1
|
Preston R, Chrisp R, Dudek M, Morais MRPT, Tian P, Williams E, Naylor RW, Davenport B, Pathiranage DRJ, Benson E, Spiller DG, Bagnall J, Zeef L, Lawless C, Baker SM, Meng QJ, Lennon R. The glomerular circadian clock temporally regulates basement membrane dynamics and the podocyte glucocorticoid response. Kidney Int 2025; 107:99-115. [PMID: 39515644 DOI: 10.1016/j.kint.2024.10.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 09/19/2024] [Accepted: 10/02/2024] [Indexed: 11/16/2024]
Abstract
Kidney physiology shows diurnal variation, and a disrupted circadian rhythm is associated with kidney disease. However, it remains largely unknown whether glomeruli, the filtering units in the kidney, are under circadian control. Here, we investigated core circadian clock components in glomeruli, together with their rhythmic targets and modes of regulation. With clock gene reporter mice, cell-autonomous glomerular clocks which likely govern rhythmic fluctuations in glomerular physiology were identified. Using circadian time-series transcriptomic profiling, the first circadian glomerular transcriptome with 375 rhythmic transcripts, enriched for extracellular matrix and glucocorticoid receptor signaling ontologies, were identified. Subsets of rhythmic matrix-related genes required for basement membrane assembly and turnover, and circadian variation in matrix ultrastructure, coinciding with peak abundance of rhythmic basement membrane proteins, were uncovered. This provided multiomic evidence for interactions between glomerular matrix and intracellular time-keeping mechanisms. Furthermore, glucocorticoids, which are frequently used to treat glomerular disease, reset the podocyte clock and induce rhythmic expression of potential glomerular disease genes associated with nephrotic syndrome that included Nphs1 (nephrin) and Nphs2 (podocin). Disruption of the clock with pharmacological inhibition altered the expression of these disease genes, indicating an interplay between clock gene expression and key genes required for podocyte health. Thus, our results provide a strong basis for future investigations of the functional implications and therapeutic potential of chronotherapy in glomerular health and disease.
Collapse
Affiliation(s)
- Rebecca Preston
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Ruby Chrisp
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Michal Dudek
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Mychel R P T Morais
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Pinyuan Tian
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Emily Williams
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Richard W Naylor
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Bernard Davenport
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Dharshika R J Pathiranage
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Emma Benson
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - David G Spiller
- Bioimaging Core Facility, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester, UK
| | - James Bagnall
- Bioimaging Core Facility, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester, UK
| | - Leo Zeef
- Bioinformatics Core Facility, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester, UK
| | - Craig Lawless
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Syed Murtuza Baker
- Bioinformatics Core Facility, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester, UK
| | - Qing-Jun Meng
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK.
| | - Rachel Lennon
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK; Department of Pediatric Nephrology, Royal Manchester Children's Hospital, Manchester University Hospitals National Health Service Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK.
| |
Collapse
|
2
|
Locatelli F, Del Vecchio L, Ponticelli C. Systemic and targeted steroids for the treatment of IgA nephropathy. Clin Kidney J 2023; 16:ii40-ii46. [PMID: 38053978 PMCID: PMC10695509 DOI: 10.1093/ckj/sfad224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Indexed: 12/07/2023] Open
Abstract
Immunoglobulin A nephropathy (IgAN) is a common glomerulonephritis partially correlated with mucosal immune system dysfunction. Progressive renal failure occurs in many patients, with about 30-50% of the patients with IgAN developing end-stage kidney disease (ESKD). Many treatments have been used for decades, despite uncertainty about their effectiveness and the ideal dose. Randomised controlled trials reported that systemic glucocorticoids can be an effective treatment for patients with persistent and significant proteinuria despite renin-angiotensin system inhibitors use possibly causing systemic side effects. The primary focus of IgAN management should be based on optimised supportive care, including renin-angiotensin system (RAS) blockade and now SGLT2 inhibitors. The novel targeted-release formulation (TRF) of budesonide has been tested to reduce the adverse events of systemic steroids by delivering the drug to the distal ileum. The local efficacy of TRF-budesonide may represent a novel and promising approach to treating IgAN. Two clinical trials showed that TRF-budesonide could significantly reduce proteinuria and haematuria and possibly preserve renal function while significantly reducing the side effects. However, the limited number of treated patients and the relatively short follow-up suggest caution before considering budesonide superior to the current six-months steroid pulses scheme. Long-term data on the efficacy and safety of TRF budesonide are awaited, together with the design of trials with a head-to-head comparison with systemic steroids before considering TRF-budesonide as the standard of care treatment for IgAN nephropathy.
Collapse
Affiliation(s)
- Francesco Locatelli
- Department of Nephrology and Dialysis, Alessandro Manzoni Hospital, Lecco, Italy
| | - Lucia Del Vecchio
- Department of Nephrology and Dialysis, Sant’ Anna Hospital, ASST Lariana, Como, Italy
| | | |
Collapse
|
3
|
Ren Q, Yu S, Zeng H, Xia H. The role of PTEN in puromycin aminonucleoside-induced podocyte injury. Int J Med Sci 2022; 19:1451-1459. [PMID: 36035365 PMCID: PMC9413557 DOI: 10.7150/ijms.72988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 07/28/2022] [Indexed: 11/27/2022] Open
Abstract
Podocytes are specialized cells of the glomerulus that play important structural and functional roles in maintaining the filtration barrier. Loss and injury of podocytes are leading factors of glomerular disease and kidney failure. Recent studies found that phosphatase and tensin homolog (PTEN) may play a critical role in maintaining the normal structure and function in podocytes. However, we still understand very little about how PTEN is regulated under podocyte injury conditions. In this study, We therefore investigated whether PTEN could play a role in podocyte injury induced by puromycin aminonucleoside (PAN), and whether dexamethasone (DEX) alleviates podocyte injury by PTEN/PI3K/Akt signaling. Our results showed that PI3K/Akt pathway was activated in podocytes exposed to PAN conditions, accompanied by down-regulation of the PTEN and microtubule-associated light chain 3 (LC3) expression.podocyte-specific knockout of PTEN significantly promoted podocyte injury, The potential renoprotection of overexpressed PTEN in podocytes was partly attributed with an improvement in autophagy and the inhibition of apoptosis.These novel findings also suggest that targeting PTEN might be a novel and promising therapeutic strategy against podocyte injury.
Collapse
Affiliation(s)
- Qi Ren
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, P.R. China
- Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong, P.R. China
| | - Shengyou Yu
- Department of Pediatrics, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, P.R.China
| | - Huasong Zeng
- Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong, P.R. China
| | - Huimin Xia
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, P.R. China
- Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong, P.R. China
| |
Collapse
|
4
|
Wu X, Ren L, Yang Q, Song H, Tang Q, Zhang M, Zhang J, Tang Z, Shi S. Glucocorticoids Inhibit EGFR Signaling Activation in Podocytes in Anti-GBM Crescentic Glomerulonephritis. Front Med (Lausanne) 2022; 9:697443. [PMID: 35223886 PMCID: PMC8866651 DOI: 10.3389/fmed.2022.697443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 01/18/2022] [Indexed: 11/24/2022] Open
Abstract
Glucocorticoids are commonly used to treat anti-GBM crescentic glomerulonephritis, however, the mechanism underlying its therapeutic effectiveness is not completely understood. Since podocyte EGFR/STAT3 signaling is known to mediate the development of anti-GBM glomerulonephritis, we investigated the effect of glucocorticoids on EGFR/STAT3 signaling in podocytes. We found that the levels of phosphorylated (activated) EGFR and STAT3 in podocytes were markedly elevated in anti-GBM patients without glucocorticoids treatment, but were normalized in patients with glucocorticoids treatment. In a rat model of anti-GBM glomerulonephritis, glucocorticoids treatment significantly attenuated the proteinuria, crescent formation, parietal epithelial cell (PEC) activation and proliferation, accompanied by elimination of podocyte EGFR/STAT3 signaling activation. In cultured podocytes, glucocorticoids were found to inhibit HB-EGF-induced EGFR and STAT3 activation. The conditioned medium from podocytes treated with HB-EGF in the absence but not presence of glucocorticoids was capable of activating Notch signaling (which is known to be involved in PEC proliferation and crescent formation) and enhancing proliferative activity in primary PECs, suggesting that glucocorticoids prevent podocytes from producing secreted factors that cause PEC proliferation and crescent formation. Furthermore, we found that glucocorticoids can downregulate the expression of EGFR ligands, EGF and HB-EGF, while upregulate the expression of EGFR inhibitor, Gene 33, explaining how glucocorticoids suppress EGFR signaling. Taken together, glucocorticoids exert therapeutic effect on anti-GBM crescentic glomerulonephritis through inhibiting podocyte EGFR/STAT3 signaling and the downstream pathway that leads to PEC proliferation and crescent formation.
Collapse
Affiliation(s)
- Xiaomei Wu
- National Clinical Research Center for Kidney Diseases, Jingling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Lu Ren
- National Clinical Research Center of Kidney Diseases, Jinling Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Qianqian Yang
- National Clinical Research Center of Kidney Diseases, Jinling Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Hui Song
- National Clinical Research Center for Kidney Diseases, Jingling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Qiaoli Tang
- National Clinical Research Center for Kidney Diseases, Jingling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Mingchao Zhang
- National Clinical Research Center for Kidney Diseases, Jingling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Jiong Zhang
- National Clinical Research Center for Kidney Diseases, Jingling Hospital, Nanjing University School of Medicine, Nanjing, China
- *Correspondence: Jiong Zhang
| | - Zheng Tang
- National Clinical Research Center for Kidney Diseases, Jingling Hospital, Nanjing University School of Medicine, Nanjing, China
- Zheng Tang
| | - Shaolin Shi
- National Clinical Research Center for Kidney Diseases, Jingling Hospital, Nanjing University School of Medicine, Nanjing, China
- Shaolin Shi
| |
Collapse
|
5
|
Role of Endothelial Glucocorticoid Receptor in the Pathogenesis of Kidney Diseases. Int J Mol Sci 2021; 22:ijms222413295. [PMID: 34948091 PMCID: PMC8706765 DOI: 10.3390/ijms222413295] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/05/2021] [Accepted: 12/07/2021] [Indexed: 01/12/2023] Open
Abstract
Glucocorticoids, as multifunctional hormones, are widely used in the treatment of various diseases including nephrological disorders. They are known to affect immunological cells, effectively treating many autoimmune and inflammatory processes. Furthermore, there is a growing body of evidence demonstrating the potent role of glucocorticoids in non-immune cells such as podocytes. Moreover, novel data show additional pathways and processes affected by glucocorticoids, such as the Wnt pathway or autophagy. The endothelium is currently considered as a key organ in the regulation of numerous kidney functions such as glomerular filtration, vascular tone and the regulation of inflammation and coagulation. In this review, we analyse the literature concerning the effects of endothelial glucocorticoid receptor signalling on kidney function in health and disease, with special focus on hypertension, diabetic kidney disease, glomerulopathies and chronic kidney disease. Recent studies demonstrate the potential role of endothelial GR in the prevention of fibrosis of kidney tissue and cell metabolism through Wnt pathways, which could have a protective effect against disease progression. Another important aspect covered in this review is blood pressure regulation though GR and eNOS. We also briefly cover potential therapies that might affect the endothelial glucocorticoid receptor and its possible clinical implications, with special interest in selective or local GR stimulation and potential mitigation of GC treatment side effects.
Collapse
|
6
|
Huang Y, Wang J, Jiang K, Chung EJ. Improving kidney targeting: The influence of nanoparticle physicochemical properties on kidney interactions. J Control Release 2021; 334:127-137. [PMID: 33892054 DOI: 10.1016/j.jconrel.2021.04.016] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 12/23/2022]
Abstract
Kidney-targeted nanoparticles have become of recent interest due to their potential to deliver drugs directly to diseased tissue, decrease off-target adverse effects, and increase overall tolerability to patients with chronic kidney disease that require lifelong drug exposure. Given the physicochemical properties of nanoparticles can drastically affect their ability to extravasate past cellular and biological barriers and access the kidneys, we surveyed the literature from the past decade and analyzed how nanoparticle size, charge, shape, and material density affects passage and interaction with the kidneys. Specifically, we found that nanoparticle size impacted the mechanism of nanoparticle entry into the kidneys such as glomerular filtration or tubular secretion. In addition, we found charge, aspect ratio, and material density influences nanoparticle renal retention and provide insights for designing nanoparticles for passive kidney targeting. Finally, we conclude by highlighting active targeting strategies that bolster kidney retention and discuss the clinical status of nanomedicine for kidney diseases.
Collapse
Affiliation(s)
- Yi Huang
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Jonathan Wang
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Kairui Jiang
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Eun Ji Chung
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA; Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, USA; Department of Medicine, Division of Nephrology and Hypertension, University of Southern California, Los Angeles, CA, USA; Department of Surgery, Division of Vascular Surgery and Endovascular Therapy, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
7
|
de Assis Pinheiro J, Freitas FV, Borçoi AR, Mendes SO, Conti CL, Arpini JK, Dos Santos Vieira T, de Souza RA, Dos Santos DP, Barbosa WM, Archanjo AB, de Oliveira MM, Dos Santos JG, Sorroche BP, Casali-da-Rocha JC, Trivilin LO, Borloti EB, Louro ID, Arantes LMRB, Alvares-da-Silva AM. Alcohol consumption, depression, overweight and cortisol levels as determining factors for NR3C1 gene methylation. Sci Rep 2021; 11:6768. [PMID: 33762648 PMCID: PMC7990967 DOI: 10.1038/s41598-021-86189-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 03/09/2021] [Indexed: 02/06/2023] Open
Abstract
The NR3C1 glucocorticoid receptor (GR) gene is a component of the stress response system, which can be regulated by epigenetic mechanisms. NR3C1 methylation has been associated with trauma and mental issues, including depression, post-traumatic stress, anxiety, and personality disorders. Previous studies have reported that stressful events are involved in NR3C1 gene methylation, suggesting that its regulation under environmental effects is complex. The present study aimed to analyze associations involving stressors such as socioeconomic status, health conditions, and lifestyle in relation to NR3C1 methylation in adults. This study included 386 individual users of the Brazilian Public Unified Health System (SUS), and evaluated socioeconomic and health conditions, body mass index, cortisol levels, and lifestyle. Data were correlated with NR3C1 methylation, determined using DNA pyrosequencing. The results showed that alcohol consumption, overweight, and high cortisol levels were related to NR3C1 demethylation, while depression was related to its methylation. Habits, lifestyle, and health status may influence NR3C1 gene regulation via methylation, revealing the complexity of environmental impacts on NR3C1 methylation.
Collapse
Affiliation(s)
- Júlia de Assis Pinheiro
- Biotechnology/Renorbio Graduate Program, Universidade Federal do Espirito Santo, Vitoria, ES, Brazil
| | - Flávia Vitorino Freitas
- Biotechnology/Renorbio Graduate Program, Universidade Federal do Espirito Santo, Vitoria, ES, Brazil.,Department of Pharmacy and Nutrition, Universidade Federal do Espirito Santo, Alegre, ES, Brazil
| | - Aline Ribeiro Borçoi
- Biotechnology/Renorbio Graduate Program, Universidade Federal do Espirito Santo, Vitoria, ES, Brazil
| | - Suzanny Oliveira Mendes
- Biotechnology/Renorbio Graduate Program, Universidade Federal do Espirito Santo, Vitoria, ES, Brazil
| | - Catarine Lima Conti
- Biotechnology/Renorbio Graduate Program, Universidade Federal do Espirito Santo, Vitoria, ES, Brazil
| | - Juliana Krüger Arpini
- Graduate Program in Forest Sciences, Universidade Federal do Espirito Santo, Alegre, ES, Brazil
| | - Tamires Dos Santos Vieira
- Biotechnology/Renorbio Graduate Program, Universidade Federal do Espirito Santo, Vitoria, ES, Brazil
| | | | | | - Wagner Miranda Barbosa
- Department of Pharmacy and Nutrition, Universidade Federal do Espirito Santo, Alegre, ES, Brazil
| | - Anderson Barros Archanjo
- Biotechnology/Renorbio Graduate Program, Universidade Federal do Espirito Santo, Vitoria, ES, Brazil
| | - Mayara Mota de Oliveira
- Biotechnology/Renorbio Graduate Program, Universidade Federal do Espirito Santo, Vitoria, ES, Brazil
| | | | - Bruna Pereira Sorroche
- Molecular Oncology Research Center, Hospital do Câncer de Barretos, Barretos, SP, Brazil
| | | | | | - Elizeu Batista Borloti
- Department of Social and Developmental Psychology, Universidade Federal do Espírito Santo, Vitoria, ES, Brazil
| | - Iuri Drumond Louro
- Biotechnology/Renorbio Graduate Program, Universidade Federal do Espirito Santo, Vitoria, ES, Brazil
| | | | - Adriana Madeira Alvares-da-Silva
- Biotechnology/Renorbio Graduate Program, Universidade Federal do Espirito Santo, Vitoria, ES, Brazil. .,Department of Morphology, Health Science Center, Universidade Federal do Espirito Santo, Vitoria, ES, Brazil. .,Departamento de Biologia, Universidade Federal do Espirito Santo, Alto Universitário Sem Número, Alegre, ES, 29500000, Brazil.
| |
Collapse
|
8
|
Ohno Y, Sone M, Inagaki N, Takeda Y, Kurihara I, Tsuiki M, Ichijo T, Wada N, Katabami T, Ogawa Y, Okamura S, Fukuoka T, Kai T, Izawa S, Yoshikawa Y, Hashimoto S, Yamada M, Chiba Y, Naruse M. Latent Autonomous Cortisol Secretion From Apparently Nonfunctioning Adrenal Tumor in Nonlateralized Hyperaldosteronism. J Clin Endocrinol Metab 2019; 104:4382-4389. [PMID: 31058960 DOI: 10.1210/jc.2018-02790] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 04/30/2019] [Indexed: 02/13/2023]
Abstract
CONTEXT Adrenal tumors (ATs), even those diagnosed as nonfunctioning, may cause metabolic disorders. Some primary aldosteronism (PA) patients with ATs are diagnosed with bilateral PA based on adrenal venous sampling (AVS), and their ATs are apparently nonfunctioning. OBJECTIVE To clarify the influence of apparently nonfunctioning ATs, we compared hormone levels and clinical complications between bilateral PA cases with and without ATs. DESIGN, SETTING, AND PARTICIPANTS After retrospectively assessing 2814 patients with PA in the multicenter Japan PA study, bilateral PA cases on AVS were divided into cases with and without ATs by computed tomography findings. Importantly, patients with cortisol levels >1.8 µg/dL after the 1-mg dexamethasone suppression test (DST) were excluded. Clinical characteristics and biochemical data were compared between them. The correlation between AT size and hormone levels was also analyzed. MAIN OUTCOME MEASURES Analyzed were 196 bilateral PA patients with ATs and 331 those without ATs. Although basal cortisol and aldosterone levels were similar between them, cortisol levels after the 1-mg DST and the prevalences of diabetes mellitus and proteinuria were significantly higher and ACTH levels and plasma renin activity were significantly lower in cases with ATs than in those without. After adjusting for patients' backgrounds, cortisol levels after the 1-mg DST and plasma renin activity remained significantly different between them. Moreover, cortisol levels after the 1-mg DST and ACTH levels correlated with AT size. CONCLUSIONS Apparently nonfunctioning ATs in bilateral PA cases may cause latent autonomous cortisol secretion, inducing diabetes and proteinuria.
Collapse
Affiliation(s)
- Youichi Ohno
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University, Kyoto, Japan
| | - Masakatsu Sone
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University, Kyoto, Japan
| | - Nobuya Inagaki
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University, Kyoto, Japan
| | - Yoshiyu Takeda
- Department of Internal Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Isao Kurihara
- Department of Endocrinology, Metabolism and Nephrology, School of Medicine Keio University, Tokyo, Japan
| | - Mika Tsuiki
- Department of Endocrinology and Metabolism, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Takamasa Ichijo
- Department of Diabetes and Endocrinology, Saiseikai Yokohamashi Tobu Hospital, Yokohama, Japan
| | - Norio Wada
- Department of Diabetes and Endocrinology, Sapporo City General Hospital, Sapporo, Japan
| | - Takuyuki Katabami
- Division of Metabolism and Endocrinology, Department of Internal Medicine, St. Marianna University School of Medicine Yokohama City Seibu Hospital, Yokohama, Japan
| | - Yoshihiro Ogawa
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | - Tomikazu Fukuoka
- Department of Internal Medicine, Matsuyama Red Cross Hospital, Matsuyama, Japan
| | - Tatsuya Kai
- Department of Cardiology, Saiseikai Tondabayashi Hospital, Tondabayashi, Japan
| | - Shoichiro Izawa
- Division of Cardiovascular Medicine, Endocrinology and Metabolism, Department of Molecular Medicine and Therapeutics, Tottori University Faculty of Medicine, Yonago, Japan
| | - Yuichiro Yoshikawa
- Department of Endocrinology and Diabetes Mellitus, Misato Kenwa Hospital, Misato, Japan
| | - Shigeatsu Hashimoto
- Division of Nephrology, Hypertension, Endocrinology, and Diabetology/Metabolism, Fukushima Medical University Hospital, Fukushima, Japan
| | - Masanobu Yamada
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yoshiro Chiba
- Endovascular Treatment Group, Mito Saiseikai General Hospital, Mito, Japan
| | - Mitsuhide Naruse
- Clinical Research Institute of Endocrinology and Metabolism, Kyoto Medical Center, National Hospital Organization, Kyoto, Japan
- Endocrinology Center, Ijinkai Takeda General Hospital, Kyoto, Japan
| | | |
Collapse
|
9
|
Tang TT, Lv LL, Wang B, Cao JY, Feng Y, Li ZL, Wu M, Wang FM, Wen Y, Zhou LT, Ni HF, Chen PS, Gu N, Crowley SD, Liu BC. Employing Macrophage-Derived Microvesicle for Kidney-Targeted Delivery of Dexamethasone: An Efficient Therapeutic Strategy against Renal Inflammation and Fibrosis. Am J Cancer Res 2019; 9:4740-4755. [PMID: 31367254 PMCID: PMC6643445 DOI: 10.7150/thno.33520] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 05/15/2019] [Indexed: 01/13/2023] Open
Abstract
Although glucocorticoids are the mainstays in the treatment of renal diseases for decades, the dose dependent side effects have largely restricted their clinical use. Microvesicles (MVs) are small lipid-based membrane-bound particles generated by virtually all cells. Here we show that RAW 264.7 macrophage cell-derived MVs can be used as vectors to deliver dexamethasone (named as MV-DEX) targeting the inflamed kidney efficiently. Methods: RAW macrophages were incubated with dexamethasone and then MV-DEX was isolated from the supernatants by centrifugation method. Nanoparticle tracking analysis, transmission electron microscopy, western blot and high-performance liquid chromatography were used to analyze the properties of MV-DEX. The LC-MS/MS was applied to investigate the protein compositions of MV-DEX. Based on the murine models of LPS- or Adriamycin (ADR)-induced nephropathy or in-vitro culture of glomerular endothelial cells, the inflammation-targeting characteristics and the therapeutic efficacy of MV-DEX was examined. Finally, we assessed the side effects of chronic glucocorticoid therapy in MV-DEX-treated mice. Results: Proteomic analysis revealed distinct integrin expression patterns on the MV-DEX surface, in which the integrin αLβ2 (LFA-1) and α4β1 (VAL-4) enabled them to adhere to the inflamed kidney. Compared to free DEX treatment, equimolar doses of MV-DEX significantly attenuated renal injury with an enhanced therapeutic efficacy against renal inflammation and fibrosis in murine models of LPS- or ADR-induced nephropathy. In vitro, MV-DEX with about one-fifth of the doses of free DEX achieved significant anti-inflammatory efficacy by inhibiting NF-κB activity. Mechanistically, MV-DEX could package and deliver glucocorticoid receptors to renal cells, thereby, increasing cellular levels of the receptor and improving cell sensitivity to glucocorticoids. Notably, delivering DEX in MVs significantly reduced the side effects of chronic glucocorticoid therapy (e.g., hyperglycemia, suppression of HPA axis). Conclusion: In summary, macrophage-derived MVs efficiently deliver DEX into the inflamed kidney and exhibit a superior capacity to suppress renal inflammation and fibrosis without apparent glucocorticoid adverse effects. Our findings demonstrate the effectiveness and security of a novel drug delivery strategy with promising clinical applications.
Collapse
Affiliation(s)
- Tao-Tao Tang
- Institute of Nephrology, Zhong Da Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Lin-Li Lv
- Institute of Nephrology, Zhong Da Hospital, School of Medicine, Southeast University, Nanjing, China,✉ Corresponding authors: Bi-Cheng Liu () or Lin-Li Lv ()
| | - Bin Wang
- Institute of Nephrology, Zhong Da Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Jing-Yuan Cao
- Institute of Nephrology, Zhong Da Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Ye Feng
- Institute of Nephrology, Zhong Da Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Zuo-Lin Li
- Institute of Nephrology, Zhong Da Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Min Wu
- Institute of Nephrology, Zhong Da Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Feng-Mei Wang
- Institute of Nephrology, Zhong Da Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Yi Wen
- Institute of Nephrology, Zhong Da Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Le-Ting Zhou
- Institute of Nephrology, Zhong Da Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Hai-Feng Ni
- Institute of Nephrology, Zhong Da Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Ping-Sheng Chen
- Institute of Nephrology, Zhong Da Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Ning Gu
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, China
| | - Steven D. Crowley
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, North Carolina, United States
| | - Bi-Cheng Liu
- ✉ Corresponding authors: Bi-Cheng Liu () or Lin-Li Lv ()
| |
Collapse
|
10
|
Abstract
Glucocorticoids are potent anti-inflammatory agents that are commonly used in the treatment of various glomerular diseases. Data from in vitro and in vivo studies, in both animals and humans, convincingly demonstrate that glucocorticoids have many beneficial direct effects on glomeruli, including podocytes, suggesting that, in theory, systemic administration is not necessary to achieve therapeutic benefit. Indeed, it is increasingly recognized that systemic steroids often have an unfavorable risk-to-benefit ratio. As we move into an age of personalized medicine, strategies to develop targeted steroid delivery systems and individualized risk assessment algorithms are desirable in clinicians' efforts to "first, do no harm."
Collapse
Affiliation(s)
- Julie E Goodwin
- Department of Pediatrics, Yale University School of Medicine , New Haven, Connecticut ; and Vascular Biology and Therapeutics Program, Yale University School of Medicine , New Haven, Connecticut
| |
Collapse
|
11
|
Schijvens AM, Ter Heine R, de Wildt SN, Schreuder MF. Pharmacology and pharmacogenetics of prednisone and prednisolone in patients with nephrotic syndrome. Pediatr Nephrol 2019; 34:389-403. [PMID: 29549463 PMCID: PMC6349812 DOI: 10.1007/s00467-018-3929-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 01/19/2018] [Accepted: 02/19/2018] [Indexed: 01/29/2023]
Abstract
Nephrotic syndrome is one of the most common glomerular disorders in childhood. Glucocorticoids have been the cornerstone of the treatment of childhood nephrotic syndrome for several decades, as the majority of children achieves complete remission after prednisone or prednisolone treatment. Currently, treatment guidelines for the first manifestation and relapse of nephrotic syndrome are mostly standardized, while large inter-individual variation is present in the clinical course of disease and side effects of glucocorticoid treatment. This review describes the mechanisms of glucocorticoid action and clinical pharmacokinetics and pharmacodynamics of prednisone and prednisolone in nephrotic syndrome patients. However, these mechanisms do not account for the large inter-individual variability in the response to glucocorticoid treatment. Previous research has shown that genetic factors can have a major influence on the pharmacokinetic and dynamic profile of the individual patient. Therefore, pharmacogenetics may have a promising role in personalized medicine for patients with nephrotic syndrome. Currently, little is known about the impact of genetic polymorphisms on glucocorticoid response and steroid-related toxicities in children with nephrotic syndrome. Although the evidence is limited, the data summarized in this study do suggest a role for pharmacogenetics to improve individualization of glucocorticoid therapy. Therefore, studies in larger cohorts with nephrotic syndrome patients are necessary to draw final conclusions about the influence of genetic polymorphisms on the glucocorticoid response and steroid-related toxicities to ultimately implement pharmacogenetics in clinical practice.
Collapse
Affiliation(s)
- Anne M Schijvens
- Department of Pediatric Nephrology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Amalia Children's Hospital, 804, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Rob Ter Heine
- Department of Pharmacy, Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - Saskia N de Wildt
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, The Netherlands
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Michiel F Schreuder
- Department of Pediatric Nephrology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Amalia Children's Hospital, 804, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
12
|
Zhao X, Hwang DY, Kao HY. The Role of Glucocorticoid Receptors in Podocytes and Nephrotic Syndrome. NUCLEAR RECEPTOR RESEARCH 2018; 5. [PMID: 30417008 PMCID: PMC6224173 DOI: 10.11131/2018/101323] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Glucocorticoid receptor (GC), a founding member of the nuclear hormone receptor superfamily, is a glucocorticoid-activated transcription factor that regulates gene expression and controls the development and homeostasis of human podocytes. Synthetic glucocorticoids are the standard treatment regimens for proteinuria (protein in the urine) and nephrotic syndrome (NS) caused by kidney diseases. These include minimal change disease (MCD), focal segmental glomerulosclerosis (FSGS), membranous nephropathy (MN) and immunoglobulin A nephropathy (IgAN) or subsequent complications due to diabetes mellitus or HIV infection. However, unwanted side effects and steroid-resistance remain major issues for their long-term use. Furthermore, the mechanism by which glucocorticoids elicit their renoprotective activity in podocyte and glomeruli is poorly understood. Podocytes are highly differentiated epithelial cells that contribute to the integrity of kidney glomerular filtration barrier. Injury or loss of podocytes leads to proteinuria and nephrotic syndrome. Recent studies in multiple experimental models have begun to explore the mechanism of GC action in podocytes. This review will discuss progress in our understanding of the role of glucocorticoid receptor and glucocorticoids in podocyte physiology and their renoprotective activity in nephrotic syndrome.
Collapse
Affiliation(s)
- Xuan Zhao
- Department of Biochemistry, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, USA
| | - Daw-Yang Hwang
- Division of Nephrology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hung-Ying Kao
- Department of Biochemistry, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, USA
| |
Collapse
|
13
|
Wu X, Zhang M, Huang X, Zhang L, Zeng C, Zhang J, Liu Z, Tang Z. Therapeutic Mechanism of Glucocorticoids on Cellular Crescent Formation in Patients With Antiglomerular Basement Membrane Disease. Am J Med Sci 2017; 354:145-151. [PMID: 28864372 DOI: 10.1016/j.amjms.2017.04.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 04/07/2017] [Accepted: 04/20/2017] [Indexed: 11/13/2022]
Abstract
BACKGROUND This study aimed to explore the therapeutic mechanism of glucocorticoids (GCs) in antiglomerular basement membrane disease. MATERIALS AND METHODS Thirty-four patients with biopsy-proven antiglomerular basement membrane nephritis were divided into the following 2 groups: group 1 (patients treated with GCs, n = 22) and group 2 (patients who were not treated with GCs, n = 12). The expression of parietal epithelial cells (PECs), activated PECs and glucocorticoid receptors (GRs) was examined quantitatively and compared between the 2 groups. Correlations between GR expression in glomeruli and patients' clinicopathological indices were also analyzed. RESULTS Compared with patients in group 2, patients in group 1 showed lower levels of serum creatinine (SCr) (P = 0.03), average cellular crescent percentage (P = 0.005) and macrophages infiltrating in renal interstitium (P = 0.03). PECs (P = 0.007) and activated PECs (P = 0.03) were strongly detected in the cellular components of classic crescents, and both were significantly reduced in group 1 compared to group 2. GR expression either in glomeruli (P = 0.01) or interstitium (P = 0.009) was lower in group 1 after GCs treatment than in group 2. Additionally, GR expression in glomeruli was strongly correlated with renal function (SCr: r = 0.45, P = 0.009; eGFR: r = -0.35, P = 0.046), the proportion of cellular crescents (r = 0.67, P < 0.001), PECs (r = 0.64, P < 0.001) and activated PECs (r = 0.72, P < 0.001), and the degree of interstitial (r = 0.50, P = 0.004) and glomerular (r = 0.49, P = 0.007) macrophage infiltration. CONCLUSIONS GCs might exert their therapeutic effects via inhibiting the activation and proliferation of PECs, as well as macrophage infiltration, which could contribute to crescent formation and determine renal survival. GRs are involved in this process as well.
Collapse
Affiliation(s)
- Xiaomei Wu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Mingchao Zhang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Xiao Huang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Lihua Zhang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Caihong Zeng
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Jiong Zhang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China.
| | - Zhihong Liu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Zheng Tang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China.
| |
Collapse
|
14
|
Loss of the podocyte glucocorticoid receptor exacerbates proteinuria after injury. Sci Rep 2017; 7:9833. [PMID: 28852159 PMCID: PMC5575043 DOI: 10.1038/s41598-017-10490-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 08/09/2017] [Indexed: 12/11/2022] Open
Abstract
Nephrotic syndrome is a common disorder in adults and children whose etiology is largely unknown. Glucocorticoids remain the mainstay of therapy in most cases, though their mechanism of action remains poorly understood. Emerging evidence suggests that immunomodulatory therapies used in nephrotic syndrome directly target the podocytes. To study how steroids directly affect the podocytes in the treatment of proteinuria, we created a mouse model with podocyte-specific deletion of the glucocorticoid receptor. The podocyte-specific glucocorticoid receptor (GR) knockout mice had similar renal function and protein excretion compared to wild type. However, after glomerular injury induced by either LPS or nephrotoxic serum, the podocyte GR knockout mice demonstrated worsened proteinuria compared to wild type. Ultrastructural examination of podocytes confirmed more robust foot process effacement in the knockout animals. Expression of several key slit diaphragm protein was down regulated in pGR KO mice. Primary podocytes isolated from wild type and podocyte GR knockout mice showed similar actin stress fiber staining patterns in unstimulated conditions. Yet, when exposed to LPS, GR knockout podocytes demonstrated fewer stress fibers and impaired migration compared to wild type podocytes. We conclude that the podocyte glucocorticoid receptor is important for limiting proteinuria in settings of podocyte injury.
Collapse
|
15
|
Short courses of daily prednisolone during upper respiratory tract infections reduce relapse frequency in childhood nephrotic syndrome. Pediatr Nephrol 2017; 32:1377-1382. [PMID: 28341877 DOI: 10.1007/s00467-017-3640-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 03/02/2017] [Accepted: 03/03/2017] [Indexed: 10/19/2022]
Abstract
BACKGROUND Relapses of childhood nephrotic syndrome (NS) are frequently precipitated by viral upper respiratory tract infections (URTIs). A review of the literature reveals that in patients with steroid-dependent NS on alternate day corticosteroids, a short course of daily corticosteroid therapy during the course of an URTI may reduce relapse frequency. OBJECTIVE To assess the effect of a short course of low-dose corticosteroid therapy during the course of an URTI on relapse frequency in patients with steroid-sensitive NS who have not been taking any treatment for a minimum period of 3 months. METHODS A double-blind placebo-controlled crossover trial was conducted on 48 patients with idiopathic NS who had not been receiving corticosteroid therapy for a minimum of 3 months. Patients were randomized into two groups. Group A received 5 days of daily prednisolone at 0.5 mg/kg at the onset of an URTI while group B received 5 days of placebo. Both groups were followed up for 1 year and the URTI-induced relapse frequency was noted. A crossover was performed during the next year, with group A receiving placebo and group B receiving prednisolone. RESULTS Thirty-three patients completed the study. In the treatment group, 115 episodes of URTI led to 11 relapses while in the control group 101 episodes of URTI led to 25 relapses. There was no significant difference between the mean number of URTIs between the treatment and control groups. The treatment group had significantly less relapses compared to the control group (p = 0.014). Within the treatment group, 65.6% did not relapse, while the remainder had a single relapse. In contrast, only 40.6% of the control group remained in remission while 40.6% suffered a single relapse and 18.8% had two or more relapses. CONCLUSIONS Prescribing a short course of daily corticosteroids during an URTI significantly reduces the frequency of URTI-induced relapse in patients with steroid-responsive NS who are off corticosteroid therapy.
Collapse
|
16
|
Müller-Deile J, Schiffer M. Podocytes from the diagnostic and therapeutic point of view. Pflugers Arch 2017; 469:1007-1015. [PMID: 28508947 DOI: 10.1007/s00424-017-1993-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 05/04/2017] [Indexed: 01/23/2023]
Abstract
The central role of podocytes in glomerular diseases makes this cell type an interesting diagnostic tool as well as a therapeutic target. In this review, we discuss the current literature on the use of podocytes and podocyte-specific markers as non-invasive diagnostic tools in different glomerulopathies. Furthermore, we highlight the direct effects of drugs currently used to treat primary glomerular diseases and describe their direct cellular effects on podocytes. A new therapeutic potential is seen in drugs targeting the podocytic actin cytoskeleton which is essential for podocyte foot process structure and function. Incubation of cultured human podocyte cell lines with sera from patients with active glomerular diseases is currently also used to identify novel circulating factors with pathophysiological relevance for the glomerular filtration barrier. In addition, treatment of detached urinary podocytes from patients with substances that restore their cytoskeleton might serve as a novel personalized tool to estimate their potential for podocyte recovery ex vivo.
Collapse
Affiliation(s)
- Janina Müller-Deile
- Department of Nephrology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| | - Mario Schiffer
- Department of Nephrology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
17
|
Albumin-based nanoparticles as methylprednisolone carriers for targeted delivery towards the neonatal Fc receptor in glomerular podocytes. Int J Mol Med 2017; 39:851-860. [PMID: 28259932 PMCID: PMC5360426 DOI: 10.3892/ijmm.2017.2902] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 02/16/2017] [Indexed: 01/08/2023] Open
Abstract
Glucocorticoids (GCs) are commonly used in the treatment of nephrotic syndrome. However, high doses and long periods of GC therapy can result in severe side effects. The present study aimed to selectively deliver albumin-methylprednisolone (MP) nanoparticles towards glomerular podocytes, which highly express the specific neonatal Fc receptor (FcRn) of albumin. Bovine serum albumin (BSA) was labeled with a fluorescent dye and linked with modified MP via an amide bond. The outcome nanoparticle named BSA633-MP showed a uniform size with a diameter of approximately 10 nm and contained 12 drug molecules on average. The nanoconjugates were found to be stable at pH 7.4 and acid-sensitive at pH 4.0, with approximately 72% release of the MP drug after 48 h of incubation. The nanoparticle demonstrated a 36-fold uptake in receptor-specific cellular delivery in the FcRn-expressing human podocytes compared to the uptake in the non-FcRn-expressing control cells. Co-localization further confirmed that uptake of the nanoconjugates involved receptor-mediated endocytosis followed by lysosome associated transportation. In vitro cellular experiments indicated that the BSA633-MP ameliorated puromycin aminonucleoside-induced podocyte apoptosis. Moreover, in vivo fluorescence molecular imaging showed that BSA633-MP was mainly accumulated in the liver and kidney after intravenous dosing for 24 h. Collectively, this study may provide an approach for the effective and safe therapy of nephrotic syndrome.
Collapse
|
18
|
Kuppe C, van Roeyen C, Leuchtle K, Kabgani N, Vogt M, Van Zandvoort M, Smeets B, Floege J, Gröne HJ, Moeller MJ. Investigations of Glucocorticoid Action in GN. J Am Soc Nephrol 2016; 28:1408-1420. [PMID: 27895155 DOI: 10.1681/asn.2016010060] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 10/04/2016] [Indexed: 12/21/2022] Open
Abstract
For several decades, glucocorticoids have been used empirically to treat rapid progressive GN. It is commonly assumed that glucocorticoids act primarily by dampening the immune response, but the mechanisms remain incompletely understood. In this study, we inactivated the glucocorticoid receptor (GR) specifically in kidney epithelial cells using Pax8-Cre/GRfl/fl mice. Pax8-Cre/GRfl/fl mice did not exhibit an overt spontaneous phenotype. In mice treated with nephrotoxic serum to induce crescentic nephritis (rapidly progressive GN), this genetic inactivation of the GR in kidney epithelial cells exerted renal benefits, including inhibition of albuminuria and cellular crescent formation, similar to the renal benefits observed with high-dose prednisolone in control mice. However, genetic inactivation of the GR in kidney epithelial cells did not induce the immunosuppressive effects observed with prednisolone. In vitro, prednisolone and the pharmacologic GR antagonist mifepristone each acted directly on primary cultures of parietal epithelial cells, inhibiting cellular outgrowth and proliferation. In wild-type mice, pharmacologic treatment with the GR antagonist mifepristone also attenuated disease as effectively as high-dose prednisolone without the systemic immunosuppressive effects. Collectively, these data show that glucocorticoids act directly on activated glomerular parietal epithelial cells in crescentic nephritis. Furthermore, we identified a novel therapeutic approach in crescentic nephritis, that of glucocorticoid antagonism, which was at least as effective as high-dose prednisolone with potentially fewer adverse effects.
Collapse
Affiliation(s)
- Christoph Kuppe
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule Aachen University, Aachen, Germany
| | - Claudia van Roeyen
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule Aachen University, Aachen, Germany
| | - Katja Leuchtle
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule Aachen University, Aachen, Germany.,Interdisciplinary Centre for Clinical Research, Rheinisch-Westfälische Technische Hochschule Aachen University Hospital, Aachen, Germany
| | - Nazanin Kabgani
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule Aachen University, Aachen, Germany
| | - Michael Vogt
- Interdisciplinary Centre for Clinical Research, Rheinisch-Westfälische Technische Hochschule Aachen University Hospital, Aachen, Germany
| | - Marc Van Zandvoort
- Interdisciplinary Centre for Clinical Research, Rheinisch-Westfälische Technische Hochschule Aachen University Hospital, Aachen, Germany.,Department of Genetics and Cell Biology, Sector Molecular Cell Biology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Bart Smeets
- Department of Pathology, Radboud University, Nijmegen, The Netherlands; and
| | - Jürgen Floege
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule Aachen University, Aachen, Germany
| | - Hermann-Josef Gröne
- Cellular and Molecular Pathology, German Cancer Research Center, Heidelberg, Germany
| | - Marcus J Moeller
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule Aachen University, Aachen, Germany; .,Interdisciplinary Centre for Clinical Research, Rheinisch-Westfälische Technische Hochschule Aachen University Hospital, Aachen, Germany
| |
Collapse
|
19
|
Mallipattu SK, He JC. The podocyte as a direct target for treatment of glomerular disease? Am J Physiol Renal Physiol 2016; 311:F46-51. [PMID: 27097894 DOI: 10.1152/ajprenal.00184.2016] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 04/18/2016] [Indexed: 11/22/2022] Open
Abstract
The Centers for Disease Control and Prevention estimates more than 10% of adults in the United States, over 20 million Americans, have chronic kidney disease (CKD). A failure to maintain the glomerular filtration barrier directly contributes to the onset of CKD. The visceral epithelial cells, podocytes, are integral to the maintenance of this renal filtration barrier. Direct podocyte injury contributes to the onset and progression of glomerular diseases such as minimal change disease (MCD), focal segmental glomerular sclerosis (FSGS), diabetic nephropathy, and HIV-associated nephropathy (HIVAN). Since podocytes are terminally differentiated with minimal capacity to self-replicate, they are extremely sensitive to cellular injury. In the past two decades, our understanding of the mechanism(s) by which podocyte injury occurs has greatly expanded. With this newfound knowledge, therapeutic strategies have shifted to identifying targets directed specifically at the podocyte. Although the systemic effects of these agents are important, their direct effect on the podocyte proves to be essential in ameliorating glomerular disease. In this review, we highlight the mechanisms by which these agents directly target the podocyte independent of its systemic effects.
Collapse
Affiliation(s)
- Sandeep K Mallipattu
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York
| | - John C He
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York; and Renal Section, James J. Peters VA Medical Center, New York, New York
| |
Collapse
|
20
|
Podocyte directed therapy of nephrotic syndrome-can we bring the inside out? Pediatr Nephrol 2016; 31:393-405. [PMID: 25939817 DOI: 10.1007/s00467-015-3116-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 04/08/2015] [Accepted: 04/09/2015] [Indexed: 12/15/2022]
Abstract
Several of the drugs currently used for the treatment of glomerular diseases are prescribed for their immunotherapeutic or anti-inflammatory properties, based on the current understanding that glomerular diseases are mediated by immune responses. In recent years our understanding of podocytic signalling pathways and the crucial role of genetic predispositions in the pathology of glomerular diseases has broadened. Delineation of those signalling pathways supports the hypothesis that several of the medications and immunosuppressive agents used to treat glomerular diseases directly target glomerular podocytes. Several central downstream signalling pathways merge into regulatory pathways of the podocytic actin cytoskeleton and its connection to the slit diaphragm. The slit diaphragm and the cytoskeleton of the foot process represent a functional unit. A breakdown of the cytoskeletal backbone of the foot processes leads to internalization of slit diaphragm molecules, and internalization of slit diaphragm components in turn negatively affects cytoskeletal signalling pathways. Podocytes display a remarkable ability to recover from complete effacement and to re-form interdigitating foot processes and intact slit diaphragms after pharmacological intervention. This ability indicates an active inside-out signalling machinery which stabilizes integrin complex formations and triggers the recycling of slit diaphragm molecules from intracellular compartments to the cell surface. In this review we summarize current evidence from patient studies and model organisms on the direct impact of immunosuppressive and supportive drugs on podocyte signalling pathways. We highlight new therapeutic targets that may open novel opportunities to enhance and stabilize inside-out pathways in podocytes.
Collapse
|
21
|
Dexamethasone-dependent modulation of cyclic GMP synthesis in podocytes. Mol Cell Biochem 2015; 409:243-53. [PMID: 26272337 PMCID: PMC4589550 DOI: 10.1007/s11010-015-2528-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Accepted: 08/06/2015] [Indexed: 12/28/2022]
Abstract
Podocytes may be direct target for glucocorticoid therapy in glomerular proteinuric disease. Permeability of podocytes largely depends on their capacity to migrate which involves the contractile apparatus in their foot processes. In this study, we examined the effect of synthetic glucocorticoid dexamethasone (DEX) on the ability of podocytes to produce cyclic guanosine monophosphate (cGMP) in the presence of vasoactive factors, atrial natriuretic peptide (ANP), nitric oxide (NO), and angiotensin II (Ang II). We investigated also the effects of cGMP and DEX on podocyte motility. Primary rat podocytes and immortalized mouse podocytes were pretreated with 1 µM DEX for 4 or 24 h. Glomerular hypertension was mimicked by subjecting the cells to mechanical stress. Total and subcellular cGMP levels were determined in podocytes incubated with 0.1 µM ANP, 1 µM S-nitroso-N-acetyl penicillamine (SNAP), and 1 µM Ang II. Cell motility was estimated by a wound-healing assay. The ANP-dependent production of cGMP increased after 4 h exposition to DEX, but was attenuated after 24 h. Adversely, a 24-h pretreatment with DEX augmented the NO-dependent cGMP synthesis. Ang II suppressed the ANP-dependent cGMP production and the effect was enhanced by DEX in mechanical stress conditions. Mechanical stress reduced total cGMP production in the presence of all stimulators, whereas extracellular to total cGMP ratio increased. 8-Br cGMP enhanced podocyte migration which was accompanied by F-actin disassembly. In the presence of DEX these effects were prevented. We conclude that DEX modulates the production of cGMP in podocytes stimulated with vasoactive factors such as Ang II, ANP, and NO, and the effect is time-dependent. cGMP increases podocyte motility, which is prevented by DEX. This mechanism may account for the antiproteinuric effect of glucocorticoids.
Collapse
|
22
|
Sugano Y, Lindenmeyer MT, Auberger I, Ziegler U, Segerer S, Cohen CD, Neuhauss SCF, Loffing J. The Rho-GTPase binding protein IQGAP2 is required for the glomerular filtration barrier. Kidney Int 2015; 88:1047-56. [PMID: 26154927 DOI: 10.1038/ki.2015.197] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 04/27/2015] [Accepted: 05/07/2015] [Indexed: 01/09/2023]
Abstract
Podocyte dysfunction impairs the size selectivity of the glomerular filter, leading to proteinuria, hypoalbuminuria, and edema, clinically defined as nephrotic syndrome. Hereditary forms of nephrotic syndrome are linked to mutations in podocyte-specific genes. To identify genes contributing to podocyte dysfunction in acquired nephrotic syndrome, we studied human glomerular gene expression data sets for glomerular-enriched gene transcripts differentially regulated between pretransplant biopsy samples and biopsies from patients with nephrotic syndrome. Candidate genes were screened by in situ hybridization for expression in the zebrafish pronephros, an easy-to-use in vivo assay system to assess podocyte function. One glomerulus-enriched product was the Rho-GTPase binding protein, IQGAP2. Immunohistochemistry found a strong presence of IQGAP2 in normal human and zebrafish podocytes. In zebrafish larvae, morpholino-based knockdown of iqgap2 caused a mild foot process effacement of zebrafish podocytes and a cystic dilation of the urinary space of Bowman's capsule upon onset of urinary filtration. Moreover, the glomerulus of zebrafish morphants showed a glomerular permeability for injected high-molecular-weight dextrans, indicating an impaired size selectivity of the glomerular filter. Thus, IQGAP2 is a Rho-GTPase binding protein, highly abundant in human and zebrafish podocytes, which controls normal podocyte structure and function as evidenced in the zebrafish pronephros.
Collapse
Affiliation(s)
- Yuya Sugano
- Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.,Institute of Anatomy, University of Zurich, Zurich, Switzerland.,Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | | | - Ines Auberger
- Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.,Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Urs Ziegler
- Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.,Center for Microscopy and Image Analysis, University of Zurich, Zurich, Switzerland
| | - Stephan Segerer
- Institute of Physiology, University of Zurich, Zurich, Switzerland.,Division of Nephrology, University Hospital, Zurich, Switzerland
| | - Clemens D Cohen
- Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.,Institute of Physiology, University of Zurich, Zurich, Switzerland.,Division of Nephrology, Klinikum Harlaching, Munich, Germany
| | - Stephan C F Neuhauss
- Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.,Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Johannes Loffing
- Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.,Institute of Anatomy, University of Zurich, Zurich, Switzerland
| |
Collapse
|
23
|
Tanaka R, Kakuta T, Koiwa F, Fukagawa M, Saito A. Long-term prognosis of parathyroid function in chronic dialysis patients after PEIT-a single-centre trial. NDT Plus 2015; 1:iii29-iii34. [PMID: 25983970 PMCID: PMC4421133 DOI: 10.1093/ndtplus/sfn084] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2008] [Accepted: 03/07/2008] [Indexed: 01/29/2023] Open
Abstract
Background. Secondary hyperparathyroidism (SHPT) is a common complication observed in long-term dialysis patients. Percutaneous ethanol injection therapy (PEIT) of parathyroid glands (PTGs) is now established in Japan as a treatment option for SHPT. In this study, to elucidate the factors influencing efficacy in 1 year and relapse following PEIT, we analysed the long-term prognosis of parathyroid function that is known to have the greatest effect on therapeutic results. Methods. The study design was a retrospective cohort study. We studied 104 patients with SHPT, who underwent PEIT at Tokai University Hospital between January 1993 and December 2002, and we followed them up until January 2008. The effective group reached intact parathyroid hormone of 200 pg/ml or less, corrected calcium (Ca) of 10.5 mg/dl or less and phosphate (P) of 6.0 mg/dl or less. The ineffective group failed to achieve these criteria. Results. Among the 104 patients, 66 patients (63%) fulfilled the criteria for the effective group within the first year of PEIT. Using the multivariate logistic regression analysis, the number of PTGs before PEIT was a significant risk factor to deviate from the criteria. At the end of the surveillance period, 31 patients (30%) fulfilled the criteria, and their SHPT was controlled with PEIT. Using the multivariate logistic regression analysis, more than three PTGs at the beginning, and the increase in PTGs during the observation period were significant risk factors to deviate from the criteria. In conclusion, superior results with PEIT are obtained in terms of efficacy, remission period and risk of relapse, regardless of the size of the gland.
Collapse
Affiliation(s)
- Reika Tanaka
- Department of Internal Medicine , Tokai University School of Medicine
| | - Takatoshi Kakuta
- Department of Internal Medicine , Tokai University School of Medicine
| | - Fumihiko Koiwa
- Department of Internal Medicine, Division of Nephrology , Showa University Fujigaoka Hospital
| | - Masafumi Fukagawa
- Division of Nephrology and Dialysis Center , Kobe University School of Medicine , Japan
| | - Akira Saito
- Department of Internal Medicine , Tokai University School of Medicine
| |
Collapse
|
24
|
Goodwin JE. Glucocorticoids and the Cardiovascular System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015. [DOI: 10.1007/978-1-4939-2895-8_13] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
25
|
Yu M, Ren Q, Yu SY. Role of nephrin phosphorylation inducted by dexamethasone and angiotensin II in podocytes. Mol Biol Rep 2014; 41:3591-3595. [PMID: 24515388 DOI: 10.1007/s11033-014-3222-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 01/28/2014] [Indexed: 11/27/2022]
Abstract
The phosphorylation of nephrin plays an important role in maintaining the normal structure and function in podocytes. Dexamethasone (Dex) is usually used to treat glomerular diseases with proteinuria. In this study, we observated the effect of Dex and angiotensin II (AngII) on the change of nephrin phosphorylation in cultured podocytes. In vitro, cultured podocytes were exposed to AngII (10(-6) mol/L) pretreated with or without Dex (100 nM) for different time periods. Nck or Fyn were silenced by small interfering RNA (siRNA), nephrin and its phosphorylation expression were analyzed by Western blotting. In vitro, the phosphorylation of nephrin was significantly reduced after AngII stimulation (P < 0.05). Dex significantly resisted podocyte injury inducted by AngII via increasing the phosphorylation of nephrin (P < 0.05), siRNA silencing Nck can partially inhibited nephrin phosphorylation, siRNA silencing Fyn can completely inhibited nephrin phosphorylation. Phosphorylation of nephrin is important for the survival status of podocytes. Glucocorticoid treatment for human glomerulonephritis may exert its function by regulating Nck and Fyn complex to promote phosphorylation of nephrin. These results elucidate a novel mechanism of glucocorticoid treatment for glomerulonephritis.
Collapse
Affiliation(s)
- Meng Yu
- Nephrology Department of the Frist Hospital Affiliated to Ji'nan University, Guangzhou, China
| | | | | |
Collapse
|
26
|
He FF, Chen S, Su H, Meng XF, Zhang C. Actin-associated Proteins in the Pathogenesis of Podocyte Injury. Curr Genomics 2014; 14:477-84. [PMID: 24396279 PMCID: PMC3867723 DOI: 10.2174/13892029113146660014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2013] [Revised: 09/24/2013] [Accepted: 09/25/2013] [Indexed: 02/07/2023] Open
Abstract
Podocytes have a complex cellular architecture with interdigitating processes maintained by a precise organization of actin filaments. The actin-based foot processes of podocytes and the interposed slit diaphragm form the final barrier to proteinuria. The function of podocytes is largely based on the maintenance of the normal foot process structure with actin cytoskeleton. Cytoskeletal dynamics play important roles during normal podocyte development, in maintenance of the healthy glomerular filtration barrier, and in the pathogenesis of glomerular diseases. In this review, we focused on recent findings on the mechanisms of organization and reorganization of these actin-related molecules in the pathogenesis of podocyte injury and potential therapeutics targeting the regulation of actin cytoskeleton in podocytopathies.
Collapse
Affiliation(s)
- Fang-Fang He
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shan Chen
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hua Su
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xian-Fang Meng
- Department of Neurobiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
27
|
Long DA, Kolatsi-Joannou M, Price KL, Dessapt-Baradez C, Huang JL, Papakrivopoulou E, Hubank M, Korstanje R, Gnudi L, Woolf AS. Albuminuria is associated with too few glomeruli and too much testosterone. Kidney Int 2013; 83:1118-29. [PMID: 23447063 PMCID: PMC3674403 DOI: 10.1038/ki.2013.45] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 12/15/2012] [Accepted: 12/21/2012] [Indexed: 12/12/2022]
Abstract
Normally, the glomerular filtration barrier almost completely excludes circulating albumin from entering the urine. Genetic variation and both pre- and postnatal environmental factors may affect albuminuria in humans. Here we determine whether glomerular gene expression in mouse strains with naturally occurring variations in albuminuria would allow identification of proteins deregulated in relatively 'leaky' glomeruli. Albuminuria increased in female B6 to male B6 to female FVB/N to male FVB/N mice, whereas the number of glomeruli/kidney was the exact opposite. Testosterone administration led to increased albuminuria in female B6 but not female FVB/N mice. A common set of 39 genes, many expressed in podocytes, were significantly differentially expressed in each of the four comparisons: male versus female B6 mice, male versus female FVB/N mice, male FVB/N versus male B6 mice, and female FVB/N versus female B6 mice. The transcripts encoded proteins involved in oxidation/reduction reactions, ion transport, and enzymes involved in detoxification. These proteins may represent novel biomarkers and even therapeutic targets for early kidney and cardiovascular disease.
Collapse
Affiliation(s)
- David A Long
- Nephro-Urology Unit, UCL Institute of Child Health, London, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Yu-Shengyou, Li Y. Dexamethasone inhibits podocyte apoptosis by stabilizing the PI3K/Akt signal pathway. BIOMED RESEARCH INTERNATIONAL 2013; 2013:326986. [PMID: 23710442 PMCID: PMC3655450 DOI: 10.1155/2013/326986] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2013] [Revised: 03/16/2013] [Accepted: 03/28/2013] [Indexed: 02/01/2023]
Abstract
Corticosteroids like dexamethasone (DEX) are well-established treatments for the glomerular disease that sustain renal function, at least in part, by protecting podocytes from apoptotic death. In this study, we found that PAN causes abnormal expression of the PI3K-binding protein CD2AP, reducing PI3K/Akt signaling and promoting podocyte apoptosis. In contrast, DEX restores CD2AP-PI3K-Akt-GSK3 β signaling, which promotes the activity of antiapoptotic proteins and inhibits the activity of proapoptotic proteins. In addition, we also found that CD2AP was aberrantly colocalized with p85. Normal CD2AP mRNA expression and subcellular protein distribution were maintained in the PAN + DEX group, and DEX coapplication also reduced CD2AP-p85 colocalization. PAN evoked a concentration-dependent decrease in p-Akt and p-GSK3 β expressions, with p-Akt expression reaching a nadir at 15 min and p-GSK3 β expression at 30 min. DEX treatment induced a concentration-dependent reversal of PAN-induced p-Akt and p-GSK3 β downregulation. The PI3K inhibitor LY294002 blocked p-Akt and p-GSK3 β expressions in podocytes. Cells treated with PAN exhibited a significantly higher apoptosis rate than untreated or vehicle-treated cells. Furthermore, LY294002 exacerbated PAN-induced apoptosis. DEX cotreatment caused a significant concentration-dependent decrease in PAN-induced apoptosis. These results strongly suggest that DEX protects podocytes by stabilizing the expression and subcellular distribution of CD2AP and by maintaining the expression of phosphor-activated Akt and GSK3β .
Collapse
Affiliation(s)
- Yu-Shengyou
- Department of Pediatrics, Guangzhou First People's Hospital, Affiliate to Guangzhou Medical College, Guangzhou, Guangdong 510180, China
| | - Yu Li
- Department of Pediatrics, Guangzhou First People's Hospital, Affiliate to Guangzhou Medical College, Guangzhou, Guangdong 510180, China
| |
Collapse
|
29
|
Yu SY, Qi R. Role of bad in podocyte apoptosis induced by puromycin aminonucleoside. Transplant Proc 2013; 45:569-573. [PMID: 23498794 DOI: 10.1016/j.transproceed.2012.07.160] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Revised: 07/14/2012] [Accepted: 07/27/2012] [Indexed: 11/20/2022]
Abstract
Apoptosis, which is usually a response to the microenvironment, requires inactivation of prosurvival molecules. Apoptosis contributes to loss of podocytes in the course of renal injury, an event closely associated with the development of proteinuria. Dexamethasone (DEX) is the standard of care for most forms of nephrotic syndrome. However, the precise mechanisms of DEX action on podocytes are unknown. This study examined the hypothesis that cultured podocytes exposed to puromycin aminonucleoside (PAN) showed a reduced rate of apoptosis upon DEX exposure. Apoptotic podocytes seemed to be related to increased Bad mRNA and protein expressions. DEX reduced apoptosis by decreasing Bad mRNA and protein expressions, thereby protecting podocytes. These findings provided insights into the beneficial effects of DEX directly on podocytes. The present study illustrated the signal transduction mechanism of podocyte apoptosis induced by PAN.
Collapse
Affiliation(s)
- S Y Yu
- Department of Core Laboratory, GuangZhou Medical University, Guangzhou first people hospital, China.
| | | |
Collapse
|
30
|
Che R, Zhang A. Mechanisms of Glucocorticoid Resistance in Idiopathic Nephrotic Syndrome. ACTA ACUST UNITED AC 2013; 37:360-78. [DOI: 10.1159/000350163] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2013] [Indexed: 11/19/2022]
|
31
|
LIU HAIMEI, GAO XIA, XU HONG, FENG CHUN, KUANG XINYU, LI ZENGXIA, ZHA XILIANG. α-Actinin-4 is involved in the process by which dexamethasone protects actin cytoskeleton stabilization from adriamycin-induced podocyte injury. Nephrology (Carlton) 2012; 17:669-75. [DOI: 10.1111/j.1440-1797.2012.01645.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
32
|
Khurana S, Bruggeman LA, Kao HY. Nuclear hormone receptors in podocytes. Cell Biosci 2012; 2:33. [PMID: 22995171 PMCID: PMC3543367 DOI: 10.1186/2045-3701-2-33] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Accepted: 08/10/2012] [Indexed: 11/14/2022] Open
Abstract
Nuclear receptors are a family of ligand-activated, DNA sequence-specific transcription factors that regulate various aspects of animal development, cell proliferation, differentiation, and homeostasis. The physiological roles of nuclear receptors and their ligands have been intensively studied in cancer and metabolic syndrome. However, their role in kidney diseases is still evolving, despite their ligands being used clinically to treat renal diseases for decades. This review will discuss the progress of our understanding of the role of nuclear receptors and their ligands in kidney physiology with emphasis on their roles in treating glomerular disorders and podocyte injury repair responses.
Collapse
Affiliation(s)
- Simran Khurana
- Department of Biochemistry, School of Medicine, Case Western Reserve University (CWRU) and the Comprehensive Cancer Center of CWRU, 10900 Euclid Avenue, Cleveland, Ohio 44106, USA.
| | | | | |
Collapse
|
33
|
Glucocorticoid-induced hypertension. Pediatr Nephrol 2012; 27:1059-66. [PMID: 21744056 DOI: 10.1007/s00467-011-1928-4] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Revised: 05/12/2011] [Accepted: 05/13/2011] [Indexed: 10/18/2022]
Abstract
Glucocorticoid-induced hypertension is a common clinical problem that is poorly understood, thus rendering treatment strategies sub-optimal. This form of hypertension has been commonly thought to be mediated by excess sodium and water reabsorption by the renal mineralocorticoid receptor. However, experimental and clinical data in both humans and animal models suggest important roles for the glucocorticoid receptor as well, in both the pathogenesis and maintenance of this hypertension. The glucocorticoid receptor is widely expressed in a number of organ systems relevant to blood pressure regulation, including the kidney, the brain and the vasculature. In vitro studies in isolated kidney tissues as well as in vascular smooth muscle and vascular endothelial cells have attempted to elucidate the molecular physiology of glucocorticoid-induced hypertension, but have generally been limited by the inability to study signaling pathways in an intact organism. More recently, the power of mouse genetics has been employed to examine the tissue-specific contributions of vascular and extra-vascular tissues to this form of hypertension. Here we review recent developments in our understanding of the pathogenesis of glucocorticoid-induced hypertension.
Collapse
|
34
|
Yan K, Ito N, Nakajo A, Kurayama R, Fukuhara D, Nishibori Y, Kudo A, Akimoto Y, Takenaka H. The struggle for energy in podocytes leads to nephrotic syndrome. Cell Cycle 2012; 11:1504-11. [PMID: 22433955 DOI: 10.4161/cc.19825] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Podocytes are terminally differentiated post-mitotic cells similar to neurons, and their damage leads to nephrotic syndrome, which is characterized by massive proteinuria associated with generalized edema. A recent functional genetic approach has identified the pathological relevance of several mutated proteins in glomerular podocytes to the mechanism of proteinuria in hereditary nephrotic syndrome. In contrast, the pathophysiology of acquired primary nephrotic syndrome, including minimal change disease, is still largely unknown. We recently demonstrated the possible linkage of an energy-consuming process in glomerular podocytes to the mechanism of proteinuria. Puromycin aminonucleoside nephrosis, a rat model of minimal change disease, revealed the activation of the unfolded protein response (UPR) in glomerular podocytes to be a cause of proteinuria. The pretreatment of puromycin aminonucleoside rat podocytes and cultured podocytes with the mammalian target of rapamycin (mTOR) inhibitor everolimus further revealed that mTOR complex 1 consumed energy, which was followed by UPR activation. In this paper, we will review nutritional transporters to summarize the energy uptake process in podocytes and review the involvement of the UPR in the pathogenesis of glomerular diseases. We will also present additional data that reveal how mTOR complex 1 acts upstream of the UPR. Finally, we will discuss the potential significance of targeting the energy metabolism of podocytes to develop new therapeutic interventions for acquired nephrotic syndrome.
Collapse
Affiliation(s)
- Kunimasa Yan
- Department of Pediatrics, Kyorin University School of Medicine, Mitaka, Tokyo, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Schönenberger E, Ehrich JH, Haller H, Schiffer M. The podocyte as a direct target of immunosuppressive agents. Nephrol Dial Transplant 2010; 26:18-24. [PMID: 20937691 DOI: 10.1093/ndt/gfq617] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Podocytes play a key role in maintaining the blood-urine barrier for high-molecular-weight proteins. They are considered to be terminally differentiated, and podocyte loss cannot be compensated by regenerative proliferation. Various diseases leading to podocyte damage and loss result in proteinuria and cause nephrotic syndrome. Therefore, direct therapeutical strategies to protect podocytes in disease situations are a logical concept to prevent disease or to delay disease progression. Acquired podocytopathies like idiopathic focal segmental glomerulosclerosis and minimal change disease are historically considered as immunological diseases. Therefore, immunosuppressive agents such as steroids and calcineurin inhibitors are the commonly used treatment strategies. However, the causative disease mechanisms behind these treatment strategies remain elusive. Recent evidence shows that immunosuppressive agents, in addition to the effect on the immune system, directly influence the unique structure and function of podocytes. In this context, the actin cytoskeleton of the podocyte and cytokines such as vascular endothelial growth factor play a pivotal role. In this review, we summarize the direct effects on podocytes obtained in vivo and in vitro after treatment with calcineurin inhibitors, mTOR inhibitors and glucocorticoids. These direct effects could play a key role in the treatment concepts of podocytopathies with an important impact on the long-term renal function in patients with pharmacological immunosuppression.
Collapse
|
36
|
Guess A, Agrawal S, Wei CC, Ransom RF, Benndorf R, Smoyer WE. Dose- and time-dependent glucocorticoid receptor signaling in podocytes. Am J Physiol Renal Physiol 2010; 299:F845-53. [PMID: 20630936 DOI: 10.1152/ajprenal.00161.2010] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Glucocorticoids (GC) are the primary therapy for idiopathic nephrotic syndrome (NS). Recent evidence has identified glomerular podocytes as a potential site of GC action in this disease. The objectives of this study were to determine the presence of key components of the glucocorticoid receptor (GR) complex and the functionality of this signaling pathway in podocytes and to explore potential opportunities for manipulation of GC responsiveness. Here, we show that cultured murine podocytes express key components of the GR complex, including the GR, heat shock protein 90, and the immunophilins FKBP51 and FKBP52. The functionality of GR-mediated signaling was verified by measuring several GC (dexamethasone)-induced responses, including 1) increases in mRNA and protein levels of selected GC-regulated genes (FKBP51, phenol sulfotransferase 1, αB-crystallin); 2) downregulation of the GR protein; 3) increased phosphorylation of the GR; and 4) translocation of the GR into the nuclear fraction. Dexamethasone-induced phosphorylation and downregulation of GR protein were also demonstrated in isolated rat glomeruli. Podocyte gene expression in response to dexamethasone was regulated at both the transcriptional and posttranscriptional levels, the latter also including protein degradation. Short-term, high-dose GC treatment resulted in similar changes in gene expression and GR phosphorylation to that of long-term, low-dose GC treatment, thus providing a molecular rationale for the known efficacy of pulse GC therapy in NS. Induction of FKBP51 and downregulation of the GR represent negative feedback mechanisms that can potentially be exploited to improve clinical GC efficacy. Collectively, these findings demonstrate the presence of key molecular components of the GR signaling pathway and its functionality in podocytes and identify novel opportunities for improving clinical GC efficacy in the treatment of NS.
Collapse
Affiliation(s)
- Adam Guess
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, 700 Children's Dr., Columbus, OH 43205, USA
| | | | | | | | | | | |
Collapse
|
37
|
Sekine Y, Nishibori Y, Akimoto Y, Kudo A, Ito N, Fukuhara D, Kurayama R, Higashihara E, Babu E, Kanai Y, Asanuma K, Nagata M, Majumdar A, Tryggvason K, Yan K. Amino acid transporter LAT3 is required for podocyte development and function. J Am Soc Nephrol 2009; 20:1586-96. [PMID: 19443642 DOI: 10.1681/asn.2008070809] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
LAT3 is a Na+-independent neutral l-amino acid transporter recently isolated from a human hepatocellular carcinoma cell line. Although liver, skeletal muscle, and pancreas are known to express LAT3, the tissue distribution and physiologic function of this transporter are not completely understood. Here, we observed that glomeruli express LAT3. Immunofluorescence, confocal microscopy, and immunoelectron microscopy revealed that LAT3 localizes to the apical plasma membrane of podocyte foot processes. In mice, starvation upregulated glomerular LAT3, phosphorylated AKT1, reconstituted the actin network, and elongated foot processes. In the fetal kidney, we observed intense LAT3 expression at the capillary loops stage of renal development. Finally, zebrafish morphants lacking lat3 function showed collapsed glomeruli with thickened glomerular basement membranes. Permeability studies of the glomerular filtration barrier in these zebrafish morphants demonstrated a disruption of selective glomerular permeability. Our data suggest that LAT3 may play a crucial role in the development and maintenance of podocyte structure and function by regulating protein synthesis and the actin cytoskeleton.
Collapse
Affiliation(s)
- Yuji Sekine
- Kyorin University School of Medicine, Department of Pediatrics, Mitaka, Tokyo 181-8611, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Bruneau S, Dantal J. New insights into the pathophysiology of idiopathic nephrotic syndrome. Clin Immunol 2009; 133:13-21. [PMID: 19410518 DOI: 10.1016/j.clim.2009.03.532] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2009] [Accepted: 03/26/2009] [Indexed: 01/08/2023]
Abstract
Corticoresistant idiopathic nephrotic syndrome (INS) is a glomerulopathy of unknown etiology whose original aspect is its recurrence after kidney transplantation in 30 to 50% of patients with end-stage renal disease. This suggests the involvement of circulating factors that would alter the glomerular filtration barrier, but whose nature remains elusive. Although a T cell immune origin has been suggested, the actual role of these cells in INS recurrence is still unclear. Here we present an 8-year-old patient with corticoresistant INS who developed a recurrence of her initial disease after kidney transplantation. Rituximab therapy was proposed 11 months after transplantation; although no immediate effect was induced, a slow but persistent decrease in proteinuria began a few months after Rituximab infusions despite cessation of plasma exchanges and steroid therapy. The pathophysiology of INS and the putative mechanisms of action of Rituximab are discussed.
Collapse
|
39
|
Expression of galectin-1, a new component of slit diaphragm, is altered in minimal change nephrotic syndrome. J Transl Med 2009; 89:178-95. [PMID: 19079321 DOI: 10.1038/labinvest.2008.125] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Nephrin is an essential structural component of the glomerular slit diaphragm (SD), a highly organized intercellular junction that constitutes the ultrafiltration barrier of the kidney. Recent studies have identified two additional nephrin-interacting SD proteins (NEPH1 and NEPH2), suggesting that the zipper-like pattern of the SD is formed through complex intra- and intermolecular interactions of these proteins. However, the complexity of the SD structure suggests that additional SD components remain to be discovered. In this study, we identified galectin-1 (Gal-1) as a new component of the SD, binding to the ectodomain of nephrin. Using dual-immunofluorescence and confocal microscopy and dual-immunoelectron microscopy, we found Gal-1 co-localizing with the ectodomain of nephrin at the SD in normal human kidney. By immunoprecipitation and surface plasmon resonance, we confirmed a direct molecular interaction between Gal-1 and nephrin. Moreover, recombinant Gal-1 induced tyrosine phosphorylation of the cytoplasmic domain of nephrin and activation of the extracellular signal-regulated kinase 1/2 in podocytes. We also showed that podocytes are a major site of biosynthesis of Gal-1 in the glomerulus and that the normal expression patterns and levels of Gal-1 are altered in patients with minimal change nephrotic syndrome. Finally, in puromycin aminonucleoside-induced rat nephrosis, an apparent reduction in the levels of Gal-1 and nephrin around the onset of heavy proteinuria was also revealed. Our data present Gal-1 as a new extracellular ligand of nephrin localized at the glomerular SD, and provide further insight into the complex molecular organization, interaction, and structure of the SD, which is an active site of intracellular signaling necessary for podocyte function.
Collapse
|
40
|
Kakuta T, Fukagawa M, Kitaoka M, Koiwa F, Onoda N, Tominaga Y, Akizawa T, Kurokawa K. Percutaneous ethanol injection therapy for advanced renal hyperparathyroidism in Japan: 2004 survey by the Japanese Society for Parathyroid Intervention. Clin Kidney J 2008; 1:iii21-iii25. [PMID: 25983968 PMCID: PMC4421128 DOI: 10.1093/ndtplus/sfn082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2008] [Accepted: 03/08/2008] [Indexed: 11/12/2022] Open
Abstract
Background. Marked hyperplasia of the parathyroid gland (PTG) is a characteristic feature of severe hyperparathyroidism in patients under chronic haemodialysis treatment. Percutaneous ethanol injection therapy (PEIT) is now becoming popular in Japan as a treatment option for secondary hyperparathyroidism (SHPT) and its cost is covered by the National Health Insurance (NHI) System. The Japanese Society for Parathyroid Intervention surveyed its membership in 2004 to revise the guidelines for the use of PEIT. Methods. The project was approved by the Executive Committee of the Society, and the primary questionnaire was addressed to 3268 centres (departments) affiliated with the Japanese Society for Dialysis Therapy. A follow-up questionnaire was sent to all the centres that responded. Results. Although the number of centres to which the questionnaire was sent in 2004 was 3268, compared with 2653 in 1998, the number of responses decreased from 1425 (53.7%) in 1998 to 962 (29.4%) in 2004. To the question of whether the centre performed PEIT, 114 (11.9%) answered ‘Yes’ and 848 (88.1%) answered ‘No’ in 2004. It was an increase from 1998 when only 83 (5.8%) of 1425 centres answered ‘Yes’. In the 1998 survey, 612 patients underwent PEIT at 74 centres, and in 2004, 2098 patients underwent PEIT at 111 centres. Conclusions. PEIT may become the frequently performed treatment for SHPT patients who become resistant to medical therapy. However, the same problems as in 1998 remain unsolved; that is, recurrent nerve paralysis, difficulty of post-PEIT PTx and lack of evidence showing the long-term effectiveness of PEIT.
Collapse
Affiliation(s)
- Takatoshi Kakuta
- Department of Internal Medicine, Tokai University School of Medicine
| | - Masafumi Fukagawa
- Division of Nephrology and Dialysis Center, Kobe University School of Medicine
| | | | - Fumihiko Koiwa
- Department of Internal Medicine Division of Nephrology, Showa University Fujigaoka Hospital
| | | | | | - Tadao Akizawa
- Department of Internal Medicine Division of Nephrology, Showa University School of Medicine
| | | | | |
Collapse
|
41
|
NPHS3: new clues for understanding idiopathic nephrotic syndrome. Pediatr Nephrol 2008; 23:847-50. [PMID: 18270750 DOI: 10.1007/s00467-008-0747-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2007] [Revised: 11/26/2007] [Accepted: 12/14/2007] [Indexed: 01/15/2023]
Abstract
Hereditary forms of childhood nephrotic syndrome (H-CHNS) have long been counted as rare variants of steroid-resistant nephrotic syndrome (SRNS). This concept must be specified by two new findings: First, a study on nephrotic syndrome manifesting in the first year of life documents that H-CHNS are actually the predominant cause of nephrotic syndrome in infants. Second, the recent identification of autosomal recessive nephrotic syndrome type 3 (NPHS3) caused by mutations in the phospholipase PLCE1 gene has, for the first time, shown steroid responsiveness in H-CHNS. NPHS3 is a severe form of isolated nephrotic syndrome with rapid progression to terminal renal failure. NPHS3 is caused by a developmental rather than structural podocyte dysfunction and is a major cause of diffuse mesangial sclerosis. Therapy response in NPHS3 is documented and could open insights into direct genomic and nongenomic effects of glucocorticoids on podocytes. The findings on NPHS3 support the idea that both clinical course and histology in H-CHNS are subject to genotypic variability and that mutational analysis is the most reliable diagnostic tool. Future studies are needed to determine the clinical implications of NPHS3. Identification of further variants of H-CHNS can be anticipated and may include steroid-responsive hereditary diseases.
Collapse
|
42
|
Nakajo A, Khoshnoodi J, Takenaka H, Hagiwara E, Watanabe T, Kawakami H, Kurayama R, Sekine Y, Bessho F, Takahashi S, Swiatecka-Urban A, Tryggvason K, Yan K. Mizoribine corrects defective nephrin biogenesis by restoring intracellular energy balance. J Am Soc Nephrol 2007; 18:2554-64. [PMID: 17687078 DOI: 10.1681/asn.2006070732] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Proteins are modified and folded within the endoplasmic reticulum (ER). When the influx of proteins exceeds the capacity of the ER to handle the load, the ER is "stressed" and protein biogenesis is affected. We have previously shown that the induction of ER stress by ATP depletion in podocytes leads to mislocalization of nephrin and subsequent injury of podocytes. The aim of the present study was to determine whether ER stress is associated with proteinuria in vivo and whether the immunosuppressant mizoribine may exert its antiproteinuric effect by restoring normal nephrin biogenesis. Induction of nephrotic-range proteinuria with puromycin aminonucleoside in mice increased expression of the ER stress marker GRP78 in podocytes, and led to the mislocalization of nephrin to the cytoplasm. In vitro, mizoribine, through a mechanism likely dependent on the inhibition of inosine 5'-monophosphate dehydrogenase (IMPDH) activity in podocytes, restored the intracellular energy balance by increasing levels of ATP and corrected the posttranslational processing of nephrin. Therefore, we speculate that mizoribine may induce remission of proteinuria, at least in part, by restoring the biogenesis of slit diaphragm proteins in injured podocytes. Further understanding of the ER microenvironment may lead to novel approaches to treat diseases in which abnormal handling of proteins plays a role in pathogenesis.
Collapse
Affiliation(s)
- Aya Nakajo
- Department of Pediatrics, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka, Tokyo 181-8611, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Marwick JA, Ito K, Adcock IM, Kirkham PA. Oxidative stress and steroid resistance in asthma and COPD: pharmacological manipulation of HDAC-2 as a therapeutic strategy. Expert Opin Ther Targets 2007; 11:745-55. [PMID: 17504013 DOI: 10.1517/14728222.11.6.745] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Insensitivity to corticosteroid treatment in inflammatory conditions, such as asthma and chronic obstructive pulmonary disease, present considerable management problems and cost burdens to health services. Oxidative stress is a major component of chronic inflammation and can have a significant suppressive effect on corticosteroid efficacy. Recent advances in the understanding of both the mechanisms of corticosteroid action and corticosteroid insensitivity have provided hope for a therapeutic strategy of restoring corticosteroid sensitivity. Histone deacetylase 2 (HDAC-2) plays a pivotal role in corticosteroid action and is reduced in many cases of steroid insensitivity. Moreover, it has shown that oxidative stress can be responsible for this reduction in HDAC-2 activity. Two structurally different compounds; methyl-xanthine theophylline and polyphenol curcumin restore HDAC activity, thereby restoring corticosteroid function. Low, subbronchodilator doses of theophylline can also act as corticosteroid-sparing drugs in asthmatics. Although these compounds appear to restore corticosteroid function and may initially provide therapeutic potential, they lack specificity and the mechanism of their action is unknown. Once their mechanisms of action are established, it is likely that derivatives of these compounds may be used as a therapeutic strategy to restore corticosteroid insensitivity in the future.
Collapse
Affiliation(s)
- John A Marwick
- Novartis Institute For Biomedical Research, Respiratory Disease Area, Wimblehurst Road, Horsham, West Sussex, RH12 5AB, UK
| | | | | | | |
Collapse
|
44
|
Fukuhara D, Kanai Y, Chairoungdua A, Babu E, Bessho F, Kawano T, Akimoto Y, Endou H, Yan K. Protein characterization of NA+-independent system L amino acid transporter 3 in mice: a potential role in supply of branched-chain amino acids under nutrient starvation. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 170:888-98. [PMID: 17322374 PMCID: PMC1864869 DOI: 10.2353/ajpath.2007.060428] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
We recently cloned the human Na(+)-independent system L neutral amino acid transporter LAT3. The aim of the present study was to characterize the molecular nature of mouse LAT3 at the protein level. Isolated mouse LAT3 showed 83% identity to human LAT3. Xenopus oocytes injected with mouse LAT3 cRNA showed the same functional property as human LAT3. Reverse transcriptase-polymerase chain reaction revealed apparent transcripts of mouse LAT3 in the liver, skeletal muscle, and pancreas, an expression pattern identical to that found in humans. Antibody generated against mouse LAT3 detected both approximately 58-kd and 48-kd bands in the sample from liver and only a 48-kd band in skeletal muscle and pancreas. Immunohistochemical study showed its clear localization in the plasma membrane of liver and skeletal muscle, whereas it was only detectable in the endoplasmic reticulum and in crystalline inclusions in pancreatic acinar cells. Starvation induced up-regulation of mouse LAT3 protein and mRNA in both liver and skeletal muscle but not in pancreas. These results suggest that LAT3 may indeed function as an amino acid transporter, transporting branched-chain amino acids from liver and skeletal muscle to the bloodstream and thereby participating in the regulatory system of interorgan amino acid nutrition.
Collapse
MESH Headings
- Amino Acid Transport Systems, Basic/genetics
- Amino Acid Transport Systems, Basic/metabolism
- Amino Acids, Branched-Chain/metabolism
- Animals
- Base Sequence
- Blotting, Western
- Cells, Cultured
- Cloning, Molecular
- Fluorescent Antibody Technique
- Humans
- Immunohistochemistry
- Male
- Mice
- Mice, Inbred ICR
- Microscopy, Confocal
- Microscopy, Electron, Transmission
- Microscopy, Immunoelectron
- Molecular Sequence Data
- Reverse Transcriptase Polymerase Chain Reaction
- Sequence Homology, Nucleic Acid
- Starvation/metabolism
Collapse
Affiliation(s)
- Daisuke Fukuhara
- Department of Pediatrics, Kyorin University School of Medicine, Mitaka, Tokyo 181-8611, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Xing CY, Saleem MA, Coward RJ, Ni L, Witherden IR, Mathieson PW. Direct effects of dexamethasone on human podocytes. Kidney Int 2006; 70:1038-45. [PMID: 16837924 DOI: 10.1038/sj.ki.5001655] [Citation(s) in RCA: 133] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Glucocorticoids are widely used in the treatment of human glomerular diseases, but their mode of action is poorly understood particularly in steroid-sensitive nephrotic syndrome, which is most common in childhood and is characterized by a lack of inflammation in the kidney. The podocyte is a key cell in the glomerulus in health and disease: until recently, human podocytes have been difficult to study in vitro. We have developed a conditionally immortalized human podocyte cell line transfected with a temperature-sensitive simian virus 40 transgene: when the transgene is inactivated in vitro, these cells adopt the phenotype of differentiated podocytes. We have used these cells to evaluate, using immunocytochemistry, reverse transcriptase-polymerase chain reaction, and Western blotting, direct effects of the glucocorticoid dexamethasone at concentrations designed to mimic in vivo therapeutic corticosteroid levels. Dexamethasone upregulated expression of nephrin and tubulin-alpha, and downregulated vascular endothelial growth factor. Effects on cell cycle were complex with downregulation of cyclin kinase inhibitor p21 and augmentation of podocyte survival, without any effect on apoptosis. We report cytokine production by human podocytes, especially interleukin (IL)-6 and -8; IL-6 expression was suppressed by dexamethasone. These potent direct effects on podocytes illustrate a novel mode of action of glucocorticoids and suggest potential new therapeutic strategies for glomerular disease.
Collapse
Affiliation(s)
- C-Y Xing
- Academic Renal Unit, University of Bristol, Southmead Hospital, Bristol, UK
| | | | | | | | | | | |
Collapse
|
46
|
Fujii Y, Khoshnoodi J, Takenaka H, Hosoyamada M, Nakajo A, Bessho F, Kudo A, Takahashi S, Arimura Y, Yamada A, Nagasawa T, Ruotsalainen V, Tryggvason K, Lee AS, Yan K. The effect of dexamethasone on defective nephrin transport caused by ER stress: a potential mechanism for the therapeutic action of glucocorticoids in the acquired glomerular diseases. Kidney Int 2006; 69:1350-9. [PMID: 16531978 DOI: 10.1038/sj.ki.5000317] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The mechanism by which glucocorticoids govern antiproteinuric effect in nephrotic syndrome remains unknown. Present study examined the protective role of dexamethasone (DEX) in the intracellular trafficking of nephrin under endoplasmic reticulum (ER) stress. Human embryonic kidney-293 cell line expressing a full-length human nephrin was cultured in mediums containing 5.5 or 25 mM glucose with or without DEX. The result revealed that glucose starvation evoked a rapid ER stress leading to formation of underglycosylated nephrin that was remained in the ER as a complex with calreticulin/calnexin. DEX rescued this interfered trafficking through binding to its receptor and stimulating the mitochondrial transcripts and adenosine 5' triphosphate (ATP) production, leading to synthesis of fully glycosylated nephrin. These results suggest that ER-stress in podocytes may cause alteration of nephrin N-glycosylation, which may be an underlying factor in the pathomechanism of the proteinuria in nephrotic syndrome. DEX may restore this imbalance by stimulating expression of mitochondrial genes, resulted in the production of ATP that is essential factor for proper folding machinery aided by the ER chaperones.
Collapse
Affiliation(s)
- Y Fujii
- Department of Internal Medicine, Kyorin University School of Medicine, Mitaka, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Xing Y, Ding J, Fan Q, Guan N. Diversities of podocyte molecular changes induced by different antiproteinuria drugs. Exp Biol Med (Maywood) 2006; 231:585-93. [PMID: 16636307 DOI: 10.1177/153537020623100513] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Nephrin, podocin, CD2AP, and alpha-actinin-4 are important podocyte proteins that help maintain the integrity of the slit diaphragm and prevent proteinuria. Studies have shown that angiotensin-converting enzyme inhibitors, glucocorticoids, and all-trans retinoic acid (ATRA) have antiproteinuric effects. However, it is still unclear whether these drugs, with different pharmacological mechanisms, lead to a reduction in proteinuria by changing the expression and distribution of these important podocyte proteins. In this study, changes in the expression and distribution of nephrin, podocin, CD2AP, and alpha-actinin-4 were dynamically detected in Adriamycin-induced nephrotic (ADR) rats treated with three different drugs: lisinopril, prednisone, and ATRA. Nephropathy was induced by an intravenous injection of Adriamycin. After Adriamycin injection, rats received lisinopril, prednisone, and ATRA treatment, respectively. Renal tissues were collected at Days 3, 7, 14, and 28. The distribution and the expression of messenger RNA and protein of nephrin, podocin, CD2AP, and alpha-actinin-4 were detected by indirect immunofluorescence, real-time polymerase chain reaction, and Western blotting, respectively. With the intervention of lisinopril, prednisone, and ATRA, changes in the expression of nephrin, podocin, and CD2AP were diverse, which was different from that detected in ADR rats. After lisinopril and prednisone intervention, podocin exhibited prominent earlier changes compared with those of nephrin and CD2AP, whereas CD2AP showed more prominent changes after ATRA intervention. There was no change in the expression of alpha-actinin-4 molecule. In summary, we conclude that the antiproteinuric effects of lisinopril, prednisone, and ATRA were achieved by changes in the expression and distribution of the important podocyte molecules nephrin, podocin, CD2AP, and alpha-actinin-4. The pattern in the change of podocyte molecules after lisinopril and prednisone intervention was similar, but the pattern in the change of podocyte molecules after ATRA intervention was different from that of lisinopril or prednisone intervention.
Collapse
Affiliation(s)
- Yan Xing
- Department of Pediatrics, Peking University First Hospital, Beijing, People's Republic of China
| | | | | | | |
Collapse
|
48
|
Matsuo H, Tamura M, Kabashima N, Serino R, Tokunaga M, Shibata T, Matsumoto M, Aijima M, Oikawa S, Anai H, Nakashima Y. Prednisolone inhibits hyperosmolarity-induced expression of MCP-1 via NF-κB in peritoneal mesothelial cells. Kidney Int 2006; 69:736-46. [PMID: 16518329 DOI: 10.1038/sj.ki.5000131] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The mechanism of peritoneal fibrosis in patients on continuous ambulatory peritoneal dialysis (CAPD) is poorly elucidated. We investigated the cellular mechanism of high-glucose-induced expression of monocyte chemoattractant protein-1 (MCP-1), which is important in recruiting monocytes into the peritoneum and progression of peritoneal fibrosis, and examined the inhibitory mechanism of glucocorticoids. Rat peritoneal mesothelial cells were cultured in high-glucose-containing medium and then analyzed for phosphorylation levels of p42/44 and p38 mitogen-activated protein (MAP) kinases (MAPK), MAPK or extracellular signal-regulated kinase kinase (MEK)1/2, c-Jun N-terminal kinase (JNK)1/2, and protein kinase C (PKC) by Western blotting. Expression of MCP-1 was examined by reverse transcription-polymerase chain reaction and enzyme-linked immunosorbent assay, respectively. DNA-binding activity of nuclear factor (NF)-kappaB was measured by electrophoretic mobility shift assay. High glucose increased MCP-1 mRNA and MCP-1 protein expression. Although glucose increased phosphorylation of MEK1/2, p42/44 MAPK, p38 MAPK, JNK1/2, and PKC, and DNA-binding activity of NF-kappaB, its effect on MCP-1 expression was suppressed only by PKC and NF-kappaB inhibitors. Mannitol caused a similar increase in PKC and NF-kappaB activation and MCP-1 synthesis. Prednisolone increased I-kappaB-alpha expression and inhibited glucose/mannitol-induced NF-kappaB DNA binding and MCP-1 expression without affecting PKC phosphorylation. The inhibitory effects of prednisolone on MCP-1 expression were reversed by mifepristone, a glucocorticoid receptor antagonist. Our results indicate that glucose induces MCP-1 mainly through hyperosmolarity by activating PKC and its downstream NF-kappaB, and that such effect was inhibited by prednisolone, suggesting the efficacy of prednisolone in preventing peritoneal fibrosis in patients on CAPD.
Collapse
Affiliation(s)
- H Matsuo
- The Second Department of Internal Medicine, University of Occupational and Environmental Health School of Medicine, and Kidney Center, University Hospital, Kitakyushu, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Ransom RF, Lam NG, Hallett MA, Atkinson SJ, Smoyer WE. Glucocorticoids protect and enhance recovery of cultured murine podocytes via actin filament stabilization. Kidney Int 2006; 68:2473-83. [PMID: 16316324 DOI: 10.1111/j.1523-1755.2005.00723.x] [Citation(s) in RCA: 125] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Nephrotic syndrome is a common kidney disease in both children and adults that is characterized by dramatic structural changes in the actin-rich foot processes of glomerular podocytes. Although glucocorticoids are the primary treatment for nephrotic syndrome, neither the target cell nor mechanism of action of glucocorticoids in nephrotic syndrome is known. For the last 30 years glucocorticoids have been presumed to act by reducing the release of soluble mediators of disease by circulating lymphocytes. In contrast, we hypothesized that glucocorticoids exert their beneficial effects in nephrotic syndrome by direct action on podocytes. METHODS Cultured murine podocytes were treated with glucocorticoids in the presence and absence of mifepristone (to inhibit glucocorticoid-induced transcriptional activation) and challenged using our previously reported in vitro model of puromycin aminonucleoside (PAN)-induced podocyte injury, as well as by direct disruption of actin filaments with latrunculin and cytochalasin. Cell viability, actin filament distribution, total polymerized actin content, and actin-regulating guanine triphosphatase (GTPase) activities were measured. RESULTS We demonstrated that treatment of cultured murine podocytes with the glucocorticoid dexamethasone both protected and enhanced recovery from PAN-induced injury. Dexamethasone also increased total cellular polymerized actin, stabilized actin filaments against disruption by PAN, latrunculin, or cytochalasin, and induced a significant increase in the activity of the actin-regulating GTPase RhoA. CONCLUSION These data suggest that, contrary to the current therapeutic paradigm, the beneficial effects of glucocorticoids in nephrotic syndrome may result, at least in part, from direct effects on podocytes leading to enhanced actin filament stability.
Collapse
Affiliation(s)
- Richard F Ransom
- Pediatric Nephrology Division, University of Michigan, Ann Arbor, MI 48109, USA.
| | | | | | | | | |
Collapse
|
50
|
Abstract
Nuclear receptors are transcription factors that are essential in embryonic development, maintenance of differentiated cellular phenotypes, metabolism, and apoptosis. Dysfunction of nuclear receptor signaling leads to a wide spectra of proliferative, reproductive, and metabolic diseases, including cancers, infertility, obesity, and diabetes. In addition, many proteins have been identified as coregulators which can be recruited by DNA-binding nuclear receptors to affect transcriptional regulation. The cellular level of coregulators is crucial for nuclear receptor-mediated transcription and many coregulators have been shown to be targets for diverse intracellular signaling pathways and posttranslational modifications. This review provides a general overview of the roles and mechanism of action of nuclear receptors and their coregulators. Since progression of renal diseases is almost always associated with inflammatory processes and/or involve metabolic disorders of lipid and glucose, cell proliferation, hypertrophy, apoptosis, and hypertension, the importance of nuclear receptors and their coregulators in these contexts will be addressed.
Collapse
Affiliation(s)
- Xiong Z Ruan
- Centre for Nephrology, Royal Free and University College Medical School, University College London, Royal Free Campus, Rowland Hill Street, London, United Kingdom.
| | | | | | | |
Collapse
|