1
|
Busselman BW, Ratnayake I, Terasaki MR, Thakkar VP, Ilyas A, Otterpohl KL, Zimmerman JL, Chandrasekar I. Actin cytoskeleton and associated myosin motors within the renal epithelium. Am J Physiol Renal Physiol 2024; 327:F553-F565. [PMID: 39052845 PMCID: PMC11483076 DOI: 10.1152/ajprenal.00078.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/09/2024] [Accepted: 07/24/2024] [Indexed: 07/27/2024] Open
Abstract
This review highlights the complexity of renal epithelial cell membrane architectures and organelles through careful review of ultrastructural and physiological studies published over the past several decades. We also showcase the vital roles played by the actin cytoskeleton and actin-associated myosin motor proteins in regulating cell type-specific physiological functions within the cells of the renal epithelium. The purpose of this review is to provide a fresh conceptual framework to explain the structure-function relationships that exist between the actin cytoskeleton, organelle structure, and cargo transport within the mammalian kidney. With recent advances in technologies to visualize the actin cytoskeleton and associated proteins within intact kidneys, it has become increasingly imperative to reimagine the functional roles of these proteins in situ to provide a rationale for their unique, cell type-specific functions that are necessary to establish and maintain complex physiological processes.
Collapse
Affiliation(s)
- Brook W Busselman
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, United States
- Basic Biomedical Sciences Graduate Program, University of South Dakota, Vermillion, South Dakota, United States
| | | | - Mark R Terasaki
- Department of Cell Biology, University of Connecticut, Farmington, Connecticut, United States
- Department of Pediatrics, University of South Dakota Sanford School of Medicine, Sioux Falls, South Dakota, United States
| | - Vedant P Thakkar
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, United States
| | - Arooba Ilyas
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, United States
- Basic Biomedical Sciences Graduate Program, University of South Dakota, Vermillion, South Dakota, United States
| | - Karla L Otterpohl
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, United States
| | - Jenna L Zimmerman
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, United States
| | - Indra Chandrasekar
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, United States
- Department of Cell Biology, University of Connecticut, Farmington, Connecticut, United States
- Department of Pediatrics, University of South Dakota Sanford School of Medicine, Sioux Falls, South Dakota, United States
| |
Collapse
|
2
|
Shaughnessey EM, Kann SH, Charest JL, Vedula EM. Human Kidney Proximal Tubule-Microvascular Model Facilitates High-Throughput Analyses of Structural and Functional Effects of Ischemia-Reperfusion Injury. Adv Biol (Weinh) 2024; 8:e2300127. [PMID: 37786311 DOI: 10.1002/adbi.202300127] [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: 03/28/2023] [Revised: 08/27/2023] [Indexed: 10/04/2023]
Abstract
Kidney ischemia reperfusion injury (IRI) poses a major global healthcare burden, but effective treatments remain elusive. IRI involves a complex interplay of tissue-level structural and functional changes caused by interruptions in blood and filtrate flow and reduced oxygenation. Existing in vitro models poorly replicate the in vivo injury environment and lack means of monitoring tissue function during the injury process. Here, a high-throughput human primary kidney proximal tubule (PT)-microvascular model is described, which facilitates in-depth structural and rapid functional characterization of IRI-induced changes in the tissue barrier. The PREDICT96 (P96) microfluidic platform's user-controlled fluid flow can mimic the conditions of IR to induce pronounced changes in cell structure that resemble clinical and in vivo phenotypes. High-throughput trans-epi/endo-thelial electrical resistance (TEER) sensing is applied to non-invasively track functional changes in the PT-microvascular barrier during the two-stage injury process and over repeated episodes of injury. Notably, ischemia causes an initial increase in tissue TEER followed by a sudden increase in permeability upon reperfusion, and this biphasic response occurs only with the loss of both fluid flow and oxygenation. This study demonstrates the potential of the P96 kidney IRI model to enhance understanding of IRI and fuel therapeutic development.
Collapse
Affiliation(s)
- Erin M Shaughnessey
- Draper Scholar, The Charles Stark Draper Laboratory Inc., 555 Technology Square, Cambridge, MA, 02139, USA
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA, 02155, USA
| | - Samuel H Kann
- Draper Scholar, The Charles Stark Draper Laboratory Inc., 555 Technology Square, Cambridge, MA, 02139, USA
- Department of Mechanical Engineering, Boston University, 110 Cummington Mall, Boston, MA, 02215, USA
| | - Joseph L Charest
- The Charles Stark Draper Laboratory Inc., 555 Technology Square, Cambridge, MA, 02139, USA
| | - Else M Vedula
- The Charles Stark Draper Laboratory Inc., 555 Technology Square, Cambridge, MA, 02139, USA
| |
Collapse
|
3
|
Luther T, Bülow-Anderberg S, Persson P, Franzén S, Skorup P, Wernerson A, Hultenby K, Palm F, Schiffer TA, Frithiof R. Renal mitochondrial dysfunction in ovine experimental sepsis-associated acute kidney injury. Am J Physiol Renal Physiol 2023; 324:F571-F580. [PMID: 37102685 DOI: 10.1152/ajprenal.00294.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 03/31/2023] [Accepted: 04/19/2023] [Indexed: 04/28/2023] Open
Abstract
Sheep develop sepsis-associated acute kidney injury (SA-AKI) during experimental sepsis despite normal to increased renal oxygen delivery. A disturbed relation between oxygen consumption (V̇o2) and renal Na+ transport has been demonstrated in sheep and in clinical studies of AKI, which could be explained by mitochondrial dysfunction. We investigated the function of isolated renal mitochondria compared with renal oxygen handling in an ovine hyperdynamic model of SA-AKI. Anesthetized sheep were randomized to either an infusion of live Escherichia coli with resuscitative measures (sepsis group; n = 13 animals) or served as controls (n = 8 animals) for 28 h. Renal V̇o2 and Na+ transport were repeatedly measured. Live cortical mitochondria were isolated at baseline and at the end of the experiment and assessed in vitro with high-resolution respirometry. Sepsis markedly reduced creatinine clearance, and the relation between Na+ transport and renal V̇o2 was decreased in septic sheep compared with control sheep. Cortical mitochondrial function was altered in septic sheep with a reduced respiratory control ratio (6.0 ± 1.5 vs. 8.2 ± 1.6, P = 0.006) and increased complex II-to-complex I ratio during state 3 (1.6 ± 0.2 vs. 1.3 ± 0.1, P = 0.0014) mainly due to decreased complex I-dependent state 3 respiration (P = 0.016). However, no differences in renal mitochondrial efficiency or mitochondrial uncoupling were found. In conclusion, renal mitochondrial dysfunction composed of a reduction of the respiratory control ratio and an increased complex II/complex I relation in state 3 was demonstrated in an ovine model of SA-AKI. However, the disturbed relation between renal V̇o2 and renal Na+ transport could not be explained by a change in renal cortical mitochondrial efficiency or uncoupling.NEW & NOTEWORTHY We studied the function of renal cortical mitochondria in relation to oxygen consumption in an ovine model of sepsis with acute kidney injury. We demonstrated changes in the electron transport chain induced by sepsis consisting of a reduced respiratory control ratio mainly by a reduced complex I-mediated respiration. Neither an increase in mitochondrial uncoupling nor a reduction in mitochondrial efficiency was demonstrated and cannot explain why oxygen consumption was unaffected despite reduced tubular transport.
Collapse
Affiliation(s)
- Tomas Luther
- Department of Surgical Sciences, Anesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden
| | - Sara Bülow-Anderberg
- Department of Surgical Sciences, Anesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden
| | - Patrik Persson
- Section of Integrative Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Stephanie Franzén
- Department of Surgical Sciences, Anesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden
| | - Paul Skorup
- Section of Infectious Diseases, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Annika Wernerson
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Kjell Hultenby
- Division of Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Fredrik Palm
- Section of Integrative Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Tomas A Schiffer
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Robert Frithiof
- Department of Surgical Sciences, Anesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden
| |
Collapse
|
4
|
Hurtado KA, Janda J, Schnellmann RG. Lasmiditan promotes recovery from acute kidney injury through induction of mitochondrial biogenesis. Am J Physiol Renal Physiol 2023; 324:F56-F63. [PMID: 36326468 PMCID: PMC9762961 DOI: 10.1152/ajprenal.00249.2022] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 10/27/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022] Open
Abstract
Acute kidney injury (AKI) involves rapid loss of renal function and occurs in 8-16% of hospitalized patients. AKI can be induced by drugs, sepsis, and ischemia-reperfusion (I/R). Hallmarks of AKI include mitochondrial and microvasculature dysfunction as well as renal tubular injury. There is currently no available therapeutic for AKI. Previously, our group identified that serotonin (5-HT)1F receptor agonism with lasmiditan accelerated endothelial cell recovery and induced mitochondrial biogenesis (MB) in vitro. We hypothesized that lasmiditan, a Federal Drug Administration-approved drug, would induce MB and improve microvascular and renal function in a mouse model of AKI. Male mice were subjected to renal I/R and treated with lasmiditan (0.3 mg/kg) or vehicle beginning 24 h after injury and then daily until euthanasia at 6 or 12 days. Serum creatinine was measured to estimate glomerular filtration rate. The renal cortex was assessed for mitochondrial density, vascular permeability and integrity, tubular damage, and interstitial fibrosis. Lasmiditan increased mitochondrial number (1.4-fold) in renal cortices. At 6 days, serum creatinine decreased 41% in the I/R group and 72% with lasmiditan. At 6 or 12 days, kidney injury molecule-1 increased in the I/R group and decreased 50% with lasmiditan. At 12 days, interstitial fibrosis decreased with lasmiditan by 50% and collagen type 1 by 38%. Evan's blue dye leakage increased 2.5-fold in the I/R group and was restored with lasmiditan. The tight junction proteins zonula occludens-1, claudin-2, and claudin-5 decreased in the I/R group and recovered with lasmiditan. At 6 or 12 days, peroxisome proliferator-activated receptor-γ coactivator-1α and electron transport chain complexes increased only with lasmiditan. In conclusion, lasmiditan treatment beginning AKI induces MB, attenuated vascular and tubular injury, decreased interstitial fibrosis, and lowered serum creatinine. Given that lasmiditan is a Federal Drug Administration-approved drug, these preclinical data support repurposing lasmiditan as a therapeutic for AKI.NEW & NOTEWORTHY AKI pathology involves a rapid decline in kidney function and occurs in 8-16% of hospitalized patients. There is currently no therapeutic for AKI. AKI results in mitochondria dysfunction, microvasculature injury, and loss of renal tubular function. In an I/R-induced AKI mouse model, treatment with the FDA-approved 5-HT1F receptor-selective agonist lasmiditan induced mitochondrial biogenesis, improved vascular integrity, reduced fibrosis, and reduced proximal tubule damage. These data support repurposing lasmiditan for the treatment of AKI.
Collapse
Affiliation(s)
- Kevin A Hurtado
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona
| | - Jaroslav Janda
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona
| | - Rick G Schnellmann
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona
- Southern Arizona Veterans Affairs Health Care System, Tucson, Arizona
- Southwest Environmental Health Science Center, University of Arizona, Tucson, Arizona
| |
Collapse
|
5
|
Madan S, Uttekar B, Chowdhary S, Rikhy R. Mitochondria Lead the Way: Mitochondrial Dynamics and Function in Cellular Movements in Development and Disease. Front Cell Dev Biol 2022; 9:781933. [PMID: 35186947 PMCID: PMC8848284 DOI: 10.3389/fcell.2021.781933] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 12/16/2021] [Indexed: 01/09/2023] Open
Abstract
The dynamics, distribution and activity of subcellular organelles are integral to regulating cell shape changes during various physiological processes such as epithelial cell formation, cell migration and morphogenesis. Mitochondria are famously known as the powerhouse of the cell and play an important role in buffering calcium, releasing reactive oxygen species and key metabolites for various activities in a eukaryotic cell. Mitochondrial dynamics and morphology changes regulate these functions and their regulation is, in turn, crucial for various morphogenetic processes. In this review, we evaluate recent literature which highlights the role of mitochondrial morphology and activity during cell shape changes in epithelial cell formation, cell division, cell migration and tissue morphogenesis during organism development and in disease. In general, we find that mitochondrial shape is regulated for their distribution or translocation to the sites of active cell shape dynamics or morphogenesis. Often, key metabolites released locally and molecules buffered by mitochondria play crucial roles in regulating signaling pathways that motivate changes in cell shape, mitochondrial shape and mitochondrial activity. We conclude that mechanistic analysis of interactions between mitochondrial morphology, activity, signaling pathways and cell shape changes across the various cell and animal-based model systems holds the key to deciphering the common principles for this interaction.
Collapse
|
6
|
Bernhardt A, Häberer S, Xu J, Damerau H, Steffen J, Reichardt C, Wolters K, Steffen H, Isermann B, Borucki K, Artelt N, Endlich N, Kozyraki R, Brandt S, Lindquist JA, Mertens PR. High salt diet-induced proximal tubular phenotypic changes and sodium-glucose cotransporter-2 expression are coordinated by cold shock Y-box binding protein-1. FASEB J 2021; 35:e21912. [PMID: 34533842 DOI: 10.1096/fj.202100667rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/06/2021] [Accepted: 08/25/2021] [Indexed: 11/11/2022]
Abstract
High salt diet (HSD) is a hallmark of blood pressure elevations, weight gain and diabetes onset in the metabolic syndrome. In kidney, compensatory mechanisms are activated to balance salt turnover and maintain homeostasis. Data on the long-term effects of HSD with respect to tubular cell functions and kidney architecture that exclude confounding indirect blood pressure effects are scarce. Additionally we focus on cold shock Y-box binding protein-1 as a tubular cell protective factor. A HSD model (4% NaCl in chow; 1% NaCl in water) was compared to normal salt diet (NSD, standard chow) over 16 months using wild type mice and an inducible conditional whole body knockout for cold shock Y-box binding protein-1 (BL6J/N, Ybx1). HSD induced no difference in blood pressure over 16 months, comparing NSD/HSD and Ybx1 wild type/knockout. Nevertheless, marked phenotypic changes were detected. Glucosuria and subnephrotic albuminuria ensued in wild type animals under HSD, which subsided in Ybx1-deficient animals. At the same time megalin receptors were upregulated. The sodium-glucose cotransporter-2 (SGLT2) was completely downregulated in wild type HSD animals that developed glucosuria. In Ybx1 knockouts, expression of AQP1 and SGLT2 was maintained under HSD; proximal tubular widening and glomerular tubularization developed. Concurrently, amino aciduria of neutral and hydrophobic amino acids was seen. In vitro translation confirmed that YB-1 translationally represses Sglt2 transcripts. Our data reveal profound effects of HSD primarily within glomeruli and proximal tubular segments. YB-1 is regulated by HSD and orchestrates HSD-dependent changes; notably, sets reabsorption thresholds for amino acids, proteins and glucose.
Collapse
Affiliation(s)
- Anja Bernhardt
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Saskia Häberer
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - JingJing Xu
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Hannah Damerau
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Johannes Steffen
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Charlotte Reichardt
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Katharina Wolters
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Hannes Steffen
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Berend Isermann
- Institute of Clinical Chemistry and Pathobiochemistry, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Katrin Borucki
- Institute of Clinical Chemistry and Pathobiochemistry, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Nadine Artelt
- Institute of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany.,NIPOKA GmbH, Greifswald, Germany
| | - Nicole Endlich
- Institute of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany.,NIPOKA GmbH, Greifswald, Germany
| | - Renata Kozyraki
- Centre de Recherche des Cordeliers, INSERM, UMRS-1138, Université de Paris, Paris, France
| | - Sabine Brandt
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Jonathan A Lindquist
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Peter R Mertens
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| |
Collapse
|
7
|
Ricksten SE, Bragadottir G, Lannemyr L, Redfors B, Skytte J. Renal Hemodynamics, Function, and Oxygenation in Critically Ill Patients and after Major Surgery. KIDNEY360 2021; 2:894-904. [PMID: 35373068 PMCID: PMC8791344 DOI: 10.34067/kid.0007012020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/02/2021] [Indexed: 02/04/2023]
Abstract
This review outlines the available data from the work of our group on renal hemodynamics, function, and oxygenation in patients who are critically ill with acute renal dysfunction, such as those with postoperative AKI, those in early clinical septic shock, in patients undergoing cardiac surgery with cardiopulmonary bypass, or in patients undergoing liver transplantation. We also provide information on renal hemodynamics, function, and oxygenation in patients with chronic renal impairment due to congestive heart failure. This review will argue that, for all of these groups of patients, the common denominator is that renal oxygenation is impaired due to a lower renal oxygen delivery or a pronounced increase in renal oxygen consumption.
Collapse
|
8
|
Tutunea-Fatan E, Lee JC, Denker BM, Gunaratnam L. Heterotrimeric Gα 12/13 proteins in kidney injury and disease. Am J Physiol Renal Physiol 2020; 318:F660-F672. [PMID: 31984793 DOI: 10.1152/ajprenal.00453.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Gα12 and Gα13 are ubiquitous members of the heterotrimeric guanine nucleotide-binding protein (G protein) family that play central and integrative roles in the regulation of signal transduction cascades within various cell types in the kidney. Gα12/Gα13 proteins enable the kidney to adapt to an ever-changing environment by transducing stimuli from cell surface receptors and accessory proteins to effector systems. Therefore, perturbations in Gα12/Gα13 levels or their activity can contribute to the pathogenesis of various renal diseases, including renal cancer. This review will highlight and discuss the complex and expanding roles of Gα12/Gα13 proteins on distinct renal pathologies, with emphasis on more recently reported findings. Deciphering how the different Gα12/Gα13 interaction networks participate in the onset and development of renal diseases may lead to the discovery of new therapeutic strategies.
Collapse
Affiliation(s)
- Elena Tutunea-Fatan
- Matthew Mailing Centre for Translational Transplant Studies, Lawson Health Research Institute, London, Ontario, Canada
| | - Jasper C Lee
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Bradley M Denker
- Beth Israel Deaconess Medical Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Lakshman Gunaratnam
- Matthew Mailing Centre for Translational Transplant Studies, Lawson Health Research Institute, London, Ontario, Canada.,Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada.,Division of Nephrology, Department of Medicine, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
9
|
Nensén O, Hansell P, Palm F. Role of carbonic anhydrase in acute recovery following renal ischemia reperfusion injury. PLoS One 2019; 14:e0220185. [PMID: 31465457 PMCID: PMC6715224 DOI: 10.1371/journal.pone.0220185] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/10/2019] [Indexed: 11/18/2022] Open
Abstract
Ischemia reperfusion (IR) injury can cause acute kidney injury. It has previously been reported that kidney oxygen consumption (QO2) in relation to glomerular filtration rate (GFR), and thus tubular sodium load, is markedly increased following IR injury, indicating reduced electrolyte transport efficiency. Since proximal tubular sodium reabsorption (TNa) is a major contributor to overall kidney QO2, we investigated whether inhibition of proximal tubular sodium transport through carbonic anhydrase (CA) inhibition would improve renal oxygenation following ischemia reperfusion. Anesthetized adult male Sprague Dawley rats were administered the CA inhibitor acetazolamide (50 mg/kg bolus iv), or volume-matched vehicle, and kidney function, hemodynamics and QO2 were estimated before and after 45 minutes of unilateral complete warm renal ischemia. CA inhibition per se reduced GFR (-20%) and TNa (-22%), while it increased urine flow and urinary sodium excretion (36-fold). Renal blood flow was reduced (-31%) due to increased renal vascular resistance (+37%) without affecting QO2. IR per se resulted in similar decrease in GFR and TNa, independently of CA activity. However, the QO2/TNa ratio following ischemia-reperfusion was profoundly increased in the group receiving CA inhibition, indicating a significant contribution of basal oxygen metabolism to the total kidney QO2 following inhibition of proximal tubular function after IR injury. Ischemia increased urinary excretion of kidney injury molecule-1, an effect that was unaffected by CA. In conclusion, this study demonstrates that CA inhibition further impairs renal oxygenation and does not protect tubular function in the acute phase following IR injury. Furthermore, these results indicate a major role of the proximal tubule in the acute recovery from an ischemic insult.
Collapse
Affiliation(s)
- Oskar Nensén
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- Uppsala University Hospital, Uppsala, Sweden
| | - Peter Hansell
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Fredrik Palm
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- * E-mail:
| |
Collapse
|
10
|
Abstract
The term blood-bile barrier (BBlB) refers to the physical structure within a hepatic lobule that compartmentalizes and hence segregates sinusoidal blood from canalicular bile. Thus, this barrier provides physiological protection in the liver, shielding the hepatocytes from bile toxicity and restricting the mixing of blood and bile. BBlB is primarily composed of tight junctions; however, adherens junction, desmosomes, gap junctions, and hepatocyte bile transporters also contribute to the barrier function of the BBlB. Recent findings also suggest that disruption of BBlB is associated with major hepatic diseases characterized by cholestasis and aberrations in BBlB thus may be a hallmark of many chronic liver diseases. Several molecular signaling pathways have now been shown to play a role in regulating the structure and function and eventually contribute to regulation of the BBlB function within the liver. In this review, we will discuss the structure and function of the BBlB, summarize the methods to assess the integrity and function of BBlB, discuss the role of BBlB in liver pathophysiology, and finally, discuss the mechanisms of BBlB regulation. Collectively, this review will demonstrate the significance of the BBlB in both liver homeostasis and hepatic dysfunction.
Collapse
Affiliation(s)
- Tirthadipa Pradhan-Sundd
- *Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- †Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Satdarshan Pal Monga
- *Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- †Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- ‡Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|
11
|
van Smaalen TC, Ellis SR, Mascini NE, Siegel TP, Cillero-Pastor B, Hillen LM, van Heurn LWE, Peutz-Kootstra CJ, Heeren RMA. Rapid Identification of Ischemic Injury in Renal Tissue by Mass-Spectrometry Imaging. Anal Chem 2019; 91:3575-3581. [PMID: 30702282 PMCID: PMC6581420 DOI: 10.1021/acs.analchem.8b05521] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 01/31/2019] [Indexed: 12/14/2022]
Abstract
The increasing analytical speed of mass-spectrometry imaging (MSI) has led to growing interest in the medical field. Acute kidney injury is a severe disease with high morbidity and mortality. No reliable cut-offs are known to estimate the severity of acute kidney injury. Thus, there is a need for new tools to rapidly and accurately assess acute ischemia, which is of clinical importance in intensive care and in kidney transplantation. We investigated the value of MSI to assess acute ischemic kidney tissue in a porcine model. A perfusion model was developed where paired kidneys received warm (severe) or cold (minor) ischemia ( n = 8 per group). First, ischemic tissue damage was systematically assessed by two blinded pathologists. Second, MALDI-MSI of kidney tissues was performed to study the spatial distributions and compositions of lipids in the tissues. Histopathological examination revealed no significant difference between kidneys, whereas MALDI-MSI was capable of a detailed discrimination of severe and mild ischemia by differential expression of characteristic lipid-degradation products throughout the tissue within 2 h. In particular, lysolipids, including lysocardiolipins, lysophosphatidylcholines, and lysophosphatidylinositol, were dramatically elevated after severe ischemia. This study demonstrates the significant potential of MSI to differentiate and identify molecular patterns of early ischemic injury in a clinically acceptable time frame. The observed changes highlight the underlying biochemical processes of acute ischemic kidney injury and provide a molecular classification tool that can be deployed in assessment of acute ischemic kidney injury.
Collapse
Affiliation(s)
- T. C. van Smaalen
- Department
of Surgery, Maastricht University Medical
Center+, 6229 HX Maastricht, The Netherlands
| | - S. R. Ellis
- The
Maastricht Multimodal Molecular Imaging Institute (M4I), Division
of Imaging Mass Spectrometry, Maastricht
University, 6200 MD Maastricht, The Netherlands
| | - N. E. Mascini
- The
Maastricht Multimodal Molecular Imaging Institute (M4I), Division
of Imaging Mass Spectrometry, Maastricht
University, 6200 MD Maastricht, The Netherlands
| | - T. Porta Siegel
- The
Maastricht Multimodal Molecular Imaging Institute (M4I), Division
of Imaging Mass Spectrometry, Maastricht
University, 6200 MD Maastricht, The Netherlands
| | - B. Cillero-Pastor
- The
Maastricht Multimodal Molecular Imaging Institute (M4I), Division
of Imaging Mass Spectrometry, Maastricht
University, 6200 MD Maastricht, The Netherlands
| | - L. M. Hillen
- Department
of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center+, 6229 HX Maastricht, The Netherlands
- GROW-School
for Oncology and Developmental Biology, Maastricht University Medical Center+, 6229 HX Maastricht, The Netherlands
| | - L. W. E. van Heurn
- Department
of Surgery, Maastricht University Medical
Center+, 6229 HX Maastricht, The Netherlands
| | - C. J. Peutz-Kootstra
- Department
of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center+, 6229 HX Maastricht, The Netherlands
| | - R. M. A. Heeren
- The
Maastricht Multimodal Molecular Imaging Institute (M4I), Division
of Imaging Mass Spectrometry, Maastricht
University, 6200 MD Maastricht, The Netherlands
| |
Collapse
|
12
|
Renal Blood Flow, Glomerular Filtration Rate, and Renal Oxygenation in Early Clinical Septic Shock*. Crit Care Med 2018. [DOI: 10.1097/ccm.0000000000003088] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
13
|
Setyaningsih WAW, Arfian N, Suryadi E, Romi MM, Tranggono U, Sari DCR. Hyperuricemia Induces Wnt5a/Ror2 Gene Expression, Epithelial-Mesenchymal Transition, and Kidney Tubular Injury in Mice. IRANIAN JOURNAL OF MEDICAL SCIENCES 2018; 43:164-173. [PMID: 29749985 PMCID: PMC5936848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
BACKGROUND Hyperuricemia contributes to kidney injury, characterized by tubular injury with epithelial-mesenchymal transition (EMT). Wnt5a/Ror2 signaling drives EMT in many kidney pathologies. This study sought to evaluate the involvement of Wnt5a/Ror2 in hyperuricemia-induced EMT in kidney tubular injury. METHODS A hyperuricemia model was performed in male Swiss background mice (3 months old, 30-40 g) with daily intraperitoneal injections of 125 mg/kg body weight (BW) of uric acid. The mice were terminated on day 7 (UA7, n=5) and on day 14 (UA14, n=5). Allopurinol groups (UAl7 and UAl14, each n=5) were added with oral 50 mg/kg BW of allopurinol treatment. The serum uric acid level was quantified, and tubular injury was assessed based on PAS staining. Reverse transcriptase-PCR was done to quantify Wnt5a, Ror2, E-cadherin, and vimentin expressions. IHC staining was done for E-cadherin and collagen I. We used the Shapiro-Wilk for normality testing and one-way ANOVA for variance analysis with a P<0.05 as significance level using SPSS 22 software. RESULTS The hyperuricemia groups had a higher uric acid level, which was associated with a higher tubular injury score. Meanwhile, the allopurinol groups had a significantly lower uric acid level and tubular injury than the uric acid groups. Reverse transcriptase-PCR revealed downregulation of the E-cadherin expression. While vimentin and collagen I expression are upregulated, which was associated with a higher Wnt5a expression. However, the allopurinol groups had reverse results. Immunostaining revealed a reduction in E-cadherin staining in the epithelial cells and collagen I positive staining in the epithelial cells and the interstitial areas. CONCLUSION Hyperuricemia induced tubular injury, which might have been mediated by EMT through the activation of Wnt5a.
Collapse
Affiliation(s)
| | - Nur Arfian
- Department of Anatomy, Medical Faculty, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Efrayim Suryadi
- Department of Anatomy, Medical Faculty, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Muhammad Mansyur Romi
- Department of Anatomy, Medical Faculty, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Untung Tranggono
- Department of Surgery, Medical Faculty, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | | |
Collapse
|
14
|
Nilsson KF, Sandin J, Gustafsson LE, Frithiof R. The novel nitric oxide donor PDNO attenuates ovine ischemia-reperfusion induced renal failure. Intensive Care Med Exp 2017; 5:29. [PMID: 28600797 PMCID: PMC5466578 DOI: 10.1186/s40635-017-0143-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 05/26/2017] [Indexed: 01/18/2023] Open
Abstract
Background Renal ischemia-reperfusion injury is a common cause of acute kidney injury in intensive care and surgery. Recently, novel organic mononitrites of 1,2-propanediol (PDNO) were synthesized and shown to rapidly and controllably deploy nitric oxide in the circulation when administered intravenously. We hypothesized that intravenous infusion of PDNO during renal ischemia reperfusion would improve post-ischemic renal function and microcirculation. Methods Sixteen sheep were anesthetized, mechanically ventilated, and surgically instrumented. The left renal artery was clamped for 90 min, and the effects of ischemia were studied for a total of 8 h. Fifteen minutes prior to the release of the clamp, intravenous infusions of PDNO (n = 8) or vehicle (1,2 propanediol + inorganic nitrite, n = 8) were initiated (180 nmol/kg/min for 30 min, thereafter 60 nmol/kg/min for the remainder of the experiment). Results Renal artery blood flow, cortical and medullary perfusion, and diuresis and creatinine clearance decreased in the left kidney post ischemia. However, in the sheep treated with PDNO, diuresis and creatinine clearance in the left kidney were significantly higher post ischemia compared to vehicle-treated animals (1.7 ± 0.5 vs 0.7 ± 0.3 ml/kg/h, p = 0.04 and 7.5 ± 2.1 vs 1.7 ± 0.6 ml/min, p = 0.02, respectively). Left renal medullary perfusion and oxygen uptake were higher in the PDNO group (73 ± 9 vs 37 ± 5% of baseline, p = 0.004 and 2.6 ± 0.4 vs 1.6 ± 0.3 ml/min, p = 0.02, respectively). PDNO significantly increased renal oxygen consumption and reduced the oxygen utilization for sodium reabsorption (p = 0.03 for both). Mean arterial blood pressure was significantly reduced by PDNO (83 ± 3 vs 94 ± 3 mmHg, p = 0.02) but was still within normal limits. Total renal blood flow was not affected, and there were no signs of increased blood methemoglobin concentrations or tachyphylaxis. Conclusions The novel nitric oxide donor PDNO improved renal function after ischemia. PDNO also prevented the persistent reduction in medullary perfusion during reperfusion and improved renal oxygen utilization without severe side effects.
Collapse
Affiliation(s)
- Kristofer F Nilsson
- Department of Cardiothoracic and Vascular Surgery, Faculty of Medicine and Health, Örebro University, Örebro, Sweden.,Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - John Sandin
- Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Lars E Gustafsson
- Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Robert Frithiof
- Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden. .,Department of Surgical Sciences, Section of Anesthesia and Intensive Care, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
15
|
Coskun A, Baykal AT, Kazan D, Akgoz M, Senal MO, Berber I, Titiz I, Bilsel G, Kilercik H, Karaosmanoglu K, Cicek M, Yurtsever I, Yazıcı C. Proteomic Analysis of Kidney Preservation Solutions Prior to Renal Transplantation. PLoS One 2016; 11:e0168755. [PMID: 28036361 PMCID: PMC5201308 DOI: 10.1371/journal.pone.0168755] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 12/06/2016] [Indexed: 12/01/2022] Open
Abstract
One of the main issues in kidney transplantation is the optimal functional preservation of the organ until its transplantation into the appropriate recipient. Despite intensive efforts, the functional preservation period remains limited to hours. During this time, as a result of cellular injury, various proteins, peptides, and other molecules are released by the organ into the preservation medium. In this study, we used proteomic techniques to analyze the protein profiles of preservation solutions in which organs had been preserved prior to their transplantation. Samples were obtained from the preservation solutions of 25 deceased donor kidneys scheduled for transplantation. The protein profiles of the solutions were analyzed using 2D gel electrophoresis/MALDI-TOF and LC-MS/MS. We identified and quantified 206 proteins and peptides belonging to 139 different groups. Of these, 111 proteins groups were belonging to kidney tissues. This study used proteomic techniques to analyze the protein profiles of organ preservation solutions. These findings will contribute to the development of improved preservation solutions to effectively protect organs for transplantation.
Collapse
Affiliation(s)
- Abdurrahman Coskun
- Acibadem University School of Medicine, Department of Medical Biochemistry, Istanbul, Turkey
- * E-mail:
| | - Ahmet Tarik Baykal
- Acibadem University School of Medicine, Department of Medical Biochemistry, Istanbul, Turkey
| | - Dilek Kazan
- Marmara University Engineering Faculty, Department of Bioengineering, Istanbul, Turkey
| | | | | | - Ibrahim Berber
- Acibadem University School of Medicine Department of General Surgery, Istanbul, Turkey
| | - Izzet Titiz
- Haydarpasa Numune Research and Training Hospital, Department of General Surgery, Istanbul, Turkey
| | | | - Hakan Kilercik
- Yeni Yuzyil University, Gaziosmanpasa Hospital, Department of Anesthesiology, Istanbul, Turkey
| | - Kubra Karaosmanoglu
- Marmara University Engineering Faculty, Department of Bioengineering, Istanbul, Turkey
| | - Muslum Cicek
- Yeni Yuzyil University, Gaziosmanpasa Hospital, Department of Anesthesiology, Istanbul, Turkey
| | - Ilknur Yurtsever
- Istanbul Medipol University, Regenerative and Restorative Medicine Research Center, Beykoz/Istanbul, Turkey
| | - Cevat Yazıcı
- Erciyes University, School of Medicine, Department of Medical Biochemistry, Kayseri, Turkey
| |
Collapse
|
16
|
Vallon V. Tubular Transport in Acute Kidney Injury: Relevance for Diagnosis, Prognosis and Intervention. Nephron Clin Pract 2016; 134:160-166. [PMID: 27238156 DOI: 10.1159/000446448] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 04/24/2016] [Indexed: 12/19/2022] Open
Abstract
The clinical diagnosis and recovery of acute kidney injury (AKI) are mainly based on the rapid decline of glomerular filtration rate (GFR) and its subsequent recovery. The factors that determine kidney recovery and reduce the risk of subsequent progression to chronic kidney disease (CKD), however, are poorly understood. Thus, there is a need to better define the magnitude and time pattern of changes in kidney function during AKI and its recovery that go beyond GFR. Tubular transport regulates body homeostasis and the associated transport work is a primary determinant of the kidneys' energy needs. The tubular system is at the center of the pathophysiology of AKI and its recovery. In particular, proximal tubules and thick ascending limbs have been proposed to act as sensors, effectors and injury recipients of AKI stimuli. Surprisingly little attention has been given to aspects of tubular transport function in AKI and the relevance for kidney recovery. This review aims to outline changes in tubular transport function in AKI, discusses their potential consequences and relevance for the diagnosis and prognosis of AKI and its recovery, including changes in GFR, and poses the question whether tubular transport provides an opportunity for intervention to rest the tubular system, which may have consequences for the progression to CKD. © 2016 S. Karger AG, Basel.
Collapse
Affiliation(s)
- Volker Vallon
- Departments of Medicine and Pharmacology, University of California, San Diego, La Jolla, Calif., USA
| |
Collapse
|
17
|
Bennett J, Cassidy H, Slattery C, Ryan MP, McMorrow T. Tacrolimus Modulates TGF-β Signaling to Induce Epithelial-Mesenchymal Transition in Human Renal Proximal Tubule Epithelial Cells. J Clin Med 2016; 5:jcm5050050. [PMID: 27128949 PMCID: PMC4882479 DOI: 10.3390/jcm5050050] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Revised: 04/16/2016] [Accepted: 04/19/2016] [Indexed: 01/05/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT), a process which describes the trans-differentiation of epithelial cells into motile mesenchymal cells, is pivotal in stem cell behavior, development and wound healing, as well as contributing to disease processes including fibrosis and cancer progression. Maintenance immunosuppression with calcineurin inhibitors (CNIs) has become routine management for renal transplant patient, but unfortunately the nephrotoxicity of these drugs has been well documented. HK-2 cells were exposed to Tacrolimus (FK506) and EMT markers were assessed by RT PCR and western blot. FK506 effects on TGF-β mRNA were assessed by RT PCR and TGF-β secretion was measured by ELISA. The impact of increased TGF-β secretion on Smad signaling pathways was investigated. The impact of inhibition of TGF-β signaling on EMT processes was assessed by scratch-wound assay. The results presented in this study suggest that FK506 initiates EMT processes in the HK-2 cell line, with altered expression of epithelial and myofibroblast markers evident. Additionally, the study demonstrates that FK506 activation of the TGF-β/ SMAD pathways is an essential step in the EMT process. Overall the results demonstrate that EMT is heavily involved in renal fibrosis associated with CNI nephrotoxicity.
Collapse
Affiliation(s)
- Jason Bennett
- Centre for Cell Signaling and Inflammation, Department of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK.
| | - Hilary Cassidy
- Renal Disease Research Group, School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin 4, Ireland.
| | - Craig Slattery
- Renal Disease Research Group, School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin 4, Ireland.
| | - Michael P Ryan
- Renal Disease Research Group, School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin 4, Ireland.
| | - Tara McMorrow
- Renal Disease Research Group, School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin 4, Ireland.
| |
Collapse
|
18
|
Tögel FE, Ahlstrom JD, Yang Y, Hu Z, Zhang P, Westenfelder C. Carbamylated Erythropoietin Outperforms Erythropoietin in the Treatment of AKI-on-CKD and Other AKI Models. J Am Soc Nephrol 2016; 27:3394-3404. [PMID: 26984884 DOI: 10.1681/asn.2015091059] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 02/05/2016] [Indexed: 12/31/2022] Open
Abstract
Erythropoietin (EPO) may be a beneficial tissue-protective cytokine. However, high doses of EPO are associate with adverse effects, including thrombosis, tumor growth, and hypertension. Carbamylated erythropoietin (CEPO) lacks both erythropoietic and vasoconstrictive actions. In this study, we compared the renoprotective, hemodynamic, and hematologic activities and survival effects of identical EPO and CEPO doses in rat models of clinically relevant AKI presentations, including ischemia-reperfusion-induced AKI superimposed on CKD (5000 U/kg EPO or CEPO; three subcutaneous injections) and ischemia-reperfusion-induced AKI in old versus young animals and male versus female animals (1000 U/kg EPO or CEPO; three subcutaneous injections). Compared with EPO therapy, CEPO therapy induced greater improvements in renal function and body weight in AKI on CKD animals, with smaller increases in hematocrit levels and similarly improved survival. Compared with EPO therapy in the other AKI groups, CEPO therapy induced greater improvements in protection and recovery of renal function and survival, with smaller increases in systolic BP and hematocrit levels. Overall, old or male animals had more severe loss in kidney function and higher mortality rates than young or female animals, respectively. Notably, mRNA and protein expression analyses confirmed the renal expression of the heterodimeric EPO receptor/CD131 complex, which is required for the tissue-protective effects of CEPO signaling. In conclusion, CEPO improves renal function, body and kidney weight, and survival in AKI models without raising hematocrit levels and BP as substantially as EPO. Thus, CEPO therapy may be superior to EPO in improving outcomes in common forms of clinical AKI.
Collapse
Affiliation(s)
- Florian E Tögel
- Department of Medicine, Massachusetts General Hospital Medicine Group, Boston, Massachusetts
| | - Jon D Ahlstrom
- Department of Medicine, Division of Nephrology and.,Department of Medicine, Section of Nephrology, Veterans Affairs Medical Center Salt Lake City, Salt Lake City, Utah
| | - Ying Yang
- Department of Medicine, Division of Nephrology and
| | - Zhuma Hu
- Department of Medicine, Division of Nephrology and
| | - Ping Zhang
- Department of Medicine, Division of Nephrology and
| | - Christof Westenfelder
- Department of Medicine, Division of Nephrology and .,Department of Medicine, Section of Nephrology, Veterans Affairs Medical Center Salt Lake City, Salt Lake City, Utah.,Department of Physiology, University of Utah, Salt Lake City, Utah; and
| |
Collapse
|
19
|
Alves DS, Thulin G, Loffing J, Kashgarian M, Caplan MJ. Akt Substrate of 160 kD Regulates Na+,K+-ATPase Trafficking in Response to Energy Depletion and Renal Ischemia. J Am Soc Nephrol 2015; 26:2765-76. [PMID: 25788531 DOI: 10.1681/asn.2013101040] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Accepted: 01/06/2015] [Indexed: 01/26/2023] Open
Abstract
Renal ischemia and reperfusion injury causes loss of renal epithelial cell polarity and perturbations in tubular solute and fluid transport. Na(+),K(+)-ATPase, which is normally found at the basolateral plasma membrane of renal epithelial cells, is internalized and accumulates in intracellular compartments after renal ischemic injury. We previously reported that the subcellular distribution of Na(+),K(+)-ATPase is modulated by direct binding to Akt substrate of 160 kD (AS160), a Rab GTPase-activating protein that regulates the trafficking of glucose transporter 4 in response to insulin and muscle contraction. Here, we investigated the effect of AS160 on Na(+),K(+)-ATPase trafficking in response to energy depletion. We found that AS160 is required for the intracellular accumulation of Na(+),K(+)-ATPase that occurs in response to energy depletion in cultured epithelial cells. Energy depletion led to dephosphorylation of AS160 at S588, which was required for the energy depletion-induced accumulation of Na,K-ATPase in intracellular compartments. In AS160-knockout mice, the effects of renal ischemia on the distribution of Na(+),K(+)-ATPase were substantially reduced in the epithelial cells of distal segments of the renal tubules. These data demonstrate that AS160 has a direct role in linking the trafficking of Na(+),K(+)-ATPase to the energy state of renal epithelial cells.
Collapse
Affiliation(s)
| | - Gunilla Thulin
- Pathology, Yale University School of Medicine, New Haven, Connecticut; and
| | | | - Michael Kashgarian
- Pathology, Yale University School of Medicine, New Haven, Connecticut; and
| | | |
Collapse
|
20
|
Fogelgren B, Zuo X, Buonato JM, Vasilyev A, Baek JI, Choi SY, Chacon-Heszele MF, Palmyre A, Polgar N, Drummond I, Park KM, Lazzara MJ, Lipschutz JH. Exocyst Sec10 protects renal tubule cells from injury by EGFR/MAPK activation and effects on endocytosis. Am J Physiol Renal Physiol 2014; 307:F1334-41. [PMID: 25298525 DOI: 10.1152/ajprenal.00032.2014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Acute kidney injury is common and has a high mortality rate, and no effective treatment exists other than supportive care. Using cell culture models, we previously demonstrated that exocyst Sec10 overexpression reduced damage to renal tubule cells and speeded recovery and that the protective effect was mediated by higher basal levels of mitogen-activated protein kinase (MAPK) signaling. The exocyst, a highly-conserved eight-protein complex, is known for regulating protein trafficking. Here we show that the exocyst biochemically interacts with the epidermal growth factor receptor (EGFR), which is upstream of MAPK, and Sec10-overexpressing cells express greater levels of phosphorylated (active) ERK, the final step in the MAPK pathway, in response to EGF stimulation. EGFR endocytosis, which has been linked to activation of the MAPK pathway, increases in Sec10-overexpressing cells, and gefitinib, a specific EGFR inhibitor, and Dynasore, a dynamin inhibitor, both reduce EGFR endocytosis. In turn, inhibition of the MAPK pathway reduces ligand-mediated EGFR endocytosis, suggesting a potential feedback of elevated ERK activity on EGFR endocytosis. Gefitinib also decreases MAPK signaling in Sec10-overexpressing cells to levels seen in control cells and, demonstrating a causal role for EGFR, reverses the protective effect of Sec10 overexpression following cell injury in vitro. Finally, using an in vivo zebrafish model of acute kidney injury, morpholino-induced knockdown of sec10 increases renal tubule cell susceptibility to injury. Taken together, these results suggest that the exocyst, acting through EGFR, endocytosis, and the MAPK pathway is a candidate therapeutic target for acute kidney injury.
Collapse
Affiliation(s)
- Ben Fogelgren
- Departments of Anatomy, Biochemistry, and Physiology, University of Hawaii at Manoa, Honolulu, Hawaii
| | - Xiaofeng Zuo
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Janine M Buonato
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Jeong-In Baek
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Soo Young Choi
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | | | - Aurélien Palmyre
- Department of Pathology, Harvard Medical School, Boston, Massachusetts
| | - Noemi Polgar
- Departments of Anatomy, Biochemistry, and Physiology, University of Hawaii at Manoa, Honolulu, Hawaii
| | - Iain Drummond
- Departments of Medicine and Genetics, Harvard Medical School, Boston, Massachusetts
| | - Kwon Moo Park
- Department of Anatomy and BK21 Plus, Kyungpook National University School of Medicine, Junggu, Daegu, Republic of Korea; and
| | - Matthew J Lazzara
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Joshua H Lipschutz
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina; Department of Medicine, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| |
Collapse
|
21
|
Ince C. The central role of renal microcirculatory dysfunction in the pathogenesis of acute kidney injury. Nephron Clin Pract 2014; 127:124-8. [PMID: 25343835 DOI: 10.1159/000363203] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Acute kidney injury (AKI) is a rapidly developing condition often associated with critical illness, with a high degree of morbidity and mortality, whose pathophysiology is ill understood. Recent investigations have identified the dysfunction of the renal microcirculation and its cellular and subcellular constituents as being central to the etiology of AKI. Injury is caused by inflammatory activation involving endothelial leucocyte interactions in combination with dysregulation of the homeostatis between oxygen, nitric oxide, and reactive oxygen species. Effective therapies expected to resolve AKI will have to control inflammation and restore this homeostasis. In order to apply and guide these therapies effectively, diagnostic tools aimed at physiological biomarkers of AKI for monitoring renal microcirculatory function in advance of changes in pharmacological biomarkers associated with structural damage of the kidney will need to be developed.
Collapse
Affiliation(s)
- Can Ince
- Department of Translational Physiology, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
22
|
Yang Y, Wei RB, Zheng XY, Qiu Q, Cui SY, Yin Z, Shi SZ, Chen XM. Effects of compound Shenhua tablet on renal tubular Na+-K+-ATPase in rats with acute ischemic reperfusion injury. Chin J Integr Med 2014; 20:200-8. [PMID: 24464369 DOI: 10.1007/s11655-014-1740-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To observe the effect of Compound Shenhua Tablet (, SHT) on the sodium-potassium- exchanging adenosinetriphosphatase (Na(+)-K(+)-ATPase) in the renal tubular epithelial cells of rats with acute ischemic reperfusion and to investigate the mechanisms underlying the effects of SHT on renal ischemic reperfusion injury (RIRI). METHODS Fifty male Wistar rats were randomly divided into the sham surgery group, model group, astragaloside group [150 mg/(kg·d)], SHT low-dose group [1.5 g/(kg·d)] and SHT high-dose group [3.0 g/(kg·d)], with 10 rats in each group. After 1 week of continuous intragastric drug administration, surgery was performed to establish the model. At either 24 or 72 h after the surgery, 5 rats in each group were sacrificed, blood biochemistry, renal pathology, immunoblot and immunohistochemical examinations were performed, and double immunofluorescence staining was observed under a laser confocal microscope. RESULTS Compared with the sham surgery group, the serum creatinine (SCr) and blood urea nitrogen (BUN) levels were significantly increased, Na(+)-K(+)-ATPase protein level was decreased, and kidney injury molecule-1 (KIM-1) protein level was increased in the model group after the surgery (P<0.01 or P<0.05). Compared with the model group, the SCr, BUN, pathological scores, Na(+)-K(+)-ATPase, and the KIM-1 protein level of the three treatment groups were significantly improved at 72 h after the surgery (P<0.05 or P<0.01). And the SCr, BUN of the SHT low- and high-dose groups, and the pathological scores of the SHT high-dose group were significantly lower than those of the astragaloside group (P<0.05). The localizations of Na(+)-K(+)-ATPase and megalin of the model group were disrupted, with the distribution areas overlapping with each other and alternately arranged. The severity of the disruption was slightly milder in three treatment groups compared with that of the model group. The results of immunofluorescence staining showed that the SHT high-dose group had a superior effect as compared with the astragaloside group and the SHT low-dose group. CONCLUSIONS The SHT effectively alleviated RIRI caused by ischemic reperfusion, promoted the recovery of the polarity of renal tubular epithelial cells, and protected the renal tubules. The therapeutic effects of SHT were superior to those of astragaloside as a single agent.
Collapse
Affiliation(s)
- Yue Yang
- State Discipline and State Key Laboratory of Kidney Disease (Chinese PLA General Hospital, 2011DAV00088), Beijing, 100853, China
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Hansell P, Welch WJ, Blantz RC, Palm F. Determinants of kidney oxygen consumption and their relationship to tissue oxygen tension in diabetes and hypertension. Clin Exp Pharmacol Physiol 2013. [PMID: 23181475 DOI: 10.1111/1440-1681.12034] [Citation(s) in RCA: 212] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The high renal oxygen (O(2) ) demand is associated primarily with tubular O(2) consumption (Qo(2) ) necessary for solute reabsorption. Increasing O(2) delivery relative to demand via increased blood flow results in augmented tubular electrolyte load following elevated glomerular filtration, which, in turn, increases metabolic demand. Consequently, elevated kidney metabolism results in decreased tissue oxygen tension. The metabolic efficiency for solute transport (Qo(2) /T(Na) ) varies not only between different nephron sites, but also under different conditions of fluid homeostasis and disease. Contributing mechanisms include the presence of different Na(+) transporters, different levels of oxidative stress and segmental tubular dysfunction. Sustained hyperglycaemia results in increased kidney Qo(2) , partly due to mitochondrial dysfunction and reduced electrolyte transport efficiency. This results in intrarenal tissue hypoxia because the increased Qo(2) is not matched by a similar increase in O(2) delivery. Hypertension leads to renal hypoxia, mediated by increased angiotensin receptor tonus and oxidative stress. Reduced uptake in the proximal tubule increases load to the thick ascending limb. There, the increased load is reabsorbed, but at greater O(2) cost. The combination of hypertension, angiotensin II and oxidative stress initiates events leading to renal damage and reduced function. Tissue hypoxia is now recognized as a unifying pathway to chronic kidney disease. We have gained good knowledge about major changes in O(2) metabolism occurring in diabetic and hypertensive kidneys. However, further efforts are needed to elucidate how these alterations can be prevented or reversed before translation into clinical practice.
Collapse
Affiliation(s)
- Peter Hansell
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden.
| | | | | | | |
Collapse
|
24
|
Singh P, Ricksten SE, Bragadottir G, Redfors B, Nordquist L. Renal oxygenation and haemodynamics in acute kidney injury and chronic kidney disease. Clin Exp Pharmacol Physiol 2013; 40:138-47. [PMID: 23360244 DOI: 10.1111/1440-1681.12036] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Revised: 11/19/2012] [Accepted: 11/20/2012] [Indexed: 11/29/2022]
Abstract
Acute kidney injury (AKI) is a major burden on health systems and may arise from multiple initiating insults, including ischaemia-reperfusion injury, cardiovascular surgery, radiocontrast administration and sepsis. Similarly, the incidence and prevalence of chronic kidney disease (CKD) continues to increase, with significant morbidity and mortality. Moreover, an increasing number of AKI patients survive to develop CKD and end-stage renal disease. Although the mechanisms for the development of AKI and progression to CKD remain poorly understood, initial impairment of oxygen balance likely constitutes a common pathway, causing renal tissue hypoxia and ATP starvation that, in turn, induce extracellular matrix production, collagen deposition and fibrosis. Thus, possible future strategies for one or both conditions may involve dopamine, loop diuretics, atrial natriuretic peptide and inhibitors of inducible nitric oxide synthase, substances that target kidney oxygen consumption and regulators of renal oxygenation, such as nitric oxide and heme oxygenase-1.
Collapse
Affiliation(s)
- Prabhleen Singh
- Division of Nephrology-Hypertension, VA San Diego Healthcare System, University of California San Diego, San Diego, CA, USA
| | | | | | | | | |
Collapse
|
25
|
Lee SY, Han SM, Kim JE, Chung KY, Han KH. Expression of E-cadherin in pig kidney. J Vet Sci 2013; 14:381-6. [PMID: 23820247 PMCID: PMC3885730 DOI: 10.4142/jvs.2013.14.4.381] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Accepted: 12/24/2012] [Indexed: 12/24/2022] Open
Abstract
E-cadherin is a cell adhesion molecule that plays an important role in maintaining renal epithelial polarity and integrity. The purpose of this study was to determine the exact cellular localization of E-cadherin in pig kidney. Kidney tissues from pigs were processed for light and electron microscopy immunocytochemistry, and immunoblot analysis. E-cadhedrin bands of the same size were detected by immunoblot of samples from rat and pig kidneys. In pig kidney, strong E-cadherin expression was observed in the basolateral plasma membrane of the tubular epithelial cells. E-cadherin immunolabeling was not detected in glomeruli or blood vessels of pig kidney. Double-labeling results demonstrated that E-cadherin was expressed in the calbindin D28k-positive distal convoluted tubule and H(+)-ATPase- positive collecting duct, but not in the aquaporin 1-positive, N-cadherin-positive proximal tubule. In contrast to rat, E-cadherin immunoreactivity was not expressed at detectable levels in the Tamm-Horsfall protein-positive thick ascending limb of pig kidney. Immunoelectron microscopy confirmed that E-cadherin was localized in both the lateral membranes and basal infoldings of the collecting duct. These results suggest that E-cadherin may be a critical adhesion molecule in the distal convoluted tubule and collecting duct cells of pig kidney.
Collapse
Affiliation(s)
- Su-Youn Lee
- Departments of Anatomy, Ewha Womans University School of Medicine, Seoul 158-710, Korea
| | | | | | | | | |
Collapse
|
26
|
Ricksten SE, Bragadottir G, Redfors B. Renal oxygenation in clinical acute kidney injury. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2013; 17:221. [PMID: 23514538 PMCID: PMC3672481 DOI: 10.1186/cc12530] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
27
|
H2O2 activates G protein, α 12 to disrupt the junctional complex and enhance ischemia reperfusion injury. Proc Natl Acad Sci U S A 2012; 109:6680-5. [PMID: 22493269 DOI: 10.1073/pnas.1116800109] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The epithelial cell tight junction separates apical and basolateral domains and is essential for barrier function. Disruption of the tight junction is a hallmark of epithelial cell damage and can lead to end organ damage including renal failure. Herein, we identify Gα12 activation by H(2)O(2) leading to tight junction disruption and demonstrate a critical role for Gα12 activation during bilateral renal ischemia/reperfusion injury. Madin-Darby canine kidney (MDCK) cells with inducible Gα12 (Gα12-MDCK) and silenced Gα12 (shGα12-MDCK) were subjected to ATP depletion/repletion and H(2)O(2)/catalase as models of tight junction disruption and recovery by monitoring transepithelial resistance. In ATP depleted cells, barrier disruption and recovery was not affected by Gα12, but reassembly was accelerated by Gα12 depletion. In contrast, silencing of Gα12 completely protected cells from H(2)O(2)-stimulated barrier disruption, a response that rapidly occurred in control cells. H(2)O(2) activated Src and Rho, and Src inhibition (by PP2), but not Rho (by Y27632), protected cells from H(2)O(2)-mediated barrier disruption. Immunofluorescent and biochemical analysis showed that H(2)O(2) led to increased tyrosine phosphorylation of numerous proteins and altered membrane localization of tight junction proteins through Gα12/Src signaling pathway. Gα12 and Src were activated in vivo during ischemia/reperfusion injury, and transgenic mice with renal tubular QLα12 (activated mutant) expression were delayed in recovery and showed more extensive injury. Conversely, Gα12 knockout mice were nearly completely protected from ischemia/reperfusion injury. Taken together, these studies reveal that ROS stimulates Gα12 to activate injury pathways and identifies a therapeutic target for ameliorating ROS mediated injury.
Collapse
|
28
|
Abstract
Acute kidney injury (AKI) is the leading cause of nephrology consultation and is associated with high mortality rates. The primary causes of AKI include ischemia, hypoxia, or nephrotoxicity. An underlying feature is a rapid decline in glomerular filtration rate (GFR) usually associated with decreases in renal blood flow. Inflammation represents an important additional component of AKI leading to the extension phase of injury, which may be associated with insensitivity to vasodilator therapy. It is suggested that targeting the extension phase represents an area potential of treatment with the greatest possible impact. The underlying basis of renal injury appears to be impaired energetics of the highly metabolically active nephron segments (i.e., proximal tubules and thick ascending limb) in the renal outer medulla, which can trigger conversion from transient hypoxia to intrinsic renal failure. Injury to kidney cells can be lethal or sublethal. Sublethal injury represents an important component in AKI, as it may profoundly influence GFR and renal blood flow. The nature of the recovery response is mediated by the degree to which sublethal cells can restore normal function and promote regeneration. The successful recovery from AKI depends on the degree to which these repair processes ensue and these may be compromised in elderly or chronic kidney disease (CKD) patients. Recent data suggest that AKI represents a potential link to CKD in surviving patients. Finally, earlier diagnosis of AKI represents an important area in treating patients with AKI that has spawned increased awareness of the potential that biomarkers of AKI may play in the future.
Collapse
Affiliation(s)
- David P Basile
- Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA.
| | | | | |
Collapse
|
29
|
Lee SY, Shin JA, Kwon HM, Weiner ID, Han KH. Renal ischemia-reperfusion injury causes intercalated cell-specific disruption of occludin in the collecting duct. Histochem Cell Biol 2011; 136:637-47. [PMID: 22048282 PMCID: PMC3214267 DOI: 10.1007/s00418-011-0881-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2011] [Indexed: 11/30/2022]
Abstract
Renal ischemic events open tight junctions and disrupt epithelial polarity. The purpose of this study was to examine the effects of ischemia–reperfusion (IR) injury on expression and distribution of the tight junction proteins, occludin and ZO-1, in the rat kidney. IR injury was induced by clamping both renal pedicles for 30 min and animals were killed at 6 h after the reperfusion. IR injury decreased blood bicarbonate level, but did not persistently alter pH, Na+, K+, or Cl−. In control kidneys, occludin immunoreactivity was intense in the tight junctions in the thick ascending limb, distal convoluted tubule, and collecting duct, moderate in the thin limbs of the loop of Henle, and was not detected in the proximal tubule, glomerulus, and blood vessels. ZO-1 was expressed in the same sites in which occludin was expressed, and additionally was also expressed in the proximal tubule, glomerulus, and vascular endothelial cells. IR kidneys exhibited damaged renal tubular epithelial cells in both proximal tubule and collecting duct segments in the outer medulla. In the collecting duct, the response of intercalated cells and principal cells differed. Following IR injury, intercalated cells, but not principal cells, lost their normal epithelial polarity and were frequently extruded into the tubule lumen. Occludin, instead of being localized to tight junctions, was localized diffusely in the cytoplasm in intercalated cells of IR kidneys. Principal cells, in contrast, were not detectably affected and neither occludin nor ZO-1 expression were altered in response to IR injury. The normal localization of ZO-1 expression to tight junction sites in both the proximal tubule and collecting duct was altered in response to IR, and, instead, ZO-1 expression was present diffusely in the cytoplasm. IR injury did not alter detectably either occludin or ZO-1 localization to the tight junction of the thick ascending limb cells. The abundance of total occludin protein by immunoblot analysis was not changed with IR injury. These results demonstrate that renal IR injury causes tight junction disruptions in both the proximal tubule and the collecting duct, and that altered distribution of the tight junction protein, occludin, may play a critical role in the collecting duct dysfunction which IR induces.
Collapse
Affiliation(s)
- Su-Youn Lee
- Department of Anatomy, Ewha Womans University School of Medicine, 911-1 Mok-6-dong, Yangcheon-ku, Seoul, 158-710 Korea
| | - Jung-A Shin
- Department of Anatomy, Ewha Womans University School of Medicine, 911-1 Mok-6-dong, Yangcheon-ku, Seoul, 158-710 Korea
| | - H. Moo Kwon
- Division of Nephrology, University of Maryland School of Medicine, Baltimore, MD USA
| | - I. David Weiner
- Division of Nephrology, University of Florida College of Medicine, Gainesville, FL USA
- Nephrology Section, NF/SGVHS, Gainesville, FL USA
| | - Ki-Hwan Han
- Department of Anatomy, Ewha Womans University School of Medicine, 911-1 Mok-6-dong, Yangcheon-ku, Seoul, 158-710 Korea
| |
Collapse
|
30
|
Li L, Black R, Ma Z, Yang Q, Wang A, Lin F. Use of mouse hematopoietic stem and progenitor cells to treat acute kidney injury. Am J Physiol Renal Physiol 2011; 302:F9-F19. [PMID: 21937606 DOI: 10.1152/ajprenal.00377.2011] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
New and effective treatment for acute kidney injury remains a challenge. Here, we induced mouse hematopoietic stem and progenitor cells (HSPC) to differentiate into cells that partially resemble a renal cell phenotype and tested their therapeutic potential. We sequentially treated HSPC with a combination of protein factors for 1 wk to generate a large number of cells that expressed renal developmentally regulated genes and protein. Cell fate conversion was associated with increased histone acetylation on promoters of renal-related genes. Further treatment of the cells with a histone deacetylase inhibitor improved the efficiency of cell conversion by sixfold. Treated cells formed tubular structures in three-dimensional cultures and were integrated into tubules of embryonic kidney organ cultures. When injected under the renal capsule, they integrated into renal tubules of postischemic kidneys and expressed the epithelial marker E-cadherin. No teratoma formation was detected 2 and 6 mo after cell injection, supporting the safety of using these cells. Furthermore, intravenous injection of the cells into mice with renal ischemic injury improved kidney function and morphology by increasing endogenous renal repair and decreasing tubular cell death. The cells produced biologically effective concentrations of renotrophic factors including VEGF, IGF-1, and HGF to stimulate epithelial proliferation and tubular repair. Our study indicates that hematopoietic stem and progenitor cells can be converted to a large number of renal-like cells within a short period for potential treatment of acute kidney injury.
Collapse
Affiliation(s)
- Ling Li
- Dept. of Pediatrics, Columbia Univ. College of Physicians and Surgeons, 630 West 168th St., New York, NY 10032, USA
| | | | | | | | | | | |
Collapse
|
31
|
Histological and functional renal alterations caused by Bothrops alternatus snake venom: Expression and activity of Na+/K+-ATPase. Biochim Biophys Acta Gen Subj 2011; 1810:895-906. [DOI: 10.1016/j.bbagen.2011.06.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Revised: 05/23/2011] [Accepted: 06/01/2011] [Indexed: 11/21/2022]
|
32
|
Abstract
The integrity and function of many vertebrate organs depend on cellular barriers that are mainly formed by intercellular protein complexes of the plasma membrane. These cell-cell contacts, tight junctions (TJs), exhibit the most apical localization in the lateral membrane; they regulate the permeability of the paracellular space between opposing epithelial and endothelial cells. This Forum reviews the currently available data on the influence of oxidative stress and the effects of antioxidative mechanisms on TJ proteins and on tissue barrier functions inseparably linked to these proteins. The contributions are focused on the most important transmembranal and membrane-associated TJ proteins and on tissue barriers characterized by predominant involvement of the TJs, and alterations at the molecular and functional levels induced by redox signaling are also discussed. This Forum demonstrates that cell barriers are highly sensitive to oxidative stress but also respond to antioxidative intervention. However, our knowledge of the molecular basis of the specific mechanisms responsible for functional disturbances remains limited and needs further investigations.
Collapse
|
33
|
Tyritzis SI, Zachariades M, Evangelou K, Gorgoulis VG, Kyroudi-Voulgari A, Pavlakis K, Troupis TG, Constantinides CA. Effects of Prolonged Warm and Cold Ischemia in a Solitary Kidney Animal Model after Partial Nephrectomy: An Ultrastructural Investigation. Ultrastruct Pathol 2011; 35:60-5. [DOI: 10.3109/01913123.2010.542880] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
34
|
Acute renal failure is NOT an "acute renal success"--a clinical study on the renal oxygen supply/demand relationship in acute kidney injury. Crit Care Med 2010; 38:1695-701. [PMID: 20512036 DOI: 10.1097/ccm.0b013e3181e61911] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVES Acute kidney injury occurs frequently after cardiac or major vascular surgery and is believed to be predominantly a consequence of impaired renal oxygenation. However, in patients with acute kidney injury, data on renal oxygen consumption (RVO2), renal blood flow, glomerular filtration, and renal oxygenation, i.e., the renal oxygen supply/demand relationship, are lacking and current views on renal oxygenation in the clinical situation of acute kidney injury are presumptive and largely based on experimental studies. DESIGN Prospective, two-group comparative study. SETTING Cardiothoracic intensive care unit of a tertiary center. PATIENTS Postcardiac surgery patients with (n = 12) and without (n = 37) acute kidney injury were compared with respect to renal blood flow, glomerular filtration, RVO2, and renal oxygenation. INTERVENTIONS None MEASUREMENTS AND MAIN RESULTS Data on systemic hemodynamics (pulmonary artery catheter) and renal variables were obtained during two 30-min periods. Renal blood flow was measured using two independent techniques: the renal vein thermodilution technique and the infusion clearance of paraaminohippuric acid, corrected for renal extraction of paraaminohippuric acid. The filtration fraction was measured by the renal extraction of Cr-EDTA and the renal sodium resorption was measured as the difference between filtered and excreted sodium. Renal oxygenation was estimated from the renal oxygen extraction. Cardiac index and mean arterial pressure did not differ between the two groups. In the acute kidney injury group, glomerular filtration (-57%), renal blood flow (-40%), filtration fraction (-26%), and sodium resorption (-59%) were lower, renal vascular resistance (52%) and renal oxygen extraction (68%) were higher, whereas there was no difference in renal oxygen consumption between groups. Renal oxygen consumption for one unit of reabsorbed sodium was 2.4 times higher in acute kidney injury. CONCLUSIONS Renal oxygenation is severely impaired in acute kidney injury after cardiac surgery, despite the decrease in glomerular filtration and tubular workload. This was caused by a combination of renal vasoconstriction and tubular sodium resorption at a high oxygen demand.
Collapse
|
35
|
Park KM, Fogelgren B, Zuo X, Kim J, Chung DC, Lipschutz JH. Exocyst Sec10 protects epithelial barrier integrity and enhances recovery following oxidative stress, by activation of the MAPK pathway. Am J Physiol Renal Physiol 2010; 298:F818-26. [PMID: 20053792 DOI: 10.1152/ajprenal.00596.2009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Cell-cell contacts are essential for epithelial cell function, and disruption is associated with pathological conditions including ischemic kidney injury. We hypothesize that the exocyst, a highly-conserved eight-protein complex that targets secretory vesicles carrying membrane proteins, is involved in maintaining renal epithelial barrier integrity. Accordingly, increasing exocyst expression in renal tubule cells may protect barrier function from oxidative stress resulting from ischemia and reperfusion (I/R) injury. When cultured on plastic, Madin-Darby canine kidney (MDCK) cells overexpressing Sec10, a central exocyst component, formed domes showing increased resistance to hydrogen peroxide (H2O2). Transepithelial electric resistance (TER) of Sec10-overexpressing MDCK cells grown on Transwell filters was higher than in control MDCK cells, and the rate of TER decrease following H2O2 treatment was less in Sec10-overexpressing MDCK cells compared with control MDCK cells. After removal of H2O2, TER returned to normal more rapidly in Sec10-overexpressing compared with control MDCK cells. In collagen culture MDCK cells form cysts, and H2O2 treatment damaged Sec10-overexpressing MDCK cell cysts less than control MDCK cell cysts. The MAPK pathway has been shown to protect animals from I/R injury. Levels of active ERK, the final MAPK pathway step, were higher in Sec10-overexpressing compared with control MDCK cells. U0126 inhibited ERK activation, exacerbated the H2O2-induced decrease in TER and cyst disruption, and delayed recovery of TER following H2O2 removal. Finally, in mice with renal I/R injury, exocyst expression decreased early and returned to normal concomitant with functional recovery, suggesting that the exocyst may be involved in the recovery following I/R injury.
Collapse
Affiliation(s)
- Kwon Moo Park
- Department of Medicine, University of Pennsylvania, and the Veterans Administration Medical Center, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | |
Collapse
|
36
|
Kwon O, Hong SM, Sutton TA, Temm CJ. Preservation of peritubular capillary endothelial integrity and increasing pericytes may be critical to recovery from postischemic acute kidney injury. Am J Physiol Renal Physiol 2008; 295:F351-9. [PMID: 18562634 DOI: 10.1152/ajprenal.90276.2008] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Decreased renal blood flow following an ischemic insult contributes to a reduction in glomerular filtration. However, little is known about the underlying cellular or subcellular mechanisms mediating reduced renal blood flow in human ischemic acute kidney injury (AKI) or acute renal failure (ARF). To examine renal vascular injury following ischemia, intraoperative graft biopsies were performed after reperfusion in 21 cadaveric renal allografts. Confocal fluorescence microscopy was utilized to examine vascular smooth muscle and endothelial cell integrity as well as peritubular interstitial pericytes in the biopsies. The reperfused, transplanted kidneys exhibited postischemic injury to the renal vasculature, as demonstrated by disorganization/disarray of the actin cytoskeleton in vascular smooth muscle cells and disappearance of von Willebrand factor from vascular endothelial cells. Damage to peritubular capillary endothelial cells was more severe in subjects destined to have sustained ARF than in those with rapid recovery of their graft function. In addition, peritubular pericytes/myofibroblasts were more pronounced in recipients destined to recover than those with sustained ARF. Taken together, these data suggest damage to the renal vasculature occurs after ischemia-reperfusion in human kidneys. Preservation of peritubular capillary endothelial integrity and increasing pericytes may be critical to recovery from postischemic AKI.
Collapse
Affiliation(s)
- Osun Kwon
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA.
| | | | | | | |
Collapse
|
37
|
Zhu L, Hatakeyama J, Chen C, Shastri A, Poon K, Forte JG. Comparative study of ezrin phosphorylation among different tissues: more is good; too much is bad. Am J Physiol Cell Physiol 2008; 295:C192-202. [PMID: 18480298 DOI: 10.1152/ajpcell.00159.2008] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In a comparison of three different tissues, the membrane cytoskeleton linker protein ezrin was found to assume high levels of phosphorylation on threonine-567 (T567) in the brush border membranes of renal proximal tubule cells and small intestine enterocytes, in contrast to the apical canalicular membrane of gastric parietal cells. Together with an earlier observation that increased T567 phosphorylation is associated with more elaborate microvilli in parietal cells, this comparative study suggested a higher phosphorylation level requirement for the denser and more uniform distribution of microvilli at brush border surfaces. Using a kinase inhibitor, staurosporin, and metabolic inhibitor, sodium azide, relatively high turnover of ezrin T567 phosphorylation was observed in all three epithelia. Aiming to understand the role of phosphorylation turnover in these tissues, detergent extraction analysis of gastric glands and proximal tubules revealed that an increased phosphorylation on ezrin T567 greatly enhanced its association with F-actin, while ezrin-membrane interaction persisted regardless of the changes of phosphorylation level on ezrin T567. Finally, expression of Thr567Asp mutant ezrin, which mimics the phospho-ezrin state but does not allow turnover, caused aberrant growth of membrane projections in cultured proximal tubule cells, consistent with what had previously been observed in several cell lines and gastric parietal cells. These results fit into a model of surface plasticity, which posits that the turnover of phosphorylation on T567 empowers ezrin to relax and reposition membrane to the underlying cytoskeleton under varying conditions of filament growth or rapid membrane expansion (or depletion).
Collapse
Affiliation(s)
- Lixin Zhu
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720-3200, USA
| | | | | | | | | | | |
Collapse
|
38
|
Li L, Truong P, Igarashi P, Lin F. Renal and Bone Marrow Cells Fuse after Renal Ischemic Injury. J Am Soc Nephrol 2007; 18:3067-77. [DOI: 10.1681/asn.2007030284] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
|
39
|
Myers BD. Pathogenetic Processes in Human Acute Renal Failure. Semin Dial 2007. [DOI: 10.1111/j.1525-139x.1996.tb00305.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
40
|
Tuğtepe H, Sener G, Biyikli NK, Yüksel M, Cetinel S, Gedik N, Yeğen BC. The protective effect of oxytocin on renal ischemia/reperfusion injury in rats. ACTA ACUST UNITED AC 2007; 140:101-8. [PMID: 17261335 DOI: 10.1016/j.regpep.2006.11.026] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2006] [Revised: 11/09/2006] [Accepted: 11/11/2006] [Indexed: 11/16/2022]
Abstract
AIM Oxytocin was previously shown to have anti-inflammatory effects in different inflammation models. The major objective of the present study was to evaluate the protective role of oxytocin (OT) in protecting the kidney against ischemia/reperfusion (I/R) injury. MATERIALS AND METHODS Male Wistar albino rats (250-300 g) were unilaterally nephrectomized, and subjected to 45 min of renal pedicle occlusion followed by 6 h of reperfusion. OT (1 mg/kg, ip) or vehicle was administered 15 min prior to ischemia and was repeated immediately before the reperfusion period. At the end of the reperfusion period, rats were decapitated and kidney samples were taken for histological examination or determination of malondialdehyde (MDA), an end product of lipid peroxidation; glutathione (GSH), a key antioxidant; and myeloperoxidase (MPO) activity, an index of tissue neutrophil infiltration. Creatinine and urea concentrations in blood were measured for the evaluation of renal function, while TNF-alpha and lactate dehydrogenase (LDH) levels were determined to evaluate generalized tissue damage. Formation of reactive oxygen species in renal tissue samples was monitored by chemiluminescence technique using luminol and lucigenin probes. RESULTS The results revealed that I/R injury increased (p<0.01-0.001) serum urea, creatinine, TNF-alpha and LDH levels, as well as MDA, MPO and reactive oxygen radical levels in the renal tissue, while decreasing renal GSH content. However, alterations in these biochemical and histopathological indices due to I/R injury were attenuated by OT treatment (p<0.05-0.001). CONCLUSIONS Since OT administration improved renal function and microscopic damage, along with the alleviation of oxidant tissue responses, it appears that oxytocin protects renal tissue against I/R-induced oxidative damage.
Collapse
Affiliation(s)
- Halil Tuğtepe
- Marmara University, School of Medicine, Department of Pediatric Surgery, Istanbul, Turkey.
| | | | | | | | | | | | | |
Collapse
|
41
|
Prozialeck WC, Edwards JR. Cell adhesion molecules in chemically-induced renal injury. Pharmacol Ther 2007; 114:74-93. [PMID: 17316817 PMCID: PMC1913814 DOI: 10.1016/j.pharmthera.2007.01.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2007] [Accepted: 01/05/2007] [Indexed: 01/15/2023]
Abstract
Cell adhesion molecules are integral cell-membrane proteins that maintain cell-cell and cell-substrate adhesion and in some cases act as regulators of intracellular signaling cascades. In the kidney, cell adhesion molecules, such as the cadherins, the catenins, the zonula occludens protein-1 (ZO-1), occludin and the claudins are essential for maintaining the epithelial polarity and barrier integrity that are necessary for the normal absorption/excretion of fluid and solutes. A growing volume of evidence indicates that these cell adhesion molecules are important early targets for a variety of nephrotoxic substances including metals, drugs, and venom components. In addition, it is now widely appreciated that molecules, such as intracellular adhesion molecule-1 (ICAM-1), integrins, and selectins play important roles in the recruitment of leukocytes and inflammatory responses that are associated with nephrotoxic injury. This review summarizes the results of recent in vitro and in vivo studies indicating that these cell adhesion molecules may be primary molecular targets in many types of chemically-induced renal injury. Some of the specific agents that are discussed include cadmium (Cd), mercury (Hg), bismuth (Bi), cisplatin, aminoglycoside antibiotics, S-(1,2-dichlorovinyl)-l-cysteine (DCVC), and various venom toxins. This review also includes a discussion of the various mechanisms, by which these substances can affect cell adhesion molecules in the kidney.
Collapse
Affiliation(s)
- Walter C Prozialeck
- Department of Pharmacology, Midwestern University, 555 31st Street, Downers Grove, IL 60515, United States.
| | | |
Collapse
|
42
|
A novel method for the evaluation of proximal tubule epithelial cellular necrosis in the intact rat kidney using ethidium homodimer. BMC PHYSIOLOGY 2007; 7:1. [PMID: 17319948 PMCID: PMC1810561 DOI: 10.1186/1472-6793-7-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2006] [Accepted: 02/23/2007] [Indexed: 01/08/2023]
Abstract
BACKGROUND Ethidium homodimer is a cell-membrane impermeant nuclear fluorochrome that has been widely used to identify necrotic cells in culture. Here, we describe a novel technique for evaluating necrosis of epithelial cells in the proximal tubule that involves perfusing ethidium homodimer through the intact rat kidney. As a positive control for inducing necrosis, rats were treated with 3.5, 1.75, 0.87 and 0.43 mg/kg mercuric chloride (Hg2+, intraperitoneal), treatments which have previously been shown to rapidly cause dose-dependent necrosis of the proximal tubule. Twenty-four h after the administration of Hg2+, ethidium homodimer (5 microM) was perfused through the intact left kidney while the animal was anesthetized. The kidney was then removed, placed in embedding medium, frozen and cryosectioned at a thickness of 5 microm. Sections were permeabilized with -20 degrees C methanol and then stained with 4',6-diamidino-2-phenylindole (DAPI) to label total nuclei. Total cell number was determined from the DAPI staining in random microscopic fields and the number of necrotic cells in the same field was determined by ethidium homodimer labeling. RESULTS The Hg2+-treated animals showed a dose-dependent increase in the number of ethidium labeled cells in the proximal tubule, but not in other segments of the nephron. Other results showed that a nephrotoxic dose of gentamicin also caused a significant increase in the number of ethidium labeled cells in the proximal tubule. CONCLUSION These results indicate that this simple and sensitive perfusion technique can be used to evaluate cellular necrosis in the proximal tubule with the three-dimensional cyto-architecture intact.
Collapse
|
43
|
Genescà M, Sola A, Hotter G. Actin cytoskeleton derangement induces apoptosis in renal ischemia/reperfusion. Apoptosis 2006; 11:563-71. [PMID: 16528472 DOI: 10.1007/s10495-006-4937-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
UNLABELLED This study evaluated whether cytoskeletal alterations during the ischemic conditions associated with kidney preservation could determine apoptosis. Cytoskeletal alterations are among the main effects of ischemia and may induce apoptosis. Rat kidneys were preserved in University of Wisconsin (UW) solution for 24 h. Some groups of animals underwent 45 min of warm ischemia (WI) to evaluate its effect on both the actin cytoskeleton and apoptosis (assessed by caspase-3 activity and TUNEL staining). Swinholide A (SwinA) and Latrunculin B (LB), two actin cytoskeleton-targeted agents, were administered to assess the effect of direct actin disruption on apoptosis. Jasplakinolide (JP), a compound that stabilizes actin filaments, was administered to evaluate the effect of actin stabilization. Apoptosis was evaluated at 3 h of ex vivo reperfusion using the isolated perfused rat kidney (IPK) model. RESULTS Apoptosis increased during reperfusion with WI or administration of actin disruptor agents. Administration of stabilizing agents reversed apoptosis in kidneys that had previously undergone WI or had received an actin disruptor agent. CONCLUSION The disruption of the actin cytoskeleton during ischemic conditions associated with kidney preservation induces apoptosis upon reperfusion through caspase-3 activation.
Collapse
Affiliation(s)
- M Genescà
- Department of Experimental Pathology, IIBB-CSIC, IDIBAPS, Barcelona, Spain
| | | | | |
Collapse
|
44
|
Abstract
Albumin is the most abundant plasmaprotein serving multiple functions as a carrier of metabolites, hormones, vitamins, and drugs, as an acid/base buffer, as antioxidant and by supporting the oncotic pressure and volume of the blood. The presence of albumin in urine is considered to be the result of the balance between glomerular filtration and tubular reabsorption. Albuminuria has been accepted as an independent risk factor and a marker for renal as well as cardiovascular disease, and during the past decade, evidence has suggested that albumin itself may cause progression of renal disease. Thus, the reduction of proteinuria and, in particular, albuminuria has become a target in itself to prevent deterioration of renal function. Studies have shown albumin and its ligands to induce expression of inflammatory and fibrogenic mediators, and it has been hypothesized that increased filtration of albumin causes excessive tubular reabsorption, resulting in inflammation and fibrosis, resulting in the loss of renal function. In addition, it is known that tubular dysfunction in itself may cause albuminuria owing to decreased reabsorption of filtered albumin, and, recently, it has been suggested that significant amounts of albumin fragments are excreted in the urine as a result of tubular degradation. Thus, although both tubular and glomerular dysfunction influences renal handling of albumin, it appears that tubular reabsorption plays a central role in mediating the effects of albumin on renal function. The present paper will review the mechanisms for tubular albumin uptake and the possible implications for the development of renal disease.
Collapse
Affiliation(s)
- H Birn
- Department of Cell Biology, Institute of Anatomy, University of Aarhus, Aarhus C, Denmark
| | | |
Collapse
|
45
|
Abstract
The epithelial tight junction (TJ) has three major functions. As a "gate," it serves as a regulatory barrier separating and maintaining biological fluid compartments of different composition. As a "fence," it generates and maintains the apicobasal polarity of cells that form the confluent epithelium. Finally, the TJ proteins form a trafficking and signaling platform that regulates cell growth, proliferation, differentiation, and dedifferentiation. Six examples are selected that illustrate the emerging link between TJ dysfunction and kidney disease. First, the glomerular slit diaphragm (GSD) is evolved, in part, from the TJ and, on maturation, exhibits all three functions of the TJ. GSD dysfunction leads to proteinuria and, in some instances, podocyte dedifferentiation and proliferation. Second, accumulating evidence supports epithelial-mesenchymal transformation (EMT) as a major player in renal fibrosis, the final common pathway that leads to end-stage renal failure. EMT is characterized by a loss of cell-cell contact and apicobasal polarity, which are hallmarks of TJ dysfunction. Third, in autosomal dominant polycystic kidney disease, mutations of the polycystins may disrupt their known interactions with the apical junction complex, of which the TJ is a major component. This can lead to disturbances in epithelial polarity regulation with consequent abnormal tubulogenesis and cyst formation. Fourth, evidence for epithelial barrier and polarity dysregulation in the pathogenesis of ischemic acute renal failure will be summarized. Fifth, the association between mutations of paracellin-1, the first TJ channel identified, and clinical disorders of magnesium and calcium wasting and bovine renal fibrosis will be used to highlight an integral TJ protein that can serve multiple TJ functions. Finally, the role of WNK4 protein kinase in shunting chloride across the TJ of the distal nephron will be addressed.
Collapse
Affiliation(s)
- David B N Lee
- Division of Nephrology, Veterans Affairs Greater Los Angeles Healthcare System, California, USA.
| | | | | |
Collapse
|
46
|
Trumper L, Coux G, Monasterolo LA, Molinas S, García VMC, Elías MM. Effect of acetaminophen on the membrane anchoring of Na+, K+ATPase of rat renal cortical cells. Biochim Biophys Acta Mol Basis Dis 2005; 1740:332-9. [PMID: 15949700 DOI: 10.1016/j.bbadis.2004.09.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2003] [Revised: 09/20/2004] [Accepted: 09/24/2004] [Indexed: 10/26/2022]
Abstract
In previous works we reported that the administration of a toxic dose of acetaminophen (APAP) induces acute renal failure (ARF) and promotes changes on Na(+), K(+)ATPase distribution in renal proximal plasma membranes. In the present work, we analyzed if APAP could promote the dissociation of Na(+), K(+)ATPase from its membrane anchorage. The participation of calpain activation was also evaluated. We analyzed the Triton X-100 extractability of Na(+), K(+)ATPase in freshly isolated cortical cell suspensions incubated with different APAP concentrations (0.1, 1, 10 and 100 mM). Both alpha(1) and beta(1) subunits were studied by Western blot. APAP promoted the increment of both subunits abundance in the Triton-soluble fraction. Calpain activation was detected in the membrane fractions of cells incubated with APAP. Incubation with APAP 0.1, 1 and 10 mM did not promote an increment in LDH release compared with controls, while APAP 100 mM promoted an increased LDH release. Our results show that incubation of proximal cells with sublethal and lethal APAP concentrations promotes the detachment of Na(+), K(+)ATPase from its membrane anchoring. Inhibition of calpain activation by SJA 7029 protected against APAP-induced membrane damage but not against APAP-induced increase of the Triton X-100 extractability of Na(+), K(+)ATPase.
Collapse
Affiliation(s)
- Laura Trumper
- Consejo de Investigaciones de la Universidad Nacional de Rosario (CIUNR), República Argentina.
| | | | | | | | | | | |
Collapse
|
47
|
Runembert I, Couette S, Federici P, Colucci-Guyon E, Babinet C, Briand P, Friedlander G, Terzi F. Recovery of Na-glucose cotransport activity after renal ischemia is impaired in mice lacking vimentin. Am J Physiol Renal Physiol 2004; 287:F960-8. [PMID: 15238351 DOI: 10.1152/ajprenal.00064.2004] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Vimentin, an intermediate filament protein mainly expressed in mesenchyma-derived cells, is reexpressed in renal tubular epithelial cells under many pathological conditions, characterized by intense cell proliferation. Whether vimentin reexpression is only a marker of cell dedifferentiation or is instrumental in the maintenance of cell structure and/or function is still unknown. Here, we used vimentin knockout mice ( Vim−/−) and an experimental model of acute renal injury (30-min bilateral renal ischemia) to explore the role of vimentin. Bilateral renal ischemia induced an initial phase of acute tubular necrosis that did not require vimentin and was similar, in terms of morphological and functional changes, in Vim+/+and Vim−/−mice. However, vimentin was essential to favor Na-glucose cotransporter 1 localization to brush-border membranes and to restore Na-glucose cotransport activity in regenerating tubular cells. We show that the effect of vimentin inactivation is specific and results in persistent glucosuria. We propose that vimentin is part of a structural network that favors carrier localization to plasma membranes to restore transport activity in injured kidneys.
Collapse
Affiliation(s)
- Isabelle Runembert
- INSERM U426, Faculté de Médecine Xavier Bichat, 16 Rue Henri Huchard, BP 416, 75870 Paris Cedex 18, France
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Affiliation(s)
- Bruce A Molitoris
- Indiana University School of Medicine, Nephrology Division, Indianapolis, Indiana 46202, USA.
| |
Collapse
|
49
|
George SK, Meyer TN, Abdeen O, Bush KT, Nigam SK. Tunicamycin preserves intercellular junctions, cytoarchitecture, and cell-substratum interactions in ATP-depleted epithelial cells. Biochem Biophys Res Commun 2004; 322:223-31. [PMID: 15313195 DOI: 10.1016/j.bbrc.2004.07.097] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2004] [Indexed: 11/29/2022]
Abstract
Pretreatment with the nucleoside antibiotic tunicamycin was found to protect cultured renal epithelial cells in the face of ATP-depletion, in large part by preserving junctional and cellular architecture. Tunicamycin pretreatment of Madin-Darby canine kidney cells not only preserved E-cadherin staining at the plasma membrane, but also inhibited ATP-depletion-mediated E-cadherin degradation. Electron microscopic analysis, together with the preservation of the staining patterns of the tight junction marker ZO-1, the apical/microvillar marker gp135, and basolateral marker Na/K-ATPase suggested that tunicamycin preserved the junctional complex and the polarized epithelial cell phenotype. Tunicamycin pretreatment also prevented reductions in the filamentous actin content of the cells, as well as preserving Golgi architecture. Moreover, a quantitative measure of cell adhesion demonstrated that tunicamycin pretreatment resulted in a fivefold increase in attachment of cells to the substratum (77% versus 16%). Thus, pretreatment with tunicamycin protects polarized epithelial cells from ischemic injury through the preservation of epithelial cell architecture, intercellular junctions, and cell-substratum interactions in the setting of intracellular ATP-depletion.
Collapse
Affiliation(s)
- Sathish K George
- Department of Medicine, School of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0693, USA
| | | | | | | | | |
Collapse
|
50
|
Schrier RW, Wang W, Poole B, Mitra A. Acute renal failure: definitions, diagnosis, pathogenesis, and therapy. J Clin Invest 2004. [DOI: 10.1172/jci200422353] [Citation(s) in RCA: 518] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|