhanced the use of glutamine for energy consumption. Furthermore, the omics datasets revealed that VPS34 depletion triggered an antiviral response that included a decrease in the abundance of apically localized virus receptors such as ACE2. VPS34 inhibition abrogated SARS-CoV-2 infection in human kidney organoids and cultured proximal tubule cells in a glutamine-dependent manner. Thus, our results demonstrate that VPS34 adjusts endocytosis, nutrient transport, autophagy, and antiviral responses in proximal tubule cells in the kidney.
Collapse
Affiliation(s)
- Markus M Rinschen
- Scripps Center for Metabolomics, Scripps Research, La Jolla, CA 92037, USA
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
- Department II of Internal Medicine and Center for Molecular Medicine, University of Cologne, 50937 Cologne, Germany
- Aarhus Institute for Advanced Studies, Aarhus University, 8000 Aarhus, Denmark
| | - Jennifer L Harder
- Division of Nephrology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | | | | | - Carmen Mirabelli
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Fatih Demir
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | | | | | - Madlen Kunke
- Department of Anatomy, Christian-Albrechts-University Kiel, 24118 Kiel, Germany
| | - Yifan Tan
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | - Anja Billing
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | - Eileen Dahlke
- Department of Anatomy, Christian-Albrechts-University Kiel, 24118 Kiel, Germany
| | - Alexey A Larionov
- Department of Medicine, University of Fribourg, 1700 Fribourg, Switzerland
| | - Wibke Bechtel-Walz
- IV Department of Medicine and Faculty of Medicine, University Medical Center Freiburg, 79110 Freiburg, Germany
| | - Ute Aukschun
- IV Department of Medicine and Faculty of Medicine, University Medical Center Freiburg, 79110 Freiburg, Germany
| | - Marlen Grabbe
- IV Department of Medicine and Faculty of Medicine, University Medical Center Freiburg, 79110 Freiburg, Germany
| | - Rikke Nielsen
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | | | - Matthias Kretzler
- Division of Nephrology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Tobias B Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Christiane E Wobus
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - David Olagnier
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | - Gary Siuzdak
- Scripps Center for Metabolomics, Scripps Research, La Jolla, CA 92037, USA
| | - Florian Grahammer
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Franziska Theilig
- Department of Anatomy, Christian-Albrechts-University Kiel, 24118 Kiel, Germany
- Department of Medicine, University of Fribourg, 1700 Fribourg, Switzerland
| |
Collapse
|
13
|
Polesel M, Kaminska M, Haenni D, Bugarski M, Schuh C, Jankovic N, Kaech A, Mateos JM, Berquez M, Hall AM. Spatiotemporal organisation of protein processing in the kidney. Nat Commun 2022; 13:5732. [PMID: 36175561 PMCID: PMC9522658 DOI: 10.1038/s41467-022-33469-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 09/13/2022] [Indexed: 11/09/2022] Open
Abstract
The kidney regulates plasma protein levels by eliminating them from the circulation. Proteins filtered by glomeruli are endocytosed and degraded in the proximal tubule and defects in this process result in tubular proteinuria, an important clinical biomarker. However, the spatiotemporal organization of renal protein metabolism in vivo was previously unclear. Here, using functional probes and intravital microscopy, we track the fate of filtered proteins in real time in living mice, and map specialized processing to tubular structures with singular value decomposition analysis and three-dimensional electron microscopy. We reveal that degradation of proteins requires sequential, coordinated activity of distinct tubular sub-segments, each adapted to specific tasks. Moreover, we leverage this approach to pinpoint the nature of endo-lysosomal disorders in disease models, and show that compensatory uptake in later regions of the proximal tubule limits urinary protein loss. This means that measurement of proteinuria likely underestimates severity of endocytotic defects in patients. Polesel et al. visualize plasma protein filtration, uptake and metabolism in the kidneys of living mice in real-time. They reveal coordinated activity of different specialized tubular segments, with major compensatory adaptations occurring in disease states.
Collapse
Affiliation(s)
| | - Monika Kaminska
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Dominik Haenni
- Center for Microscopy and Image Analysis, 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
| | - Nevena Jankovic
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Andres Kaech
- Center for Microscopy and Image Analysis, University of Zurich, Zurich, Switzerland
| | - Jose M Mateos
- Center for Microscopy and Image Analysis, University of Zurich, Zurich, Switzerland
| | - Marine Berquez
- Institute of Physiology, 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
|
14
|
Imenez Silva PH, Mohebbi N. Kidney metabolism and acid-base control: back to the basics. Pflugers Arch 2022; 474:919-934. [PMID: 35513635 PMCID: PMC9338915 DOI: 10.1007/s00424-022-02696-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 01/18/2023]
Abstract
Kidneys are central in the regulation of multiple physiological functions, such as removal of metabolic wastes and toxins, maintenance of electrolyte and fluid balance, and control of pH homeostasis. In addition, kidneys participate in systemic gluconeogenesis and in the production or activation of hormones. Acid-base conditions influence all these functions concomitantly. Healthy kidneys properly coordinate a series of physiological responses in the face of acute and chronic acid-base disorders. However, injured kidneys have a reduced capacity to adapt to such challenges. Chronic kidney disease patients are an example of individuals typically exposed to chronic and progressive metabolic acidosis. Their organisms undergo a series of alterations that brake large detrimental changes in the homeostasis of several parameters, but these alterations may also operate as further drivers of kidney damage. Acid-base disorders lead not only to changes in mechanisms involved in acid-base balance maintenance, but they also affect multiple other mechanisms tightly wired to it. In this review article, we explore the basic renal activities involved in the maintenance of acid-base balance and show how they are interconnected to cell energy metabolism and other important intracellular activities. These intertwined relationships have been investigated for more than a century, but a modern conceptual organization of these events is lacking. We propose that pH homeostasis indissociably interacts with central pathways that drive progression of chronic kidney disease, such as inflammation and metabolism, independent of etiology.
Collapse
Affiliation(s)
- Pedro Henrique Imenez Silva
- Institute of Physiology, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland.
- National Center of Competence in Research NCCR Kidney.CH, Zurich, Switzerland.
| | - Nilufar Mohebbi
- National Center of Competence in Research NCCR Kidney.CH, Zurich, Switzerland
- Praxis Und Dialysezentrum Zurich, Zurich, Switzerland
| |
Collapse
|
15
|
Hall AM, de Seigneux S. Metabolic mechanisms of acute proximal tubular injury. Pflugers Arch 2022; 474:813-827. [PMID: 35567641 PMCID: PMC9338906 DOI: 10.1007/s00424-022-02701-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 04/12/2022] [Accepted: 05/02/2022] [Indexed: 12/11/2022]
Abstract
Damage to the proximal tubule (PT) is the most frequent cause of acute kidney injury (AKI) in humans. Diagnostic and treatment options for AKI are currently limited, and a deeper understanding of pathogenic mechanisms at a cellular level is required to rectify this situation. Metabolism in the PT is complex and closely coupled to solute transport function. Recent studies have shown that major changes in PT metabolism occur during AKI and have highlighted some potential targets for intervention. However, translating these insights into effective new therapies still represents a substantial challenge. In this article, in addition to providing a brief overview of the current state of the field, we will highlight three emerging areas that we feel are worthy of greater attention. First, we will discuss the role of axial heterogeneity in cellular function along the PT in determining baseline susceptibility to different metabolic hits. Second, we will emphasize that elucidating insult specific pathogenic mechanisms will likely be critical in devising more personalized treatments for AKI. Finally, we will argue that uncovering links between tubular metabolism and whole-body homeostasis will identify new strategies to try to reduce the considerable morbidity and mortality associated with AKI. These concepts will be illustrated by examples of recent studies emanating from the authors' laboratories and performed under the auspices of the Swiss National Competence Center for Kidney Research (NCCR Kidney.ch).
Collapse
Affiliation(s)
- Andrew M Hall
- Institute of Anatomy, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland.
- Department of Nephrology, University Hospital Zurich, Zurich, Switzerland.
| | - Sophie de Seigneux
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
- Department of Medicine, Service of Nephrology, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
16
|
SGLT2 Inhibitors in Type 2 Diabetes Mellitus and Heart Failure-A Concise Review. J Clin Med 2022; 11:jcm11061470. [PMID: 35329796 PMCID: PMC8952302 DOI: 10.3390/jcm11061470] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 02/28/2022] [Accepted: 03/06/2022] [Indexed: 01/25/2023] Open
Abstract
The incidence of both diabetes mellitus type 2 and heart failure is rapidly growing, and the diseases often coexist. Sodium-glucose co-transporter 2 inhibitors (SGLT2i) are a new antidiabetic drug class that mediates epithelial glucose transport at the renal proximal tubules, inhibiting glucose absorption—resulting in glycosuria—and therefore improving glycemic control. Recent trials have proven that SGLT2i also improve cardiovascular and renal outcomes, including reduced cardiovascular mortality and fewer hospitalizations for heart failure. Reduced preload and afterload, improved vascular function, and changes in tissue sodium and calcium handling may also play a role. The expected paradigm shift in treatment strategies was reflected in the most recent 2021 guidelines published by the European Society of Cardiology, recommending dapagliflozin and empagliflozin as first-line treatment for heart failure patients with reduced ejection fraction. Moreover, the recent results of the EMPEROR-Preserved trial regarding empagliflozin give us hope that there is finally an effective treatment for patients with heart failure with preserved ejection fraction. This review aims to assess the efficacy and safety of these new anti-glycemic oral agents in the management of diabetic and heart failure patients.
Collapse
|
17
|
Edwards A, Long KR, Baty CJ, Shipman KE, Weisz OA. Modeling normal and nephrotic axial uptake of albumin and other filtered proteins along the proximal tubule. J Physiol 2022; 600:1933-1952. [PMID: 35178707 PMCID: PMC9012691 DOI: 10.1113/jp282885] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 02/10/2022] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS We used new and published data to develop a mathematical model that predicts the profile of albumin uptake in the mouse proximal tubule (PT) in normal and nephrotic states, and partially accounts for competitive inhibition of uptake by normally filtered and pathologic ligands. Three pathways, consisting of high-affinity uptake by cubilin receptors, low-affinity uptake by megalin receptors, and fluid phase uptake, contribute to the overall retrieval of filtered proteins. The axial profile and efficiency of protein uptake depend on the initial filtrate composition and the individual protein affinities for megalin and cubilin. Under normal conditions, the majority of albumin is retrieved in S1 but shifts to S2 under nephrotic conditions. Other proteins exhibit different uptake profiles. Our model explains how tubular proteinuria can occur despite a large excess in potential PT uptake capacity. ABSTRACT Recent studies indicate that filtered albumin is retrieved in the proximal tubule (PT) via three pathways: receptor-mediated endocytosis via cubilin (high affinity) and megalin (low affinity), and fluid-phase uptake. Expression of megalin is required to maintain all three pathways, making it challenging to determine their respective contributions. Moreover, uptake of filtered molecules varies between the sub-segments (S1, S2, and S3) that make up the PT. Here we used new and published data to develop a mathematical model that predicts the rates of albumin uptake in mouse PT sub-segments in normal and nephrotic states, and partially accounts for competition by β2-microglobulin (β2m) and Immunoglobulin G (IgG). Our simulations indicate that receptor-mediated, rather than fluid-phase uptake, accounts for the vast majority of ligand recovery. Our model predicts that ∼75% of normally filtered albumin is reabsorbed via cubilin; however, megalin-mediated uptake predominates under nephrotic conditions. Our results also suggest that ∼80% of albumin is normally recovered in S1, whereas nephrotic conditions or knockout of cubilin shifts the bulk of albumin uptake to S2. The model predicts β2m and IgG axial recovery profiles qualitatively similar to those of albumin under normal conditions. In contrast with albumin however, the bulk of IgG and β2m uptake still occurs in S1 under nephrotic conditions. Overall, our model provides a kinetic rationale for why tubular proteinuria can occur even though a large excess in potential PT uptake capacity exists, and suggests testable predictions to expand our understanding of the recovery profile of filtered proteins along the PT. Abstract figure legend. Data from mouse models and from cultured proximal tubule (PT) cells were used to create a mathematical model that predicts the uptake profile of albumin and other filtered ligands along the mouse PT in normal and nephrotic states. The distinct contributions of cubilin receptors (magenta), megalin receptors (green), and fluid phase uptake (blue) to total albumin retrieval (black) in S1, S2, and S3 subsegments of the PT are delineated. Under normal conditions, albumin is primarily recovered in the S1 segment by cubilin, whereas the majority is retrieved in S2 under nephrotic conditions. Other proteins exhibit strikingly different uptake profiles. Our model explains how the distribution and capacity of high-affinity and low-affinity uptake pathways enable uptake of albumin over a broad range of filtered concentrations, and how tubular proteinuria can occur despite a large excess in potential PT uptake capacity. Created with BioRender.com. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Aurélie Edwards
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
| | - Kimberly R Long
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Catherine J Baty
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Katherine E Shipman
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Ora A Weisz
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| |
Collapse
|
18
|
Hall AM, Trepiccione F, Unwin RJ. Drug toxicity in the proximal tubule: new models, methods and mechanisms. Pediatr Nephrol 2022; 37:973-982. [PMID: 34050397 PMCID: PMC9023418 DOI: 10.1007/s00467-021-05121-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 03/23/2021] [Accepted: 05/05/2021] [Indexed: 10/28/2022]
Abstract
The proximal tubule (PT) reabsorbs most of the glomerular filtrate and plays an important role in the uptake, metabolism and excretion of xenobiotics. Some therapeutic drugs are harmful to the PT, and resulting nephrotoxicity is thought to be responsible for approximately 1 in 6 of cases of children hospitalized with acute kidney injury (AKI). Clinically, PT dysfunction leads to urinary wasting of important solutes normally reabsorbed by this nephron segment, leading to systemic complications such as bone demineralization and a clinical scenario known as the renal Fanconi syndrome (RFS). While PT defects can be diagnosed using a combination of blood and urine markers, including urinary excretion of low molecular weight proteins (LMWP), standardized definitions of what constitutes clinically significant toxicity are lacking, and identifying which patients will go on to develop progressive loss of kidney function remains a major challenge. In addition, much of our understanding of cellular mechanisms of drug toxicity is still limited, partly due to the constraints of available cell and animal models. However, advances in new and more sophisticated in vitro models of the PT, along with the application of high-content analytical methods that can provide readouts more relevant to the clinical manifestations of nephrotoxicity, are beginning to extend our knowledge. Such technical progress should help in discovering new biomarkers that can better detect nephrotoxicity earlier and predict its long-term consequences, and herald a new era of more personalized medicine.
Collapse
Affiliation(s)
- Andrew M. Hall
- grid.7400.30000 0004 1937 0650Institute of Anatomy, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland ,grid.412004.30000 0004 0478 9977Department of Nephrology, University Hospital Zurich, Zurich, Switzerland
| | - Francesco Trepiccione
- grid.9841.40000 0001 2200 8888Department of Translational Medical Science, University of Campania ‘Luigi Vanvitelli’, Naples, Italy ,grid.428067.f0000 0004 4674 1402Biogem Research Institute, Ariano Irpino, Italy
| | - Robert J. Unwin
- grid.83440.3b0000000121901201Department of Renal Medicine, University College London, London, UK
| |
Collapse
|
19
|
Dzgoev SG. Effect of Vasopressin on Collagenase Activity in Rat Papillary Renal Cells. J EVOL BIOCHEM PHYS+ 2021. [DOI: 10.1134/s0022093021060028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
20
|
Weisz OA. Endocytic adaptation to functional demand by the kidney proximal tubule. J Physiol 2021; 599:3437-3446. [PMID: 34036593 DOI: 10.1113/jp281599] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 05/06/2021] [Indexed: 12/28/2022] Open
Abstract
The kidney proximal tubule (PT) efficiently recovers the low level of albumin and other proteins that normally escape the glomerular filtration barrier. Two large receptors, megalin and cubilin/amnionless (CUBAM), bind to and efficiently retrieve these predominantly low molecular-weight proteins via clathrin-mediated endocytosis. Studies in cell culture models suggest that PT cells may sense changes in shear stress to modulate recovery of filtered proteins in response to normal variations in filtration rate. Impairments in PT endocytic function lead to the excretion of filtered proteins into the urine (tubular proteinuria). Remarkably, when the glomerular filtration barrier is breached, the PT is able to recover excess albumin with a capacity that is orders of magnitude higher than normal. What mediates this excess capacity for albumin uptake under nephrotic conditions, and why doesn't it compensate to prevent tubular proteinuria? Here we propose an integrated new working model to describe the PT recovery of filtered proteins under normal and nephrotic states. We hypothesize that uptake via the fluid phase provides excess capacity to recover high concentrations of filtered proteins under nephrotic conditions. Further, concentration of tubular fluid along the tubule axis will enhance the efficiency of uptake in more distal regions of the PT. By contrast to cells where fluid phase and receptor-mediated uptake are independent pathways, expression of megalin is required to maintain apical endocytic pathway integrity and is essential for both uptake mechanisms. This model accounts for both the high-affinity and the high-capacity responses to filtration load in physiological and pathological states.
Collapse
Affiliation(s)
- Ora A Weisz
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| |
Collapse
|
21
|
Festa BP, Berquez M, Nieri D, Luciani A. Endolysosomal Disorders Affecting the Proximal Tubule of the Kidney: New Mechanistic Insights and Therapeutics. Rev Physiol Biochem Pharmacol 2021; 185:233-257. [PMID: 33649992 DOI: 10.1007/112_2020_57] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Epithelial cells that line the proximal tubule of the kidney rely on an intertwined ecosystem of vesicular membrane trafficking pathways to ensure the reabsorption of essential nutrients. To function effectively and to achieve homeostasis, these specialized cells require the sorting and recycling of a wide array of cell surface proteins within the endolysosomal network, including signaling receptors, nutrient transporters, ion channels, and polarity markers. The dysregulation of the endolysosomal system can lead to a generalized proximal tubule dysfunction, ultimately causing severe metabolic complications and kidney disease.In this chapter, we highlight the biological functions of the genes that code endolysosomal proteins from the perspective of understanding - and potentially reversing - the pathophysiology of endolysosomal disorders affecting the proximal tubule of the kidney. These insights might ultimately lead to potential treatments for currently intractable diseases and transform our ability to regulate kidney homeostasis and health.
Collapse
Affiliation(s)
- Beatrice Paola Festa
- Institute of Physiology, Mechanisms of Inherited Kidney Disorders Group, University of Zurich, Zurich, Switzerland
| | - Marine Berquez
- Institute of Physiology, Mechanisms of Inherited Kidney Disorders Group, University of Zurich, Zurich, Switzerland
| | - Daniela Nieri
- Institute of Physiology, Mechanisms of Inherited Kidney Disorders Group, University of Zurich, Zurich, Switzerland
| | - Alessandro Luciani
- Institute of Physiology, Mechanisms of Inherited Kidney Disorders Group, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
22
|
Edwards A, Christensen EI, Unwin RJ, Norden AGW. Predicting the protein composition of human urine in normal and pathological states: Quantitative description based on Dent1 disease (
CLCN5
mutation). J Physiol 2020; 599:323-341. [DOI: 10.1113/jp280740] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/16/2020] [Indexed: 01/25/2023] Open
Affiliation(s)
- Aurélie Edwards
- Department of Biomedical Engineering Boston University Boston MA USA
| | | | - Robert J. Unwin
- Department of Renal Medicine Royal Free Campus University College London London UK
| | - Anthony G. W. Norden
- Department of Renal Medicine Royal Free Campus University College London London UK
| |
Collapse
|
23
|
Park HJ, Fan Z, Bai Y, Ren Q, Rbaibi Y, Long KR, Gliozzi ML, Rittenhouse N, Locker JD, Poholek AC, Weisz OA. Transcriptional Programs Driving Shear Stress-Induced Differentiation of Kidney Proximal Tubule Cells in Culture. Front Physiol 2020; 11:587358. [PMID: 33192601 PMCID: PMC7662153 DOI: 10.3389/fphys.2020.587358] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 09/28/2020] [Indexed: 12/16/2022] Open
Abstract
Cultured cell models are an essential complement to dissecting kidney proximal tubule (PT) function in health and disease but do not fully recapitulate key features of this nephron segment. We recently determined that culture of opossum kidney (OK) cells under continuous orbital shear stress (OSS) significantly augments their morphological and functional resemblance to PTs in vivo. Here we used RNASeq to identify temporal transcriptional changes upon cell culture under static or shear stress conditions. Comparison of gene expression in cells cultured under static or OSS conditions with a database of rat nephron segment gene expression confirms that OK cells cultured under OSS are more similar to the PT in vivo compared with cells maintained under static conditions. Both improved oxygenation and mechanosensitive stimuli contribute to the enhanced differentiation in these cells, and we identified temporal changes in gene expression of known mechanosensitive targets. We observed changes in mRNA and protein levels of membrane trafficking components that may contribute to the enhanced endocytic capacity of cells cultured under OSS. Our data reveal pathways that may be critical for PT differentiation in vivo and validate the utility of this improved cell culture model as a tool to study PT function.
Collapse
Affiliation(s)
- Hyun Jung Park
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Zhenjiang Fan
- Department of Computer Science, University of Pittsburgh, Pittsburgh, PA, United States
| | - Yulong Bai
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Qidong Ren
- School of Medicine, Tsinghua University, Beijing, China
| | - Youssef Rbaibi
- Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Kimberly R Long
- Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Megan L Gliozzi
- Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Natalie Rittenhouse
- Division of Pediatric Rheumatology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Joseph D Locker
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Amanda C Poholek
- Division of Pediatric Rheumatology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Ora A Weisz
- Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
24
|
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
|
25
|
Genini A, Mohebbi N, Daryadel A, Bettoni C, Wagner CA. Adaptive response of the murine collecting duct to alkali loading. Pflugers Arch 2020; 472:1079-1092. [DOI: 10.1007/s00424-020-02423-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 05/31/2020] [Accepted: 06/19/2020] [Indexed: 01/14/2023]
|
26
|
Abstract
Acid-base balance is critical for normal life. Acute and chronic disturbances impact cellular energy metabolism, endocrine signaling, ion channel activity, neuronal activity, and cardiovascular functions such as cardiac contractility and vascular blood flow. Maintenance and adaptation of acid-base homeostasis are mostly controlled by respiration and kidney. The kidney contributes to acid-base balance by reabsorbing filtered bicarbonate, regenerating bicarbonate through ammoniagenesis and generation of protons, and by excreting acid. This review focuses on acid-base disorders caused by renal processes, both inherited and acquired. Distinct rare inherited monogenic diseases affecting acid-base handling in the proximal tubule and collecting duct have been identified. In the proximal tubule, mutations of solute carrier 4A4 (SLC4A4) (electrogenic Na+/HCO3--cotransporter Na+/bicarbonate cotransporter e1 [NBCe1]) and other genes such as CLCN5 (Cl-/H+-antiporter), SLC2A2 (GLUT2 glucose transporter), or EHHADH (enoyl-CoA, hydratase/3-hydroxyacyl CoA dehydrogenase) causing more generalized proximal tubule dysfunction can cause proximal renal tubular acidosis resulting from bicarbonate wasting and reduced ammoniagenesis. Mutations in adenosine triphosphate ATP6V1 (B1 H+-ATPase subunit), ATPV0A4 (a4 H+-ATPase subunit), SLC4A1 (anion exchanger 1), and FOXI1 (forkhead transcription factor) cause distal renal tubular acidosis type I. Carbonic anhydrase II mutations affect several nephron segments and give rise to a mixed proximal and distal phenotype. Finally, mutations in genes affecting aldosterone synthesis, signaling, or downstream targets can lead to hyperkalemic variants of renal tubular acidosis (type IV). More common forms of renal acidosis are found in patients with advanced stages of chronic kidney disease and are owing, at least in part, to a reduced capacity for ammoniagenesis.
Collapse
Affiliation(s)
- Carsten A Wagner
- Institute of Physiology, University of Zurich, Zurich, Switzerland; National Center for Competence in Research Kidney, Switzerland.
| | - Pedro H Imenez Silva
- Institute of Physiology, University of Zurich, Zurich, Switzerland; National Center for Competence in Research Kidney, Switzerland
| | - Soline Bourgeois
- Institute of Physiology, University of Zurich, Zurich, Switzerland; National Center for Competence in Research Kidney, Switzerland
| |
Collapse
|
27
|
Gottwald EM, Schuh CD, Drücker P, Haenni D, Pearson A, Ghazi S, Bugarski M, Polesel M, Duss M, Landau EM, Kaech A, Ziegler U, Lundby AKM, Lundby C, Dittrich PS, Hall AM. The iron chelator Deferasirox causes severe mitochondrial swelling without depolarization due to a specific effect on inner membrane permeability. Sci Rep 2020; 10:1577. [PMID: 32005861 PMCID: PMC6994599 DOI: 10.1038/s41598-020-58386-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 01/15/2020] [Indexed: 12/13/2022] Open
Abstract
The iron chelator Deferasirox (DFX) causes severe toxicity in patients for reasons that were previously unexplained. Here, using the kidney as a clinically relevant in vivo model for toxicity together with a broad range of experimental techniques, including live cell imaging and in vitro biophysical models, we show that DFX causes partial uncoupling and dramatic swelling of mitochondria, but without depolarization or opening of the mitochondrial permeability transition pore. This effect is explained by an increase in inner mitochondrial membrane (IMM) permeability to protons, but not small molecules. The movement of water into mitochondria is prevented by altering intracellular osmotic gradients. Other clinically used iron chelators do not produce mitochondrial swelling. Thus, DFX causes organ toxicity due to an off-target effect on the IMM, which has major adverse consequences for mitochondrial volume regulation.
Collapse
Affiliation(s)
| | - Claus D Schuh
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Patrick Drücker
- Department of Biosystems Science and Engineering, ETH Zurich, Zurich, Switzerland
| | - Dominik Haenni
- Institute of Anatomy, University of Zurich, Zurich, Switzerland.,Center for Microscopy and Image Analysis, University of Zurich, Zurich, Switzerland
| | - Adam Pearson
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Susan Ghazi
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Milica Bugarski
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | | | - Michael Duss
- Department of Chemistry, University of Zurich, Zurich, Switzerland
| | - Ehud M Landau
- Department of Chemistry, University of Zurich, Zurich, Switzerland
| | - Andres Kaech
- Center for Microscopy and Image Analysis, University of Zurich, Zurich, Switzerland
| | - Urs Ziegler
- Center for Microscopy and Image Analysis, University of Zurich, Zurich, Switzerland
| | - Anne K M Lundby
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Carsten Lundby
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Petra S Dittrich
- Department of Biosystems Science and Engineering, ETH Zurich, Zurich, Switzerland
| | - Andrew M Hall
- Institute of Anatomy, University of Zurich, Zurich, Switzerland. .,Department of Nephrology, University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
28
|
Polesel M, Hall AM. Axial differences in endocytosis along the kidney proximal tubule. Am J Physiol Renal Physiol 2019; 317:F1526-F1530. [DOI: 10.1152/ajprenal.00459.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The proximal tubule (PT) reabsorbs filtered proteins via receptor-mediated endocytosis to prevent energetically inefficient wasting in the urine. Recent intravital imaging studies have suggested that protein reabsorption occurs in early (S1) segments, which have a very high capacity. In contrast, uptake of fluid phase substrates also occurs in distal (S2) segments. In this article, we will review these findings and their implications for understanding integrated proximal tubular function, patterns of damage caused by endocytosed toxins, and the origins of proteinuria. We will also discuss whether compensatory downstream increases in protein uptake might occur in disease states, and the environmental factors that could drive these changes.
Collapse
Affiliation(s)
| | - Andrew M. Hall
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
- Department of Nephrology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
29
|
Janssens V, Gaide Chevronnay HP, Marie S, Vincent MF, Van Der Smissen P, Nevo N, Vainio S, Nielsen R, Christensen EI, Jouret F, Antignac C, Pierreux CE, Courtoy PJ. Protection of Cystinotic Mice by Kidney-Specific Megalin Ablation Supports an Endocytosis-Based Mechanism for Nephropathic Cystinosis Progression. J Am Soc Nephrol 2019; 30:2177-2190. [PMID: 31548351 DOI: 10.1681/asn.2019040371] [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] [Received: 04/12/2019] [Accepted: 07/31/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Deletions or inactivating mutations of the cystinosin gene CTNS lead to cystine accumulation and crystals at acidic pH in patients with nephropathic cystinosis, a rare lysosomal storage disease and the main cause of hereditary renal Fanconi syndrome. Early use of oral cysteamine to prevent cystine accumulation slows progression of nephropathic cystinosis but it is a demanding treatment and not a cure. The source of cystine accumulating in kidney proximal tubular cells and cystine's role in disease progression are unknown. METHODS To investigate whether receptor-mediated endocytosis by the megalin/LRP2 pathway of ultrafiltrated, disulfide-rich plasma proteins could be a source of cystine in proximal tubular cells, we used a mouse model of cystinosis in which conditional excision of floxed megalin/LRP2 alleles in proximal tubular cells of cystinotic mice was achieved by a Cre-LoxP strategy using Wnt4-CRE. We evaluated mice aged 6-9 months for kidney cystine levels and crystals; histopathology, with emphasis on swan-neck lesions and proximal-tubular-cell apoptosis and proliferation (turnover); and proximal-tubular-cell expression of the major apical transporters sodium-phosphate cotransporter 2A (NaPi-IIa) and sodium-glucose cotransporter-2 (SGLT-2). RESULTS Wnt4-CRE-driven megalin/LRP2 ablation in cystinotic mice efficiently blocked kidney cystine accumulation, thereby preventing lysosomal deformations and crystal deposition in proximal tubular cells. Swan-neck lesions were largely prevented and proximal-tubular-cell turnover was normalized. Apical expression of the two cotransporters was also preserved. CONCLUSIONS These observations support a key role of the megalin/LRP2 pathway in the progression of nephropathic cystinosis and provide a proof of concept for the pathway as a therapeutic target.
Collapse
Affiliation(s)
- Virginie Janssens
- Cell Biology Unit, de Duve Institute and Université Catholique de Louvain, Brussels, Belgium
| | | | - Sandrine Marie
- Biochemical Genetics, Academic Hospital Saint-Luc, Brussels, Belgium
| | | | - Patrick Van Der Smissen
- Cell Biology Unit, de Duve Institute and Université Catholique de Louvain, Brussels, Belgium
| | - Nathalie Nevo
- Laboratory of Hereditary Kidney Diseases, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Imagine Institute, Paris Descartes University, Paris, France
| | - Seppo Vainio
- Faculty of Biochemistry and Molecular Medicine, Laboratory of Developmental Biology, Oulu Center for Cell-Matrix Research, Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Rikke Nielsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; and
| | | | - François Jouret
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA), Cardiovascular Sciences, University of Liège, Liège, Belgium
| | - Corinne Antignac
- Laboratory of Hereditary Kidney Diseases, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Imagine Institute, Paris Descartes University, Paris, France
| | - Christophe E Pierreux
- Cell Biology Unit, de Duve Institute and Université Catholique de Louvain, Brussels, Belgium;
| | - Pierre J Courtoy
- Cell Biology Unit, de Duve Institute and Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
30
|
A single-nucleus RNA-sequencing pipeline to decipher the molecular anatomy and pathophysiology of human kidneys. Nat Commun 2019; 10:2832. [PMID: 31249312 PMCID: PMC6597610 DOI: 10.1038/s41467-019-10861-2] [Citation(s) in RCA: 194] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 05/31/2019] [Indexed: 12/17/2022] Open
Abstract
Defining cellular and molecular identities within the kidney is necessary to understand its organization and function in health and disease. Here we demonstrate a reproducible method with minimal artifacts for single-nucleus Droplet-based RNA sequencing (snDrop-Seq) that we use to resolve thirty distinct cell populations in human adult kidney. We define molecular transition states along more than ten nephron segments spanning two major kidney regions. We further delineate cell type-specific expression of genes associated with chronic kidney disease, diabetes and hypertension, providing insight into possible targeted therapies. This includes expression of a hypertension-associated mechano-sensory ion channel in mesangial cells, and identification of proximal tubule cell populations defined by pathogenic expression signatures. Our fully optimized, quality-controlled transcriptomic profiling pipeline constitutes a tool for the generation of healthy and diseased molecular atlases applicable to clinical samples. Single-cell studies in solid tissues remain challenging and have benefited from the development of single-nuclei RNA sequencing strategies. Here Lake et al. apply single-nucleus RNA sequencing to human kidney tissues to provide a comprehensive molecular and cellular atlas of the human kidney, with potential implications for the understanding of kidney physiology and disease.
Collapse
|
31
|
Establishment of renal proximal tubule cell lines derived from the kidney of p53 knockout mice. Cytotechnology 2019; 71:45-56. [PMID: 30603921 DOI: 10.1007/s10616-018-0261-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 09/19/2018] [Indexed: 12/14/2022] Open
Abstract
The human cell line HK-2 is most commonly used as a model of renal proximal tubular epithelial cells (PTECs) for various studies despite the absence or low expression of transporters characteristic of parental PTECs. In an effort to develop reliable PTEC models, several human cell lines have been newly established over the last decade. In contrast, reliable mouse PTEC models are still unavailable. In this study, we established immortalized renal cortex tubule cell lines derived from p53 knockout mice and evaluated various PTEC characteristics toward the development of reliable mouse PTEC models. Here, we focus on MuRTE61, one of 13 newly established clonal cell lines. Albumin uptake in MuRTE61 cells was verified by incubation with fluorescent dye-labeled albumin. RT-PCR confirmed the expression of efflux transporter genes characteristic of PTECs in the MuRTE61 cells. MuRTE61 cells exhibited high sensitivity to treatment with cisplatin, a nephrotoxic agent, accompanied by upregulated expression of the uptake transporter Slc22a2 gene. Furthermore, MuRTE61 cells consistently formed spheroids with a lumen and apicobasal polarity in three-dimensional Matrigel cultures. Apical brush border microvilli were also observed in the spheroids by transmission electron microscopy. These data validate that MuRTE61 can be characterized as a reliable mouse PTEC line. In future, detailed analysis of reliable mouse and human PTEC lines will provide an accurate extrapolation of results of experiments using mice and humans, and may help resolve apparent inconsistencies between mouse and human nephrotoxicity.
Collapse
|
32
|
Nawata CM, Pannabecker TL. Mammalian urine concentration: a review of renal medullary architecture and membrane transporters. J Comp Physiol B 2018; 188:899-918. [PMID: 29797052 PMCID: PMC6186196 DOI: 10.1007/s00360-018-1164-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 04/23/2018] [Accepted: 05/14/2018] [Indexed: 01/10/2023]
Abstract
Mammalian kidneys play an essential role in balancing internal water and salt concentrations. When water needs to be conserved, the renal medulla produces concentrated urine. Central to this process of urine concentration is an osmotic gradient that increases from the corticomedullary boundary to the inner medullary tip. How this gradient is generated and maintained has been the subject of study since the 1940s. While it is generally accepted that the outer medulla contributes to the gradient by means of an active process involving countercurrent multiplication, the source of the gradient in the inner medulla is unclear. The last two decades have witnessed advances in our understanding of the urine-concentrating mechanism. Details of medullary architecture and permeability properties of the tubules and vessels suggest that the functional and anatomic relationships of these structures may contribute to the osmotic gradient necessary to concentrate urine. Additionally, we are learning more about the membrane transporters involved and their regulatory mechanisms. The role of medullary architecture and membrane transporters in the mammalian urine-concentrating mechanism are the focus of this review.
Collapse
Affiliation(s)
- C Michele Nawata
- Department of Physiology, Banner University Medical Center, University of Arizona, 1501 N. Campbell Avenue, Tucson, AZ, 85724-5051, USA.
| | - Thomas L Pannabecker
- Department of Physiology, Banner University Medical Center, University of Arizona, 1501 N. Campbell Avenue, Tucson, AZ, 85724-5051, USA
| |
Collapse
|
33
|
Schuh CD, Polesel M, Platonova E, Haenni D, Gassama A, Tokonami N, Ghazi S, Bugarski M, Devuyst O, Ziegler U, Hall AM. Combined Structural and Functional Imaging of the Kidney Reveals Major Axial Differences in Proximal Tubule Endocytosis. J Am Soc Nephrol 2018; 29:2696-2712. [PMID: 30301861 DOI: 10.1681/asn.2018050522] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 09/18/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The kidney proximal convoluted tubule (PCT) reabsorbs filtered macromolecules via receptor-mediated endocytosis (RME) or nonspecific fluid phase endocytosis (FPE); endocytosis is also an entry route for disease-causing toxins. PCT cells express the protein ligand receptor megalin and have a highly developed endolysosomal system (ELS). Two PCT segments (S1 and S2) display subtle differences in cellular ultrastructure; whether these translate into differences in endocytotic function has been unknown. METHODS To investigate potential differences in endocytic function in S1 and S2, we quantified ELS protein expression in mouse kidney PCTs using real-time quantitative polymerase chain reaction and immunostaining. We also used multiphoton microscopy to visualize uptake of fluorescently labeled ligands in both living animals and tissue cleared using a modified CLARITY approach. RESULTS Uptake of proteins by RME occurs almost exclusively in S1. In contrast, dextran uptake by FPE takes place in both S1 and S2, suggesting that RME and FPE are discrete processes. Expression of key ELS proteins, but not megalin, showed a bimodal distribution; levels were far higher in S1, where intracellular distribution was also more polarized. Tissue clearing permitted imaging of ligand uptake at single-organelle resolution in large sections of kidney cortex. Analysis of segmented tubules confirmed that, compared with protein uptake, dextran uptake occurred over a much greater length of the PCT, although individual PCTs show marked heterogeneity in solute uptake length and three-dimensional morphology. CONCLUSIONS Striking axial differences in ligand uptake and ELS function exist along the PCT, independent of megalin expression. These differences have important implications for understanding topographic patterns of kidney diseases and the origins of proteinuria.
Collapse
Affiliation(s)
| | | | | | - Dominik Haenni
- Institute of Anatomy.,Center for Microscopy and Image Analysis, and
| | - Alkaly Gassama
- Institute of Physiology, University of Zurich, Zurich, Switzerland; and
| | - Natsuko Tokonami
- Institute of Physiology, University of Zurich, Zurich, Switzerland; and
| | | | | | - Olivier Devuyst
- Institute of Physiology, University of Zurich, Zurich, Switzerland; and
| | - Urs Ziegler
- Center for Microscopy and Image Analysis, and
| | - Andrew M Hall
- Institute of Anatomy, .,Department of Nephrology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
34
|
Vps34/PI3KC3 deletion in kidney proximal tubules impairs apical trafficking and blocks autophagic flux, causing a Fanconi-like syndrome and renal insufficiency. Sci Rep 2018; 8:14133. [PMID: 30237523 PMCID: PMC6148293 DOI: 10.1038/s41598-018-32389-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 07/01/2018] [Indexed: 12/21/2022] Open
Abstract
Kidney proximal tubular cells (PTCs) are highly specialized for ultrafiltrate reabsorption and serve as paradigm of apical epithelial differentiation. Vps34/PI3-kinase type III (PI3KC3) regulates endosomal dynamics, macroautophagy and lysosomal function. However, its in vivo role in PTCs has not been evaluated. Conditional deletion of Vps34/PI3KC3 in PTCs by Pax8-Cre resulted in early (P7) PTC dysfunction, manifested by Fanconi-like syndrome, followed by kidney failure (P14) and death. By confocal microscopy, Vps34∆/∆ PTCs showed preserved apico-basal specification (brush border, NHERF-1 versus Na+/K+-ATPase, ankyrin-G) but basal redistribution of late-endosomes/lysosomes (LAMP-1) and mis-localization to lysosomes of apical recycling endocytic receptors (megalin, cubilin) and apical non-recycling solute carriers (NaPi-IIa, SGLT-2). Defective endocytosis was confirmed by Texas-red-ovalbumin tracing and reduced albumin content. Disruption of Rab-11 and perinuclear galectin-3 compartments suggested mechanistic clues for defective receptor recycling and apical biosynthetic trafficking. p62-dependent autophagy was triggered yet abortive (p62 co-localization with LC3 but not LAMP-1) and PTCs became vacuolated. Impaired lysosomal positioning and blocked autophagy are known causes of cell stress. Thus, early trafficking defects show that Vps34 is a key in vivo component of molecular machineries governing apical vesicular trafficking, thus absorptive function in PTCs. Functional defects underline the essential role of Vps34 for PTC homeostasis and kidney survival.
Collapse
|
35
|
Bugarski M, Martins JR, Haenni D, Hall AM. Multiphoton imaging reveals axial differences in metabolic autofluorescence signals along the kidney proximal tubule. Am J Physiol Renal Physiol 2018; 315:F1613-F1625. [PMID: 30132348 DOI: 10.1152/ajprenal.00165.2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Kidney proximal tubules (PTs) are densely packed with mitochondria, and defects in mitochondrial function are implicated in many kidney diseases. However, little is known about intrinsic mitochondrial function within PT cells. Here, using intravital multiphoton microscopy and live slices of mouse kidney cortex, we show that autofluorescence signals provide important functional readouts of redox state and substrate metabolism and that there are striking axial differences in signals along the PT. Mitochondrial NAD(P)H intensity was similar in both PT segment (S)1 and S2 and was sensitive to changes in respiratory chain (RC) redox state, whereas cytosolic NAD(P)H intensity was significantly higher in S2. Mitochondrial NAD(P)H increased in response to lactate and butyrate but decreased in response to glutamine and glutamate. Cytosolic NAD(P)H was sensitive to lactate and pyruvate and decreased dramatically in S2 in response to inhibition of glucose metabolism. Mitochondrial flavoprotein (FP) intensity was markedly higher in S2 than in S1 but was insensitive to changes in RC redox state. Mitochondrial FP signal increased in response to palmitate but decreased in response to glutamine and glutamate. Fluorescence lifetime decays were similar in both S1 and S2, suggesting that intensity differences are explained by differences in abundance of the same molecular species. Expression levels of known fluorescent mitochondrial FPs were higher in S2 than S1. In summary, substantial metabolic information can be obtained in kidney tissue using a label-free live imaging approach, and our findings suggest that metabolism is tailored to the specialized functions of S1 and S2 PT segments.
Collapse
Affiliation(s)
- Milica Bugarski
- Institute of Anatomy, University of Zurich , Zurich , Switzerland
| | | | - Dominik Haenni
- Institute of Anatomy, University of Zurich , Zurich , Switzerland.,Center for Microscopy and Image Analysis, 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
|
36
|
Schäffers OJM, Hoenderop JGJ, Bindels RJM, de Baaij JHF. The rise and fall of novel renal magnesium transporters. Am J Physiol Renal Physiol 2018; 314:F1027-F1033. [DOI: 10.1152/ajprenal.00634.2017] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Body Mg2+ balance is finely regulated in the distal convoluted tubule (DCT), where a tight interplay among transcellular reabsorption, mitochondrial exchange, and basolateral extrusion takes place. In the last decades, several research groups have aimed to identify the molecular players in these processes. A multitude of proteins have been proposed to function as Mg2+ transporter in eukaryotes based on phylogenetic analysis, differential gene expression, and overexpression studies. However, functional evidence for many of these proteins is lacking. The aim of this review is, therefore, to critically reconsider all putative Mg2+ transporters and put their presumed function in context of the renal handling of Mg2+. Sufficient experimental evidence exists to acknowledge transient receptor potential melastatin (TRPM) 6 and TRPM7, solute carrier family 41 (SLC41) A1 and SLC41A3, and mitochondrial RNA splicing 2 (MRS2) as Mg2+ transporters. TRPM6/7 facilitate Mg2+ influx, SLC41A1 mediates Mg2+ extrusion, and MRS2 and SLC41A3 are implicated in mitochondrial Mg2+ homeostasis. These proteins are highly expressed in the DCT. The function of cyclin M (CNNM) proteins is still under debate. For the other proposed Mg2+ transporters including Mg2+ transporter subtype 1 (MagT1), nonimprinted in Prader-Willi/Angelman syndrome (NIPA), membrane Mg2+ transport (MMgT), Huntingtin-interacting protein 14 (HIP14), and ATP13A4, functional evidence is limited, or functions alternative to Mg2+ transport have been suggested. Additional characterization of their Mg2+ transport proficiency should be provided before further claims about their role as Mg2+ transporter can be made.
Collapse
Affiliation(s)
- Olivier J. M. Schäffers
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joost G. J. Hoenderop
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - René J. M. Bindels
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jeroen H. F. de Baaij
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
37
|
Pathare G, Dhayat NA, Mohebbi N, Wagner CA, Bobulescu IA, Moe OW, Fuster DG. Changes in V-ATPase subunits of human urinary exosomes reflect the renal response to acute acid/alkali loading and the defects in distal renal tubular acidosis. Kidney Int 2018; 93:871-880. [PMID: 29310826 DOI: 10.1016/j.kint.2017.10.018] [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] [Received: 06/28/2017] [Revised: 10/02/2017] [Accepted: 10/05/2017] [Indexed: 12/26/2022]
Abstract
In the kidney, final urinary acidification is achieved by V-ATPases expressed in type A intercalated cells. The B1 subunit of the V-ATPase is required for maximal urinary acidification, while the role of the homologous B2 subunit is less clear. Here we examined the effect of acute acid/alkali loading in humans on B1 and B2 subunit abundance in urinary exosomes in normal individuals and of acid loading in patients with distal renal tubular acidosis (dRTA). Specificities of B1 and B2 subunit antibodies were verified by yeast heterologously expressing human B1 and B2 subunits, and murine wild-type and B1-deleted kidney lysates. Acute ammonium chloride loading elicited systemic acidemia, a drop in urinary pH, and increased urinary ammonium excretion. Nadir urinary pH was achieved at four to five hours, and exosomal B1 abundance was significantly increased at two through six hours after ammonium chloride loading. After acute equimolar sodium bicarbonate loading, blood and urinary pH rose rapidly, with a concomitant reduction of exosomal B1 abundance within two hours, which remained lower throughout the test. In contrast, no change in exosomal B2 abundance was found following acid or alkali loading. In patients with inherited or acquired distal RTA, the urinary B1 subunit was extremely low or undetectable and did not respond to acid loading in urine, whereas no change in B2 subunit was found. Thus, both B1 and B2 subunits of the V-ATPase are detectable in human urinary exosomes, and acid and alkali loading or distal RTA cause changes in the B1 but not B2 subunit abundance in urinary exosomes.
Collapse
Affiliation(s)
- Ganesh Pathare
- Division of Nephrology and Hypertension, Bern University Hospital, University of Bern, Bern, Switzerland; Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland; National Centre of Competence in Research Transcure, University of Bern, Bern, Switzerland
| | - Nasser A Dhayat
- Division of Nephrology and Hypertension, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Nilufar Mohebbi
- Division of Nephrology, University Hospital Zurich, Zurich, Switzerland
| | - Carsten A Wagner
- Institute of Physiology, University of Zurich, Zurich, Switzerland; National Center for Competence in Research Kidney.CH, Zurich, Switzerland
| | - Ion A Bobulescu
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Orson W Moe
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Daniel G Fuster
- Division of Nephrology and Hypertension, Bern University Hospital, University of Bern, Bern, Switzerland; Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland; National Centre of Competence in Research Transcure, University of Bern, Bern, Switzerland.
| |
Collapse
|
38
|
Vedula EM, Alonso JL, Arnaout MA, Charest JL. A microfluidic renal proximal tubule with active reabsorptive function. PLoS One 2017; 12:e0184330. [PMID: 29020011 PMCID: PMC5636065 DOI: 10.1371/journal.pone.0184330] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 08/22/2017] [Indexed: 11/18/2022] Open
Abstract
In the kidney, the renal proximal tubule (PT) reabsorbs solutes into the peritubular capillaries through active transport. Here, we replicate this reabsorptive function in vitro by engineering a microfluidic PT. The microfluidic PT architecture comprises a porous membrane with user-defined submicron surface topography separating two microchannels representing a PT filtrate lumen and a peritubular capillary lumen. Human PT epithelial cells and microvascular endothelial cells in respective microchannels created a PT-like reabsorptive barrier. Co-culturing epithelial and endothelial cells in the microfluidic architecture enhanced viability, metabolic activity, and compactness of the epithelial layer. The resulting tissue expressed tight junctions, kidney-specific morphology, and polarized expression of kidney markers. The microfluidic PT actively performed sodium-coupled glucose transport, which could be modulated by administration of a sodium-transport inhibiting drug. The microfluidic PT reproduces human physiology at the cellular and tissue levels, and measurable tissue function which can quantify kidney pharmaceutical efficacy and toxicity.
Collapse
Affiliation(s)
- Else M. Vedula
- Biomedical Microsystems Group, Draper, Cambridge, Massachusetts, United States of America
| | - José Luis Alonso
- Leukocyte Biology and Inflammation Program, Department of Medicine, Nephrology Division, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, United States of America
| | - M. Amin Arnaout
- Leukocyte Biology and Inflammation Program, Department of Medicine, Nephrology Division, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, United States of America
- * E-mail: (JLC); (MAA)
| | - Joseph L. Charest
- Biomedical Microsystems Group, Draper, Cambridge, Massachusetts, United States of America
- * E-mail: (JLC); (MAA)
| |
Collapse
|
39
|
Dibb R, Xie L, Wei H, Liu C. Magnetic susceptibility anisotropy outside the central nervous system. NMR IN BIOMEDICINE 2017; 30:10.1002/nbm.3544. [PMID: 27199082 PMCID: PMC5112155 DOI: 10.1002/nbm.3544] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 03/29/2016] [Accepted: 03/30/2016] [Indexed: 06/01/2023]
Abstract
Magnetic-susceptibility-based MRI has made important contributions to the characterization of tissue microstructure, chemical composition, and organ function. This has motivated a number of studies to explore the link between microstructure and susceptibility in organs and tissues throughout the body, including the kidney, heart, and connective tissue. These organs and tissues have anisotropic magnetic susceptibility properties and cellular organizations that are distinct from the lipid organization of myelin in the brain. For instance, anisotropy is traced to the epithelial lipid orientation in the kidney, the myofilament proteins in the heart, and the collagen fibrils in the knee cartilage. The magnetic susceptibility properties of these and other tissues are quantified using specific MRI tools: susceptibility tensor imaging (STI), quantitative susceptibility mapping (QSM), and individual QSM measurements with respect to tubular and filament directions determined from diffusion tensor imaging. These techniques provide complementary and supplementary information to that produced by traditional MRI methods. In the kidney, STI can track tubules in all layers including the cortex, outer medulla, and inner medulla. In the heart, STI detected myofibers throughout the myocardium. QSM in the knee revealed three unique layers in articular cartilage by exploiting the anisotropic susceptibility features of collagen. While QSM and STI are promising tools to study tissue susceptibility, certain technical challenges must be overcome in order to realize routine clinical use. This paper reviews essential experimental findings of susceptibility anisotropy in the body, the underlying mechanisms, and the associated MRI methodologies. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Russell Dibb
- Center for In Vivo Microscopy, Department of Radiology, Duke University Medical Center, Durham, North Carolina 27710
| | - Luke Xie
- Center for In Vivo Microscopy, Department of Radiology, Duke University Medical Center, Durham, North Carolina 27710
- Utah Center for Advanced Imaging Research, Department of Radiology, University of Utah, Salt Lake City, Utah 84108
| | - Hongjiang Wei
- Brain Imaging and Analysis Center, Duke University Medical Center, Durham, North Carolina, 27710
| | - Chunlei Liu
- Center for In Vivo Microscopy, Department of Radiology, Duke University Medical Center, Durham, North Carolina 27710
- Brain Imaging and Analysis Center, Duke University Medical Center, Durham, North Carolina, 27710
| |
Collapse
|
40
|
Bland SK, Schmiedt CW, Clark ME, DeLay J, Bienzle D. Expression of Kidney Injury Molecule-1 in Healthy and Diseased Feline Kidney Tissue. Vet Pathol 2017; 54:490-510. [DOI: 10.1177/0300985817690213] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Sensitive markers to detect acute kidney injury (AKI) in cats are lacking. Kidney injury molecule-1 (KIM-1) is a promising marker of acute tubular injury in humans, and sequence and structure of feline KIM-1 have been determined. KIM-1 is shed into urine of cats with natural AKI. The objectives of this study were to characterize temporal and cellular expression of KIM-1 in kidneys from cats without and with experimental and natural AKI using histopathology and immunohistochemistry. Tissue sections from 8 cats without kidney disease, 3 to 4 cats with experimentally induced AKI on each day 1, 3, 6, and 12 after unilateral ischemia/reperfusion, and 9 cats with natural AKI were assessed. In sections from cats without kidney disease, patterns of periodic acid–Schiff and aquaporin-1 staining allowed identification of 3 distinct segments of the proximal tubule. KIM-1 staining was absent in segments 1 (S1) and S2, and faint in S3. Injury of S3 in cats with experimental and natural AKI was characterized by cell loss and necrosis, and remaining intact cells had cytoplasmic blebs and reduced brush borders. In experimental AKI, intensity of KIM-1 expression increased in proportion to the severity of injury and was consistently present in S3 but only transiently in other segments. Vimentin was absent in proximal tubules of healthy cats but expressed in injured S3. These findings indicate that S3 is the proximal tubular segment most susceptible to ischemic injury and that KIM-1 is a sensitive tissue indicator of AKI in cats.
Collapse
Affiliation(s)
- S. K. Bland
- Department of Pathobiology, University of Guelph, Guelph, Ontario, Canada
| | - C. W. Schmiedt
- Department of Small Animal Medicine and Surgery, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - M. E. Clark
- Department of Pathobiology, University of Guelph, Guelph, Ontario, Canada
| | - J. DeLay
- Animal Health Laboratory, University of Guelph, Guelph, Ontario, Canada
| | - D. Bienzle
- Department of Pathobiology, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
41
|
Daryadel A, Haubitz M, Figueiredo M, Steubl D, Roos M, Mäder A, Hettwer S, Wagner CA. The C-Terminal Fragment of Agrin (CAF), a Novel Marker of Renal Function, Is Filtered by the Kidney and Reabsorbed by the Proximal Tubule. PLoS One 2016; 11:e0157905. [PMID: 27380275 PMCID: PMC4933355 DOI: 10.1371/journal.pone.0157905] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 06/07/2016] [Indexed: 02/07/2023] Open
Abstract
Agrin, a multidomain proteoglycan and neurotrypsin, a neuronal serine protease, are important for forming (neuromuscular) synapses. Proteolytical activity of neurotrypsin produces a C-terminal fragment of agrin, termed CAF, of approximately 22 kDA molecular size which also circulates in blood. The presence of CAF in urine suggests either glomerular filtration or secretion into urine. Blood levels of CAF have been identified as a potential novel marker of kidney function. Here we describe that several nephron segments in the mouse kidney express agrin and neutrotrypsin in addition to the localization of both protein in the glomerulum. Agrin mRNA and protein was detected in almost all nephron segments and mRNA abundance was highest in the inner medullary collecting duct. Neurotrypsin mRNA was mostly detected in the thick ascending limb of the loop of Henle, the distal convoluted tubule, and the inner medullary collecting duct. Moreover, we show that the proximal tubule absorbs injected recombinant CAF by a process shared with receptor-mediated and fluid phase endocytosis. Co-injection of CAF with recombinant human transferrin, a substrate of the receptor-mediated endocytic pathway as well as with FITC-labelled dextran (10 kDa), a marker of fluid phase endocytosis, showed partial colocalization of CAF with both markers. Further colocalization of CAF with the lysosomal marker cathepsin B suggested degradation of CAF by the lysosome in the proximal tubule. Thus, the murine kidney expresses agrin and neurotrypsin in nephron segments beyond the glomerulum. CAF is filtered by the glomerulum and is reabsorbed by endocytosis by the proximal tubule. Thus, impaired kidney function could impair glomerular clearance of CAF and thereby increase circulating CAF levels.
Collapse
Affiliation(s)
- Arezoo Daryadel
- Institute of Physiology and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | | | - Marta Figueiredo
- Institute of Physiology and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Dominik Steubl
- Department of Nephrology, Klinikum rechts der Isar, Munich, Germany
| | - Marcel Roos
- Department of Nephrology, Klinikum rechts der Isar, Munich, Germany
| | | | | | - Carsten A. Wagner
- Institute of Physiology and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
- * E-mail:
| |
Collapse
|
42
|
|
43
|
Daryadel A, Bourgeois S, Figueiredo MFL, Gomes Moreira A, Kampik NB, Oberli L, Mohebbi N, Lu X, Meima ME, Danser AHJ, Wagner CA. Colocalization of the (Pro)renin Receptor/Atp6ap2 with H+-ATPases in Mouse Kidney but Prorenin Does Not Acutely Regulate Intercalated Cell H+-ATPase Activity. PLoS One 2016; 11:e0147831. [PMID: 26824839 PMCID: PMC4732657 DOI: 10.1371/journal.pone.0147831] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 01/08/2016] [Indexed: 12/11/2022] Open
Abstract
The (Pro)renin receptor (P)RR/Atp6ap2 is a cell surface protein capable of binding and non-proteolytically activate prorenin. Additionally, (P)RR is associated with H+-ATPases and alternative functions in H+-ATPase regulation as well as in Wnt signalling have been reported. Kidneys express very high levels of H+-ATPases which are involved in multiple functions such as endocytosis, membrane protein recycling as well as urinary acidification, bicarbonate reabsorption, and salt absorption. Here, we wanted to localize the (P)RR/Atp6ap2 along the murine nephron, exmaine whether the (P)RR/Atp6ap2 is coregulated with other H+-ATPase subunits, and whether acute stimulation of the (P)RR/Atp6ap2 with prorenin regulates H+-ATPase activity in intercalated cells in freshly isolated collecting ducts. We localized (P)PR/Atp6ap2 along the murine nephron by qPCR and immunohistochemistry. (P)RR/Atp6ap2 mRNA was detected in all nephron segments with highest levels in the collecting system coinciding with H+-ATPases. Further experiments demonstrated expression at the brush border membrane of proximal tubules and in all types of intercalated cells colocalizing with H+-ATPases. In mice treated with NH4Cl, NaHCO3, KHCO3, NaCl, or the mineralocorticoid DOCA for 7 days, (P)RR/Atp6ap2 and H+-ATPase subunits were regulated but not co-regulated at protein and mRNA levels. Immunolocalization in kidneys from control, NH4Cl or NaHCO3 treated mice demonstrated always colocalization of PRR/Atp6ap2 with H+-ATPase subunits at the brush border membrane of proximal tubules, the apical pole of type A intercalated cells, and at basolateral and/or apical membranes of non-type A intercalated cells. Microperfusion of isolated cortical collecting ducts and luminal application of prorenin did not acutely stimulate H+-ATPase activity. However, incubation of isolated collecting ducts with prorenin non-significantly increased ERK1/2 phosphorylation. Our results suggest that the PRR/Atp6ap2 may form a complex with H+-ATPases in proximal tubule and intercalated cells but that prorenin has no acute effect on H+-ATPase activity in intercalated cells.
Collapse
MESH Headings
- Ammonium Chloride/pharmacology
- Animals
- Anion Transport Proteins/genetics
- Anion Transport Proteins/metabolism
- Aquaporin 2/genetics
- Aquaporin 2/metabolism
- Cell Membrane/drug effects
- Cell Membrane/metabolism
- Dogs
- Gene Expression Regulation
- Kidney Cortex/cytology
- Kidney Cortex/drug effects
- Kidney Cortex/metabolism
- Kidney Medulla/cytology
- Kidney Medulla/drug effects
- Kidney Medulla/metabolism
- Kidney Tubules, Collecting/cytology
- Kidney Tubules, Collecting/drug effects
- Kidney Tubules, Collecting/metabolism
- Kidney Tubules, Proximal/cytology
- Kidney Tubules, Proximal/drug effects
- Kidney Tubules, Proximal/metabolism
- Madin Darby Canine Kidney Cells
- Male
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/metabolism
- Mice
- Mice, Inbred C57BL
- Proton-Translocating ATPases/genetics
- Proton-Translocating ATPases/metabolism
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Renin/pharmacology
- Renin-Angiotensin System/drug effects
- Signal Transduction
- Sodium Bicarbonate/pharmacology
- Sodium Chloride/pharmacology
- Sodium-Phosphate Cotransporter Proteins, Type IIa/genetics
- Sodium-Phosphate Cotransporter Proteins, Type IIa/metabolism
- Solute Carrier Family 12, Member 1/genetics
- Solute Carrier Family 12, Member 1/metabolism
- Solute Carrier Family 12, Member 3/genetics
- Solute Carrier Family 12, Member 3/metabolism
- Sulfate Transporters
Collapse
Affiliation(s)
- Arezoo Daryadel
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Soline Bourgeois
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | | | | | - Nicole B. Kampik
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Lisa Oberli
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Nilufar Mohebbi
- Institute of Physiology, University of Zurich, Zurich, Switzerland
- Divison of Nephrology, University Hospital Zurich, Zurich, Switzerland
| | - Xifeng Lu
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Marcel E. Meima
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - A. H. Jan Danser
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Carsten A. Wagner
- Institute of Physiology, University of Zurich, Zurich, Switzerland
- * E-mail:
| |
Collapse
|
44
|
Penton D, Czogalla J, Loffing J. Dietary potassium and the renal control of salt balance and blood pressure. Pflugers Arch 2015; 467:513-30. [PMID: 25559844 DOI: 10.1007/s00424-014-1673-1] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 12/10/2014] [Accepted: 12/11/2014] [Indexed: 01/09/2023]
Abstract
Dietary potassium (K(+)) intake has antihypertensive effects, prevents strokes, and improves cardiovascular outcomes. The underlying mechanism for these beneficial effects of high K(+) diets may include vasodilation, enhanced urine flow, reduced renal renin release, and negative sodium (Na(+)) balance. Indeed, several studies demonstrate that dietary K(+) intake induces renal Na(+) loss despite elevated plasma aldosterone. This review briefly highlights the epidemiological and experimental evidences for the effects of dietary K(+) on arterial blood pressure. It discusses the pivotal role of the renal distal tubule for the regulation of urinary K(+) and Na(+) excretion and blood pressure and highlights that it depends on the coordinated interaction of different nephron portions, epithelial cell types, and various ion channels, transporters, and ATPases. Moreover, we discuss the relevance of aldosterone and aldosterone-independent factors in mediating the effects of an altered K(+) intake on renal K(+) and Na(+) handling. Particular focus is given to findings suggesting that an aldosterone-independent downregulation of the thiazide-sensitive NaCl cotransporter significantly contributes to the natriuretic and antihypertensive effect of a K(+)-rich diet. Last but not least, we refer to the complex signaling pathways enabling the kidney to adapt its function to the homeostatic needs in response to an altered K(+) intake. Future work will have to further address the underlying cellular and molecular mechanism and to elucidate, among others, how an altered dietary K(+) intake is sensed and how this signal is transmitted to the different epithelial cells lining the distal tubule.
Collapse
Affiliation(s)
- David Penton
- Institute of Anatomy, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | | | | |
Collapse
|
45
|
|
46
|
Xie L, Dibb R, Cofer GP, Li W, Nicholls PJ, Johnson GA, Liu C. Susceptibility tensor imaging of the kidney and its microstructural underpinnings. Magn Reson Med 2014; 73:1270-81. [PMID: 24700637 DOI: 10.1002/mrm.25219] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 02/14/2014] [Accepted: 02/19/2014] [Indexed: 12/14/2022]
Abstract
PURPOSE The purpose of this study was to determine whether susceptibility tensor imaging (STI) could overcome limitations of current techniques to detect tubules throughout the kidney. METHODS Normal mouse kidneys (n = 4) were imaged at 9.4T using a three-dimensional gradient multi-echo sequence (55-micron isotropic resolution). Phase images from 12 orientations were obtained to compute the susceptibility tensor. Diffusion tensor imaging (DTI) with 12 encoding directions was compared with STI. Tractography was performed to visualize and track the course of tubules with DTI and STI. Confocal microscopy was used to identify which tubular segments of the nephron were detected by DTI and STI. RESULTS Diffusion anisotropy was limited to the inner medulla of the kidney. DTI did not find a significant number of coherent tubular tracks in the outer medulla or cortex. With STI, we found strong susceptibility anisotropy and many tracks in the inner and outer medulla and in limited areas of the cortex. CONCLUSION STI was able to track tubules throughout the kidney, whereas DTI was limited to the inner medulla. STI provides a novel contrast mechanism related to local tubule microstructure and may offer a powerful method to study the nephron.
Collapse
Affiliation(s)
- Luke Xie
- Center for In Vivo Microscopy, Department of Radiology, Duke University Medical Center, Durham, North Carolina, USA; Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Gaide Chevronnay HP, Janssens V, Van Der Smissen P, N'Kuli F, Nevo N, Guiot Y, Levtchenko E, Marbaix E, Pierreux CE, Cherqui S, Antignac C, Courtoy PJ. Time course of pathogenic and adaptation mechanisms in cystinotic mouse kidneys. J Am Soc Nephrol 2014; 25:1256-69. [PMID: 24525030 DOI: 10.1681/asn.2013060598] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Cystinosis, a main cause of Fanconi syndrome, is reproduced in congenic C57BL/6 cystinosin knockout (KO) mice. To identify the sequence of pathogenic and adaptation mechanisms of nephropathic cystinosis, we defined the onset of Fanconi syndrome in KO mice between 3 and 6 months of age and analyzed the correlation with structural and functional changes in proximal tubular cells (PTCs), with focus on endocytosis of ultrafiltrated disulfide-rich proteins as a key source of cystine. Despite considerable variation between mice at the same age, typical event sequences were delineated. At the cellular level, amorphous lysosomal inclusions preceded cystine crystals and eventual atrophy without crystals. At the nephron level, lesions started at the glomerulotubular junction and then extended distally. In situ hybridization and immunofluorescence revealed progressive loss of expression of megalin, cubilin, sodium-glucose cotransporter 2, and type IIa sodium-dependent phosphate cotransporter, suggesting apical dedifferentiation accounting for Fanconi syndrome before atrophy. Injection of labeled proteins revealed that defective endocytosis in S1 PTCs led to partial compensatory uptake by S3 PTCs, suggesting displacement of endocytic load and injury by disulfide-rich cargo. Increased PTC apoptosis allowed luminal shedding of cystine crystals and was partially compensated for by tubular proliferation. We conclude that lysosomal storage triggered by soluble cystine accumulation induces apical PTC dedifferentiation, which causes transfer of the harmful load of disulfide-rich proteins to more distal cells, possibly explaining longitudinal progression of swan-neck lesions. Furthermore, our results suggest that subsequent adaptation mechanisms include lysosomal clearance of free and crystalline cystine into urine and ongoing tissue repair.
Collapse
Affiliation(s)
| | - Virginie Janssens
- Cell Biology Unit, de Duve Institute and Université Catholique de Louvain, Brussels, Belgium
| | - Patrick Van Der Smissen
- Cell Biology Unit, de Duve Institute and Université Catholique de Louvain, Brussels, Belgium
| | - Francisca N'Kuli
- Cell Biology Unit, de Duve Institute and Université Catholique de Louvain, Brussels, Belgium
| | - Nathalie Nevo
- Inserm, U574, Hôpital Necker-Enfants Malades and Université Paris Descartes, Sorbonne Paris Cité, Institut Imagine, Paris, France
| | - Yves Guiot
- Pathology Department, Saint-Luc University Clinics, Brussels, Belgium
| | - Elena Levtchenko
- Department of Pediatric Nephrology, University Hospitals Leuven, Leuven, Belgium; and
| | - Etienne Marbaix
- Cell Biology Unit, de Duve Institute and Université Catholique de Louvain, Brussels, Belgium; Pathology Department, Saint-Luc University Clinics, Brussels, Belgium
| | - Christophe E Pierreux
- Cell Biology Unit, de Duve Institute and Université Catholique de Louvain, Brussels, Belgium
| | - Stéphanie Cherqui
- Department of Pediatrics, Division of Genetics, University of California, San Diego, California
| | - Corinne Antignac
- Inserm, U574, Hôpital Necker-Enfants Malades and Université Paris Descartes, Sorbonne Paris Cité, Institut Imagine, Paris, France
| | - Pierre J Courtoy
- Cell Biology Unit, de Duve Institute and Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
48
|
Christensen EI, Grann B, Kristoffersen IB, Skriver E, Thomsen JS, Andreasen A. Three-dimensional reconstruction of the rat nephron. Am J Physiol Renal Physiol 2014; 306:F664-71. [PMID: 24477686 DOI: 10.1152/ajprenal.00522.2013] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
This study gives a three-dimensional (3D) structural analysis of rat nephrons and their connections to collecting ducts. Approximately 4,500 2.5-μm-thick serial sections from the renal surface to the papillary tip were obtained from each of 3 kidneys of Wistar rats. Digital images were recorded and aligned into three image stacks and traced from image to image. Short-loop nephrons (SLNs), long-loop nephrons (LLNs), and collecting ducts (CDs) were reconstructed in 3D. We identified a well-defined boundary between the outer stripe and the inner stripe of the outer medulla corresponding to the transition of descending thick limbs to descending thin limbs and between the inner stripe and the inner medulla, i.e., the transition of ascending thin limbs into ascending thick limbs of LLNs. In all nephrons, a mosaic pattern of proximal tubule (PT) cells and descending thin limb (DTL) cells was observed at the transition between the PT and the DTL. The course of the LLNs revealed tortuous proximal "straight" tubules and winding of the DTLs within the outer half of the inner stripe. The localization of loop bends of SLNs in the inner stripe of the outer medulla and the bends of LLNs in the inner medulla reflected the localization of their glomeruli; i.e., the deeper the glomerulus, the deeper the bend. Each CD drained approximately three to six nephrons with a different pattern than previously established in mice. This information will provide a basis for evaluation of structural changes within nephrons as a result of physiological or pharmaceutical intervention.
Collapse
Affiliation(s)
- Erik I Christensen
- Dept. of Biomedicine, Anatomy Section of Cell Biology, Univ. of Aarhus, DK-8000 Aarhus C, Denmark.
| | | | | | | | | | | |
Collapse
|
49
|
Christensen EI, Birn H, Storm T, Weyer K, Nielsen R. Endocytic Receptors in the Renal Proximal Tubule. Physiology (Bethesda) 2012; 27:223-36. [DOI: 10.1152/physiol.00022.2012] [Citation(s) in RCA: 170] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Protein reabsorption is a predominant feature of the renal proximal tubule. Animal studies show that the ability to rescue plasma proteins relies on the endocytic receptors megalin and cubilin. Recently, studies of patients with syndromes caused by dysfunctional receptors have supported the importance of these for protein clearance of human ultrafiltrate. This review focuses on the molecular biology and physiology of the receptors and their involvement in renal pathological conditions.
Collapse
Affiliation(s)
- Erik I. Christensen
- Department of Biomedicine, Faculty of Health Sciences, Aarhus University, Aarhus, Denmark
| | - Henrik Birn
- Department of Biomedicine, Faculty of Health Sciences, Aarhus University, Aarhus, Denmark
| | - Tina Storm
- Department of Biomedicine, Faculty of Health Sciences, Aarhus University, Aarhus, Denmark
| | - Kathrin Weyer
- Department of Biomedicine, Faculty of Health Sciences, Aarhus University, Aarhus, Denmark
| | - Rikke Nielsen
- Department of Biomedicine, Faculty of Health Sciences, Aarhus University, Aarhus, Denmark
| |
Collapse
|