1
|
Kira S, Namba T, Hiraishi M, Nakamura T, Otani Y, Kon Y, Ichii O. Species-specific histological characterizations of renal tubules and collecting ducts in the kidneys of cats and dogs. PLoS One 2024; 19:e0306479. [PMID: 38959226 PMCID: PMC11221681 DOI: 10.1371/journal.pone.0306479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 06/18/2024] [Indexed: 07/05/2024] Open
Abstract
The histomorphological features of normal kidneys in cats and dogs have been revealed despite the high susceptibility of cats to tubulointerstitial damage. Herein, the histological characteristics of the two species were compared. Cytoplasmic lipid droplets (LDs) were abundant in the proximal convoluted tubules (PCTs) of cats aged 23-27 months but scarce in dogs aged 24-27 months. LDs were rarely observed in the distal tubules (DTs) and collecting ducts (CDs) of either species, as visualized by the expression of Tamm-Horsfall protein 1, calbindin-D28K, and aquaporin 2. The occupational area ratio of proximal tubules (PTs) in the renal cortex was higher, but that of DTs or CDs was significantly lower in adult cats than in dogs. Single PT epithelial cells were larger, but PCT, DT, and CD lumens were significantly narrower in adult cats than in dogs. Unlike adults, young cats at 6 months exhibited significantly abundant cytoplasmic LDs in proximal straight tubules, indicating lipid metabolism-related development. Histochemistry of the 21 lectins also revealed variations in glycosylation across different renal tubules and CDs in both species. Sodium-glucose cotransporter 2 was expressed only in PTs, excluding the proximal straight tubules with few LDs in adult cats or the PCTs of young cats and adult dogs. These findings are crucial for understanding species-specific characteristics of renal histomorphology and pathogenesis.
Collapse
Affiliation(s)
- Shunnosuke Kira
- Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Takashi Namba
- Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Masaya Hiraishi
- Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Teppei Nakamura
- Laboratory of Laboratory Animal Science and Medicine, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Yuki Otani
- Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
- One Health Research Center, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Yasuhiro Kon
- Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Osamu Ichii
- Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
- One Health Research Center, Hokkaido University, Sapporo, Hokkaido, Japan
| |
Collapse
|
2
|
Roskosch J, Huynh-Do U, Rudloff S. Lectin-mediated, time-efficient, and high-yield sorting of different morphologically intact nephron segments. Pflugers Arch 2024; 476:379-393. [PMID: 38091061 PMCID: PMC10847228 DOI: 10.1007/s00424-023-02894-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 11/20/2023] [Accepted: 11/24/2023] [Indexed: 02/08/2024]
Abstract
The kidney is a highly complex organ equipped with a multitude of miniscule filter-tubule units called nephrons. Each nephron can be subdivided into multiple segments, each with its own morphology and physiological function. To date, conventional manual approaches to isolate specific nephron segments are very laborious, time-consuming, often limited to only a specific segment, and typically have low yield. Here, we describe a novel, unconventional method that is superior in many aspects to previous protocols by combining low-cost fluorophore-conjugated lectins or agglutinins (Flaggs) with flow sorting. This allows the simultaneous separation of different nephron segments with preserved 3D morphology from mouse or human samples in under 3 h. Using a 200-µm nozzle and 5 psi, glomeruli, proximal, or distal convoluted tubules are sorted with Cy3-labeled Sambucus Nigra agglutinin (SNA-Cy3), Fluorescein-labeled Lotus Tetragonolobus lectin (LTL-FITC), or Pacific Blue-labeled soybean agglutinin (SBA-PB), respectively. Connecting tubules and collecting ducts are sorted by double-positive SBA-PB and SNA-Cy3 signals, while thick ascending limb segments are characterized by the absence of any Flaggs labeling. From two mouse kidneys, this yields 37-521 ng protein/s or 0.71-16.71 ng RNA/s, depending on the specific nephron segment. The purity of sorted segments, as assessed by mRNA expression level profiling of 15 genes, is very high with a 96.1-fold median enrichment across all genes and sorted segments. In summary, our method represents a simple, straightforward, cost-effective, and widely applicable tool yielding high amounts of pure and morphologically largely intact renal tubule materials with the potential to propel nephron segment-specific research.
Collapse
Affiliation(s)
- Jessica Roskosch
- Division of Nephrology and Hypertension, University of Bern and University Hospital Bern, Freiburgstrasse 15, CH-3010, Bern, Switzerland
| | - Uyen Huynh-Do
- Division of Nephrology and Hypertension, University of Bern and University Hospital Bern, Freiburgstrasse 15, CH-3010, Bern, Switzerland
| | - Stefan Rudloff
- Division of Nephrology and Hypertension, University of Bern and University Hospital Bern, Freiburgstrasse 15, CH-3010, Bern, Switzerland.
| |
Collapse
|
3
|
Freund P, Skopnik CM, Metzke D, Goerlich N, Klocke J, Grothgar E, Prskalo L, Hiepe F, Enghard P. Addition of formaldehyde releaser imidazolidinyl urea and MOPS buffer to urine samples enables delayed processing for flow cytometric analysis of urinary cells: A simple, two step conservation method of urinary cells for flow cytometry. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2023; 104:417-425. [PMID: 36880455 DOI: 10.1002/cyto.b.22117] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 02/03/2023] [Accepted: 02/13/2023] [Indexed: 03/08/2023]
Abstract
INTRODUCTION Kidney diseases are a major health concern worldwide. Currently there is a large unmet need for novel biomarkers to non-invasively diagnose and monitor kidney diseases. Urinary cells are promising biomarkers and their analysis by flow cytometry has demonstrated its utility in diverse clinical settings. However, up to date this methodology depends on fresh samples, as cellular event counts and the signal-to-noise-ratio deter over time. Here we developed an easy-to-use two-step preservation method for conservation of urine samples for subsequent flow cytometry. METHODS The protocol utilizes a combination of the formaldehyde releasing agent imidazolidinyl urea (IU) and MOPS buffer, leading to gentle fixation of urinary cells. RESULTS The preservation method increases acceptable storing time of urine samples from several hours to up to 6 days. Cellular event counts and staining properties of cells remain comparable to fresh untreated samples. OUTLOOK The hereby presented preservation method facilitates future investigations on flow cytometry of urinary cells as potential biomarkers and may enable broad implementation in clinical practice.
Collapse
Affiliation(s)
- Paul Freund
- Department of Nephrology and Medical Intensive Care, Charité - Universital Hospital Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Rheumatism Research Center Berlin (DRFZ), An Institute of the Leibniz Foundation, Berlin, Germany
| | - Christopher M Skopnik
- Department of Nephrology and Medical Intensive Care, Charité - Universital Hospital Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Rheumatism Research Center Berlin (DRFZ), An Institute of the Leibniz Foundation, Berlin, Germany
| | - Diana Metzke
- Department of Nephrology and Medical Intensive Care, Charité - Universital Hospital Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Rheumatism Research Center Berlin (DRFZ), An Institute of the Leibniz Foundation, Berlin, Germany
| | - Nina Goerlich
- Department of Nephrology and Medical Intensive Care, Charité - Universital Hospital Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Rheumatism Research Center Berlin (DRFZ), An Institute of the Leibniz Foundation, Berlin, Germany
| | - Jan Klocke
- Department of Nephrology and Medical Intensive Care, Charité - Universital Hospital Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Rheumatism Research Center Berlin (DRFZ), An Institute of the Leibniz Foundation, Berlin, Germany
| | - Emil Grothgar
- Department of Nephrology and Medical Intensive Care, Charité - Universital Hospital Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Rheumatism Research Center Berlin (DRFZ), An Institute of the Leibniz Foundation, Berlin, Germany
| | - Luka Prskalo
- Department of Nephrology and Medical Intensive Care, Charité - Universital Hospital Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Rheumatism Research Center Berlin (DRFZ), An Institute of the Leibniz Foundation, Berlin, Germany
| | - Falk Hiepe
- German Rheumatism Research Center Berlin (DRFZ), An Institute of the Leibniz Foundation, Berlin, Germany
- Department of Rheumatology and Clinical Immunology, Charite - Universital Hospital Berlin, Berlin, Germany
| | - Philipp Enghard
- Department of Nephrology and Medical Intensive Care, Charité - Universital Hospital Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Rheumatism Research Center Berlin (DRFZ), An Institute of the Leibniz Foundation, Berlin, Germany
| |
Collapse
|
4
|
Buvall L, Menzies RI, Williams J, Woollard KJ, Kumar C, Granqvist AB, Fritsch M, Feliers D, Reznichenko A, Gianni D, Petrovski S, Bendtsen C, Bohlooly-Y M, Haefliger C, Danielson RF, Hansen PBL. Selecting the right therapeutic target for kidney disease. Front Pharmacol 2022; 13:971065. [DOI: 10.3389/fphar.2022.971065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022] Open
Abstract
Kidney disease is a complex disease with several different etiologies and underlying associated pathophysiology. This is reflected by the lack of effective treatment therapies in chronic kidney disease (CKD) that stop disease progression. However, novel strategies, recent scientific breakthroughs, and technological advances have revealed new possibilities for finding novel disease drivers in CKD. This review describes some of the latest advances in the field and brings them together in a more holistic framework as applied to identification and validation of disease drivers in CKD. It uses high-resolution ‘patient-centric’ omics data sets, advanced in silico tools (systems biology, connectivity mapping, and machine learning) and ‘state-of-the-art‘ experimental systems (complex 3D systems in vitro, CRISPR gene editing, and various model biological systems in vivo). Application of such a framework is expected to increase the likelihood of successful identification of novel drug candidates based on strong human target validation and a better scientific understanding of underlying mechanisms.
Collapse
|
5
|
SGLT2 Inhibitors in Type 2 Diabetes Mellitus and Heart Failure-A Concise Review. J Clin Med 2022; 11:jcm11061470. [PMID: 35329796 PMCID: PMC8952302 DOI: 10.3390/jcm11061470] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 02/28/2022] [Accepted: 03/06/2022] [Indexed: 01/25/2023] Open
Abstract
The incidence of both diabetes mellitus type 2 and heart failure is rapidly growing, and the diseases often coexist. Sodium-glucose co-transporter 2 inhibitors (SGLT2i) are a new antidiabetic drug class that mediates epithelial glucose transport at the renal proximal tubules, inhibiting glucose absorption—resulting in glycosuria—and therefore improving glycemic control. Recent trials have proven that SGLT2i also improve cardiovascular and renal outcomes, including reduced cardiovascular mortality and fewer hospitalizations for heart failure. Reduced preload and afterload, improved vascular function, and changes in tissue sodium and calcium handling may also play a role. The expected paradigm shift in treatment strategies was reflected in the most recent 2021 guidelines published by the European Society of Cardiology, recommending dapagliflozin and empagliflozin as first-line treatment for heart failure patients with reduced ejection fraction. Moreover, the recent results of the EMPEROR-Preserved trial regarding empagliflozin give us hope that there is finally an effective treatment for patients with heart failure with preserved ejection fraction. This review aims to assess the efficacy and safety of these new anti-glycemic oral agents in the management of diabetic and heart failure patients.
Collapse
|
6
|
DNA repair factor KAT5 prevents ischemic acute kidney injury through glomerular filtration regulation. iScience 2021; 24:103436. [PMID: 34877495 PMCID: PMC8633972 DOI: 10.1016/j.isci.2021.103436] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 09/10/2021] [Accepted: 11/10/2021] [Indexed: 01/03/2023] Open
Abstract
The “preconditioning effect” in AKI is a phenomenon in which an episode of ischemia-reperfusion results in tolerance to subsequent ischemia-reperfusion injury. However, its relationship between DNA damage repair has not been elucidated. Here, we show the role of KAT5 in the preconditioning effect. Preconditioning attenuated DNA damage in proximal tubular cells with elevated KAT5 expression. Ischemia-reperfusion (IR) injuries were exacerbated, and preconditioning effect vanished in proximal tubular-cell-specific KAT5 knockout mice. Investigation of tubuloglomerular feedback (TGF) by MALDI-IMS and urinary adenosine revealed that preconditioning caused attenuated TGF at least in part via KAT5. In addition, K-Cl cotransporter 3 (KCC3) expression decreased in damaged proximal tubular cells, which may be involved in accelerated TGF following IR. Furthermore, KAT5 induced KCC3 expression by maintaining chromatin accessibility and binding to the KCC3 promoter. These results suggest a novel mechanism of the preconditioning effect mediated by the promotion of DNA repair and attenuation of TGF through KAT5. KAT5-mediated DNA damage repair acts against ischemia-reperfusion (IR) injuries K-Cl cotransporter3 (KCC3) expression is decreased in damaged proximal tubular cells Decreased KCC3 may lead to AKI via acceleration of tubuloglomerular feedback KAT5 induces KCC3 expression through an epigenetic mechanism
Collapse
|
7
|
Liu Q, Kong Y, Guo X, Liang B, Xie H, Hu S, Han M, Zhao X, Feng P, Lyu Q, Dong W, Liang X, Wang W, Li C. GSK-3β inhibitor TDZD-8 prevents reduction of aquaporin-1 expression via activating autophagy under renal ischemia reperfusion injury. FASEB J 2021; 35:e21809. [PMID: 34314052 DOI: 10.1096/fj.202100549r] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/12/2021] [Accepted: 07/06/2021] [Indexed: 11/11/2022]
Abstract
Renal ischemia/reperfusion (I/R) injury is a main cause of acute kidney injury (AKI). Aquaporin (AQP)-1 water channel in the kidney is critical for the maintenance of water homeostasis and the urinary concentrating ability. Increasing evidence supports an important role of autophagy in the pathogenesis of AKI induced by renal I/R. The purpose of the present study is to investigate whether activation of autophagy prevents downregulation of AQP1 protein induced by renal I/R and potential molecular mechanisms. Renal I/R induced consistently reduced protein expression of AQP1, 2, and 3, as well as sodium cotransporters Na+ -K+ -2Cl- cotransporter and α-Na,K-ATPase, which was associated with increased urine output and decreased creatinine clearance in rats. Renal I/R also suppressed autophagy and increased inflammatory responses in the kidney. 4-Benzyl-2-methyl-1,2,4-thiadiazolidine-3,5-dione (TDZD-8), the glycogen synthase kinase-3β inhibitor, ameliorated renal injury under I/R, activated autophagy and markedly increased expression of AQPs and sodium transporters in the kidney, which was associated with improved urine output and creatinine clearance in rats. Hypoxia/reoxygenation (H/R) induced suppression of autophagy and downregulation of AQP1 in murine inner medullary collecting duct 3 (IMCD3) cells, which was fully prevented by TDZD-8 treatment. Inhibition of autophagy by 3-methyladenine or Atg5 gene knockdown attenuated recovery of AQP1 protein expression induced by TDZD-8 in IMCD3 cells with H/R. Interleukin-1 beta (IL-1β) decreased the abundance of AQP1 protein in IMCD3 cells. H/R induced increases in protein expression of nod-like receptor pyrin domain-containing 3 and IL-1β, which was reversed by TDZD-8. In conclusion, TDZD-8 treatment prevented downregulation of AQP1 expression under renal I/R injury, likely via activating autophagy and decreasing IL-1β production.
Collapse
Affiliation(s)
- Qiaojuan Liu
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yonglun Kong
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiangdong Guo
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Baien Liang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Haixia Xie
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Shan Hu
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Mengke Han
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiaoduo Zhao
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Pinning Feng
- Department of Clinical Laboratory, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qianqian Lyu
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, China
| | - Wei Dong
- Division of Nephrology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xinling Liang
- Division of Nephrology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Weidong Wang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Department of Nephrology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Chunling Li
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
8
|
Bayrasheva VK, Pchelin IY, Dobronravov VA, Babenko AY, Chefu SG, Shatalov IS, Vasilkova VN, Hudiakova NV, Ivanova AN, Andoskin PA, Grineva EN. Short-term renal and metabolic effects of low dose vildagliptin treatment added-on insulin therapy in non-proteinuric patients with type 2 diabetes: open-label randomized prospective study. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2020; 64:418-426. [PMID: 32267348 PMCID: PMC10522081 DOI: 10.20945/2359-3997000000220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 09/02/2019] [Indexed: 11/23/2022]
Abstract
Objective The aim of this randomized comparative study was to assess renal and metabolic effects of vildagliptin in insulin-treated type 2 diabetes (T2DM) patients without overt chronic kidney disease. Subjects and methods We randomized 47 insulin-treated non-proteinuric patients with satisfactory controlled T2DM and estimated glomerular filtration rate (eGFR) ≥ 60 mL/min/1.73m 2 either to continue insulin therapy (control) or to receive combined insulin-vildagliptin treatment (VIG group). We assessed eGFR using serum creatinine (eGFRcreat), cystatin C (eGFRcys), and both (eGFRcreat-cys), and urinary creatinine-adjusted excretion of albumin (UACR), type IV collagen (uCol IV/Cr), and neutrophil gelatinase-associated lipocalin (uNGAL/Cr) at baseline and after 6 months of treatment. Results Study groups were comparable in terms of age and sex (60.1 ± 6.1 years and 42.9% men in control group vs. 60.8 ± 5.2 years and 39.1% in VIG group). After 6 months of treatment, there were no significant changes in main assessed parameters in control group. VIG group demonstrated significant decrease in HbA1c, diastolic blood pressure, frequency of hypoglycemia, and high-sensitivity C-reactive protein level as compared to the changes in control group. While eGFRcreat, UACR, and uNGAL/Cr showed no significant changes after vildagliptin addition, eGFRcys, eGFRcreat-cys, and uCol IV/Cr changed significantly in comparison with control group (+7.0% [3.7;13.3]; +5.1% [1.4;8.5]; -32,8% [-55.8;-24.4], respectively, p < 0.01 each). Correlation and regression analysis revealed glucose-independent pattern of these changes. Conclusion Addition of vildagliptin to ongoing insulin therapy in patients with T2DM was associated with a reduction in uCol IV/Cr and an increase in eGFRcys and eGFRcreat-cys, independent of T2DM control parameters.
Collapse
Affiliation(s)
- Valentina K Bayrasheva
- Institute of Endocrinology, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Ivan Y Pchelin
- Department of Faculty Therapy, Saint Petersburg State University,, Saint Petersburg, Russia
| | - Vladimir A Dobronravov
- Research Institute of Nephrology, Pavlov First Saint Petersburg State Medical University, Saint Petersburg, Russia
| | - Alina Yu Babenko
- Institute of Endocrinology, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Svetlana G Chefu
- Experimental Research Laboratory, Laser Medicine Center, Pavlov First Saint Petersburg State Medical University, Saint Petersburg, Russia
| | - Ivan S Shatalov
- Scientific and Research Institute of Bioengineering, Saint Petersburg National Research University of Information, Technologies, Mechanics and Optics, Saint Petersburg, Russia
| | - Volha N Vasilkova
- Department of Internal Medicine No.1 with the Course of Endocrinology, Gomel State Medical University, Gomel, Belarus
| | - Natalia V Hudiakova
- Department of Faculty Therapy, Saint Petersburg State University,, Saint Petersburg, Russia
| | - Alexandra N Ivanova
- Department of Faculty Therapy, Saint Petersburg State University,, Saint Petersburg, Russia
| | - Pavel A Andoskin
- Laboratory of Protein Biochemistry, State Research Institute of Highly Pure BioSubstances, Saint Petersburg, Russia
| | - Elena N Grineva
- Institute of Endocrinology, Almazov National Medical Research Centre, Saint Petersburg, Russia
| |
Collapse
|
9
|
Kim J, Jeong JH, Jung J, Jeon H, Lee S, Lim JS, Go H, Oh JS, Kim YG, Lee CK, Yoo B, Hong S. Immunological characteristics and possible pathogenic role of urinary CD11c+ macrophages in lupus nephritis. Rheumatology (Oxford) 2020; 59:2135-2145. [DOI: 10.1093/rheumatology/keaa053] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 01/20/2020] [Indexed: 01/08/2023] Open
Abstract
Abstract
Objectives
Kidney-infiltrating immune cells can contribute to the pathogenesis of lupus nephritis (LN). We investigated the immunological characteristics of CD11c+ macrophages and their functions associated with the pathogenesis of LN.
Methods
CD11c+ macrophages were examined in the urine samples of patients with LN. Phenotypic markers and pro-inflammatory cytokine expression levels were analysed by flow cytometry. To determine the origin of urinary macrophages, peripheral monocytes were treated with sera from patients with systemic lupus erythematosus (SLE). The pathogenic role of CD11c+ macrophages in tubulointerstitial damage was investigated using SLE sera-treated monocytes and HK-2 cells.
Results
Urinary CD11c+ macrophages expressed pro-inflammatory cytokines, such as IL-6 and IL-1β, and resembled infiltrated monocytes rather than tissue-resident macrophages with respect to surface marker expression. CD11c+ macrophages had high expression levels of the chemokine receptor CXCR3, which were correlated with cognate chemokine IP-10 expression in urinary tubular epithelial cells. When treated with sera from SLE patients, peripheral monocytes acquired the morphological and functional characteristics of urinary CD11c+ macrophages, which was blocked by DNase treatment. Finally, SLE sera-treated monocytes induced fibronectin expression, apoptosis and cell detachment in HK-2 cells via production of IL-6.
Conclusion
CD11c+ macrophages may be involved in the pathogenesis of tubulointerstitial injury in LN.
Collapse
Affiliation(s)
- Jihye Kim
- Division of Rheumatology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Ji Hye Jeong
- Division of Rheumatology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jaehyung Jung
- Division of Rheumatology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hanwool Jeon
- Department of Neurosurgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seungjoo Lee
- Department of Neurosurgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Joon Seo Lim
- Clinical Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Heounjeong Go
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Ji Seon Oh
- Clinical Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yong-Gil Kim
- Division of Rheumatology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Chang-Keun Lee
- Division of Rheumatology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Bin Yoo
- Division of Rheumatology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seokchan Hong
- Division of Rheumatology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
10
|
Goerlich N, Brand HA, Langhans V, Tesch S, Schachtner T, Koch B, Paliege A, Schneider W, Grützkau A, Reinke P, Enghard P. Kidney transplant monitoring by urinary flow cytometry: Biomarker combination of T cells, renal tubular epithelial cells, and podocalyxin-positive cells detects rejection. Sci Rep 2020; 10:796. [PMID: 31964937 PMCID: PMC6972704 DOI: 10.1038/s41598-020-57524-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 12/31/2019] [Indexed: 01/19/2023] Open
Abstract
Creatinine and proteinuria are used to monitor kidney transplant patients. However, renal biopsies are needed to diagnose renal graft rejection. Here, we assessed whether the quantification of different urinary cells would allow non-invasive detection of rejection. Urinary cell numbers of CD4+ and CD8+ T cells, monocytes/macrophages, tubular epithelial cells (TEC), and podocalyxin(PDX)-positive cells were determined using flow cytometry and were compared to biopsy results. Urine samples of 63 renal transplant patients were analyzed. Patients with transplant rejection had higher amounts of urinary T cells than controls; however, patients who showed worsening graft function without rejection had similar numbers of T cells. T cells correlated with histological findings (interstitial inflammation p = 0.0005, r = 0.70; tubulitis p = 0.006, r = 0.58). Combining the amount of urinary T cells and TEC, or T cells and PDX+ cells, yielded a significant segregation of patients with rejection from patients without rejection (all p < 0.01, area under the curve 0.89–0.91). Urinary cell populations analyzed by flow cytometry have the potential to introduce new monitoring methods for kidney transplant patients. The combination of urinary T cells, TEC, and PDX-positive cells may allow non-invasive detection of transplant rejection.
Collapse
Affiliation(s)
| | | | | | | | | | - Benjamin Koch
- Goethe University Hospital Frankfurt, Frankfurt, Germany
| | | | | | | | | | | |
Collapse
|
11
|
Fujishiro H, Himeno S. Gene expression profiles of immortalized S1, S2, and S3 cells derived from each segment of mouse kidney proximal tubules. ACTA ACUST UNITED AC 2019. [DOI: 10.2131/fts.6.117] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Hitomi Fujishiro
- Laboratory of Molecular Nutrition and Toxicology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University
| | - Seiichiro Himeno
- Laboratory of Molecular Nutrition and Toxicology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University
| |
Collapse
|
12
|
Lyu Z, Mao Z, Li Q, Xia Y, Liu Y, He Q, Wang Y, Zhao H, Lu Z, Zhou Q. PPARγ maintains the metabolic heterogeneity and homeostasis of renal tubules. EBioMedicine 2018; 38:178-190. [PMID: 30420298 PMCID: PMC6306377 DOI: 10.1016/j.ebiom.2018.10.072] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 10/22/2018] [Accepted: 10/31/2018] [Indexed: 02/03/2023] Open
Abstract
Background The renal tubules, which have distant metabolic features and functions in different segments, reabsorb >99% of approximately 180 l of water and 25,000 mmol of Na + daily. Defective metabolism in renal tubules is involved in the pathobiology of kidney diseases. However, the mechanisms underlying the metabolic regulation in renal tubules remain to be defined. Methods We quantitatively compared the proteomes of the isolated proximal tubules (PT) and distal tubules (DT) from C57BL/6 mouse using tandem mass tag (TMT) labeling-based quantitative mass spectrometry. Bioinformatics analysis of the differentially expressed proteins revealed the significant differences between PT and DT in metabolism pathway. We also performed in vitro and in vivo assays to investigate the molecular mechanism underlying the distant metabolic features in PT and DT. Findings We demonstrate that the renal proximal tubule (PT) has high expression of lipid metabolism enzymes, which is transcriptionally upregulated by abundantly expressed PPARα/γ. In contrast, the renal distal tubule (DT) has elevated glycolytic enzyme expression, which is mediated by highly expressed c-Myc. Importantly, PPARγ transcriptionally enhances the protease iRhom2 expression in PT, which suppresses EGF expression and secretion and subsequent EGFR-dependent glycolytic gene expression and glycolysis. PPARγ inhibition reduces iRhom2 expression and increases EGF and GLUT1 expression in PT in mice, resulting in renal tubule hypertrophy, tubulointerstitial fibrosis and damaged kidney functions, which are rescued by 2-deoxy-d-glucose treatment. Interpretation These findings delineate instrumental mechanisms underlying the active lipid metabolism and suppressed glycolysis in PT and active glycolysis in DT and reveal critical roles for PPARs and c-Myc in maintaining renal metabolic homeostasis. FUND: This work was supported by the National Natural Science Foundation of China (grants 81572076 and 81873932; to Q.Z.), the Applied Development Program of the Science and Technology Committee of Chongqing (cstc2014yykfB10003; Q.Z.), the Program of Populace Creativities Workshops of the Science and Technology Committee of Chongqing (Q.Z.), the special demonstration programs for innovation and application of techniques (cstc2018jscx-mszdX0022) from the Science and Technology Committee of Chongqing (Q.Z.).
Collapse
Affiliation(s)
- Zhongshi Lyu
- The Division of Molecular Nephrology, The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The School of Laboratory Medicine, Chongqing Medical University, Chongqing, People's Republic of China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Zhaomin Mao
- The Division of Molecular Nephrology, The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The School of Laboratory Medicine, Chongqing Medical University, Chongqing, People's Republic of China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Qianyin Li
- The Division of Molecular Nephrology, The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The School of Laboratory Medicine, Chongqing Medical University, Chongqing, People's Republic of China
| | - Yan Xia
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yamin Liu
- The Division of Molecular Nephrology, The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The School of Laboratory Medicine, Chongqing Medical University, Chongqing, People's Republic of China
| | - Qingling He
- The Division of Molecular Nephrology, The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The School of Laboratory Medicine, Chongqing Medical University, Chongqing, People's Republic of China
| | - Yingchun Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Hui Zhao
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Zhimin Lu
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA
| | - Qin Zhou
- The Division of Molecular Nephrology, The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The School of Laboratory Medicine, Chongqing Medical University, Chongqing, People's Republic of China.
| |
Collapse
|
13
|
Ding W, Yousefi K, Shehadeh LA. Isolation, Characterization, And High Throughput Extracellular Flux Analysis of Mouse Primary Renal Tubular Epithelial Cells. J Vis Exp 2018. [PMID: 29985358 PMCID: PMC6101965 DOI: 10.3791/57718] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Mitochondrial dysfunction in the renal tubular epithelial cells (TECs) can lead to renal fibrosis, a major cause of chronic kidney disease (CKD). Therefore, assessing mitochondrial function in primary TECs may provide valuable insight into the bioenergetic status of the cells, providing insight into the pathophysiology of CKD. While there are a number of complex protocols available for the isolation and purification of proximal tubules in different species, the field lacks a cost-effective method optimized for tubular cell isolation without the need for purification. Here, we provide an isolation protocol that allows for studies focusing on both primary mouse proximal and distal renal TECs. In addition to cost-effective reagents and minimal animal procedures required in this protocol, the isolated cells maintain high energy levels after isolation and can be sub-cultured up to four passages, allowing for continuous studies. Furthermore, using a high throughput extracellular flux analyzer, we assess the mitochondrial respiration directly in the isolated TECs in a 96-well plate for which we provide recommendations for the optimization of cell density and compound concentration. These observations suggest that this protocol can be used for renal tubular ex vivo studies with a consistent, well-standardized production of renal TECs. This protocol may have broader future applications to study mitochondrial dysfunction associated with renal disorders for drug discovery or drug characterization purposes.
Collapse
Affiliation(s)
- Wen Ding
- Department of Molecular and Cellular Pharmacology, University of Miami Leonard M. Miller School of Medicine; Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine
| | - Keyvan Yousefi
- Department of Molecular and Cellular Pharmacology, University of Miami Leonard M. Miller School of Medicine; Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine
| | - Lina A Shehadeh
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine; Department of Medicine, Division of Cardiology, University of Miami Leonard M. Miller School of Medicine; Vascular Biology Institute, University of Miami Leonard M. Miller School of Medicine; Peggy and Harold Katz Family Drug Discovery Center, University of Miami Leonard M. Miller School of Medicine;
| |
Collapse
|
14
|
Precision toxicology based on single cell sequencing: an evolving trend in toxicological evaluations and mechanism exploration. Arch Toxicol 2017; 91:2539-2549. [DOI: 10.1007/s00204-017-1971-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 04/20/2017] [Indexed: 12/11/2022]
|
15
|
Prickett TCR, Doughty RN, Troughton RW, Frampton CM, Whalley GA, Ellis CJ, Espiner EA, Richards AM. C-Type Natriuretic Peptides in Coronary Disease. Clin Chem 2016; 63:316-324. [PMID: 28062626 DOI: 10.1373/clinchem.2016.257816] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 06/30/2016] [Indexed: 12/28/2022]
Abstract
AIMS C-type natriuretic peptide (CNP) is a paracrine growth factor expressed in the vascular endothelium. Although upregulated in atheromatous arteries, the predictive value of plasma CNP products for outcome in coronary disease is unknown. This study aimed to compare the prognostic value of plasma CNP products with those of other natriuretic peptides in individuals with coronary artery disease, and investigate their associations with cardiac and renal function. METHODS AND RESULTS Plasma concentrations of CNP and amino-terminal proCNP (NT-proCNP) were measured at baseline in 2129 individuals after an index acute coronary syndrome admission and related to cardiac and renal function, other natriuretic peptides [atrial NP (ANP) and B-type NP (BNP)] and prognosis (primary end point, mortality; secondary end point, cardiac readmission). Median follow-up was 4 years. At baseline, and in contrast to CNP, ANP, and BNP, plasma NT-proCNP was higher in males and weakly related to cardiac function but strongly correlated to plasma creatinine. All NPs were univariately associated with mortality. Resampling at 4 and 12 months in survivors showed stable concentrations of NT-proCNP whereas all other peptides declined. When studied by diagnosis (myocardial infarction, unstable angina) at index admission using a multivariate model, NT-proBNP predicted mortality and readmission in myocardial infarction. In unstable angina, only NT-proCNP predicted both mortality and cardiac readmission. CONCLUSIONS In contrast to the close association of NT-proBNP with cardiac function, and predictive value for outcome after myocardial infarction, plasma NT-proCNP is highly correlated with renal function and is an independent predictor of mortality and cardiac readmission in individuals with unstable angina.
Collapse
Affiliation(s)
| | - Robert N Doughty
- Department of Medicine, University of Auckland, Auckland, New Zealand.,Greenlane Cardiovascular Service, Auckland City Hospital, Auckland, New Zealand
| | | | - Chris M Frampton
- Department of Medicine, University of Otago, Christchurch, New Zealand
| | | | - Chris J Ellis
- Greenlane Cardiovascular Service, Auckland City Hospital, Auckland, New Zealand
| | - Eric A Espiner
- Department of Medicine, University of Otago, Christchurch, New Zealand
| | - A Mark Richards
- Department of Medicine, University of Otago, Christchurch, New Zealand
| |
Collapse
|
16
|
Endocytotic uptake of zoledronic acid by tubular cells may explain its renal effects in cancer patients receiving high doses of the compound. PLoS One 2015; 10:e0121861. [PMID: 25756736 PMCID: PMC4355483 DOI: 10.1371/journal.pone.0121861] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 02/16/2015] [Indexed: 12/22/2022] Open
Abstract
Zoledronic acid, a highly potent nitrogen-containing bisphosphonate used for the treatment of pathological bone loss, is excreted unmetabolized via the kidney if not bound to the bone. In cancer patients receiving high doses of the compound renal excretion may be associated with acute tubular necrosis. The question of how zoledronic acid is internalized by renal tubular cells has not been answered until now. In the current work, using a primary human tubular cell culture system, the pathway of cellular uptake of zoledronic acid (fluorescently/radiolabeled) and its cytotoxicity were investigated. Previous studies in our laboratory have shown that this primary cell culture model consistently mimics the physiological characteristics of molecular uptake/transport of the epithelium in vivo. Zoledronic acid was found to be taken up by tubular cells via fluid-phase-endocytosis (from apical and basolateral side) as evidenced by its co-localization with dextran. Cellular uptake and the resulting intracellular level was twice as high from the apical side compared to the basolateral side. Furthermore, the intracellular zoledronic acid level was found to be dependent on the administered concentration and not saturable. Cytotoxic effects however, were only seen at higher administration doses and/or after longer incubation times. Although zoledronic acid is taken up by tubular cells, no net tubular transport could be measured. It is concluded that fluid-phase-endocytosis of zoledronic acid and cellular accumulation at high doses may be responsible for the acute tubular necrosis observed in some cancer patients receiving high doses of the compound.
Collapse
|
17
|
Abstract
The mammalian kidney has an intrinsic ability to repair after significant injury. However, this process is inefficient: patients are at high risk for the loss of kidney function in later life. No therapy exists to treat established acute kidney injury (AKI) per se: strategies to promote endogenous repair processes and retard associated fibrosis are a high priority. Whole-organ gene expression profiling has been used to identify repair responses initiated with AKI, and factors that may promote the transition from AKI to chronic kidney disease. Transcriptional profiling has shown molecular markers and potential regulatory pathways of renal repair. Activation of a few key developmental pathways has been reported during repair. Whether these are comparable networks with similar target genes with those in earlier nephrogenesis remains unclear. Altered microRNA profiles, persistent tubular injury responses, and distinct late inflammatory responses highlight continuing kidney pathology. Additional insights into injury and repair processes will be gained by study of the repair transcriptome and cell-specific translatome using high-resolution technologies such as RNA sequencing and translational profiling tailored to specific cellular compartments within the kidney. An enhanced understanding holds promise for both the identification of novel therapeutic targets and biomarker-based evaluation of the damage-repair process.
Collapse
Affiliation(s)
- Sanjeev Kumar
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-California Institute of Regenerative Medicine (CIRM) Center for Regenerative Medicine and Stem Cell Research, The Keck School of Medicine of the University of Southern California, Los Angeles, CA
| | - Jing Liu
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-California Institute of Regenerative Medicine (CIRM) Center for Regenerative Medicine and Stem Cell Research, The Keck School of Medicine of the University of Southern California, Los Angeles, CA
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-California Institute of Regenerative Medicine (CIRM) Center for Regenerative Medicine and Stem Cell Research, The Keck School of Medicine of the University of Southern California, Los Angeles, CA.
| |
Collapse
|
18
|
Kamiyama M, Garner MK, Farragut KM, Sofue T, Hara T, Morikawa T, Konishi Y, Imanishi M, Nishiyama A, Kobori H. Detailed localization of augmented angiotensinogen mRNA and protein in proximal tubule segments of diabetic kidneys in rats and humans. Int J Biol Sci 2014; 10:530-42. [PMID: 24910532 PMCID: PMC4046880 DOI: 10.7150/ijbs.8450] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2013] [Accepted: 04/22/2014] [Indexed: 01/13/2023] Open
Abstract
In the intrarenal renin-angiotensin system, angiotensinogen levels are well known to be increased in diabetes, and these enhanced intrarenal angiotensinogen levels may initiate the development and accelerate the progression of diabetic nephropathy. However, the specific localization of the augmented angiotensinogen in proximal tubule segments in diabetes is still unknown. We investigated the detailed localization of angiotensinogen in 3 proximal tubule segments in the diabetic Otsuka Long-Evans Tokushima fatty (OLETF) rats and the control Long-Evans Tokushima Otsuka (LETO) rats. We also prepared OLETF rats treated with angiotensin II type 1 receptor blocker, olmesartan or with a combination of vasodilator agents. Moreover, biopsied samples of human kidney cortex were used to confirm the results of animal studies. We examined the co-localization of angiotensinogen with segment-specific markers by double staining using fluorescence in situ hybridization and/or immunofluorescence. Angiotensinogen mRNA expression was barely detectable in segment 1. In segment 3, the area of angiotensinogen mRNA expression was augmented in the OLETF rats compared with the LETO rats. Angiotensinogen protein expression areas in segments 1 and 3 were also increased in the OLETF rats compared with the LETO rats. Chronic treatment with olmesartan ameliorated these areas of augmented angiotensinogen expression. Biopsied human kidney samples showed similar results. These data suggest that the augmented angiotensinogen mRNA levels in segment 3 and angiotensinogen protein levels in segments 1 and 3 may contribute to the progression of diabetic nephropathy.
Collapse
Affiliation(s)
- Masumi Kamiyama
- 1. Department of Physiology, Tulane University Health Sciences Center, New Orleans, LA 70112, USA; ; 2. Hypertension and Renal Center of Excellence, Tulane University Health Sciences Center, New Orleans, LA 70112, USA
| | - Michelle K Garner
- 1. Department of Physiology, Tulane University Health Sciences Center, New Orleans, LA 70112, USA; ; 2. Hypertension and Renal Center of Excellence, Tulane University Health Sciences Center, New Orleans, LA 70112, USA
| | - Kristina M Farragut
- 1. Department of Physiology, Tulane University Health Sciences Center, New Orleans, LA 70112, USA; ; 2. Hypertension and Renal Center of Excellence, Tulane University Health Sciences Center, New Orleans, LA 70112, USA
| | - Tadashi Sofue
- 4. Department of Cardiorenal and Cerebrovascular Medicine, Kagawa University School of Medicine, Kagawa 761-0793, Japan
| | - Taiga Hara
- 4. Department of Cardiorenal and Cerebrovascular Medicine, Kagawa University School of Medicine, Kagawa 761-0793, Japan
| | - Takashi Morikawa
- 6. Department of Nephrology and Hypertension, Osaka City General Hospital, Osaka 534-0021, Japan
| | - Yoshio Konishi
- 6. Department of Nephrology and Hypertension, Osaka City General Hospital, Osaka 534-0021, Japan
| | - Masahito Imanishi
- 6. Department of Nephrology and Hypertension, Osaka City General Hospital, Osaka 534-0021, Japan
| | - Akira Nishiyama
- 5. Department of Pharmacology, Kagawa University School of Medicine, Kagawa 761-0793, Japan
| | - Hiroyuki Kobori
- 1. Department of Physiology, Tulane University Health Sciences Center, New Orleans, LA 70112, USA; ; 2. Hypertension and Renal Center of Excellence, Tulane University Health Sciences Center, New Orleans, LA 70112, USA; ; 3. Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA 70112, USA; ; 5. Department of Pharmacology, Kagawa University School of Medicine, Kagawa 761-0793, Japan
| |
Collapse
|
19
|
Jang KJ, Mehr AP, Hamilton GA, McPartlin LA, Chung S, Suh KY, Ingber DE. Human kidney proximal tubule-on-a-chip for drug transport and nephrotoxicity assessment. Integr Biol (Camb) 2014; 5:1119-29. [PMID: 23644926 DOI: 10.1039/c3ib40049b] [Citation(s) in RCA: 524] [Impact Index Per Article: 52.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Kidney toxicity is one of the most frequent adverse events reported during drug development. The lack of accurate predictive cell culture models and the unreliability of animal studies have created a need for better approaches to recapitulate kidney function in vitro. Here, we describe a microfluidic device lined by living human kidney epithelial cells exposed to fluidic flow that mimics key functions of the human kidney proximal tubule. Primary kidney epithelial cells isolated from human proximal tubule are cultured on the upper surface of an extracellular matrix-coated, porous, polyester membrane that splits the main channel of the device into two adjacent channels, thereby creating an apical 'luminal' channel and a basal 'interstitial' space. Exposure of the epithelial monolayer to an apical fluid shear stress (0.2 dyne cm(-2)) that mimics that found in living kidney tubules results in enhanced epithelial cell polarization and primary cilia formation compared to traditional Transwell culture systems. The cells also exhibited significantly greater albumin transport, glucose reabsorption, and brush border alkaline phosphatase activity. Importantly, cisplatin toxicity and Pgp efflux transporter activity measured on-chip more closely mimic the in vivo responses than results obtained with cells maintained under conventional culture conditions. While past studies have analyzed kidney tubular cells cultured under flow conditions in vitro, this is the first report of a toxicity study using primary human kidney proximal tubular epithelial cells in a microfluidic 'organ-on-a-chip' microdevice. The in vivo-like pathophysiology observed in this system suggests that it might serve as a useful tool for evaluating human-relevant renal toxicity in preclinical safety studies.
Collapse
Affiliation(s)
- Kyung-Jin Jang
- Wyss Institute for Biologically Inspired Engineering at Harvard University, CLSB Bldg. 5th floor, 3 Blackfan Circle, Boston, MA 02115, USA.
| | | | | | | | | | | | | |
Collapse
|
20
|
Mattila PE, Raghavan V, Rbaibi Y, Baty CJ, Weisz OA. Rab11a-positive compartments in proximal tubule cells sort fluid-phase and membrane cargo. Am J Physiol Cell Physiol 2013; 306:C441-9. [PMID: 24153428 DOI: 10.1152/ajpcell.00236.2013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The proximal tubule (PT) reabsorbs the majority of sodium, bicarbonate, and chloride ions, phosphate, glucose, water, and plasma proteins from the glomerular filtrate. Despite the critical importance of endocytosis for PT cell (PTC) function, the organization of the endocytic pathway in these cells remains poorly understood. We have used immunofluorescence and live-cell imaging to dissect the itinerary of apically internalized fluid and membrane cargo in polarized primary cultures of PTCs isolated from mouse kidney cortex. Cells from the S1 segment could be distinguished from those from more distal PT segments by their robust uptake of albumin and comparatively low expression of γ-glutamyltranspeptidase. Rab11a in these cells is localized to variously sized spherical compartments that resemble the apical vacuoles observed by electron microscopy analysis of PTCs in vivo. These Rab11a-positive structures are highly dynamic and receive membrane and fluid-phase cargo. In contrast, fluid-phase cargoes are largely excluded from Rab11a-positive compartments in immortalized kidney cell lines. The unusual morphology and sorting capacity of Rab11a compartments in primary PTCs may reflect a unique specialization of these cells to accommodate the functional demands of handling a high endocytic load.
Collapse
Affiliation(s)
- Polly E Mattila
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; and
| | | | | | | | | |
Collapse
|
21
|
Katagiri D, Hamasaki Y, Doi K, Okamoto K, Negishi K, Nangaku M, Noiri E. Protection of glucagon-like peptide-1 in cisplatin-induced renal injury elucidates gut-kidney connection. J Am Soc Nephrol 2013; 24:2034-43. [PMID: 24092928 DOI: 10.1681/asn.2013020134] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Accumulating evidence of the beyond-glucose lowering effects of a gut-released hormone, glucagon-like peptide-1 (GLP-1), has been reported in the context of remote organ connections of the cardiovascular system. Specifically, GLP-1 appears to prevent apoptosis, and inhibition of dipeptidyl peptidase-4 (DPP-4), which cleaves GLP-1, is renoprotective in rodent ischemia-reperfusion injury models. Whether this renoprotection involves enhanced GLP-1 signaling is unclear, however, because DPP-4 cleaves other molecules as well. Thus, we investigated whether modulation of GLP-1 signaling attenuates cisplatin (CP)-induced AKI. Mice injected with 15 mg/kg CP had increased BUN and serum creatinine and CP caused remarkable pathologic renal injury, including tubular necrosis. Apoptosis was also detected in the tubular epithelial cells of CP-treated mice using immunoassays for single-stranded DNA and activated caspase-3. Treatment with a DPP-4 inhibitor, alogliptin (AG), significantly reduced CP-induced renal injury and reduced the renal mRNA expression ratios of Bax/Bcl-2 and Bim/Bcl-2. AG treatment increased the blood levels of GLP-1, but reversed the CP-induced increase in the levels of other DPP-4 substrates such as stromal cell-derived factor-1 and neuropeptide Y. Furthermore, the GLP-1 receptor agonist exendin-4 reduced CP-induced renal injury and apoptosis, and suppression of renal GLP-1 receptor expression in vivo by small interfering RNA reversed the renoprotective effects of AG. These data suggest that enhancing GLP-1 signaling ameliorates CP-induced AKI via antiapoptotic effects and that this gut-kidney axis could be a new therapeutic target in AKI.
Collapse
|
22
|
Heer R, Hepburn AC, Williamson SC, Kennedy A, El-Sherif A, Soomro NA, Brown CDA, Robson CN. Renal differentiation from adult spermatogonial stem cells. Ren Fail 2013; 35:1387-91. [PMID: 23991628 DOI: 10.3109/0886022x.2013.828266] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
There is considerable interest in the use of multi-potent stem cells in kidney tissue regeneration. We studied if spermatogonial stem cells have the ability to undergo kidney differentiation. Spermatogonial stem cell differentiation was induced using in vitro and ex vivo co-culture techniques. Conditioned media from human kidney fibroblasts induced the expression of epithelial and endothelial lineages in spermatogonial stem cells, consistent with nephrogenesis. Furthermore, we showed that these cells up-regulated renal tubular-specific markers alkaline phosphatase, mineralocorticoid receptor, renal epithelial sodium channel and sodium-glucose transporter-2 (p<0.05). GFP-labeled spermatogonial stem cells were engrafted into metanephric kidney organ cultures harvested from E12.5 mouse embryos. After 5 days of organ culture, focal anti-GFP staining was detectable in all inoculated kidneys demonstrating integration of spermatogonial stem cells into the developing kidney (p<0.01). Histological assessment showed early nephron-like architecture. In summary, we show that spermatogonial stem cells have the potential to generate renal tissue and lay the foundations for further investigations into a novel therapeutic approach for renal insufficiency.
Collapse
Affiliation(s)
- Rakesh Heer
- Northern Institute for Cancer Research, Newcastle University , Framlington Place, Newcastle upon Tyne , UK
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Divergent localization of angiotensinogen mRNA and protein in proximal tubule segments of normal rat kidney. J Hypertens 2013; 30:2365-72. [PMID: 23032142 DOI: 10.1097/hjh.0b013e3283598eed] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Angiotensinogen in the kidneys is formed primarily in the proximal tubule cells and is secreted into the tubular fluid. Structurally, proximal tubules can be divided into three segments. The first segment, segment 1 (S1) is mainly confined to the pars convoluta, the second segment, segment 2 (S2) comprises the end of pars convoluta, and the third segment, segment 3 (S3) includes the major part of the pars recta. There are some reports describing angiotensinogen localization in kidneys; however, it remains uncertain which proximal tubule segments express angiotensinogen. To determine the detailed localization of angiotensinogen in the three proximal tubule segments, we established multistaining methods using segment-specific protein markers. METHODS Using kidneys from Wistar-Kyoto rats, we performed immunohistochemistry and double or triple staining by fluorescence in-situ hybridization and/or immunofluorescence. RESULTS Our results show that angiotensinogen mRNA and protein are expressed in the cortex and outer medulla of the normal rat kidney. Angiotensinogen mRNA was hardly detected in S1, detected weakly in S2 and strongly in S3 segments. In contrast, angiotensinogen protein was detected in S1 at high levels and less in S2 and S3 segments. CONCLUSION These data indicate divergence of angiotensinogen mRNA transcription and angiotensinogen protein synthesis and metabolism in different segments of the normal rat proximal tubules.
Collapse
|
24
|
Van der Hauwaert C, Savary G, Gnemmi V, Glowacki F, Pottier N, Bouillez A, Maboudou P, Zini L, Leroy X, Cauffiez C, Perrais M, Aubert S. Isolation and characterization of a primary proximal tubular epithelial cell model from human kidney by CD10/CD13 double labeling. PLoS One 2013; 8:e66750. [PMID: 23799132 PMCID: PMC3682988 DOI: 10.1371/journal.pone.0066750] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 05/11/2013] [Indexed: 11/27/2022] Open
Abstract
Renal proximal tubular epithelial cells play a central role in renal physiology and are among the cell types most sensitive to ischemia and xenobiotic nephrotoxicity. In order to investigate the molecular and cellular mechanisms underlying the pathophysiology of kidney injuries, a stable and well-characterized primary culture model of proximal tubular cells is required. An existing model of proximal tubular cells is hampered by the cellular heterogeneity of kidney; a method based on cell sorting for specific markers must therefore be developed. In this study, we present a primary culture model based on the mechanical and enzymatic dissociation of healthy tissue obtained from nephrectomy specimens. Renal epithelial cells were sorted using co-labeling for CD10 and CD13, two renal proximal tubular epithelial markers, by flow cytometry. Their purity, phenotypic stability and functional properties were evaluated over several passages. Our results demonstrate that CD10/CD13 double-positive cells constitute a pure, functional and stable proximal tubular epithelial cell population that displays proximal tubule markers and epithelial characteristics over the long term, whereas cells positive for either CD10 or CD13 alone appear to be heterogeneous. In conclusion, this study describes a method for establishing a robust renal proximal tubular epithelial cell model suitable for further experimentation.
Collapse
Affiliation(s)
- Cynthia Van der Hauwaert
- EA4483, Département de Biochimie et Biologie Moléculaire, Faculté de Médecine de Lille, Pôle Recherche, Lille, France
| | - Grégoire Savary
- EA4483, Département de Biochimie et Biologie Moléculaire, Faculté de Médecine de Lille, Pôle Recherche, Lille, France
| | - Viviane Gnemmi
- Institut National de la Santé et de la Recherche Médicale, U837, Centre de Recherche Jean-Pierre Aubert, Equipe 5 Mucines, Différentiation et Cancérogenèse Épithéliales, Lille, France
- Service d'Anatomie Pathologique, Centre de Biologie et Pathologie, CHRU Lille, Lille, France
- Faculté de Médecine de Lille, Université Lille 2, Lille, France
| | - François Glowacki
- EA4483, Département de Biochimie et Biologie Moléculaire, Faculté de Médecine de Lille, Pôle Recherche, Lille, France
- Faculté de Médecine de Lille, Université Lille 2, Lille, France
- Service de Néphrologie, Hôpital Huriez, CHRU Lille, Lille, France
| | - Nicolas Pottier
- EA4483, Département de Biochimie et Biologie Moléculaire, Faculté de Médecine de Lille, Pôle Recherche, Lille, France
- Faculté de Médecine de Lille, Université Lille 2, Lille, France
| | - Audrey Bouillez
- Institut National de la Santé et de la Recherche Médicale, U837, Centre de Recherche Jean-Pierre Aubert, Equipe 5 Mucines, Différentiation et Cancérogenèse Épithéliales, Lille, France
| | - Patrice Maboudou
- Service de Biochimie, Centre de Biologie et Pathologie, CHRU Lille, Lille, France
| | - Laurent Zini
- Institut National de la Santé et de la Recherche Médicale, U837, Centre de Recherche Jean-Pierre Aubert, Equipe 5 Mucines, Différentiation et Cancérogenèse Épithéliales, Lille, France
- Faculté de Médecine de Lille, Université Lille 2, Lille, France
- Service d'Urologie, Hôpital Huriez, CHRU Lille, Lille, France
| | - Xavier Leroy
- Institut National de la Santé et de la Recherche Médicale, U837, Centre de Recherche Jean-Pierre Aubert, Equipe 5 Mucines, Différentiation et Cancérogenèse Épithéliales, Lille, France
- Service d'Anatomie Pathologique, Centre de Biologie et Pathologie, CHRU Lille, Lille, France
- Faculté de Médecine de Lille, Université Lille 2, Lille, France
| | - Christelle Cauffiez
- EA4483, Département de Biochimie et Biologie Moléculaire, Faculté de Médecine de Lille, Pôle Recherche, Lille, France
- Faculté de Médecine de Lille, Université Lille 2, Lille, France
| | - Michaël Perrais
- Institut National de la Santé et de la Recherche Médicale, U837, Centre de Recherche Jean-Pierre Aubert, Equipe 5 Mucines, Différentiation et Cancérogenèse Épithéliales, Lille, France
- Faculté de Médecine de Lille, Université Lille 2, Lille, France
| | - Sébastien Aubert
- Institut National de la Santé et de la Recherche Médicale, U837, Centre de Recherche Jean-Pierre Aubert, Equipe 5 Mucines, Différentiation et Cancérogenèse Épithéliales, Lille, France
- Service d'Anatomie Pathologique, Centre de Biologie et Pathologie, CHRU Lille, Lille, France
- Faculté de Médecine de Lille, Université Lille 2, Lille, France
- * E-mail:
| |
Collapse
|
25
|
Gildea JJ, Lahiff DT, Van Sciver RE, Weiss RS, Shah N, McGrath HE, Schoeffel CD, Jose PA, Carey RM, Felder RA. A linear relationship between the ex-vivo sodium mediated expression of two sodium regulatory pathways as a surrogate marker of salt sensitivity of blood pressure in exfoliated human renal proximal tubule cells: the virtual renal biopsy. Clin Chim Acta 2013; 421:236-42. [PMID: 23454474 DOI: 10.1016/j.cca.2013.02.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Revised: 01/14/2013] [Accepted: 02/21/2013] [Indexed: 12/30/2022]
Abstract
BACKGROUND Salt sensitivity (SS) of blood pressure (BP) affects 25% of adults, shares comorbidity with hypertension, and has no convenient diagnostic test. We tested the hypothesis that urine-derived exfoliated renal proximal tubule cells (RPTCs) could diagnose the degree of an individual's SS of BP. METHODS Subjects were selected who had their SS of BP determined 5 y prior to this study (salt-sensitive: ≥7 mm Hg increase in mean arterial pressure (MAP) following transition from a random weekly diet of low (10 mmol/day) to high (300 mmol/day) sodium (Na(+)) intake, N=4; inverse salt-sensitive (ISS): ≥7 mm Hg increase in MAP transitioning from a high to low Na(+) diet, N=3, and salt-resistant (SR): <7 mm Hg change in MAP transitioned on either diet, N=5). RPTC responses to 2 independent Na(+) transport pathways were measured. RESULTS There was a negative correlation between the degree of SS and dopamine-1 receptor (D1R) plasma membrane recruitment (y=-0.0107x+0.68 relative fluorescent units (RFU), R(2)=0.88, N=12, P<0.0001) and angiotensin II-stimulated intracellular Ca(++) (y=-0.0016x+0.0336, R(2)=0.7112, P<0.001, N=10) concentration over baseline. CONCLUSIONS Isolating RPTCs from urine provides a personalized cell-based diagnostic test of SS index that offers advantages over a 2-week controlled diet with respect to cost and patient compliance. Furthermore, the linear relationship between the change in MAP and response to 2 Na(+) regulatory pathways suggests that an individual's RPTC response to intracellular Na(+) is personalized and predictive.
Collapse
Affiliation(s)
- John J Gildea
- University of Virginia, Charlottesville, VA 22903, United States
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Souma T, Abe M, Moriguchi T, Takai J, Yanagisawa-Miyazawa N, Shibata E, Akiyama Y, Toyohara T, Suzuki T, Tanemoto M, Abe T, Sato H, Yamamoto M, Ito S. Luminal alkalinization attenuates proteinuria-induced oxidative damage in proximal tubular cells. J Am Soc Nephrol 2011; 22:635-48. [PMID: 21372211 DOI: 10.1681/asn.2009111130] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
A highly acidic environment surrounds proximal tubular cells as a result of their reabsorption of HCO(3)(-). It is unclear whether this luminal acidity affects proteinuria-induced progression of tubular cell damage. Here, we investigated the contribution of luminal acidity to superoxide (O(2)(·-)) production induced by oleic acid-bound albumin (OA-Alb) in proximal tubular cells. Acidic media significantly enhanced OA-Alb-induced O(2)(·-) production in the HK-2 proximal tubular cell line. Simultaneous treatment with both OA-Alb and acidic media led to phosphorylation of the intracellular pH sensor Pyk2. Highly phosphorylated Pyk2 associated with activation of Rac1, an essential subcomponent of NAD(P)H oxidase. Furthermore, knockdown of Pyk2 with siRNA attenuated the O(2)(·-) production induced by cotreatment with OA-Alb and acid. To assess whether luminal alkalinization abrogates proteinuria-induced tubular damage, we studied a mouse model of protein-overload nephropathy. NaHCO(3) feeding selectively alkalinized the urine and dramatically attenuated the accumulation of O(2)(·-)-induced DNA damage and proximal tubular injury. Overall, these observations suggest that luminal acidity aggravates proteinuria-induced tubular damage and that modulation of this acidic environment may hold potential as a therapeutic target for proteinuric kidney disease.
Collapse
Affiliation(s)
- Tomokazu Souma
- Division of Nephrology, Endocrinology, and Vascular Medicine, Department of Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Gildea JJ, Shah I, Weiss R, Casscells ND, McGrath HE, Zhang J, Jones JE, Felder RA. HK-2 human renal proximal tubule cells as a model for G protein-coupled receptor kinase type 4-mediated dopamine 1 receptor uncoupling. Hypertension 2010; 56:505-11. [PMID: 20660820 DOI: 10.1161/hypertensionaha.110.152256] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
HK-2 human renal proximal tubule cells (RPTC) are commonly used in the in vitro study of "normal" RPTCs. We discovered recently that HK-2 cells are uncoupled from dopamine 1 receptor (D(1)R) adenylyl cyclase (AC) stimulation. We hypothesized that G protein-coupled receptor kinase type 4 (GRK4) single nucleotide polymorphisms may be responsible for the D(1)R/AC uncoupling in HK-2. This hypothesis was tested by genotyping GRK4 single nucleotide polymorphisms, measuring D(1)-like receptor agonist (fenoldopam)-stimulated cAMP accumulation, quantifying D(1)R inhibition of sodium transport, and testing the ability of GRK4 small interfering RNA to reverse the D(1)R/AC uncoupling. We compared HK-2 with 2 normally coupled human RPTC cell lines and 2 uncoupled RPTC cell lines. The HK-2 cell line was found to have 4 of 6 potential GRK4 single nucleotide polymorphisms known to uncouple the D(1)R from AC (namely, R65L, A142V, and A486V). AC response to fenoldopam stimulation was increased in the 2 normally coupled human RPTC cell lines (FEN: 2.02+/-0.05-fold and 2.33+/-0.19-fold over control; P<0.001; n=4) but not in the 2 uncoupled or HK-2 cell lines. GRK4 small interfering RNA rescued the fenoldopam-mediated AC stimulation in the uncoupled cells, including HK-2. The expected fenoldopam-mediated inhibition of sodium hydrogen exchanger type 3 was absent in HK-2 (n=6) and uncoupled RPTC cell lines (n=6) but was observed in the 2 normally coupled human RPTC cell lines (-25.41+/-4.7% and -27.36+/-2.70%; P<0.001; n=6), which express wild-type GRK4. Despite the fact that HK-2 cells retain many functional characteristics of RPTCs, they are not normal from the perspective of dopaminergic function.
Collapse
Affiliation(s)
- John J Gildea
- University of Virginia, PO Box 801400, Charlottesville, VA 22908, USA
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Epoetin delta reduces oxidative stress in primary human renal tubular cells. J Biomed Biotechnol 2010; 2010:395785. [PMID: 20454536 PMCID: PMC2864893 DOI: 10.1155/2010/395785] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2009] [Revised: 02/17/2010] [Accepted: 02/17/2010] [Indexed: 02/07/2023] Open
Abstract
Erythropoietin (EPO) exerts (renal) tissue protective effects. Since it is unclear whether this is a direct effect of EPO on the kidney or not, we investigated whether EPO is able to protect human renal tubular epithelial cells (hTECs) from oxidative stress and if so which pathways are involved. EPO (epoetin delta) could protect hTECs against oxidative stress by a dose-dependent inhibition of reactive oxygen species formation. This protective effect is possibly related to the membranous expression of the EPO receptor (EPOR) since our data point to the membranous EPOR expression as a prerequisite for this protective effect. Oxidative stress reduction went along with the upregulation of renoprotective genes. Whilst three of these, heme oxygenase-1 (HO-1), aquaporin-1 (AQP-1), and B-cell CLL/lymphoma 2 (Bcl-2) have already been associated with EPO-induced renoprotection, this study for the first time suggests carboxypeptidase M (CPM), dipeptidyl peptidase IV (DPPIV), and cytoglobin (Cygb) to play a role in this process.
Collapse
|
29
|
Characterisation of human tubular cell monolayers as a model of proximal tubular xenobiotic handling. Toxicol Appl Pharmacol 2008; 233:428-38. [PMID: 18930752 DOI: 10.1016/j.taap.2008.09.018] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2008] [Revised: 09/17/2008] [Accepted: 09/18/2008] [Indexed: 12/25/2022]
Abstract
The aim of this study was to determine whether primary human tubular cell monolayers could provide a powerful tool with which to investigate the renal proximal tubular handling of xenobiotics. Human proximal and distal tubule/collecting duct cells were grown as monolayers on permeable filter supports. After 10 days in culture, proximal tubule cells remained differentiated and expressed a wide palette of transporters at the mRNA level including NaPi-IIa, SGLT1, SGLT2, OCT2, OCTN2, OAT1, OAT3, OAT4, MDR1, MRP2 and BCRP. At the protein level, the expression of a subset of transporters including NaPi-IIa, OAT1 and OAT3 was demonstrated using immunohistochemistry. Analysis of the expression of the ATP binding cassette efflux pumps MDR1, MRP2 and BCRP confirmed their apical membrane localisation. At the functional level, tubule cell monolayers retain the necessary machinery to mediate the net secretion of the prototypic substrates; PAH and creatinine. PAH secretion across the monolayer consisted of the uptake of PAH across the basolateral membrane by OAT1 and OAT3 and the apical exit of PAH by a probenecid and MK571-sensitive route consistent with actions of MRP2 or MRP4. Creatinine secretion was by OCT2-mediated uptake at the basolateral membrane and via MDR1 at the apical membrane. Functional expression of MDR1 and BCRP at the apical membrane was also demonstrated using a Hoechst 33342 dye. Similarly, measurement of calcein efflux demonstrated the functional expression of MRP2 at the apical membrane of cell monolayers. In conclusion, human tubular cell monolayers provide a powerful tool to investigate renal xenobiotic handling.
Collapse
|
30
|
Verhulst A, Sayer R, De Broe ME, D'Haese PC, Brown CDA. Human Proximal Tubular Epithelium Actively Secretes but Does Not Retain Rosuvastatin. Mol Pharmacol 2008; 74:1084-91. [DOI: 10.1124/mol.108.047647] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
|
31
|
Huls M, Brown C, Windass A, Sayer R, van den Heuvel J, Heemskerk S, Russel F, Masereeuw R. The breast cancer resistance protein transporter ABCG2 is expressed in the human kidney proximal tubule apical membrane. Kidney Int 2008; 73:220-5. [DOI: 10.1038/sj.ki.5002645] [Citation(s) in RCA: 205] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
32
|
Schlatter P, Beglinger C, Drewe J, Gutmann H. Glucagon-like peptide 1 receptor expression in primary porcine proximal tubular cells. ACTA ACUST UNITED AC 2007; 141:120-8. [PMID: 17276524 DOI: 10.1016/j.regpep.2006.12.016] [Citation(s) in RCA: 112] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2006] [Revised: 12/21/2006] [Accepted: 12/22/2006] [Indexed: 01/11/2023]
Abstract
BACKGROUND GLP-1 is secreted into the circulation after food intake. The main biological effects of GLP-1 include stimulation of glucose dependent insulin secretion and induction of satiety feelings. Recently, it was demonstrated in rats and humans that GLP-1 can stimulate renal excretion of sodium. Based on these data, the existence of a renal GLP-1 receptor (GLP-1R) was postulated. However, the exact localization of the GLP-1R and the mechanism of this GLP-1 action have not yet been investigated. METHODS Primary porcine proximal tubular cells were isolated from porcine kidneys. Expression of GLP-1R was measured at the mRNA level by quantitative RT-PCR. Protein expression of GLP-1R was verified with immunocytochemistry, immunohistochemistry and Western blot analysis. Functional studies included transport assessments of sodium and glucose using three different GLP-1 concentrations (200 pM, 2 nM and 20 nM), 200 pM exendin-4 (GLP-1 analogue) and an inhibitor of the dipeptidylpeptidase IV (DPPIV) enzyme (P32/98 at 10 microM). Finally, the expression of NHE3, the predominant Na(+)/H(+) exchanger in proximal tubular cells, was also investigated. RESULTS GLP-1R, NHE3 and DPPIV were expressed at the mRNA level in porcine proximal tubular kidney cells. GLP-1R expression was confirmed at the protein level. Staining of human and pig kidney cortex revealed that GLP-1R was predominantly expressed in proximal tubular cells. Functional assays demonstrated an inhibition of sodium re-absorption with GLP-1 after 3 h of incubation. Exendin-4 and GLP-1 in combination with P32/98 co-administration had no clear influence on glucose and sodium uptake and transport. CONCLUSION GLP-1R is functionally expressed in porcine proximal tubular kidney cells. Addition of GLP-1 to these cells resulted in a reduced sodium re-absorption. GLP-1 had no effect on glucose re-absorption. We conclude that GLP-1 modulates sodium homeostasis in the kidney most likely through a direct action via its GLP-1R in proximal tubular cells.
Collapse
Affiliation(s)
- P Schlatter
- Department of Clinical Pharmacology and Toxicology, University Hospital of Basel, Hebelstr. 2, CH-4031 Basel, Switzerland
| | | | | | | |
Collapse
|
33
|
Verhulst A, Asselman M, Persy VP, Schepers MSJ, Helbert MF, Verkoelen CF, De Broe ME. Crystal retention capacity of cells in the human nephron: involvement of CD44 and its ligands hyaluronic acid and osteopontin in the transition of a crystal binding- into a nonadherent epithelium. J Am Soc Nephrol 2003; 14:107-15. [PMID: 12506143 DOI: 10.1097/01.asn.0000038686.17715.42] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Nephrolithiasis requires formation of crystals followed by their retention and accumulation in the kidney. Crystal retention can be caused by the association of crystals with the epithelial cells lining the renal tubules. The present study investigated the interaction between calcium oxalate monohydrate (COM) crystals and primary cultures of human proximal (PTC) and distal tubular/collecting duct cells (DTC). Both PTC and DTC were susceptible to crystal binding during the first days post-seeding (4.9 +/- 0.8 micro g COM/cm2), but DTC lost this affinity when the cultures developed into confluent monolayers with functional tight junctions (0.05 +/- 0.02 micro g COM/cm2). Confocal microscopy demonstrated the expression of the transmembrane receptor protein CD44 and its ligands osteopontin (OPN) and hyaluronic acid (HA) at the apical membrane of proliferating tubular cells; at confluence, CD44 was expressed at the basolateral membrane and OPN and HA were no longer detectable. In addition, a particle exclusion technique revealed that proliferating cells were surrounded by HA-rich pericellular matrices or "cell coats" extending several microns from the cell surface. Disintegration of these coats with hyaluronidase significantly decreased the cell surface affinity for crystals. Furthermore, CD44, OPN, and HA were also expressed in vivo at the luminal side of tubular cells in damaged kidneys. These results suggest (1) that the intact distal tubular epithelium of the human kidney does not bind crystals, and (2) that crystal retention in the human kidney may depend on the expression of CD44-, OPN-, and-HA rich cell coats by damaged distal tubular epithelium.
Collapse
Affiliation(s)
- Anja Verhulst
- Department of Nephrology-Hypertension, University of Antwerp, Antwerp, Belgium
| | | | | | | | | | | | | |
Collapse
|
34
|
Verhulst A, Persy VP, Van Rompay AR, Verstrepen WA, Helbert MF, De Broe ME. Osteopontin synthesis and localization along the human nephron. J Am Soc Nephrol 2002; 13:1210-1218. [PMID: 11961008 DOI: 10.1681/asn.v1351210] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
In normal human and rat kidneys, osteopontin (OPN) is present at the apical surface of cells in the distal nephron. After ischemic or toxic renal damage in rats, OPN is upregulated in distal tubular cells (DTC) and expressed de novo in perinuclear vesicles in proximal tubular cells (PTC). In the first phase of this study, OPN localization in ischemic human biopsies was compared with that in ischemic rat kidneys. In the second phase, cultures of PTC and DTC were used to investigate human renal OPN synthesis, secretion, and localization. OPN localization in human biopsies after renal ischemia was comparable to that in ischemic rat kidneys. Microscopic and flow cytometric detection of immunofluorescent OPN staining in tubular cell cultures demonstrated strong plasma membrane localization in DTC, whereas mainly perinuclear intracellular expression was observed in PTC. Northern blotting and reverse transcription-PCR demonstrated production of a single OPN mRNA in PTC and DTC. Detection of OPN by Western blotting and enzyme-linked immunosorbent assay demonstrated that PTC and DTC synthesized and secreted the same three molecular mass OPN forms, in comparable amounts. Finally, confocal microscopy demonstrated different staining patterns for endocytotic/lysosomal vesicles and perinuclear OPN; however, perinuclear OPN exhibited colocalization with the Golgi apparatus. In conclusion, human renal OPN localization in cell cultures demonstrated differences between PTC and DTC comparable to those observed after renal ischemia in vivo. Therefore, these cell cultures represented an excellent model for the study of human OPN synthesis, secretion, and localization in PTC versus DTC. It is reported for the first time that intracellular OPN is located in the Golgi apparatus of both PTC and DTC and that PTC and DTC are able to produce and secrete the same OPN isoforms, in comparable amounts.
Collapse
Affiliation(s)
- Anja Verhulst
- Department of Nephrology-Hypertension, University of Antwerp, Belgium
| | - Veerle P Persy
- Department of Nephrology-Hypertension, University of Antwerp, Belgium
| | - An R Van Rompay
- Department of Nephrology-Hypertension, University of Antwerp, Belgium
| | | | - Mark F Helbert
- Department of Nephrology-Hypertension, University of Antwerp, Belgium
| | - Marc E De Broe
- Department of Nephrology-Hypertension, University of Antwerp, Belgium
| |
Collapse
|
35
|
Helbert MJ, Dauwe SE, De Broe ME. Flow cytometric immunodissection of the human distal tubule and cortical collecting duct system. Kidney Int 2001; 59:554-64. [PMID: 11168937 DOI: 10.1046/j.1523-1755.2001.059002554.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND In recent years, considerable efforts were drawn to isolate human distal tubule (DT) and collecting duct (CD) cells with more or less success. Here, we present a procedure for isolating human DT cells [thick ascending limb (TAL)/distal convoluted tubule (DCT)] and CD system cells (connecting tubule/initial CD) as separate populations within the same kidney specimen, applying monoclonal antibodies in fluorescence-activated cell sorting (FACS) and culturing them. METHODS We tested antibodies directed against the DT/CD system antigens, epithelial membrane antigen (EMA) and L1-cell adhesion molecule (L1-CAM). Segmental and subsegmental expressions were first assessed by using morphologic and histotopographic criteria, and by comparing sections with adjacent sections stained for expression of well-defined distal subsegment-specific markers. Immunoreactive cells were further characterized by dual immunostaining using cell type-specific markers. As a second step, cells obtained by collagenase digestion of normal renal cortical tissue were flow sorted following labeling with aforementioned antibodies and cultured. RESULTS EMA expression was found on all cells present in the DT and in the CD system. Its expression was most abundant in TAL and from thereon decreased gradually along the course of the DT and CD system. Flow sorting of all EMA-expressing cells resulted in identification/isolation of DT and CD system cells as a heterogeneous mixture. Flow sorting of only the most strongly EMA-positive cells allowed purification of DT cells only, mainly TAL cells as shown by Tamm-Horsfall protein expression on> 80% of sorted cells. L1-CAM was expressed in only the CD system, and sorting of all L1-CAM-positive cells allowed> 95% purification of CD system cells (connecting tubule/cortical CD). Primary cultures of DT and CD system cells rapidly developed into confluent monolayers, and retained antigenic and functional properties inherent to their segments of origin. CONCLUSION Our study presents a procedure for isolating and culturing pure populations of human DT cells and CD system cells as separate populations, using antibodies to the best available markers in FACS.
Collapse
Affiliation(s)
- M J Helbert
- Department of Nephrology, University of Antwerp, Belgium
| | | | | |
Collapse
|
36
|
van Kooten C, Daha MR. Cytokine cross-talk between tubular epithelial cells and interstitial immunocompetent cells. Curr Opin Nephrol Hypertens 2001; 10:55-9. [PMID: 11195052 DOI: 10.1097/00041552-200101000-00009] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Immunostaining of renal biopsies of patients with interstitial rejection of allografts or other forms of interstitial inflammation has demonstrated the presence of activated T cells and monocytes/macrophages in the tubulointerstitial area. Cytokines that are produced by infiltrating cells are capable of activating tubular epithelial cells. In turn tubular epithelial cells can produce a wide variety of inflammatory mediators, including chemokines, which further regulate cellular influx. Interfering in this cross-talk between tubular epithelial cells and infiltrating cells might provide new options for therapeutic intervention.
Collapse
Affiliation(s)
- C van Kooten
- Department of Nephrology, Leiden University Medical Center, The Netherlands.
| | | |
Collapse
|
37
|
Pfaller W, Gstraunthaler G. Nephrotoxicity testing in vitro--what we know and what we need to know. ENVIRONMENTAL HEALTH PERSPECTIVES 1998; 106 Suppl 2:559-69. [PMID: 9599703 PMCID: PMC1533421 DOI: 10.1289/ehp.98106559] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
The kidney is affected by many chemicals. Some of the chemicals may even contribute to end-stage renal disease and thus contribute considerably to health care costs. Because of the large functional reserve of the kidney, which masks signs of dysfunction, early diagnosis of renal disease is often difficult. Although numerous studies aimed at understanding the mechanisms underlying chemicals and drugs that target various renal cell types have delivered enough understanding for a reasonable risk assessment, there is still an urgent need to better understand the mechanisms leading to renal cell injury and organ dysfunction. The increasing use of in vitro techniques using isolated renal cells, nephron fragments, or cell cultures derived from specific renal cell types has improved our insight into the molecular mechanisms involved in nephrotoxicity. A short overview is given on the various in vitro systems currently used to clarify mechanistic aspects leading to sublethal or lethal injury of the functionally most important nephron epithelial cells derived from various species. Whereas freshly isolated cells and nephron fragments appear to represent a sufficient basis to study acute effects (hours) of nephrotoxins, e.g., on cell metabolism, primary cultures of these cells are more appropriate to study long-term effects. In contrast to isolated cells and fragments, however, primary cultures tend to first lose several of their in vivo metabolic properties during culture, and second to have only a limited life span (days to weeks). Moreover, establishing such primary cultures is a time-consuming and laborious procedure. For that reason many studies have been carried out on renal cell lines, which are easy to cultivate in large quantities and which have an unlimited life span. Unfortunately, none of the lines display a state of differentiation comparable to that of freshly isolated cells or their primary cultures. Most often they lack expression of key functions (e.g., gluconeogenesis or organic anion transport) of their in vivo correspondents. Therefore, the use of cell lines for assessment of nephrotoxic mechanisms will be limited to those functions the lines express. Upcoming molecular biology approaches such as the transduction of immortalizing genes into primary cultures and the utilization of cells from transgenic animals may in the near future result in the availability of highly differentiated renal cells with markedly extended life spans and near in vivo characteristics that may facilitate the use of renal cell culture for routine screening of nephrotoxins.
Collapse
Affiliation(s)
- W Pfaller
- Institute of Physiology, University of Innsbruck, Austria.
| | | |
Collapse
|
38
|
Morin JP, De Broe ME, Pfaller W, Schmuck G. Nephrotoxicity Testing In Vitro: The Current Situation. Altern Lab Anim 1997. [DOI: 10.1177/026119299702500506] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
An ECVAM task force on nephrotoxicity has been established to advise, in particular, on the follow-up to recommendations made in the ECVAM workshop report on nephrotoxicity testing in vitro. Since this workshop was held, in 1994, there have been several improvements in the techniques used. For example, the duration of renal slice viability, and the maintenance of functional activities in slices, have been improved by using dynamic incubation systems with higher oxygen tensions and more-appropriate cell culture media. Highly differentiated primary cultures of pig, human and rabbit proximal tubule cells have been established by using specific cell isolation procedures and/or selective culture media. To date, the most comparable phenotypic expression and transepithelial transport capacities to proximal tubules in vivo have been obtained with primary cultures of rabbit proximal tubule cells which are grown on bicompartmental supports; in this system, transepithelial substrate gradients are generated and the transepithelial transport of both organic anions and cations is highly active. This in vitro system has been selected by ECVAM for further evaluation and prevalidation. Industrial needs in the area of nephrotoxicity testing have been identified, and recommendations are made at the end of this report concerning possible future initiatives.
Collapse
Affiliation(s)
- Jean-Paul Morin
- INSERM U295, School of Medicine–Pharmacy, University of Rouen, Avenue de l'Universite, 76803 Saint Etienne du Rouvray, France
| | - Marc E. De Broe
- Department of Nephrology & Hypertension, University of Antwerp, Wilrijstraat 10, 2650 Edegem Antwerpen, Belgium
| | - Walter Pfaller
- Institute of Physiology, University of Innsbruck, Fritz-Preglstrasse 3, 6010 Innsbruck, Austria
| | - Gabriele Schmuck
- Department of Research Toxicology, Bayer AG, 4296 Wuppertal, Germany
| |
Collapse
|