101
|
Martin-Sanchez D, Gallegos-Villalobos A, Fontecha-Barriuso M, Carrasco S, Sanchez-Niño MD, Lopez-Hernandez FJ, Ruiz-Ortega M, Egido J, Ortiz A, Sanz AB. Deferasirox-induced iron depletion promotes BclxL downregulation and death of proximal tubular cells. Sci Rep 2017; 7:41510. [PMID: 28139717 PMCID: PMC5282523 DOI: 10.1038/srep41510] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 12/21/2016] [Indexed: 12/21/2022] Open
Abstract
Iron deficiency has been associated with kidney injury. Deferasirox is an oral iron chelator used to treat blood transfusion-related iron overload. Nephrotoxicity is the most serious and common adverse effect of deferasirox and may present as an acute or chronic kidney disease. However, scarce data are available on the molecular mechanisms of nephrotoxicity. We explored the therapeutic modulation of deferasirox-induced proximal tubular cell death in culture. Deferasirox induced dose-dependent tubular cell death and AnexxinV/7AAD staining showed features of apoptosis and necrosis. However, despite inhibiting caspase-3 activation, the pan-caspase inhibitor zVAD-fmk failed to prevent deferasirox-induced cell death. Moreover, zVAD increased deferasirox-induced cell death, a feature sometimes found in necroptosis. Electron microscopy identified mitochondrial injury and features of necrosis. However, neither necrostatin-1 nor RIP3 knockdown prevented deferasirox-induced cell death. Deferasirox caused BclxL depletion and BclxL overexpression was protective. Preventing iron depletion protected from BclxL downregulation and deferasirox cytotoxicity. In conclusion, deferasirox promoted iron depletion-dependent cell death characterized by BclxL downregulation. BclxL overexpression was protective, suggesting a role for BclxL downregulation in iron depletion-induced cell death. This information may be used to develop novel nephroprotective strategies. Furthermore, it supports the concept that monitoring kidney tissue iron depletion may decrease the risk of deferasirox nephrotoxicity.
Collapse
Affiliation(s)
- Diego Martin-Sanchez
- Research Institute-Fundacion Jimenez Diaz, Autonoma University, Madrid, Spain
- IRSIN, Madrid, Spain
- REDINREN, Madrid, Spain
| | - Angel Gallegos-Villalobos
- Research Institute-Fundacion Jimenez Diaz, Autonoma University, Madrid, Spain
- IRSIN, Madrid, Spain
- REDINREN, Madrid, Spain
| | - Miguel Fontecha-Barriuso
- Research Institute-Fundacion Jimenez Diaz, Autonoma University, Madrid, Spain
- IRSIN, Madrid, Spain
- REDINREN, Madrid, Spain
| | - Susana Carrasco
- Research Institute-Fundacion Jimenez Diaz, Autonoma University, Madrid, Spain
- IRSIN, Madrid, Spain
- REDINREN, Madrid, Spain
| | - Maria Dolores Sanchez-Niño
- Research Institute-Fundacion Jimenez Diaz, Autonoma University, Madrid, Spain
- IRSIN, Madrid, Spain
- REDINREN, Madrid, Spain
| | - Francisco J Lopez-Hernandez
- IRSIN, Madrid, Spain
- REDINREN, Madrid, Spain
- Biomedical Research Institute of Salamanca, University of Salamanca, Salamanca, Spain
| | - Marta Ruiz-Ortega
- Research Institute-Fundacion Jimenez Diaz, Autonoma University, Madrid, Spain
- IRSIN, Madrid, Spain
- REDINREN, Madrid, Spain
| | - Jesus Egido
- Research Institute-Fundacion Jimenez Diaz, Autonoma University, Madrid, Spain
- IRSIN, Madrid, Spain
| | - Alberto Ortiz
- Research Institute-Fundacion Jimenez Diaz, Autonoma University, Madrid, Spain
- IRSIN, Madrid, Spain
- REDINREN, Madrid, Spain
| | - Ana Belén Sanz
- Research Institute-Fundacion Jimenez Diaz, Autonoma University, Madrid, Spain
- IRSIN, Madrid, Spain
- REDINREN, Madrid, Spain
| |
Collapse
|
102
|
Biological Membrane-Packed Mesenchymal Stem Cells Treat Acute Kidney Disease by Ameliorating Mitochondrial-Related Apoptosis. Sci Rep 2017; 7:41136. [PMID: 28117405 PMCID: PMC5259718 DOI: 10.1038/srep41136] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 12/15/2016] [Indexed: 12/18/2022] Open
Abstract
The mortality of rhabdomyolysis-induced AKI remains high because no effective therapy exists. We investigated a new therapeutic method using MSCs. The aim of this study was to investigate the therapeutic potential and anti-apoptotic mechanisms of action of MSCs in the treatment of AKI induced by glycerol in vivo and in vitro. We used Duragen as a biological membrane to pack MSCs on the glycerol-injured renal tissue in vivo. The anti-apoptotic mechanism was investigated. In vitro, HK-2 cells were incubated with ferrous myoglobin and MSCs-conditioned medium, followed by cell proliferation and apoptosis assays. We founded that packing MSCs on the injured renal tissue preserved renal function, ameliorated renal tubular lesions, and reduced apoptosis in the mice with glycerol-induced AKI. The MSC-conditioned medium improved HK-2 cell viability and inhibited apoptosis. These effects were reversed by the PI3K inhibitor LY294002. Biological membrane packing of MSCs on the renal tissue has a therapeutic rescue function by inhibiting cell apoptosis in vivo. MSCs protect renal cells from apoptosis induced by myoglobin in vitro. We have thus demonstrated MSCs reduced rhabdomyolysis-associated renal injury and cell apoptosis by activating the PI3K/Akt pathway and inhibiting apoptosis.
Collapse
|
103
|
Schlosser M, Löser H, Siegmund SV, Montesinos-Rongen M, Bindila L, Lutz B, Barrett DA, Sarmad S, Ortori CA, Grau V, von Brandenstein M, Fries JW. The Endocannabinoid, Anandamide, Induces Cannabinoid Receptor-Independent Cell Death in Renal Proximal Tubule Cells. Cell 2017. [DOI: 10.4236/cellbio.2017.64004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
104
|
Leci-Tahiri L, Krušlin B, Vučić M, Sonicki Z, Lovričević I. Assessment of apoptosis in the native vein used for hemodialysis access. Croat Med J 2016; 57:540-547. [PMID: 28051278 PMCID: PMC5209926 DOI: 10.3325/cmj.2016.57.540] [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] [Indexed: 12/02/2022] Open
Abstract
Aim To determine whether apoptosis is more common in previously punctured native veins than in non-punctured native veins among patients who undergo surgical creation of arteriovenous fistula (AVF) for dialysis access. Methods Cephalic vein specimens were obtained from January 1, 2013 to December 31, 2014 from 60 patients, 30 with previously punctured native veins and 30 with non-punctured native veins. Before AVF placement, a 1-cm vein segment was excised from distal part of the vein for histological, histochemical, and immunohistochemical analysis. Vein specimens were divided into two portions along the longitudinal axis and stained with hematoxylin and eosin for routine histological evaluation. Immunohistochemical analysis was used to localize Bax, p53, caspase 3, and Bcl-2 expression. Results The group with previously punctured veins showed significantly increased caspase 3 (P < 0.001, two-sided Fisher`s Exact Test) and Bax expression (P = 0.002, two-sided Fisher`s Exact Test) and significantly decreased Bcl-2 expression (P < 0.001, two-sided Fisher`s Exact Test) compared with the control group. There were no significant differences between the groups in p53 expression (χ2 = 0.071, df = 1, P = 0.791). Fistula failure was significantly more common in the study group (26.7% vs 6.7%, χ2 = 4.32, df = 1, P = 0.038). Conclusion Our study indicates a possible role of venipuncture in apoptosis development and a possible role of apoptosis in fistula failure, but we do not have sufficient evidence to conclude that it represents its main cause.
Collapse
Affiliation(s)
- Laura Leci-Tahiri
- Laura Leci-Tahiri, Clinic of Vascular Surgery, University Clinical Center of Kosovo, 10000 Prishtina, Kosovo,
| | | | | | | | | |
Collapse
|
105
|
Matsunaga N, Ikeda E, Kakimoto K, Watanabe M, Shindo N, Tsuruta A, Ikeyama H, Hamamura K, Higashi K, Yamashita T, Kondo H, Yoshida Y, Matsuda M, Ogino T, Tokushige K, Itcho K, Furuichi Y, Nakao T, Yasuda K, Doi A, Amamoto T, Aramaki H, Tsuda M, Inoue K, Ojida A, Koyanagi S, Ohdo S. Inhibition of G0/G1 Switch 2 Ameliorates Renal Inflammation in Chronic Kidney Disease. EBioMedicine 2016; 13:262-273. [PMID: 27745900 PMCID: PMC5264248 DOI: 10.1016/j.ebiom.2016.10.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 10/05/2016] [Accepted: 10/05/2016] [Indexed: 12/20/2022] Open
Abstract
Chronic kidney disease (CKD) is a global health problem, and novel therapies to treat CKD are urgently needed. Here, we show that inhibition of G0/G1 switch 2 (G0s2) ameliorates renal inflammation in a mouse model of CKD. Renal expression of chemokine (C-C motif) ligand 2 (Ccl2) was increased in response to p65 activation in the kidneys of wild-type 5/6 nephrectomy (5/6Nx) mice. Moreover, 5/6Nx Clk/Clk mice, which carry homozygous mutations in the gene encoding circadian locomotor output cycles kaput (CLOCK), did not exhibit aggravation of apoptosis or induction of F4/80-positive cells. The renal expression of G0s2 in wild-type 5/6Nx mice was important for the transactivation of Ccl2 by p65. These pathologies were ameliorated by G0s2 knockdown. Furthermore, a novel small-molecule inhibitor of G0s2 expression was identified by high-throughput chemical screening, and the inhibitor suppressed renal inflammation in 5/6Nx mice. These findings indicated that G0s2 inhibitors may have applications in the treatment of CKD.
Collapse
Affiliation(s)
- Naoya Matsunaga
- Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan; Department of Glocal Healthcare Science, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Eriko Ikeda
- Department of Molecular Biology, Daiichi University of Pharmacy, Minami-ku, Fukuoka 815-8511, Japan
| | - Keisuke Kakimoto
- Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | - Miyako Watanabe
- Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | - Naoya Shindo
- Department of Bio-Analytical Chemistry, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | - Akito Tsuruta
- Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | - Hisako Ikeyama
- Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | - Kengo Hamamura
- Department of Molecular Biology, Daiichi University of Pharmacy, Minami-ku, Fukuoka 815-8511, Japan
| | - Kazuhiro Higashi
- Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | - Tomohiro Yamashita
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Hideaki Kondo
- Center for Sleep Medicine, Saiseikai Nagasaki Hospital, Katafuchi, Nagasaki 850-0003, Japan
| | - Yuya Yoshida
- Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | - Masaki Matsuda
- Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | - Takashi Ogino
- Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | - Kazutaka Tokushige
- Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | - Kazufumi Itcho
- Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yoko Furuichi
- Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | - Takaharu Nakao
- Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | - Kaori Yasuda
- Cell-Innovator Inc., EC building, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | - Atsushi Doi
- Cell-Innovator Inc., EC building, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | - Toshiaki Amamoto
- NMedical Co. Ouryokukai, Chuo-ku, Nihombashi-Kayabacho, Tokyo 103-0025, Japan
| | - Hironori Aramaki
- Department of Molecular Biology, Daiichi University of Pharmacy, Minami-ku, Fukuoka 815-8511, Japan
| | - Makoto Tsuda
- Department of Life Innovation, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Kazuhide Inoue
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Akio Ojida
- Department of Bio-Analytical Chemistry, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | - Satoru Koyanagi
- Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan; Department of Glocal Healthcare Science, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Shigehiro Ohdo
- Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan.
| |
Collapse
|
106
|
Exercise training attenuates dexamethasone-induced hypertension by improving autonomic balance to the heart, sympathetic vascular modulation and skeletal muscle microcirculation. J Hypertens 2016; 34:1967-76. [DOI: 10.1097/hjh.0000000000001032] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
107
|
El-Sherbeeny NA, Nader MA, Attia GM, Ateyya H. Agmatine protects rat liver from nicotine-induced hepatic damage via antioxidative, antiapoptotic, and antifibrotic pathways. Naunyn Schmiedebergs Arch Pharmacol 2016; 389:1341-1351. [PMID: 27638633 DOI: 10.1007/s00210-016-1284-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Accepted: 08/05/2016] [Indexed: 01/07/2023]
Abstract
Tobacco smoking with its various forms is a global problem with proved hazardous effects to human health. The present work was planned to study the defending role of agmatine (AGM) on hepatic oxidative stress and damage induced by nicotine in rats. Thirty-two rats divided into four groups were employed: control group, nicotine-only group, AGM group, and AGM-nicotine group. Measurements of serum hepatic biochemical markers, lipid profile, and vascular cell adhesion molecule-1 were done. In addition, malondialdehyde (MDA), superoxide dismutase (SOD), glutathione (GSH) activity, and nitrate/nitrite (NOx) levels were estimated in the liver homogenates. Immunohistochemistry for Bax and transforming growth factor beta (TGF-β1) and histopathology of the liver were also included. Data of the study demonstrated that nicotine administration exhibited marked liver deterioration, an increase in liver enzymes, changes in lipid profile, and an elevation in MDA with a decline in levels of SOD, GSH, and NOx (nitrate/nitrite). Also, levels of proapoptotic Bax and profibrotic TGF-β1 showed marked elevation in the liver. AGM treatment to rats in nicotine-only group ameliorated all the previous changes. These findings indicate that AGM could successfully overcome the nicotine-evoked hepatic oxidative stress and tissue injury, apoptosis, and fibrosis.
Collapse
Affiliation(s)
- Nagla A El-Sherbeeny
- Pharmacology and Toxicology, College of Pharmacy, Taibah University, El-Madinah El-Munawarah, Saudi Arabia.,Faculty of Medicine, Suez Canal University, Ismailia Governorate, Egypt
| | - Manar A Nader
- Pharmacology and Toxicology, College of Pharmacy, Taibah University, El-Madinah El-Munawarah, Saudi Arabia.,Faculty of Pharmacy, Mansoura University, Dakahlia Governorate, Egypt
| | - Ghalia M Attia
- Department of Anatomy, Faculty of Medicine, Taibah University, El-Madinah El-Munawarah, Saudi Arabia. .,Department of Histology and Cell Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| | - Hayam Ateyya
- Pharmacology and Toxicology, College of Pharmacy, Taibah University, El-Madinah El-Munawarah, Saudi Arabia.,Faculty of Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
108
|
Kurko J, Tringham M, Tanner L, Näntö-Salonen K, Vähä-Mäkilä M, Nygren H, Pöhö P, Lietzen N, Mattila I, Olkku A, Hyötyläinen T, Orešič M, Simell O, Niinikoski H, Mykkänen J. Imbalance of plasma amino acids, metabolites and lipids in patients with lysinuric protein intolerance (LPI). Metabolism 2016; 65:1361-75. [PMID: 27506743 DOI: 10.1016/j.metabol.2016.05.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 04/26/2016] [Accepted: 05/20/2016] [Indexed: 12/26/2022]
Abstract
BACKGROUND Lysinuric protein intolerance (LPI [MIM 222700]) is an aminoaciduria with defective transport of cationic amino acids in epithelial cells in the small intestine and proximal kidney tubules due to mutations in the SLC7A7 gene. LPI is characterized by protein malnutrition, failure to thrive and hyperammonemia. Many patients also suffer from combined hyperlipidemia and chronic kidney disease (CKD) with an unknown etiology. METHODS Here, we studied the plasma metabolomes of the Finnish LPI patients (n=26) and healthy control individuals (n=19) using a targeted platform for analysis of amino acids as well as two analytical platforms with comprehensive coverage of molecular lipids and polar metabolites. RESULTS Our results demonstrated that LPI patients have a dichotomy of amino acid profiles, with both decreased essential and increased non-essential amino acids. Altered levels of metabolites participating in pathways such as sugar, energy, amino acid and lipid metabolism were observed. Furthermore, of these metabolites, myo-inositol, threonic acid, 2,5-furandicarboxylic acid, galactaric acid, 4-hydroxyphenylacetic acid, indole-3-acetic acid and beta-aminoisobutyric acid associated significantly (P<0.001) with the CKD status. Lipid analysis showed reduced levels of phosphatidylcholines and elevated levels of triacylglycerols, of which long-chain triacylglycerols associated (P<0.01) with CKD. CONCLUSIONS This study revealed an amino acid imbalance affecting the basic cellular metabolism, disturbances in plasma lipid composition suggesting hepatic steatosis and fibrosis and novel metabolites correlating with CKD in LPI. In addition, the CKD-associated metabolite profile along with increased nitrite plasma levels suggests that LPI may be characterized by increased oxidative stress and apoptosis, altered microbial metabolism in the intestine and uremic toxicity.
Collapse
Affiliation(s)
- Johanna Kurko
- Department of Medical Biochemistry and Genetics, University of Turku, Kiinamyllynkatu 10, 20520 Turku, Finland.
| | - Maaria Tringham
- Department of Medical Biochemistry and Genetics, University of Turku, Kiinamyllynkatu 10, 20520 Turku, Finland.
| | - Laura Tanner
- Department of Medical Biochemistry and Genetics, University of Turku, Kiinamyllynkatu 10, 20520 Turku, Finland; Department of Clinical Genetics, Turku University Hospital, Kiinamyllynkatu 4-8, PL 52, 20521 Turku, Finland.
| | - Kirsti Näntö-Salonen
- Department of Pediatrics, Turku University Hospital and University of Turku, Kiinamyllynkatu 4-8, PL 52, 20521 Turku, Finland.
| | - Mari Vähä-Mäkilä
- Department of Pediatrics, Turku University Hospital and University of Turku, Kiinamyllynkatu 4-8, PL 52, 20521 Turku, Finland.
| | - Heli Nygren
- VTT Technical Research Centre of Finland, Tietotie 2, P.O. Boxs 1000, Espoo 02044 VTT, Finland.
| | - Päivi Pöhö
- Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, P.O. Boxs 56, Helsinki 00014, Finland.
| | - Niina Lietzen
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Tykistökatu 6, 20520 Turku, Finland.
| | - Ismo Mattila
- Steno Diabetes Center A/S, Niels Steensens Vej 2, 2820 Gentofte, Denmark.
| | - Anu Olkku
- Eastern Finland Laboratory Centre, Puijonlaaksontie 2, 70210 Kuopio, Finland.
| | - Tuulia Hyötyläinen
- Steno Diabetes Center A/S, Niels Steensens Vej 2, 2820 Gentofte, Denmark.
| | - Matej Orešič
- Steno Diabetes Center A/S, Niels Steensens Vej 2, 2820 Gentofte, Denmark.
| | - Olli Simell
- Department of Pediatrics, Turku University Hospital and University of Turku, Kiinamyllynkatu 4-8, PL 52, 20521 Turku, Finland.
| | - Harri Niinikoski
- Department of Pediatrics, Turku University Hospital and University of Turku, Kiinamyllynkatu 4-8, PL 52, 20521 Turku, Finland.
| | - Juha Mykkänen
- Department of Pediatrics, Turku University Hospital and University of Turku, Kiinamyllynkatu 4-8, PL 52, 20521 Turku, Finland; Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Kiinamyllynkatu 10, 20520 Turku, Finland.
| |
Collapse
|
109
|
Karthivashan G, Kura AU, Arulselvan P, Md Isa N, Fakurazi S. The modulatory effect of Moringa oleifera leaf extract on endogenous antioxidant systems and inflammatory markers in an acetaminophen-induced nephrotoxic mice model. PeerJ 2016; 4:e2127. [PMID: 27441110 PMCID: PMC4941779 DOI: 10.7717/peerj.2127] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 05/21/2016] [Indexed: 12/25/2022] Open
Abstract
N-Acetyl-p-Aminophenol (APAP), also known as acetaminophen, is the most commonly used over-the counter analgesic and antipyretic medication. However, its overdose leads to both liver and kidney damage. APAP-induced toxicity is considered as one of the primary causes of acute liver failure; numerous scientific reports have focused majorly on APAP hepatotoxicity. Alternatively, not many works approach APAP nephrotoxicity focusing on both its mechanisms of action and therapeutic exploration. Moringa oleifera (MO) is pervasive in nature, is reported to possess a surplus amount of nutrients, and is enriched with several bioactive candidates including trace elements that act as curatives for various clinical conditions. In this study, we evaluated the nephro-protective potential of MO leaf extract against APAP nephrotoxicity in male Balb/c mice. A single-dose acute oral toxicity design was implemented in this study. Group 2, 3, 4 and 5 received a toxic dose of APAP (400 mg/kg of bw, i.p) and after an hour, these groups were administered with saline (10 mL/kg), silymarin—positive control (100 mg/kg of bw, i.p), MO leaf extract (100 mg/kg of bw, i.p), and MO leaf extract (200 mg/kg bw, i.p) respectively. Group 1 was administered saline (10 mL/kg) during both the sessions. APAP-treated mice exhibited a significant elevation of serum creatinine, blood urea nitrogen, sodium, potassium and chloride levels. A remarkable depletion of antioxidant enzymes such as SOD, CAT and GSH-Px with elevated MDA levels has been observed in APAP treated kidney tissues. They also exhibited a significant rise in pro-inflammatory cytokines (TNF-α, IL-1β, IL-6) and decreased anti-inflammatory (IL-10) cytokine level in the kidney tissues. Disorganized glomerulus and dilated tubules with inflammatory cell infiltration were clearly observed in the histology of APAP treated mice kidneys. All these pathological changes were reversed in a dose-dependent manner after MO leaf extract treatment. Therefore, MO leaf extract has demonstrated some therapeutic effectiveness against APAP-induced nephrotoxicity through enhancement of the endogenous antioxidant system and a modulatory effect on specific inflammatory cytokines in kidney tissues.
Collapse
Affiliation(s)
| | - Aminu Umar Kura
- Institute of Bioscience, Universiti Putra Malaysia , Serdang , Selangor , Malaysia
| | | | - Norhaszalina Md Isa
- Institute of Bioscience, Universiti Putra Malaysia , Serdang , Selangor , Malaysia
| | - Sharida Fakurazi
- Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor, Malaysia; Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| |
Collapse
|
110
|
Burlaka I, Nilsson LM, Scott L, Holtbäck U, Eklöf AC, Fogo AB, Brismar H, Aperia A. Prevention of apoptosis averts glomerular tubular disconnection and podocyte loss in proteinuric kidney disease. Kidney Int 2016; 90:135-48. [PMID: 27217195 PMCID: PMC6101029 DOI: 10.1016/j.kint.2016.03.026] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 02/19/2016] [Accepted: 03/10/2016] [Indexed: 01/09/2023]
Abstract
There is a great need for treatment that arrests progression of chronic kidney disease. Increased albumin in urine leads to apoptosis and fibrosis of podocytes and tubular cells and is a major cause of functional deterioration. There have been many attempts to target fibrosis, but because of the lack of appropriate agents, few have targeted apoptosis. Our group has described an ouabain-activated Na,K-ATPase/IP3R signalosome, which protects from apoptosis. Here we show that albumin uptake in primary rat renal epithelial cells is accompanied by a time- and dose-dependent mitochondrial accumulation of the apoptotic factor Bax, down-regulation of the antiapoptotic factor Bcl-xL and mitochondrial membrane depolarization. Ouabain opposes these effects and protects from apoptosis in albumin-exposed proximal tubule cells and podocytes. The efficacy of ouabain as an antiapoptotic and kidney-protective therapeutic tool was then tested in rats with passive Heymann nephritis, a model of proteinuric chronic kidney disease. Chronic ouabain treatment preserved renal function, protected from renal cortical apoptosis, up-regulated Bax, down-regulated Bcl-xL, and rescued from glomerular tubular disconnection and podocyte loss. Thus we have identified a novel clinically feasible therapeutic tool, which has the potential to protect from apoptosis and rescue from loss of functional tissue in chronic proteinuric kidney disease.
Collapse
Affiliation(s)
- Ievgeniia Burlaka
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Linnéa M Nilsson
- Science for Life Laboratory, Department of Applied Physics, Royal Institute of Technology, Solna, Sweden
| | - Lena Scott
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden.
| | - Ulla Holtbäck
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Ann-Christine Eklöf
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Agnes B Fogo
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Hjalmar Brismar
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden; Science for Life Laboratory, Department of Applied Physics, Royal Institute of Technology, Solna, Sweden
| | - Anita Aperia
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden.
| |
Collapse
|
111
|
Urinary miR-16 transactivated by C/EBPβ reduces kidney function after ischemia/reperfusion-induced injury. Sci Rep 2016; 6:27945. [PMID: 27297958 PMCID: PMC4906401 DOI: 10.1038/srep27945] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 05/26/2016] [Indexed: 12/15/2022] Open
Abstract
Ischemia-reperfusion (I/R) induced acute kidney injury (AKI) is regulated by transcriptional factors and microRNAs (miRs). However, modulation of miRs by transcriptional factors has not been characterized in AKI. Here, we found that urinary miR-16 was 100-fold higher in AKI patients. MiR-16 was detected earlier than creatinine in mouse after I/R. Using TargetScan, the 3′UTR of B-cell lymphoma 2 (BCL-2) was found complementary to miR-16 to decrease the fluorescent reporter activity. Overexpression of miR-16 in mice significantly attenuated renal function and increased TUNEL activity in epithelium tubule cells. The CCAAT enhancer binding protein beta (C/EBP-β) increased the expression of miR-16 after I/R injury. The ChIP and luciferase promoter assay indicated that about −1.0 kb to −0.5 kb upstream of miR-16 genome promoter region containing C/EBP-β binding motif transcriptionally regulated miR-16 expression. Meanwhile, the level of pri-miR-16 was higher in mice infected with lentivirus containing C/EBP-β compared with wild-type (WT) mice and overexpression of C/EBP-β in the kidney of WT mice reduced kidney function, increased kidney apoptosis, and elevated urinary miR-16 level. Our results indicated that miR-16 was transactivated by C/EBP-β resulting in aggravated I/R induced AKI and that urinary miR-16 may serve as a potential biomarker for AKI.
Collapse
|
112
|
Iwashita Y, Kuwabara T, Hayata M, Kakizoe Y, Izumi Y, Iiyama J, Kitamura K, Mukoyama M. Mild systemic thermal therapy ameliorates renal dysfunction in a rodent model of chronic kidney disease. Am J Physiol Renal Physiol 2016; 310:F1206-15. [DOI: 10.1152/ajprenal.00519.2015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 03/28/2016] [Indexed: 12/12/2022] Open
Abstract
Thermal therapy has become a nonpharmacological therapy in clinical settings, especially for cardiovascular diseases. However, the practical role of thermal therapy on chronic kidney disease remains elusive. We performed the present study to investigate whether a modified thermal protocol, repeated mild thermal stimulation (MTS), could affect renal damages in chronic kidney disease using a mouse renal ablation model. Mice were subjected to MTS or room temperature (RT) treatment once daily for 4 wk after subtotal nephrectomy (Nx) or sham operation (Sh). We revealed that MTS alleviated renal impairment as indicated by serum creatinine and albuminuria in Nx groups. In addition, the Nx + MTS group showed attenuated tubular histological changes and reduced urinary neutrophil gelatinase-associated lipocalin excretion approximately by half compared with the Nx + RT group. Increased apoptotic signaling, such as TUNEL-positive cell count and cleavage of caspase 3, as well as enhanced oxidative stress were significantly reduced in the Nx + MTS group compared with the Nx + RT group. These changes were accompanied with the restoration of kidney Mn-SOD levels by MTS. Heat shock protein 27, a key molecular chaperone, was phosphorylated by MTS only in Nx kidneys rather than in Sh kidneys. MTS also tended to increase the phosphorylation of p38 MAPK and Akt in Nx kidneys, possibly associated with the activation of heat shock protein 27. Taken together, these results suggest that modified MTS can protect against renal injury in a rodent model of chronic kidney disease.
Collapse
Affiliation(s)
- Yoshihiro Iwashita
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
- Department of Rehabilitation, Kumamoto Health Science University, Kumamoto, Japan; and
| | - Takashige Kuwabara
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Manabu Hayata
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Yutaka Kakizoe
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Yuichiro Izumi
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Junichi Iiyama
- Department of Rehabilitation, Kumamoto Health Science University, Kumamoto, Japan; and
| | - Kenichiro Kitamura
- Third Department of Internal Medicine, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Yamanashi, Japan
| | - Masashi Mukoyama
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| |
Collapse
|
113
|
Shen J, Zhu Y, Huang K, Jiang H, Shi C, Xiong X, Zhan R, Pan J. Buyang Huanwu Decoction attenuates H2O2-induced apoptosis by inhibiting reactive oxygen species-mediated mitochondrial dysfunction pathway in human umbilical vein endothelial cells. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 16:154. [PMID: 27245599 PMCID: PMC4886416 DOI: 10.1186/s12906-016-1152-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 05/27/2016] [Indexed: 12/23/2022]
Abstract
Background Apoptosis of endothelial cells caused by reactive oxygen species plays an important role in ischemia/reperfusion injury after cerebral infarction. Buyang Huanwu Decoction (BYHWD) has been used to treat stroke and stroke-induced disability, however, the mechanism for this treatment remains unknown. In this study, we investigated whether BYHWD can protect human umbilical vein endothelial cells (HUVECs) from H2O2-induced apoptosis and explored the underlying mechanisms. Methods To investigate the effect of BYHWD on the apoptosis of HUVECs, we established a H2O2-induced oxidative stress model and detected apoptosis by Hoechst 33342 and propidium iodide staining. JC-1 and DCFH-DA assays,western blotting and electron microscopy were used to examine the mechanism of BYHWD on apoptosis. Results Pretreatment with BYHWD significantly inhibited H2O2-induced apoptosis and protein caspase-3 expression in a concentration-dependent manner. In addition, BYHWD reduced reactive oxygen species production and promoted endogenous antioxidant defenses. Furthermore, loss of mitochondrial membrane potential and structural disruption of mitochondria were both rescued by BYHWD. Conclusions BYHWD protects HUVECs from H2O2-induced apoptosis by inhibiting oxidative stress damage and mitochondrial dysfunction. These findings indicate that BYHWD is a promising treatment for cerebral ischemia diseases.
Collapse
|
114
|
Beneficial effects of nilotinib, tyrosine kinase inhibitor on cyclosporine-A induced renal damage in rats. Int Immunopharmacol 2016; 33:1-7. [DOI: 10.1016/j.intimp.2016.01.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Revised: 01/23/2016] [Accepted: 01/26/2016] [Indexed: 11/22/2022]
|
115
|
Downregulation of renal tubular Wnt/β-catenin signaling by Dickkopf-3 induces tubular cell death in proteinuric nephropathy. Cell Death Dis 2016; 7:e2155. [PMID: 27010856 PMCID: PMC4823961 DOI: 10.1038/cddis.2016.62] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 02/18/2016] [Accepted: 02/19/2016] [Indexed: 12/14/2022]
Abstract
Studies on the role of Wnt/β-catenin signaling in different forms of kidney disease have yielded discrepant results. Here, we report the biphasic change of renal β-catenin expression in mice with overload proteinuria in which β-catenin was upregulated at the early stage (4 weeks after disease induction) but abrogated at the late phase (8 weeks). Acute albuminuria was observed at 1 week after bovine serum albumin injection, followed by partial remission at 4 weeks that coincided with overexpression of renal tubular β-catenin. Interestingly, a rebound in albuminuria at 8 weeks was accompanied by downregulated tubular β-catenin expression and heightened tubular apoptosis. In addition, there was an inverse relationship between Dickkopf-3 (Dkk-3) and renal tubular β-catenin expression at these time points. In vitro, a similar trend in β-catenin expression was observed in human kidney-2 (HK-2) cells with acute (upregulation) and prolonged (downregulation) exposure to albumin. Induction of a proapoptotic phenotype by albumin was significantly enhanced by silencing β-catenin in HK-2 cells. Finally, Dkk-3 expression and secretion was increased after prolonged exposure to albumin, leading to the suppression of intracellular β-catenin signaling pathway. The effect of Dkk-3 on β-catenin signaling was confirmed by incubation with exogenous Dkk-3 in HK-2 cells. Taken together, these data suggest that downregulation of tubular β-catenin signaling induced by Dkk-3 has a detrimental role in chronic proteinuria, partially through the increase in apoptosis.
Collapse
|
116
|
Xu Y, Guo M, Jiang W, Dong H, Han Y, An XF, Zhang J. Endoplasmic reticulum stress and its effects on renal tubular cells apoptosis in ischemic acute kidney injury. Ren Fail 2016; 38:831-7. [DOI: 10.3109/0886022x.2016.1160724] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
117
|
Kuybulu AE, Öktem F, Çiriş İM, Sutcu R, Örmeci AR, Çömlekçi S, Uz E. Effects of long-term pre- and post-natal exposure to 2.45 GHz wireless devices on developing male rat kidney. Ren Fail 2016; 38:571-80. [PMID: 26905323 DOI: 10.3109/0886022x.2016.1148937] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
PURPOSE The aim of the present study was to investigate oxidative stress and apoptosis in kidney tissues of male Wistar rats that pre- and postnatally exposed to wireless electromagnetic field (EMF) with an internet frequency of 2.45 GHz for a long time. METHODS The study was conducted in three groups of rats which were pre-natal, post-natal. and sham exposed groups. Oxidative stress markers and histological evaluation of kidney tissues were studied. RESULTS Renal tissue malondialdehyde (MDA) and total oxidant (TOS) levels of pre-natal group were high and total antioxidant (TAS) and superoxide dismutase (SOD) levels were low. Spot urine NAG/creatinine ratio was significantly higher in pre- and post-natal groups (p < 0.001). Tubular injury was detected in most of the specimens in post-natal groups. Immunohistochemical analysis showed low-intensity staining with Bax in cortex, high-intensity staining with Bcl-2 in cortical and medullar areas of pre-natal group (p values, 0.000, 0.002, 0.000, respectively) when compared with sham group. Bcl2/Bax staining intensity ratios of medullar and cortical area was higher in pre-natal group than sham group (p = 0.018, p = 0.011). CONCLUSION Based on this study, it is thought that chronic pre- and post-natal period exposure to wireless internet frequency of EMF may cause chronic kidney damages; staying away from EMF source in especially pregnancy and early childhood period may reduce negative effects of exposure on kidney.
Collapse
Affiliation(s)
- Ayça Esra Kuybulu
- a Department of Pediatric Nephrology, Faculty of Medicine , Suleyman Demirel University , Isparta , Turkey
| | - Faruk Öktem
- b Department of Pediatric Nephrology, Faculty of Medicine , Bezmi Alem University , Istanbul , Turkey
| | - İbrahim Metin Çiriş
- c Department of Pathology, Faculty of Medicine , Suleyman Demirel University , Isparta , Turkey
| | - Recep Sutcu
- d Department of Biochemistry , Katip Celebi Faculty of Medicine , Izmir , Turkey
| | - Ahmet Rıfat Örmeci
- e Department of Pediatrics, Faculty of Medicine , Suleyman Demirel University , Isparta , Turkey
| | - Selçuk Çömlekçi
- f Department of Electronics and Communication, Faculty of Engineering , Suleyman Demirel University , Isparta , Turkey
| | - Efkan Uz
- g Department of Biochemistry, Faculty of Medicine , Suleyman Demirel University , Isparta , Turkey
| |
Collapse
|
118
|
|
119
|
Ismail OZ, Zhang X, Bonventre JV, Gunaratnam L. G protein α 12 (Gα 12) is a negative regulator of kidney injury molecule-1-mediated efferocytosis. Am J Physiol Renal Physiol 2015; 310:F607-F620. [PMID: 26697979 DOI: 10.1152/ajprenal.00169.2015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 12/22/2015] [Indexed: 01/01/2023] Open
Abstract
Kidney injury molecule-1 (KIM-1) is a receptor for the "eat me" signal, phosphatidylserine, on apoptotic cells. The specific upregulation of KIM-1 by injured tubular epithelial cells (TECs) enables them to clear apoptotic cells (also known as efferocytosis), thereby protecting from acute kidney injury. Recently, we uncovered that KIM-1 binds directly to the α-subunit of heterotrimeric G12 protein (Gα12) and inhibits its activation by reactive oxygen species during renal ischemia-reperfusion injury (Ismail OZ, Zhang X, Wei J, Haig A, Denker BM, Suri RS, Sener A, Gunaratnam L. Am J Pathol 185: 1207-1215, 2015). Here, we investigated the role that Gα12 plays in KIM-1-mediated efferocytosis by TECs. We showed that KIM-1 remains bound to Gα12 and suppresses its activity during phagocytosis. When we silenced Gα12 expression using small interefering RNA, KIM-1-mediated engulfment of apoptotic cells was increased significantly; in contrast overexpression of constitutively active Gα12 (QLGα12) resulted in inhibition of efferocytosis. Inhibition of RhoA, a key effector of Gα12, using a chemical inhibitor or expression of dominant-negative RhoA, had the same effect as inhibition of Gα12 on efferocytosis. Consistent with this, silencing Gα12 suppressed active RhoA in KIM-1-expressing cells. Finally, using primary TECs from Kim-1+/+ and Kim-1-/- mice, we confirmed that engulfment of apoptotic cells requires KIM-1 expression and that silencing Gα12 enhanced efferocytosis by primary TECs. Our data reveal a previously unknown role for Gα12 in regulating efferocytosis and that renal TECs require KIM-1 to mediate this process. These results may have therapeutic implications given the known harmful role of Gα12 in acute kidney injury.
Collapse
Affiliation(s)
- Ola Z Ismail
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada.,Matthew Mailing Centre for Translational Transplant Studies, Lawson Health Research Institute, London, Ontario, Canada
| | - Xizhong Zhang
- Matthew Mailing Centre for Translational Transplant Studies, Lawson Health Research Institute, London, Ontario, Canada
| | - Joseph V Bonventre
- Renal Division and Biomedical Engineering Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Lakshman Gunaratnam
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada; .,Matthew Mailing Centre for Translational Transplant Studies, Lawson Health Research Institute, London, Ontario, Canada.,Division of Nephrology, Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| |
Collapse
|
120
|
Mei W, Peng Z, Lu M, Liu C, Deng Z, Xiao Y, Liu J, He Y, Yuan Q, Yuan X, Tang D, Yang H, Tao L. Peroxiredoxin 1 inhibits the oxidative stress induced apoptosis in renal tubulointerstitial fibrosis. Nephrology (Carlton) 2015; 20:832-42. [PMID: 25989822 DOI: 10.1111/nep.12515] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2015] [Indexed: 02/06/2023]
Affiliation(s)
- Wenjuan Mei
- Division of Nephrology; Xiangya Hospital; Central South University; Changsha China
| | - Zhangzhe Peng
- Division of Nephrology; Xiangya Hospital; Central South University; Changsha China
| | - Miaomiao Lu
- Division of Nephrology; Xiangya Hospital; Central South University; Changsha China
| | - Chunyan Liu
- Division of Nephrology; Xiangya Hospital; Central South University; Changsha China
| | - Zhenghao Deng
- Division of Pathology; Xiangya Hospital; Central South University; Changsha China
| | - Yun Xiao
- Division of Nephrology; Xiangya Hospital; Central South University; Changsha China
| | - Jishi Liu
- Division of Nephrology; The Third Xiangya Hospital; Central South University; Changsha China
| | - Ying He
- Division of Gastroenterology; Xiangya Hospital; Central South University; Changsha China
| | - Qiongjing Yuan
- Division of Nephrology; Xiangya Hospital; Central South University; Changsha China
| | - Xiangning Yuan
- Division of Nephrology; Xiangya Hospital; Central South University; Changsha China
| | - Damu Tang
- Division of Nephrology; Department of Medicine; McMaster University; Hamilton Ontario Canada
| | - Huixiang Yang
- Division of Gastroenterology; Xiangya Hospital; Central South University; Changsha China
| | - Lijian Tao
- Division of Nephrology; Xiangya Hospital; Central South University; Changsha China
- State Key Laboratory of Medical Genetics of China; Central South University; Changsha China
| |
Collapse
|
121
|
Pastori S, Virzì GM, Brocca A, de Cal M, Cantaluppi V, Castellani C, Fedrigo M, Thiene G, Valente ML, Angelini A, Vescovo G, Ronco C. Cardiorenal Syndrome Type 1: Activation of Dual Apoptotic Pathways. Cardiorenal Med 2015; 5:306-15. [PMID: 26648947 DOI: 10.1159/000438831] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 07/06/2015] [Indexed: 12/17/2022] Open
Abstract
Cardiorenal syndrome type 1 (CRS1) pathophysiology is complex, and immune-mediated damage, including alterations in the immune response with monocyte apoptosis and cytokine release, has been reported as a potential mechanism. In this study, we examined the putative role of renal tubular epithelial cell (RTC) apoptosis as a pathogenic mechanism in CRS1. In particular, we investigated the caspase pathways involved in induced apoptosis. We enrolled 29 patients with acute heart failure (AHF), 11 patients with CRS1, and 15 controls (CTR) without AHF or acute kidney injury (AKI). Patients who had AKI prior to the episode of AHF or who had any other potential causes of AKI were excluded. Plasma from different groups was incubated with RTCs for 24 h. Subsequently, cell apoptosis, DNA fragmentation, and caspase-3, -8, and -9 activities were investigated in RTCs incubated with AHF, CRS1, and CTR plasma. A p value <0.5 was considered statistically significant. A quantitative analysis of apoptosis showed significantly higher apoptosis rates in CRS1 patients compared to AHF patients and CTR (p < 0.01). This increase in apoptosis was strongly confirmed by caspase-3 levels (ρ = 0.73). Caspase-8 and -9 were significantly higher in CRS1 patients compared to AHF patients and CTR (p < 0.01). Furthermore, caspase-3 levels showed a significantly positive correlation with caspase-8 (ρ = 0.57) and -9 (ρ = 0.47; p < 0.001). This study demonstrated the significantly heightened presence of dual apoptotic disequilibrium in CRS1. Our findings indicated that apoptosis may have a central role in the mechanism of CRS1, and it could be a potential therapeutic target in this syndrome.
Collapse
Affiliation(s)
- Silvia Pastori
- Department of Nephrology, Dialysis and Transplantation, Vicenza, Italy ; Department of IRRIV-International Renal Research Institute Vicenza, Vicenza, Italy ; Department of Information Engineering, University of Padua, Italy
| | - Grazia Maria Virzì
- Department of Nephrology, Dialysis and Transplantation, Vicenza, Italy ; Department of IRRIV-International Renal Research Institute Vicenza, Vicenza, Italy
| | - Alessandra Brocca
- Department of Nephrology, Dialysis and Transplantation, Vicenza, Italy ; Department of IRRIV-International Renal Research Institute Vicenza, Vicenza, Italy ; Department of Medicine DIMED, University of Padua Medical School, Padua, Italy
| | - Massimo de Cal
- Department of Nephrology, Dialysis and Transplantation, Vicenza, Italy ; Department of IRRIV-International Renal Research Institute Vicenza, Vicenza, Italy
| | - Vincenzo Cantaluppi
- Nephrology, Dialysis and Kidney Transplantation Unit, Department of Medical Sciences, University of Torino, Azienda Ospedaliera 'Città della Salute e della Scienza di Torino-Presidio Molinette', Torino, Italy
| | - Chiara Castellani
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Italy
| | - Marny Fedrigo
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Italy
| | - Gaetano Thiene
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Italy
| | - Maria Luisa Valente
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Italy
| | - Annalisa Angelini
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Italy
| | - Giorgio Vescovo
- Department of Internal Medicine, San Bortolo Hospital, Vicenza, Italy ; Internal Medicine Unit, Sant'Antonio Hospital Padua, Padua, Italy
| | - Claudio Ronco
- Department of Nephrology, Dialysis and Transplantation, Vicenza, Italy ; Department of IRRIV-International Renal Research Institute Vicenza, Vicenza, Italy
| |
Collapse
|
122
|
Arafa E, Bondzie PA, Rezazadeh K, Meyer RD, Hartsough E, Henderson JM, Schwartz JH, Chitalia V, Rahimi N. TMIGD1 is a novel adhesion molecule that protects epithelial cells from oxidative cell injury. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:2757-67. [PMID: 26342724 DOI: 10.1016/j.ajpath.2015.06.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 05/18/2015] [Accepted: 06/18/2015] [Indexed: 10/23/2022]
Abstract
Oxidative damage to renal tubular epithelial cells is a fundamental pathogenic mechanism implicated in both acute kidney injury and chronic kidney diseases. Because epithelial cell survival influences the outcome of acute kidney injury and chronic kidney diseases, identifying its molecular regulators could provide new insight into pathobiology and possible new therapeutic strategies for these diseases. We have identified transmembrane and immunoglobulin domain-containing 1 (TMIGD1) as a novel adhesion molecule, which is highly conserved in humans and other species. TMIGD1 is expressed in renal tubular epithelial cells and promotes cell survival. The extracellular domain of TMIGD1 contains two putative immunoglobulin domains and mediates self-dimerization. Our data suggest that TMIGD1 regulates transepithelial electric resistance and permeability of renal epithelial cells. TMIGD1 controls cell migration, cell morphology, and protects renal epithelial cells from oxidative- and nutrient-deprivation-induced cell injury. Hydrogen peroxide-induced oxidative cell injury downregulates TMIGD1 expression and targets it for ubiquitination. Moreover, TMIGD1 expression is significantly affected in both acute kidney injury and in deoxy-corticosterone acetate and sodium chloride (deoxy-corticosterone acetate salt)-induced chronic hypertensive kidney disease mouse models. Taken together, we have identified TMIGD1 as a novel cell adhesion molecule expressed in kidney epithelial cells that protects kidney epithelial cells from oxidative cell injury to promote cell survival.
Collapse
Affiliation(s)
- Emad Arafa
- Department of Pathology, Boston University Medical Campus, Boston, Massachusetts
| | - Philip A Bondzie
- Department of Pathology, Boston University Medical Campus, Boston, Massachusetts
| | - Kobra Rezazadeh
- Department of Pathology, Boston University Medical Campus, Boston, Massachusetts
| | - Rosana D Meyer
- Department of Pathology, Boston University Medical Campus, Boston, Massachusetts
| | - Edward Hartsough
- Department of Pathology, Boston University Medical Campus, Boston, Massachusetts
| | - Joel M Henderson
- Department of Pathology, Boston University Medical Campus, Boston, Massachusetts
| | - John H Schwartz
- Renal Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts
| | - Vipul Chitalia
- Renal Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts
| | - Nader Rahimi
- Department of Pathology, Boston University Medical Campus, Boston, Massachusetts; Department of Ophthalmology, School of Medicine, Boston University Medical Campus, Boston, Massachusetts.
| |
Collapse
|
123
|
Yi Qi Qing Re Gao formula ameliorates puromycin aminonucleoside-induced nephrosis by suppressing inflammation and apoptosis. Altern Ther Health Med 2015; 15:155. [PMID: 26014479 PMCID: PMC4443657 DOI: 10.1186/s12906-015-0673-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Accepted: 05/13/2015] [Indexed: 12/11/2022]
Abstract
Background Yi Qi Qing Re Gao (YQQRG) formula is a traditional Chinese herbal medicine used to treat chronic nephritis. This study was designed to evaluate the underlying mechanism in the use of YQQRG formula to treat nephrosis induced by puromycin aminonucleoside (PAN). Methods Thirty-six male Wistar rats were randomly divided into 3 groups of 12 rats each: a sham group, a vehicle-treated PAN model group (PAN), and a group treated with YQQRG (PAN + YQQRG). The PAN model was established by a single intravenous injection of PAN at a dose of 40 mg/kg body weight; rats in the sham group received the same volume of saline. Twenty-four hour urinary protein was measured 0, 3, 5, 10, and 15 days after the injection. The rats were sacrificed on day 10 and day 15 and the serum lipid profile examined. The renal cortex of each rat was stained with periodic acid–Schiff reagent and the pathologic alterations and ultrastructural changes were examined by transmission electron microscopy. In situ cell apoptosis was detected by a terminal deoxynucleotidyl transferase-mediated uridine 5′-triphosphate-biotin nick end-labeling (TUNEL) assay. Transcriptive levels of inflammatory markers and molecules associated with apoptosis were detected by a real-time polymerase chain reaction and expression of proteins was examined by either immunohistochemistry or Western blot analysis. Results YQQRG significantly decreased urinary protein level, and lowered serum lipid level. YQQRG also attenuated histologic lesions in the rat kidneys. Activation of inflammatory markers was largely restored by the administration of YQQRG. TUNEL assay showed that YQQRG decreased the number of apoptotic cells. Both mRNA and protein levels of caspase-3 were significantly reduced in the group treated with YQQRG, whereas expression of the Bcl-2 protein increased in the YQQRG group. Conclusions YQQRG alleviated kidney injury in PAN-treated rats, possibly through anti-inflammatory and anti-apoptotic effects.
Collapse
|
124
|
McKee RA, Wingert RA. Zebrafish Renal Pathology: Emerging Models of Acute Kidney Injury. CURRENT PATHOBIOLOGY REPORTS 2015; 3:171-181. [PMID: 25973344 PMCID: PMC4419198 DOI: 10.1007/s40139-015-0082-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The renal system is vital to maintain homeostasis in the body, where the kidneys contain nephron functional units that remove metabolic waste from the bloodstream, regulate fluids, and balance electrolytes. Severe organ damage from toxins or ischemia that occurs abruptly can cause acute kidney injury (AKI) in which there is a rapid, life-threatening loss of these activities. Humans have a limited but poorly understood ability to regenerate damaged nephrons after AKI. However, researchers studying AKI in vertebrate animal models such as mammals, and more recently the zebrafish, have documented robust regeneration within the nephron blood filter and tubule following injury. Further, zebrafish kidneys contain progenitors that create new nephrons after AKI. Here, we review investigations in zebrafish which have established a series of exciting renal pathology paradigms that complement existing AKI models and can be implemented to discover insights into kidney regeneration and the roles of stem cells.
Collapse
Affiliation(s)
- Robert A. McKee
- Department of Biological Sciences, Center for Zebrafish Research, Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, IN 46556 USA
| | - Rebecca A. Wingert
- Department of Biological Sciences, Center for Zebrafish Research, Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, IN 46556 USA
| |
Collapse
|
125
|
Development of a Model of Chronic Kidney Disease in the C57BL/6 Mouse with Properties of Progressive Human CKD. BIOMED RESEARCH INTERNATIONAL 2015; 2015:172302. [PMID: 26064882 PMCID: PMC4433637 DOI: 10.1155/2015/172302] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 12/16/2014] [Accepted: 12/29/2014] [Indexed: 12/18/2022]
Abstract
Chronic kidney disease (CKD) is a major healthcare problem with increasing prevalence in the population. CKD leads to end stage renal disease and increases the risk of cardiovascular disease. As such, it is important to study the mechanisms underlying CKD progression. To this end, an animal model was developed to allow the testing of new treatment strategies or molecular targets for CKD prevention. Many underlying risk factors result in CKD but the disease itself has common features, including renal interstitial fibrosis, tubular epithelial cell loss through apoptosis, glomerular damage, and renal inflammation. Further, CKD shows differences in prevalence between the genders with premenopausal women being relatively resistant to CKD. We sought to develop and characterize an animal model with these common features of human CKD in the C57BL/6 mouse. Mice of this genetic background have been used to produce transgenic strains that are commercially available. Thus, a CKD model in this strain would allow the testing of the effects of numerous genes on the severity or progression of CKD with minimal cost. This paper describes such a mouse model of CKD utilizing angiotensin II and deoxycorticosterone acetate as inducers.
Collapse
|
126
|
Tang C, He L, Liu J, Dong Z. Mitophagy: Basic Mechanism and Potential Role in Kidney Diseases. KIDNEY DISEASES 2015; 1:71-9. [PMID: 27536667 DOI: 10.1159/000381510] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Revised: 03/03/2015] [Indexed: 01/17/2023]
Abstract
BACKGROUND Mitochondria play fundamental roles in cellular metabolism, signaling, and viability. Disruption of mitochondria not only leads to dysfunction of the organelles but also activates mechanisms of cell injury and death, contributing to the pathogenesis of various diseases. SUMMARY Removal of damaged mitochondria is therefore crucial for cellular homeostasis and survival. Mitophagy, the selective elimination of mitochondria via autophagy, is an important mechanism of mitochondrial quality control in physiological and pathological conditions. Defects in mitophagy have been implicated in a variety of human disorders, including both acute and chronic kidney diseases. However, the role and regulatory mechanisms of mitophagy in kidney cells and tissues remain largely unknown. KEY MESSAGE This review provides updated information on mitophagy and suggests a potential role of mitophagy in renal pathophysiology.
Collapse
Affiliation(s)
- Chengyuan Tang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Liyu He
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jing Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zheng Dong
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China; Department of Cellular Biology and Anatomy, Medical College of Georgia at Georgia Regents University and Charlie Norwood VA Medical Center, Augusta, Ga., USA
| |
Collapse
|
127
|
Ramos AM, González-Guerrero C, Sanz A, Sanchez-Niño MD, Rodríguez-Osorio L, Martín-Cleary C, Fernández-Fernández B, Ruiz-Ortega M, Ortiz A. Designing drugs that combat kidney damage. Expert Opin Drug Discov 2015; 10:541-56. [PMID: 25840605 DOI: 10.1517/17460441.2015.1033394] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Kidney disease remains one of the last worldwide frontiers in the field of non-communicable human disease. From 1990 to 2013, chronic kidney disease (CKD) was the top non-communicable cause of death with a greatest increase in global years of life lost while mortality of acute kidney injury (AKI) still hovers around 50%. This reflects the paucity (for CKD) or lack of (for AKI) therapeutic approaches beyond replacing renal function. Understanding what the barriers are and what potential pathways may facilitate the design of new drugs to combat kidney disease is a key public health priority. AREAS COVERED The authors discuss the hurdles and opportunities for future drug development for kidney disease in light of experience accumulated with drugs that made it to clinical trials. EXPERT OPINION Inflammation, cell death and fibrosis are key therapeutic targets to combat kidney damage. While the specific targeting of drugs to kidney cells would be desirable, the technology is only working at the preclinical stage and with mixed success. Nanomedicines hold promise in this respect. Most drugs undergoing clinical trials for kidney disease have been repurposed from other indications. Currently, the chemokine receptor inhibitor CCX140 holds promise for CKD and the p53 inhibitor QPI-1002 for AKI.
Collapse
Affiliation(s)
- Adrián M Ramos
- Instituto de Investigación Sanitaria-Fundación Jiménez Díaz (IIS-FJD), Laboratory of Renal and Vascular Pathology and Diabetes , Av. Reyes Católicos 2, 28040, Madrid , Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
128
|
Lin HH, Huang CC, Lin TY, Lin CY. p-Cresol mediates autophagic cell death in renal proximal tubular cells. Toxicol Lett 2015; 234:20-9. [DOI: 10.1016/j.toxlet.2015.02.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 01/27/2015] [Accepted: 02/06/2015] [Indexed: 10/24/2022]
|
129
|
Ortiz A, Sanchez-Niño MD, Izquierdo MC, Martin-Cleary C, Garcia-Bermejo L, Moreno JA, Ruiz-Ortega M, Draibe J, Cruzado JM, Garcia-Gonzalez MA, Lopez-Novoa JM, Soler MJ, Sanz AB. Translational value of animal models of kidney failure. Eur J Pharmacol 2015; 759:205-20. [PMID: 25814248 DOI: 10.1016/j.ejphar.2015.03.026] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 02/08/2015] [Accepted: 03/12/2015] [Indexed: 11/28/2022]
Abstract
Acute kidney injury (AKI) and chronic kidney disease (CKD) are associated with decreased renal function and increased mortality risk, while the therapeutic armamentarium is unsatisfactory. The availability of adequate animal models may speed up the discovery of biomarkers for disease staging and therapy individualization as well as design and testing of novel therapeutic strategies. Some longstanding animal models have failed to result in therapeutic advances in the clinical setting, such as kidney ischemia-reperfusion injury and diabetic nephropathy models. In this regard, most models for diabetic nephropathy are unsatisfactory in that they do not evolve to renal failure. Satisfactory models for additional nephropathies are needed. These include anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis, IgA nephropathy, anti-phospholipase-A2-receptor (PLA2R) membranous nephropathy and Fabry nephropathy. However, recent novel models hold promise for clinical translation. Thus, the AKI to CKD translation has been modeled, in some cases with toxins of interest for human CKD such as aristolochic acid. Genetically modified mice provide models for Alport syndrome evolving to renal failure that have resulted in clinical recommendations, polycystic kidney disease models that have provided clues for the development of tolvaptan, that was recently approved for the human disease in Japan; and animal models also contributed to target C5 with eculizumab in hemolytic uremic syndrome. Some ongoing trials explore novel concepts derived from models, such TWEAK targeting as tissue protection for lupus nephritis. We now review animal models reproducing diverse, genetic and acquired, causes of AKI and CKD evolving to kidney failure and discuss the contribution to clinical translation and prospects for the future.
Collapse
Affiliation(s)
- Alberto Ortiz
- Nephrology, IIS-Fundacion Jimenez Diaz, Madrid, Spain; REDinREN, Madrid, Spain; Universidad Autonoma de Madrid, Madrid, Spain; IRSIN, Madrid, Spain
| | | | - Maria C Izquierdo
- Nephrology, IIS-Fundacion Jimenez Diaz, Madrid, Spain; REDinREN, Madrid, Spain
| | | | - Laura Garcia-Bermejo
- REDinREN, Madrid, Spain; Dpt. of Pathology, Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS, Madrid, Spain
| | - Juan A Moreno
- Nephrology, IIS-Fundacion Jimenez Diaz, Madrid, Spain
| | - Marta Ruiz-Ortega
- Nephrology, IIS-Fundacion Jimenez Diaz, Madrid, Spain; REDinREN, Madrid, Spain; Universidad Autonoma de Madrid, Madrid, Spain
| | - Juliana Draibe
- REDinREN, Madrid, Spain; Nephrology Department, Hospital Universitari de Bellvitge, IDIBELL, L׳Hospitalet de Llobregat, Barcelona, Spain
| | - Josep M Cruzado
- REDinREN, Madrid, Spain; Nephrology Department, Hospital Universitari de Bellvitge, IDIBELL, L׳Hospitalet de Llobregat, Barcelona, Spain
| | - Miguel A Garcia-Gonzalez
- REDinREN, Madrid, Spain; Laboratorio de Nefrología, Complexo Hospitalario de Santiago de Compostela (CHUS), Instituto de Investigación Sanitaria (IDIS), Santiago de Compostela, Spain
| | - Jose M Lopez-Novoa
- REDinREN, Madrid, Spain; Departamento de Fisiología y Farmacología, Universidad de Salamanca, Salamnca, Spain
| | - Maria J Soler
- REDinREN, Madrid, Spain; Nephrology Department, Hospital del Mar, Barcelona, Spain
| | - Ana B Sanz
- Nephrology, IIS-Fundacion Jimenez Diaz, Madrid, Spain; REDinREN, Madrid, Spain.
| | | |
Collapse
|
130
|
Santamaría B, Ucero AC, Benito-Martin A, Vicent MJ, Orzáez M, Celdrán A, Selgas R, Ruíz-Ortega M, Ortiz A. Biocompatibility Reduces Inflammation-Induced Apoptosis in Mesothelial Cells Exposed to Peritoneal Dialysis Fluid. Blood Purif 2015; 39:200-209. [DOI: 10.1159/000374103] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 01/09/2015] [Indexed: 11/19/2022]
Abstract
Background/Aims: Peritonitis is a major complication that arises out of peritoneal dialysis (PD), leading to death and loss of mesothelium and peritoneal injury, which may impede PD. We studied the combined impact of inflammatory mediators and PD fluids on mesothelial cell death. Methods: Cultured human mesothelial cells. Results: Inflammatory cytokines (TNF-α and interferon-γ) cooperate with bioincompatible PD fluids containing high glucose degradation product (GDP) concentrations to promote mesothelial cell death. Thus, the inflammatory cytokine cocktail induced a higher rate of death in cells cultured in high GDP PD fluid than in low GDP PD fluid or cell culture medium (cell death expressed as % hypodiploid cells: TNF-α and interferon-γ in RPMI: 14.15 ± 1.68, TNF-α and interferon-γ in 4.25% low GDP PD fluid 13.16 ± 3.29, TNF-α and interferon-γ in 4.25% high GDP PD fluid 25.88 ± 2.18%, p < 0.05 vs. the other two groups). BclxL BH4 peptides, Apaf-1 inhibition or caspase inhibition failed to protect from apoptosis induced by the combination of inflammatory cytokines and bioincompatible PD fluids, although they protected from other forms of mesothelial cell apoptosis. Conclusion: Inflammation cooperates with high GDP PD fluids to promote mesothelial cell death, which is resistant to several therapeutic approaches. This information provides a framework for selection of PD fluid during peritonitis.
Collapse
|
131
|
Sanchez-Niño MD, Fernandez-Fernandez B, Perez-Gomez MV, Poveda J, Sanz AB, Cannata-Ortiz P, Ruiz-Ortega M, Egido J, Selgas R, Ortiz A. Albumin-induced apoptosis of tubular cells is modulated by BASP1. Cell Death Dis 2015; 6:e1644. [PMID: 25675304 PMCID: PMC4669784 DOI: 10.1038/cddis.2015.1] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 12/21/2014] [Accepted: 12/29/2014] [Indexed: 12/25/2022]
Abstract
Albuminuria promotes tubular injury and cell death, and is associated with faster progression of chronic kidney disease (CKD) to end-stage renal disease. However, the molecular mechanisms regulating tubular cell death in response to albuminuria are not fully understood. Brain abundant signal protein 1 (BASP1) was recently shown to mediate glucose-induced apoptosis in tubular cells. We have studied the role of BASP1 in albumin-induced tubular cell death. BASP1 expression was studied in experimental puromycin aminonucleoside-induced nephrotic syndrome in rats and in human nephrotic syndrome. The role of BASP1 in albumin-induced apoptosis was studied in cultured human HK2 proximal tubular epithelial cells. Puromycin aminonucleoside induced proteinuria and increased total kidney BASP1 mRNA and protein expression. Immunohistochemistry localized the increased BASP1 to tubular cells. BASP1 expression colocalized with deoxynucleotidyl-transferase-mediated dUTP nick-end labeling staining for apoptotic cells. Increased tubular BASP1 expression was observed in human proteinuric nephropathy by immunohistochemistry, providing evidence for potential clinical relevance. In cultured tubular cells, albumin induced apoptosis and increased BASP1 mRNA and protein expression at 6–48 h. Confocal microscopy localized the increased BASP1 expression in albumin-treated cells mainly to the perinuclear area. A peripheral location near the cell membrane was more conspicuous in albumin-treated apoptotic cells, where it colocalized with actin. Inhibition of BASP1 expression by a BASP1 siRNA protected from albumin-induced apoptosis. In conclusion, albumin-induced apoptosis in tubular cells is BASP1-dependent. This information may be used to design novel therapeutic approaches to slow CKD progression based on protection of tubular cells from the adverse consequences of albuminuria.
Collapse
Affiliation(s)
- M D Sanchez-Niño
- 1] Instituto de Investigacion Sanitaria IDIPAZ, Madrid, Spain [2] REDINREN, Madrid, Spain
| | - B Fernandez-Fernandez
- 1] REDINREN, Madrid, Spain [2] IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid and Fundación Renal Iñigo Alvarez de Toledo-IRSIN, Madrid, Spain
| | - M V Perez-Gomez
- 1] REDINREN, Madrid, Spain [2] IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid and Fundación Renal Iñigo Alvarez de Toledo-IRSIN, Madrid, Spain
| | - J Poveda
- 1] REDINREN, Madrid, Spain [2] IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid and Fundación Renal Iñigo Alvarez de Toledo-IRSIN, Madrid, Spain
| | - A B Sanz
- 1] REDINREN, Madrid, Spain [2] IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid and Fundación Renal Iñigo Alvarez de Toledo-IRSIN, Madrid, Spain
| | - P Cannata-Ortiz
- 1] REDINREN, Madrid, Spain [2] IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid and Fundación Renal Iñigo Alvarez de Toledo-IRSIN, Madrid, Spain
| | - M Ruiz-Ortega
- 1] REDINREN, Madrid, Spain [2] IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid and Fundación Renal Iñigo Alvarez de Toledo-IRSIN, Madrid, Spain
| | - J Egido
- IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid and Fundación Renal Iñigo Alvarez de Toledo-IRSIN, Madrid, Spain
| | - R Selgas
- 1] Instituto de Investigacion Sanitaria IDIPAZ, Madrid, Spain [2] REDINREN, Madrid, Spain
| | - A Ortiz
- 1] REDINREN, Madrid, Spain [2] IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid and Fundación Renal Iñigo Alvarez de Toledo-IRSIN, Madrid, Spain
| |
Collapse
|
132
|
Essential Oils from Fructus A. zerumbet Protect Human Aortic Endothelial Cells from Apoptosis Induced by Ox-LDL In Vitro. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 2014:956824. [PMID: 25610487 PMCID: PMC4290151 DOI: 10.1155/2014/956824] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 12/02/2014] [Indexed: 11/28/2022]
Abstract
Alpinia zerumbet is a miao folk medicinal plant widely used in the Guizhou Province of southwest China that contains several bioactive constituents and possesses protective effects against cardiovascular diseases. In the present study, we evaluated the protective effect of essential oils derived from Fructus Alpiniae zerumbet (EOFAZ) on oxidized lowdensity-lipoprotein- (ox-LDL-) induced apoptosis in human aortic endothelial cells (HAECs). Following exposure to ox-LDL, HAECs presented with classical characteristics of apoptosis. However, EOFAZ ameliorated these morphological alterations and also inhibited the decrease in cell viability. In addition, EOFAZ abrogated the number of TUNEL or Hoechst 33258 stained positive cells observed after ox-LDL challenge. Investigation into the mechanisms of this inhibition revealed that EOFAZ treatment resulted in a downregulation of Bax and Caspase-3 at both the protein and mRNA expression levels. Moreover, EOFAZ was found to upregulate Bcl-2 protein and mRNA levels and to attenuate ox-LDL-induced HAECs injury caused by apoptosis, revealing both its therapeutic potential for endothelial cell injury protection and its clinical application for atherosclerosis.
Collapse
|
133
|
Sun GX, Ding R, Li M, Guo Y, Fan LP, Yue LS, Li LY, Zhao M. Ghrelin attenuates renal fibrosis and inflammation of obstructive nephropathy. J Urol 2014; 193:2107-15. [PMID: 25481038 DOI: 10.1016/j.juro.2014.11.098] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/26/2014] [Indexed: 10/24/2022]
Abstract
PURPOSE Ghrelin is a gastric peptide that modulates multiple biological functions, of which the stimulation of food intake is the most well-known function. Ghrelin also exerts potential anti-inflammatory and antifibrotic properties in different organs but to our knowledge whether ghrelin inhibits the progression of renal fibrosis is unknown. Thus, we investigated the effect and underlying mechanisms of ghrelin in a rat model of renal fibrosis. MATERIALS AND METHODS Male Sprague Dawley® rats were divided into 4 groups, including vehicle or ghrelin treated sham operated groups and vehicle or ghrelin treated unilateral ureteral obstruction groups. Kidneys harvested on postoperative day 7 or 14 were evaluated for renal inflammation, fibrosis and apoptosis, and the expression of profibrotic and proinflammatory factors. RESULTS Ghrelin inhibited renal fibrosis by attenuating collagen production, extracellular matrix deposition, and α-smooth muscle actin and fibronectin expression. Ghrelin administration decreased macrophage infiltration and several proinflammatory cytokines, including tumor necrosis factor-α, interleukin-1β and monocyte chemotactic protein-1, as well as phosphorylated nuclear factor-κB p65. Ghrelin also inhibited myofibroblast accumulation by blocking the transforming growth factor-β1/Smad3 signaling pathway. Furthermore, ghrelin attenuated renal tubular cell apoptosis and epithelial-mesenchymal transition processes induced by unilateral ureteral obstruction injury. CONCLUSIONS These findings indicate that ghrelin is a potent antifibrotic agent that may have therapeutic potential in patients with obstructive nephropathy.
Collapse
Affiliation(s)
- Guang-Xi Sun
- Department of Organ Transplantation and National Key Clinic Specialty, Neurosurgery Institute of Guangdong Province, Department of Neurosurgery (RD), Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Rui Ding
- Department of Organ Transplantation and National Key Clinic Specialty, Neurosurgery Institute of Guangdong Province, Department of Neurosurgery (RD), Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Ming Li
- Department of Organ Transplantation and National Key Clinic Specialty, Neurosurgery Institute of Guangdong Province, Department of Neurosurgery (RD), Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Ying Guo
- Department of Organ Transplantation and National Key Clinic Specialty, Neurosurgery Institute of Guangdong Province, Department of Neurosurgery (RD), Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Li-Pei Fan
- Department of Organ Transplantation and National Key Clinic Specialty, Neurosurgery Institute of Guangdong Province, Department of Neurosurgery (RD), Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Liang-Sheng Yue
- Department of Organ Transplantation and National Key Clinic Specialty, Neurosurgery Institute of Guangdong Province, Department of Neurosurgery (RD), Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Liu-Yang Li
- Department of Organ Transplantation and National Key Clinic Specialty, Neurosurgery Institute of Guangdong Province, Department of Neurosurgery (RD), Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Ming Zhao
- Department of Organ Transplantation and National Key Clinic Specialty, Neurosurgery Institute of Guangdong Province, Department of Neurosurgery (RD), Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China.
| |
Collapse
|
134
|
Eirin A, Lerman A, Lerman LO. Mitochondria: a pathogenic paradigm in hypertensive renal disease. Hypertension 2014; 65:264-70. [PMID: 25403611 DOI: 10.1161/hypertensionaha.114.04598] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Alfonso Eirin
- From the Divisions of Nephrology and Hypertension (A.E., L.O.L.) and Cardiovascular Diseases (A.L., L.O.L.), Mayo Clinic, Rochester, MN
| | - Amir Lerman
- From the Divisions of Nephrology and Hypertension (A.E., L.O.L.) and Cardiovascular Diseases (A.L., L.O.L.), Mayo Clinic, Rochester, MN
| | - Lilach O Lerman
- From the Divisions of Nephrology and Hypertension (A.E., L.O.L.) and Cardiovascular Diseases (A.L., L.O.L.), Mayo Clinic, Rochester, MN.
| |
Collapse
|
135
|
Zhang B, Rong R, Li H, Peng X, Xiong L, Wang Y, Yu X, Mao H. Heat shock protein 72 suppresses apoptosis by increasing the stability of X-linked inhibitor of apoptosis protein in renal ischemia/reperfusion injury. Mol Med Rep 2014; 11:1793-9. [PMID: 25394481 PMCID: PMC4270332 DOI: 10.3892/mmr.2014.2939] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 11/03/2014] [Indexed: 12/26/2022] Open
Abstract
X-linked inhibitor of apoptosis protein (XIAP) negatively regulates apoptotic pathways at a post-mitochondrial level. XIAP functions by directly binding and inhibiting activation of specific caspases. Upon apoptotic stimuli, mitochondrial second mitochondria-derived activator of caspases (Smac)/direct IAP-binding protein with low PI (DIABLO) is released into the cytosol, which results in displacement of XIAP from caspases. Heat shock protein 72 (HSP72), an anti-apoptotic protein, prevents mitochondrial injury resulting from acute renal ischemia/reperfusion (I/R), its role in Smac/DIABLO and XIAP signaling remains to be elucidated. In the present study, the hypothesis that HSP72 prevents XIAP degradation in vivo and in vitro was assessed. To this purpose, a rat model of I/R injury was used to investigate the renoprotective role of HSP72 by treatment with geranylgeranylacetone (GGA), a specific inducer of HSP72. The mechanism of the cytoprotective properties of HSP72 was also investigated in vitro using adenovirus-mediated overexpression of HSP72 in adenosine triphosphate (ATP)-depleted human kidney 2 (HK-2) cells. Pre-conditioning rats with GGA attenuated renal tubular cell damage, reduced cell apoptosis, preserved XIAP protein content and improved renal function following I/R injury. An in vitro study was performed in which cells were transiently exposed to 5 mM sodium cyanide in a glucose-free medium in order to induce apoptosis. Compared with the control, overexpression of HSP72 inhibited Smac/DIABLO release from the mitochondria and increased levels of XIAP and pro-caspase 3 in ATP-depleted HK-2 cells. In addition, HSP72 interacted with Smac/DIABLO. The present data demonstrates that HSP72 preserves renal function in I/R injury through its anti-apoptotic effects, which act by suppressing mitochondrial Smac/DIABLO release and preserving XIAP protein content.
Collapse
Affiliation(s)
- Baiyu Zhang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat‑sen University, Key Laboratory of Nephrology, Ministry of Health of China, Guangzhou, Guangdong 510080, P.R. China
| | - Rong Rong
- Department of Nephrology, The First Affiliated Hospital, Sun Yat‑sen University, Key Laboratory of Nephrology, Ministry of Health of China, Guangzhou, Guangdong 510080, P.R. China
| | - Huiyan Li
- Department of Nephrology, The First Affiliated Hospital, Sun Yat‑sen University, Key Laboratory of Nephrology, Ministry of Health of China, Guangzhou, Guangdong 510080, P.R. China
| | - Xuan Peng
- Department of Nephrology, The First Affiliated Hospital, Sun Yat‑sen University, Key Laboratory of Nephrology, Ministry of Health of China, Guangzhou, Guangdong 510080, P.R. China
| | - Liping Xiong
- Department of Nephrology, The First Affiliated Hospital, Sun Yat‑sen University, Key Laboratory of Nephrology, Ministry of Health of China, Guangzhou, Guangdong 510080, P.R. China
| | - Yihan Wang
- Laboratory for Kidney Pathology, Inc., Nashville, TN 37203, USA
| | - Xueqing Yu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat‑sen University, Key Laboratory of Nephrology, Ministry of Health of China, Guangzhou, Guangdong 510080, P.R. China
| | - Haiping Mao
- Department of Nephrology, The First Affiliated Hospital, Sun Yat‑sen University, Key Laboratory of Nephrology, Ministry of Health of China, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
136
|
Abstract
Maternal undernutrition (MUN) results in growth-restricted newborns with reduced nephron numbers that is associated with increased risk of hypertension and renal disease. The total adult complement of nephrons is set during nephrogenesis suggesting that MUN affects the staged development of nephrons in as yet unknown manner. A possible cause may be the increased renal apoptosis; therefore, we investigated whether apoptotic signaling and cell death were increased in MUN rat kidneys. Pregnant rat dams were fed an ad libitum diet [control] or were 50% food restricted (MUN) starting at embryonic day (E) 10. Male offspring kidneys (n = 5 each, MUN and control) were analyzed for mRNA using quantitative PCR (E20) and for protein expression using Western blotting and immunohistochemistry (E20 and postnatal day 1, P1). Apoptosis was measured by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay. Upregulation of pro-apoptotic protein expression was detected at E20 (Fas receptor, caspase 9) and at P1 (caspase 3, Bax). The anti-apoptotic factor Bcl2 was significantly decreased in P1 kidneys. Kidney TUNEL showed apoptotic nuclei significantly increased in the P1 nephrogenic zone (MUN 3.3 + 0.3 v. C 1.6 + 0.5, P = 0.002). The majority of apoptotic nuclei co-localized to mesenchyme and pretubular aggregates in the nephrogenic zone. Differential regulation of apoptosis in mesenchyme and pretubular aggregates following parturition suggests a mechanism for nephropenia in gestational programming of the kidney.
Collapse
|
137
|
Abstract
Acute kidney injury (AKI) is a serious clinical condition with no effective treatment. Tubular cells are key targets in AKI. Tubular cells and, specifically, proximal tubular cells are extremely rich in mitochondria and mitochondrial changes had long been known to be a feature of AKI. However, only recent advances in understanding the molecules involved in mitochondria biogenesis and dynamics and the availability of mitochondria-targeted drugs has allowed the exploration of the specific role of mitochondria in AKI. We now review the morphological and functional mitochondrial changes during AKI, as well as changes in the expression of mitochondrial genes and proteins. Finally, we summarise the current status of novel therapeutic strategies specifically targeting mitochondria such as mitochondrial permeability transition pore (MPTP) opening inhibitors (cyclosporine A (CsA)), quinone analogues (MitoQ, SkQ1 and SkQR1), superoxide dismutase (SOD) mimetics (Mito-CP), Szeto-Schiller (SS) peptides (Bendavia) and mitochondrial division inhibitors (mdivi-1). MitoQ, SkQ1, SkQR1, Mito-CP, Bendavia and mdivi-1 have improved the course of diverse experimental models of AKI. Evidence for a beneficial effect of CsA on human cardiac ischaemia-reperfusion injury derives from a clinical trial; however, CsA is nephrotoxic. MitoQ and Bendavia have been shown to be safe for humans. Ongoing clinical trials are testing the efficacy of Bendavia in AKI prevention following renal artery percutaneous transluminal angioplasty.
Collapse
|
138
|
Inoue T, Kusano T, Tomori K, Nakamoto H, Suzuki H, Okada H. Effects of cell-type-specific expression of a pan-caspase inhibitor on renal fibrogenesis. Clin Exp Nephrol 2014; 19:350-8. [PMID: 25070875 DOI: 10.1007/s10157-014-1011-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 07/07/2014] [Indexed: 01/07/2023]
Abstract
BACKGROUND The caspase family of enzymes is grouped into two major sub-families, namely apoptotic and inflammatory caspases, which play central roles in the induction of apoptosis, regulation of inflammation and immunity, and cellular differentiation. METHODS The role of caspase activation in tubular epithelium and interstitial cells of 3 lines of transgenic mice with obstructed nephropathy was examined: p35 mice bearing the pan-caspase inhibitor protein expressed by the p35 gene separated from the universal CAG promoter by a floxed STOP sequence were crossed with γGT.Cre and FSP1.Cre mice that express Cre recombinase in the cortical tubular epithelium and FSP1(+) interstitial cells, respectively. The γGT.Cre;p35, FSP1.Cre;p35 and p35 control mice were then challenged with unilateral ureter obstruction (UUO). RESULTS Proinflammatory parameters such as protein levels of active IL-1β subunit and mRNA levels of TNF-α and NOD-like receptor pyrin domain containing-3, and profibrogenic parameters such as interstitial matrix deposition and mRNA levels of fibronectin EIIIA isoform and α1 chain of procollagen type I in the kidneys were significantly increased at 7 days in the FSP1.Cre;p35- and p35-UUO mice, but not in the γGT.Cre;p35-UUO mice. These changes paralleled the numbers of apoptotic nuclei in tubules, but not in interstitial cells, and the protein levels of active caspase-3 subunit in the kidneys of FSP1.Cre;p35-, p35- and γGT.Cre;p35-UUO mice. CONCLUSION This study provides evidence of the critical role of caspase activation in the tubular epithelium, but not in FSP1(+) interstitial cells, in apoptosis and inflammasome induction, leading to proinflammatory and profibrogenic processes in fibrous kidneys with UUO.
Collapse
Affiliation(s)
- Tsutomu Inoue
- Department of Nephrology, Faculty of Medicine, Saitama Medical University, 38 Morohongo, Moroyama-cho, Irumagun, Saitama, 350-0451, Japan
- Division of Project Research, Saitama Medical University Research Center of Genomic Medicine, 1397-1 Yamane, Hidaka, Saitama, 350-1298, Japan
| | - Takeru Kusano
- Division of Project Research, Saitama Medical University Research Center of Genomic Medicine, 1397-1 Yamane, Hidaka, Saitama, 350-1298, Japan
- Department of General Internal Medicine, Faculty of Medicine, Saitama Medical University, 38 Morohongo, Moroyama-cho, Irumagun, Saitama, 350-0451, Japan
| | - Kouji Tomori
- Department of Nephrology, Faculty of Medicine, Saitama Medical University, 38 Morohongo, Moroyama-cho, Irumagun, Saitama, 350-0451, Japan
| | - Hidetomo Nakamoto
- Department of General Internal Medicine, Faculty of Medicine, Saitama Medical University, 38 Morohongo, Moroyama-cho, Irumagun, Saitama, 350-0451, Japan
| | - Hiromichi Suzuki
- Department of Nephrology, Faculty of Medicine, Saitama Medical University, 38 Morohongo, Moroyama-cho, Irumagun, Saitama, 350-0451, Japan
- Division of Project Research, Saitama Medical University Research Center of Genomic Medicine, 1397-1 Yamane, Hidaka, Saitama, 350-1298, Japan
| | - Hirokazu Okada
- Department of Nephrology, Faculty of Medicine, Saitama Medical University, 38 Morohongo, Moroyama-cho, Irumagun, Saitama, 350-0451, Japan.
- Division of Project Research, Saitama Medical University Research Center of Genomic Medicine, 1397-1 Yamane, Hidaka, Saitama, 350-1298, Japan.
- Department of General Internal Medicine, Faculty of Medicine, Saitama Medical University, 38 Morohongo, Moroyama-cho, Irumagun, Saitama, 350-0451, Japan.
| |
Collapse
|
139
|
Martín P, Mora I, Cortes MA, Calleros L, García-Jerez A, Ortiz A, Rodríguez-Puyol M, Rodríguez-Puyol D, Olmos G. Relevant role of PKG in the progression of fibrosis induced by TNF-like weak inducer of apoptosis. Am J Physiol Renal Physiol 2014; 307:F75-85. [DOI: 10.1152/ajprenal.00398.2013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
TNF-like weak inducer of apoptosis (TWEAK) is an inflammatory cytokine that activates the FGF-inducible 14 receptor. Both TWEAK and the FGF-inducible 14 receptor are constitutively expressed in the kidney. TWEAK has been shown to modulate several biological responses, such as inflammation, proliferation, differentiation, and apoptosis, that contribute to kidney injury. However, the role of TWEAK in fibrosis and TWEAK-activated intracellular signaling pathways remain poorly understood. We tested the hypothesis that TWEAK can be a potent inducer of renal fibrosis by increasing transforming growth factor (TGF)-β1 expression (a well-known switch in the fibrosis process) through PKG-I downregulation. We showed that in human mesangial cells, TWEAK increased TGF-β1 expression and activity, leading to higher levels of the extracellular matrix protein fibronectin and decreased PKG-I expression and activity via the Ras pathway. PKG-I activation with 8-bromo-cGMP, Ras inactivation with dominant negative Ras, or Ras pathway inhibition with the ERK1/2 inhibitor PD-98059 resulted in the prevention of TWEAK-induced TGF-β1 upregulation. In vivo, exogenous administration of TWEAK to wild-type mice downregulated kidney PKG-I and increased kidney TGF-β1 expression. These effects were blunted in H-Ras knockout mice. Together, these data demonstrate, for the first time, the key role of PKG-I in TGF-β1 induction by TWEAK in kidney cells.
Collapse
Affiliation(s)
- Paloma Martín
- Department of System Biology, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
- REDinREN (Instituto de Salud Carlos III), Madrid, Spain
- Instituto Reina Sofía de Investigaciones Nefrológicas, Madrid, Spain
| | - Inés Mora
- REDinREN (Instituto de Salud Carlos III), Madrid, Spain
- Instituto Reina Sofía de Investigaciones Nefrológicas, Madrid, Spain
| | - M. Alicia Cortes
- Department of System Biology, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
- REDinREN (Instituto de Salud Carlos III), Madrid, Spain
- Instituto Reina Sofía de Investigaciones Nefrológicas, Madrid, Spain
| | - Laura Calleros
- Department of System Biology, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
- REDinREN (Instituto de Salud Carlos III), Madrid, Spain
- Instituto Reina Sofía de Investigaciones Nefrológicas, Madrid, Spain
| | - Andrea García-Jerez
- Department of System Biology, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
- REDinREN (Instituto de Salud Carlos III), Madrid, Spain
- Instituto Reina Sofía de Investigaciones Nefrológicas, Madrid, Spain
| | - Alberto Ortiz
- REDinREN (Instituto de Salud Carlos III), Madrid, Spain
- IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain
| | - Manuel Rodríguez-Puyol
- Department of System Biology, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
- REDinREN (Instituto de Salud Carlos III), Madrid, Spain
- Instituto Reina Sofía de Investigaciones Nefrológicas, Madrid, Spain
| | - Diego Rodríguez-Puyol
- REDinREN (Instituto de Salud Carlos III), Madrid, Spain
- Instituto Reina Sofía de Investigaciones Nefrológicas, Madrid, Spain
- Department of Medicine, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
- Nephrology Section and Research Unit, Hospital Príncipe de Asturias, Alcalá de Henares, Madrid, Spain; and
| | - Gemma Olmos
- Department of System Biology, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
- REDinREN (Instituto de Salud Carlos III), Madrid, Spain
- Instituto Reina Sofía de Investigaciones Nefrológicas, Madrid, Spain
| |
Collapse
|
140
|
Xu W, Shao X, Tian L, Gu L, Zhang M, Wang Q, Wu B, Wang L, Yao J, Xu X, Mou S, Ni Z. Astragaloside IV ameliorates renal fibrosis via the inhibition of mitogen-activated protein kinases and antiapoptosis in vivo and in vitro. J Pharmacol Exp Ther 2014; 350:552-62. [PMID: 24951279 DOI: 10.1124/jpet.114.214205] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Apoptosis of renal tubular cells plays a crucial role in renal fibrosis. Astragaloside IV (AS-IV), a compound extracted from Radix Astragali, has been shown to inhibit renal tubular cell apoptosis induced by high glucose, but its role in preventing chronic renal fibrosis as well as the underlying molecular mechanisms involved still remain obscure. In this study, human kidney tubular epithelial cells induced by transforming growth factor-β1 (TGF-β1) were used to investigate the protective role of AS-IV in antifibrosis. As an in vivo model, mice subjected to unilateral ureteral obstruction (UUO) were administered AS-IV (20 mg/kg) by intraperitoneal injection for 7 days. AS-IV significantly alleviated renal mass loss and reduced the expression of α-smooth muscle actin, fibronectin, and collagen IV both in vitro and in vivo, suggesting that this compound functions in the inhibition of renal tubulointerstitial fibrosis. Furthermore, transferase-mediated dUTP nick-end labeling assay results both in vivo and in vitro showed that AS-IV significantly attenuated both UUO and TGF-β1-induced cell apoptosis and prevented renal tubular epithelial cell injury in a dose-dependent manner. Western blotting results also revealed that the antiapoptotic effect of AS-IV was reflected in the inhibition of caspase-3 activation, which might be mediated primarily by the downregulation of mitogen-activated protein kinase effectors phospho-p38 and phospho-c-Jun N-terminal kinase. These data infer that AS-IV effectively attenuates the progression of renal fibrosis after UUO injury and may have a promising clinical role as a potential antifibrosis treatment in patients with chronic kidney disease.
Collapse
Affiliation(s)
- Weijia Xu
- Department of Nephrology, Molecular Cell Laboratory for Kidney Disease (W.X., X.S., L.T., L.G., M.Z., Q.W., B.W., L.W., S.M., Z.N.), Animal Centre (J.Y.), and Department of Biochemical Laboratory (X.X.), Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xinghua Shao
- Department of Nephrology, Molecular Cell Laboratory for Kidney Disease (W.X., X.S., L.T., L.G., M.Z., Q.W., B.W., L.W., S.M., Z.N.), Animal Centre (J.Y.), and Department of Biochemical Laboratory (X.X.), Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lei Tian
- Department of Nephrology, Molecular Cell Laboratory for Kidney Disease (W.X., X.S., L.T., L.G., M.Z., Q.W., B.W., L.W., S.M., Z.N.), Animal Centre (J.Y.), and Department of Biochemical Laboratory (X.X.), Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Leyi Gu
- Department of Nephrology, Molecular Cell Laboratory for Kidney Disease (W.X., X.S., L.T., L.G., M.Z., Q.W., B.W., L.W., S.M., Z.N.), Animal Centre (J.Y.), and Department of Biochemical Laboratory (X.X.), Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Minfang Zhang
- Department of Nephrology, Molecular Cell Laboratory for Kidney Disease (W.X., X.S., L.T., L.G., M.Z., Q.W., B.W., L.W., S.M., Z.N.), Animal Centre (J.Y.), and Department of Biochemical Laboratory (X.X.), Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qin Wang
- Department of Nephrology, Molecular Cell Laboratory for Kidney Disease (W.X., X.S., L.T., L.G., M.Z., Q.W., B.W., L.W., S.M., Z.N.), Animal Centre (J.Y.), and Department of Biochemical Laboratory (X.X.), Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bei Wu
- Department of Nephrology, Molecular Cell Laboratory for Kidney Disease (W.X., X.S., L.T., L.G., M.Z., Q.W., B.W., L.W., S.M., Z.N.), Animal Centre (J.Y.), and Department of Biochemical Laboratory (X.X.), Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ling Wang
- Department of Nephrology, Molecular Cell Laboratory for Kidney Disease (W.X., X.S., L.T., L.G., M.Z., Q.W., B.W., L.W., S.M., Z.N.), Animal Centre (J.Y.), and Department of Biochemical Laboratory (X.X.), Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jufang Yao
- Department of Nephrology, Molecular Cell Laboratory for Kidney Disease (W.X., X.S., L.T., L.G., M.Z., Q.W., B.W., L.W., S.M., Z.N.), Animal Centre (J.Y.), and Department of Biochemical Laboratory (X.X.), Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoping Xu
- Department of Nephrology, Molecular Cell Laboratory for Kidney Disease (W.X., X.S., L.T., L.G., M.Z., Q.W., B.W., L.W., S.M., Z.N.), Animal Centre (J.Y.), and Department of Biochemical Laboratory (X.X.), Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shan Mou
- Department of Nephrology, Molecular Cell Laboratory for Kidney Disease (W.X., X.S., L.T., L.G., M.Z., Q.W., B.W., L.W., S.M., Z.N.), Animal Centre (J.Y.), and Department of Biochemical Laboratory (X.X.), Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhaohui Ni
- Department of Nephrology, Molecular Cell Laboratory for Kidney Disease (W.X., X.S., L.T., L.G., M.Z., Q.W., B.W., L.W., S.M., Z.N.), Animal Centre (J.Y.), and Department of Biochemical Laboratory (X.X.), Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
141
|
Peritubular capillary rarefaction: a new therapeutic target in chronic kidney disease. Pediatr Nephrol 2014; 29:333-42. [PMID: 23475077 PMCID: PMC3726573 DOI: 10.1007/s00467-013-2430-y] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 12/24/2012] [Accepted: 01/24/2013] [Indexed: 02/07/2023]
Abstract
Chronic kidney disease (CKD) has reached worldwide epidemic proportions and desperately needs new therapies. Peritubular capillary (PTC) rarefaction, together with interstitial fibrosis and tubular atrophy, is one of the major hallmarks of CKD and predicts renal outcome in patients with CKD. PTC endothelial cells (ECs) undergo apoptosis during CKD, leading to capillary loss, tissue hypoxia, and oxidative stress. Although the mechanisms of PTC rarefaction are not well understood, the process of PTC rarefaction depends on multiple events that occur during CKD. These events, which lead to an antiangiogenic environment, include deprivation of EC survival factors, increased production of vascular growth inhibitors, malfunction of ECs, dysfunction of endothelial progenitor cells, and loss of EC integrity via pericyte detachment from the vasculature. In this review, we focus on major factors regulating angiogenesis and EC survival and describe the roles of these factors in PTC rarefaction during CKD and possible therapeutic applications.
Collapse
|
142
|
Virzì GM, Day S, de Cal M, Vescovo G, Ronco C. Heart-kidney crosstalk and role of humoral signaling in critical illness. Crit Care 2014; 18:201. [PMID: 24393300 PMCID: PMC4059499 DOI: 10.1186/cc13177] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Organ failure in the heart or kidney can initiate various complex metabolic, cell-mediated and humoral pathways affecting distant organs, contributing to the high therapeutic costs and significantly higher morbidity and mortality. The universal outreach of cells in an injured state has myriad consequences to distant organ cells and their milieu. Heart performance and kidney function are closely interconnected and communication between these organs occurs through a variety of bidirectional pathways. The term cardiorenal syndrome (CRS) is often used to describe this condition and represents an important model for exploring the pathophysiology of cardiac and renal dysfunction. Clinical evidence suggests that tissue injury in both acute kidney injury and heart failure has immune-mediated inflammatory consequences that can initiate remote organ dysfunction. Acute cardiorenal syndrome (CRS type 1) and acute renocardiac syndrome (CRS type 3) are particularly relevant in high-acuity medical units. This review briefly summarizes relevant research and focuses on the role of signaling in heart-kidney crosstalk in the critical care setting.
Collapse
Affiliation(s)
- Grazia Maria Virzì
- Department of Nephrology, Dialysis and Transplantation, San Bortolo Hospital, International Renal Research Institute Vicenza, Via Rodolfi 37, Vicenza 36100, Italy
- IRRIV – International Renal Resarch Institute Vicenza, Via Rodolfi 37, Vicenza 36100, Italy
- Clinical Genetics Unit, Department of Women’s and Children’s Health, University of Padua, Padua, Italy
| | - Sonya Day
- Department of Nephrology, Dialysis and Transplantation, San Bortolo Hospital, International Renal Research Institute Vicenza, Via Rodolfi 37, Vicenza 36100, Italy
- IRRIV – International Renal Resarch Institute Vicenza, Via Rodolfi 37, Vicenza 36100, Italy
| | - Massimo de Cal
- Department of Nephrology, Dialysis and Transplantation, San Bortolo Hospital, International Renal Research Institute Vicenza, Via Rodolfi 37, Vicenza 36100, Italy
- IRRIV – International Renal Resarch Institute Vicenza, Via Rodolfi 37, Vicenza 36100, Italy
| | - Giorgio Vescovo
- Internal Medicine, San Bortolo Hospital, Vicenza, Via Giustiniani, Padua 35128, Italy
| | - Claudio Ronco
- Department of Nephrology, Dialysis and Transplantation, San Bortolo Hospital, International Renal Research Institute Vicenza, Via Rodolfi 37, Vicenza 36100, Italy
- IRRIV – International Renal Resarch Institute Vicenza, Via Rodolfi 37, Vicenza 36100, Italy
| |
Collapse
|
143
|
Sanchez-Niño MD, Poveda J, Sanz AB, Carrasco S, Ruiz-Ortega M, Selgas R, Egido J, Ortiz A. 3,4-DGE is cytotoxic and decreases HSP27/HSPB1 in podocytes. Arch Toxicol 2013; 88:597-608. [PMID: 24337777 DOI: 10.1007/s00204-013-1181-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 12/04/2013] [Indexed: 01/19/2023]
Abstract
Hyperglycemia is the key driver of diabetic complications and increased concentrations of glucose degradation products. The study of peritoneal dialysis solution biocompatibility has highlighted the adverse biological effects of glucose degradation products. Recently, 3,4-dideoxyglucosone-3-ene (3,4-DGE) was identified as the most toxic glucose degradation product in peritoneal dialysis fluids. In addition, 3,4-DGE is present in high-fructose corn syrup, and its precursor 3-deoxyglucosone is increased in diabetes. The role of 3,4-DGE in glomerular injury had not been addressed. We studied the effects of 3,4-DGE on cultured human podocytes and in vivo in mice. 3,4-DGE induced apoptosis in podocytes in a dose- and time-dependent manner. 3,4-DGE promoted the release of cytochrome c from mitochondria and activation of caspase-3. While high glucose concentrations increased the levels of the podocyte intracellular antiapoptotic protein HSP27/HSPB1, 3,4-DGE decreased the expression of podocyte HSP27/HSPB1. Apoptosis induced by 3,4-DGE was caspase-dependent and could be prevented by the broad-spectrum caspase inhibitor zVAD-fmk. Antagonism of Bax by a Ku-70-derived peptide also prevented apoptosis. Intravenous administration of 3,4-DGE to healthy mice resulted in a decreased expression of HSP27/HSPB1 and caspase-3 activation in whole kidney and in podocytes in vivo. In conclusion, 3,4-DGE induces apoptotic cell death in cultured human podocytes, suggesting a potential role in glomerular injury resulting from metabolic disorders.
Collapse
|
144
|
Suski M, Olszanecki R, Stachowicz A, Madej J, Bujak-Giżycka B, Okoń K, Korbut R. The influence of angiotensin-(1–7) Mas receptor agonist (AVE 0991) on mitochondrial proteome in kidneys of apoE knockout mice. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2013; 1834:2463-9. [DOI: 10.1016/j.bbapap.2013.08.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 08/19/2013] [Indexed: 12/18/2022]
|
145
|
Endogenous NAMPT dampens chemokine expression and apoptotic responses in stressed tubular cells. Biochim Biophys Acta Mol Basis Dis 2013; 1842:293-303. [PMID: 24287278 DOI: 10.1016/j.bbadis.2013.11.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2013] [Revised: 11/12/2013] [Accepted: 11/19/2013] [Indexed: 11/23/2022]
Abstract
Diabetic nephropathy (DN) is the most common cause of end-stage renal disease and identification of new therapeutic targets is needed. Nicotinamide phosphoribosyltransferase (NAMPT) is both an extracellular and intracellular protein. Circulating NAMPT is increased in diabetics and in chronic kidney disease patients. The role of NAMPT in renal cell biology is poorly understood. NAMPT mRNA and protein were increased in the kidneys of rats with streptozotocin-induced diabetes. Immunohistochemistry localized NAMPT to glomerular and tubular cells in diabetic rats. The inflammatory cytokine TNFα increased NAMPT mRNA, protein and NAD production in cultured kidney human tubular cells. Exogenous NAMPT increased the mRNA expression of chemokines MCP-1 and RANTES. The NAMPT enzymatic activity inhibitor FK866 prevented these effects. By contrast, FK866 boosted TNFα-induced expression of MCP-1 and RANTES mRNA and endogenous NAMPT targeting by siRNA also had a proinflammatory effect. Furthermore, FK866 promoted tubular cell apoptosis in an inflammatory milieu containing the cytokines TNFα/IFNγ. In an inflammatory environment FK866 promoted tubular cell expression of the lethal cytokine TRAIL. These data are consistent with a role of endogenous NAMPT activity as an adaptive, protective response to an inflammatory milieu that differs from the proinflammatory activity of exogenous NAMPT. Thus, disruption of endogenous NAMPT function in stressed cells promotes tubular cell death and chemokine expression. This information may be relevant for the design of novel therapeutic strategies in DN.
Collapse
|
146
|
Husi H, Sanchez-Niño MD, Delles C, Mullen W, Vlahou A, Ortiz A, Mischak H. A combinatorial approach of Proteomics and Systems Biology in unravelling the mechanisms of acute kidney injury (AKI): involvement of NMDA receptor GRIN1 in murine AKI. BMC SYSTEMS BIOLOGY 2013; 7:110. [PMID: 24172336 PMCID: PMC3827826 DOI: 10.1186/1752-0509-7-110] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 10/28/2013] [Indexed: 12/23/2022]
Abstract
BACKGROUND Acute kidney injury (AKI) is a frequent condition in hospitalised patients undergoing major surgery or the critically ill and is associated with increased mortality. Based on the volume of the published literature addressing this condition, reporting both supporting as well as conflicting molecular evidence, it is apparent that a comprehensive analysis strategy is required to understand and fully delineate molecular events and pathways which can be used to describe disease induction and progression as well as lead to a more targeted approach in intervention therapies. RESULTS We used a Systems Biology approach coupled with a de-novo high-resolution proteomic analysis of kidney cortex samples from a mouse model of folic acid-induced AKI (12 animals in total) and show comprehensive mapping of signalling cascades, gene activation events and metabolite interference by mapping high-resolution proteomic datasets onto a de-novo hypothesis-free dataspace. The findings support the involvement of the glutamatergic signalling system in AKI, induced by over-activation of the N-methyl-D-aspartate (NMDA)-receptor leading to apoptosis and necrosis by Ca2+-influx, calpain and caspase activation, and co-occurring reactive oxygen species (ROS) production to DNA fragmentation and NAD-rundown. The specific over-activation of the NMDA receptor may be triggered by the p53-induced protein kinase Dapk1, which is a known non-reversible cell death inducer in a neurological context. The pathway mapping is consistent with the involvement of the Renin-Angiotensin Aldosterone System (RAAS), corticoid and TNFα signalling, leading to ROS production and gene activation through NFκB, PPARγ, SMAD and HIF1α trans-activation, as well as p53 signalling cascade activation. Key elements of the RAAS-glutamatergic axis were assembled as a novel hypothetical pathway and validated by immunohistochemistry. CONCLUSIONS This study shows to our knowledge for the first time in a molecular signal transduction pathway map how AKI is induced, progresses through specific signalling cascades that may lead to end-effects such as apoptosis and necrosis by uncoupling of the NMDA receptor. Our results can potentially pave the way for a targeted pharmacological intervention in disease progression or induction.
Collapse
Affiliation(s)
- Holger Husi
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, 126 University Place, Glasgow G12 8TA,, UK.
| | | | | | | | | | | | | |
Collapse
|
147
|
Unilateral ureteral obstruction: beyond obstruction. Int Urol Nephrol 2013; 46:765-76. [PMID: 24072452 DOI: 10.1007/s11255-013-0520-1] [Citation(s) in RCA: 129] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 07/15/2013] [Indexed: 01/10/2023]
Abstract
Unilateral ureteral obstruction is a popular experimental model of renal injury. However, the study of the kidney response to urinary tract obstruction is only one of several advantages of this model. Unilateral ureteral obstruction causes subacute renal injury characterized by tubular cell injury, interstitial inflammation and fibrosis. For this reason, it serves as a model both of irreversible acute kidney injury and of events taking place during human chronic kidney disease. Being a unilateral disease, it is not useful to study changes in global kidney function, but has the advantage of a low mortality and the availability of an internal control (the non-obstructed kidney). Experimental unilateral ureteral obstruction has illustrated the molecular mechanisms of apoptosis, inflammation and fibrosis, all three key processes in kidney injury of any cause, thus providing information beyond obstruction. Recently this model has supported key concepts on the role in kidney fibrosis of epithelial-mesenchymal transition, tubular epithelial cell G2/M arrest, the anti-aging hormone Klotho and renal innervation. We now review the experimental model and its contribution to identifying novel therapeutic targets in kidney injury and fibrosis, independently of the noxa.
Collapse
|
148
|
Benito-Martin A, Ucero AC, Zubiri I, Posada-Ayala M, Fernandez-Fernandez B, Cannata-Ortiz P, Sanchez-Nino MD, Ruiz-Ortega M, Egido J, Alvarez-Llamas G, Ortiz A. Osteoprotegerin in exosome-like vesicles from human cultured tubular cells and urine. PLoS One 2013; 8:e72387. [PMID: 24058411 PMCID: PMC3751949 DOI: 10.1371/journal.pone.0072387] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 07/09/2013] [Indexed: 12/12/2022] Open
Abstract
Urinary exosomes have been proposed as potential diagnostic tools. TNF superfamily cytokines and receptors may be present in exosomes and are expressed by proximal tubular cells. We have now studied the expression of selected TNF superfamily proteins in exosome-like vesicles from cultured human proximal tubular cells and human urine and have identified additional proteins in these vesicles by LC-MS/MS proteomics. Human proximal tubular cells constitutively released exosome-like vesicles that did not contain the TNF superfamily cytokines TRAIL or TWEAK. However, exosome-like vesicles contained osteoprotegerin (OPG), a TNF receptor superfamily protein, as assessed by Western blot, ELISA or selected reaction monitoring by nLC-(QQQ)MS/MS. Twenty-one additional proteins were identified in tubular cell exosome-like vesicles, including one (vitamin D binding protein) that had not been previously reported in exosome-like vesicles. Twelve were extracellular matrix proteins, including the basement membrane proteins type IV collagen, nidogen-1, agrin and fibulin-1. Urine from chronic kidney disease patients contained a higher amount of exosomal protein and exosomal OPG than urine from healthy volunteers. Specifically OPG was increased in autosomal dominant polycystic kidney disease urinary exosome-like vesicles and expressed by cystic epithelium in vivo. In conclusion, OPG is present in exosome-like vesicles secreted by proximal tubular epithelial cells and isolated from Chronic Kidney Disease urine.
Collapse
Affiliation(s)
- Alberto Benito-Martin
- Department of Nephrology, Instituto de Investigaciones Sanitarias-Fundación Jiménez Díaz - Universidad Autonoma de Madrid, Madrid, Spain
| | - Alvaro Conrado Ucero
- Department of Nephrology, Instituto de Investigaciones Sanitarias-Fundación Jiménez Díaz - Universidad Autonoma de Madrid, Madrid, Spain
| | - Irene Zubiri
- Department of Immunology, Instituto de Investigaciones Sanitarias-Fundación Jiménez Díaz - Universidad Autonoma de Madrid, Madrid, Spain
| | - Maria Posada-Ayala
- Department of Immunology, Instituto de Investigaciones Sanitarias-Fundación Jiménez Díaz - Universidad Autonoma de Madrid, Madrid, Spain
| | - Beatriz Fernandez-Fernandez
- Department of Nephrology, Instituto de Investigaciones Sanitarias-Fundación Jiménez Díaz - Universidad Autonoma de Madrid, Madrid, Spain
| | - Pablo Cannata-Ortiz
- Department of Pathology, Instituto de Investigaciones Sanitarias-Fundación Jiménez Díaz - Universidad Autonoma de Madrid, Madrid, Spain
| | | | - Marta Ruiz-Ortega
- Department of Nephrology, Instituto de Investigaciones Sanitarias-Fundación Jiménez Díaz - Universidad Autonoma de Madrid, Madrid, Spain
| | - Jesus Egido
- Department of Nephrology, Instituto de Investigaciones Sanitarias-Fundación Jiménez Díaz - Universidad Autonoma de Madrid, Madrid, Spain
- Instituto Reina Sofia de Investigacion Nefrologica, Madrid, Spain
| | - Gloria Alvarez-Llamas
- Department of Immunology, Instituto de Investigaciones Sanitarias-Fundación Jiménez Díaz - Universidad Autonoma de Madrid, Madrid, Spain
| | - Alberto Ortiz
- Department of Nephrology, Instituto de Investigaciones Sanitarias-Fundación Jiménez Díaz - Universidad Autonoma de Madrid, Madrid, Spain
- Instituto Reina Sofia de Investigacion Nefrologica, Madrid, Spain
- * E-mail:
| |
Collapse
|
149
|
Sanz AB, Sanchez-Niño MD, Izquierdo MC, Gonzalez-Espinoza L, Ucero AC, Poveda J, Ruiz-Andres O, Ruiz-Ortega M, Selgas R, Egido J, Ortiz A. Macrophages and recently identified forms of cell death. Int Rev Immunol 2013; 33:9-22. [PMID: 23802146 DOI: 10.3109/08830185.2013.771183] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Recent advances in cell death biology have uncovered an ever increasing range of cell death forms. Macrophages have a bidirectional relationship with cell death that modulates the immune response. Thus, macrophages engulf apoptotic cells and secrete cytokines that may promote cell death in parenchymal cells. Furthermore, the presence of apoptotic or necrotic dead cells in the microenvironment elicits differential macrophage responses. Apoptotic cells elicit anti-inflammatory responses in macrophages. By contrast macrophages may undergo a proinflammatory form of cell death (pyroptosis) in response to damage-associated molecular patterns (DAMPs) released from necrotic cells and also in response to pathogen-associated molecular patterns (PAMPs). Pyroptosis is a recently identified form of cell death that occurs predominantly in subsets of inflammatory macrophages and is associated to the release of interleukin-1β (IL-1β) and IL-18. Deregulation of these processes may result in disease. Thus, failure of macrophages to engulf apoptotic cells may be a source of autoantigens in autoimmune diseases, excessive macrophage release of proapoptotic factors or sterile pyroptosis may contribute to tissue injury and failure of pathogen-induced pyroptosis may contribute to pathogen survival. Ongoing research is exploring the therapeutic opportunities resulting this new knowledge.
Collapse
Affiliation(s)
- Ana B Sanz
- 1Laboratory of Nephrology, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid and Fundación Renal Iñigo Alvarez de Toledo, Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
150
|
Burlaka I, Liu XL, Rebetz J, Arvidsson I, Yang L, Brismar H, Karpman D, Aperia A. Ouabain protects against Shiga toxin-triggered apoptosis by reversing the imbalance between Bax and Bcl-xL. J Am Soc Nephrol 2013; 24:1413-23. [PMID: 23744887 DOI: 10.1681/asn.2012101044] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Hemolytic uremic syndrome, a life-threatening disease often accompanied by acute renal failure, usually occurs after gastrointestinal infection with Shiga toxin 2 (Stx2)-producing Escherichia coli. Stx2 binds to the glycosphingolipid globotriaosylceramide receptor, expressed by renal epithelial cells, and triggers apoptosis by activating the apoptotic factor Bax. Signaling via the ouabain/Na,K-ATPase/IP3R/NF-κB pathway increases expression of Bcl-xL, an inhibitor of Bax, suggesting that ouabain might protect renal cells from Stx2-triggered apoptosis. Here, exposing rat proximal tubular cells to Stx2 in vitro resulted in massive apoptosis, upregulation of the apoptotic factor Bax, increased cleaved caspase-3, and downregulation of the survival factor Bcl-xL; co-incubation with ouabain prevented all of these effects. Ouabain activated the NF-κB antiapoptotic subunit p65, and the inhibition of p65 DNA binding abolished the antiapoptotic effect of ouabain in Stx2-exposed tubular cells. Furthermore, in vivo, administration of ouabain reversed the imbalance between Bax and Bcl-xL in Stx2-treated mice. Taken together, these results suggest that ouabain can protect the kidney from the apoptotic effects of Stx2.
Collapse
Affiliation(s)
- Ievgeniia Burlaka
- Department of Women's and Children's Health, Karolinska Institutet, Astrid Lindgren Children's Hospital, Research Lab, Stockholm, Sweden
| | | | | | | | | | | | | | | |
Collapse
|