51
|
Hsu YH, Chuang HC, Lee YH, Lin YF, Chiu YJ, Wang YL, Wu MS, Chiu HW. Induction of Fibrosis and Autophagy in Kidney Cells by Vinyl Chloride. Cells 2019; 8:cells8060601. [PMID: 31212930 PMCID: PMC6627785 DOI: 10.3390/cells8060601] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/13/2019] [Accepted: 06/14/2019] [Indexed: 02/07/2023] Open
Abstract
Vinyl chloride (VC) is a noninfective occupational risk factor. It is found in industrial chemicals, volatile organic compounds, cigarette smoke ingredients, etc. It is a kind of toxic gas that causes many diseases. VC exposure causes an increased risk of liver fibrosis and can result in angiosarcoma of the liver. Previous studies have shown that high-doses of VC exposure in mice resulted in acute death with marked tubular necrosis of the renal cortex. In this study, we assessed the nephrotoxicity of VC in vitro and in vivo. As a result, we demonstrated that VC induced fibrosis-associated protein expression, such as connective tissue growth factor (CTGF), plasminogen activator inhibitor-1 (PAI-1) and collagen 1, and autophagy-associated protein expression, such as Beclin 1 and LC3-II, in kidney cells. The beclin1 siRNA experiments found that autophagy inhibited VC-induced fibrosis. Blood urea nitrogen (BUN) and creatinine levels were increased after VC treatment. Furthermore, VC caused glomerulosclerosis and tubular injury in mouse kidney tissues. Kidney tissue sections showed that VC induced fibrosis and autophagy in mouse kidney tissues. In summary, the results of VC-induced fibrosis suggest that autophagy plays an important role in kidney damage. VC may cause nephrotoxicity, and the results illustrate the importance of considering the toxicological hazards of VC in kidney cells.
Collapse
Affiliation(s)
- Yung-Ho Hsu
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan.
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Hsiao-Chi Chuang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan.
- School of Public Health, College of Public Health, Taipei Medical University, Taipei 11031, Taiwan.
| | - Yu-Hsuan Lee
- Department of Food Safety/Hygiene &Risk Management, College of Medicine, National Cheng Kung University, Tainan 70430, Taiwan.
| | - Yuh-Feng Lin
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan.
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Yu-Jhe Chiu
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan.
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Yung-Li Wang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Mai-Szu Wu
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan.
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Hui-Wen Chiu
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan.
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| |
Collapse
|
52
|
Zakiyanov O, Kalousová M, Zima T, Tesař V. Matrix Metalloproteinases in Renal Diseases: A Critical Appraisal. Kidney Blood Press Res 2019; 44:298-330. [PMID: 31185475 DOI: 10.1159/000499876] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 03/10/2019] [Indexed: 11/19/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are endopeptidases within the metzincin protein family that not only cleave extracellular matrix (ECM) components, but also process the non-ECM molecules, including various growth factors and their binding proteins. MMPs participate in cell to ECM interactions, and MMPs are known to be involved in cell proliferation mechanisms and most probably apoptosis. These proteinases are grouped into six classes: collagenases, gelatinases, stromelysins, matrilysins, membrane type MMPs, and other MMPs. Various mechanisms regulate the activity of MMPs, inhibition by tissue inhibitors of metalloproteinases being the most important. In the kidney, intrinsic glomerular cells and tubular epithelial cells synthesize several MMPs. The measurement of circulating MMPs can provide valuable information in patients with kidney diseases. They play an important role in many renal diseases, both acute and chronic. This review attempts to summarize the current knowledge of MMPs in the kidney and discusses recent data from patient and animal studies with reference to specific diseases. A better understanding of the MMPs' role in renal remodeling may open the way to new interventions favoring deleterious renal changes in a number of kidney diseases.
Collapse
Affiliation(s)
- Oskar Zakiyanov
- Department of Nephrology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia,
| | - Marta Kalousová
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| | - Tomáš Zima
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| | - Vladimír Tesař
- Department of Nephrology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| |
Collapse
|
53
|
Park SM, Jang HJ, Lee JH. Roles of Primary Cilia in the Developing Brain. Front Cell Neurosci 2019; 13:218. [PMID: 31139054 PMCID: PMC6527876 DOI: 10.3389/fncel.2019.00218] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 04/30/2019] [Indexed: 01/07/2023] Open
Abstract
Essential to development, primary cilia are microtubule-based cellular organelles that protrude from the surface of cells. Acting as cellular antenna, primary cilia play central roles in transducing or regulating several signaling pathways, including Sonic hedgehog (Shh) and Wnt signaling. Defects in primary cilia contribute to a group of syndromic disorders known as “ciliopathies” and can adversely affect development of the brain and other essential organs, including the kidneys, eyes, and liver. The molecular mechanisms of how defective primary cilia contribute to neurological defects, however, remain poorly understood. In this mini review, we summarize recent advances in understanding of the interactions between primary cilia and signaling pathways essential to cellular homeostasis and brain development.
Collapse
Affiliation(s)
- Sang Min Park
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Hee Jin Jang
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Jeong Ho Lee
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology, Daejeon, South Korea.,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| |
Collapse
|
54
|
Chou LF, Cheng YL, Hsieh CY, Lin CY, Yang HY, Chen YC, Hung CC, Tian YC, Yang CW, Chang MY. Effect of Trehalose Supplementation on Autophagy and Cystogenesis in a Mouse Model of Polycystic Kidney Disease. Nutrients 2018; 11:nu11010042. [PMID: 30585217 PMCID: PMC6356442 DOI: 10.3390/nu11010042] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 12/15/2018] [Accepted: 12/20/2018] [Indexed: 12/28/2022] Open
Abstract
Autophagy impairment has been demonstrated in the pathogenesis of autosomal dominant polycystic kidney disease (ADPKD) and could be a new target of treatment. Trehalose is a natural, nonreducing disaccharide that has been shown to enhance autophagy. Therefore, we investigated whether trehalose treatment reduces renal cyst formation in a Pkd1-hypomorphic mouse model. Pkd1 miRNA transgenic (Pkd1 miR Tg) mice and wild-type littermates were given drinking water supplemented with 2% trehalose from postnatal day 35 to postnatal day 91. The control groups received pure water or 2% sucrose for the control of hyperosmolarity. The effect on kidney weights, cystic indices, renal function, cell proliferation, and autophagic activities was determined. We found that Pkd1 miR Tg mice had a significantly lower renal mRNA expression of autophagy-related genes, including atg5, atg12, ulk1, beclin1, and p62, compared with wild-type control mice. Furthermore, immunohistochemical analysis showed that cystic lining cells had strong positive staining for the p62 protein, indicating impaired degradation of the protein by the autophagy-lysosome pathway. However, trehalose treatment did not improve reduced autophagy activities, nor did it reduce relative kidney weights, plasma blood urea nitrogen levels, or cystatin C levels in Pkd1 miR Tg mice. Histomorphological analysis revealed no significant differences in the renal cyst index, fibrosis score, or proliferative score among trehalose-, sucrose-, and water-treated groups. Our results demonstrate that adding trehalose to drinking water does not modulate autophagy activities and renal cystogenesis in Pkd1-deficient mice, suggesting that an oral supplement of trehalose may not affect the progression of ADPKD.
Collapse
Affiliation(s)
- Li-Fang Chou
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | - Ya-Lien Cheng
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | - Chun-Yih Hsieh
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | - Chan-Yu Lin
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | - Huang-Yu Yang
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | - Yung-Chang Chen
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | - Cheng-Chieh Hung
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | - Ya-Chung Tian
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | - Chih-Wei Yang
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | - Ming-Yang Chang
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| |
Collapse
|
55
|
Weimbs T, Shillingford JM, Torres J, Kruger SL, Bourgeois BC. Emerging targeted strategies for the treatment of autosomal dominant polycystic kidney disease. Clin Kidney J 2018; 11:i27-i38. [PMID: 30581563 PMCID: PMC6295603 DOI: 10.1093/ckj/sfy089] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 08/27/2018] [Indexed: 12/25/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a widespread genetic disease that leads to renal failure in the majority of patients. The very first pharmacological treatment, tolvaptan, received Food and Drug Administration approval in 2018 after previous approval in Europe and other countries. However, tolvaptan is moderately effective and may negatively impact a patient's quality of life due to potentially significant side effects. Additional and improved therapies are still urgently needed, and several clinical trials are underway, which are discussed in the companion paper Müller and Benzing (Management of autosomal-dominant polycystic kidney disease-state-of-the-art) Clin Kidney J 2018; 11: i2-i13. Here, we discuss new therapeutic avenues that are currently being investigated at the preclinical stage. We focus on mammalian target of rapamycin and dual kinase inhibitors, compounds that target inflammation and histone deacetylases, RNA-targeted therapeutic strategies, glucosylceramide synthase inhibitors, compounds that affect the metabolism of renal cysts and dietary restriction. We discuss tissue targeting to renal cysts of small molecules via the folate receptor, and of monoclonal antibodies via the polymeric immunoglobulin receptor. A general problem with potential pharmacological approaches is that the many molecular targets that have been implicated in ADPKD are all widely expressed and carry out important functions in many organs and tissues. Because ADPKD is a slowly progressing, chronic disease, it is likely that any therapy will have to continue over years and decades. Therefore, systemically distributed drugs are likely to lead to potentially prohibitive extra-renal side effects during extended treatment. Tissue targeting to renal cysts of such drugs is one potential way around this problem. The use of dietary, instead of pharmacological, interventions is another.
Collapse
Affiliation(s)
- Thomas Weimbs
- Department of Molecular, Cellular, and Developmental Biology; and Neuroscience Research Institute, University of California, Santa Barbara, CA, USA
| | - Jonathan M Shillingford
- Department of Internal Medicine, Division of Nephrology, University of Michigan, Ann Arbor, MI, USA
| | - Jacob Torres
- Department of Molecular, Cellular, and Developmental Biology; and Neuroscience Research Institute, University of California, Santa Barbara, CA, USA
| | - Samantha L Kruger
- Department of Molecular, Cellular, and Developmental Biology; and Neuroscience Research Institute, University of California, Santa Barbara, CA, USA
| | - Bryan C Bourgeois
- Department of Molecular, Cellular, and Developmental Biology; and Neuroscience Research Institute, University of California, Santa Barbara, CA, USA
| |
Collapse
|
56
|
Neckář J, Hsu A, Hye Khan MA, Gross GJ, Nithipatikom K, Cyprová M, Benák D, Hlaváčková M, Sotáková-Kašparová D, Falck JR, Sedmera D, Kolář F, Imig JD. Infarct size-limiting effect of epoxyeicosatrienoic acid analog EET-B is mediated by hypoxia-inducible factor-1α via downregulation of prolyl hydroxylase 3. Am J Physiol Heart Circ Physiol 2018; 315:H1148-H1158. [PMID: 30074840 PMCID: PMC6734065 DOI: 10.1152/ajpheart.00726.2017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 07/06/2018] [Accepted: 07/18/2018] [Indexed: 12/27/2022]
Abstract
Epoxyeicosatrienoic acids (EETs) decrease cardiac ischemia-reperfusion injury; however, the mechanism of their protective effect remains elusive. Here, we investigated the cardioprotective action of a novel EET analog, EET-B, in reperfusion and the role of hypoxia-inducible factor (HIF)-1α in such action of EET-B. Adult male rats were subjected to 30 min of left coronary artery occlusion followed by 2 h of reperfusion. Administration of 14,15-EET (2.5 mg/kg) or EET-B (2.5 mg/kg) 5 min before reperfusion reduced infarct size expressed as a percentage of the area at risk from 64.3 ± 1.3% in control to 42.6 ± 1.9% and 46.0 ± 1.6%, respectively, and their coadministration did not provide any stronger effect. The 14,15-EET antagonist 14,15-epoxyeicosa-5( Z)-enoic acid (2.5 mg/kg) inhibited the infarct size-limiting effect of EET-B (62.5 ± 1.1%). Similarly, the HIF-1α inhibitors 2-methoxyestradiol (2.5 mg/kg) and acriflavine (2 mg/kg) completely abolished the cardioprotective effect of EET-B. In a separate set of experiments, the immunoreactivity of HIF-1α and its degrading enzyme prolyl hydroxylase domain protein 3 (PHD3) were analyzed in the ischemic areas and nonischemic septa. At the end of ischemia, the HIF-1α immunogenic signal markedly increased in the ischemic area compared with the septum (10.31 ± 0.78% vs. 0.34 ± 0.08%). After 20 min and 2 h of reperfusion, HIF-1α immunoreactivity decreased to 2.40 ± 0.48% and 1.85 ± 0.43%, respectively, in the controls. EET-B blunted the decrease of HIF-1α immunoreactivity (7.80 ± 0.69% and 6.44 ± 1.37%, respectively) and significantly reduced PHD3 immunogenic signal in ischemic tissue after reperfusion. In conclusion, EET-B provides an infarct size-limiting effect at reperfusion that is mediated by HIF-1α and downregulation of its degrading enzyme PHD3. NEW & NOTEWORTHY The present study shows that EET-B is an effective agonistic 14,15-epoxyeicosatrienoic acid analog, and its administration before reperfusion markedly reduced myocardial infarction in rats. Most importantly, we demonstrate that increased hypoxia-inducible factor-1α levels play a role in cardioprotection mediated by EET-B in reperfusion likely by mechanisms including downregulation of the hypoxia-inducible factor -1α-degrading enzyme prolyl hydroxylase domain protein 3.
Collapse
MESH Headings
- 8,11,14-Eicosatrienoic Acid/analogs & derivatives
- 8,11,14-Eicosatrienoic Acid/pharmacology
- 8,11,14-Eicosatrienoic Acid/therapeutic use
- Animals
- Disease Models, Animal
- Down-Regulation
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Hypoxia-Inducible Factor-Proline Dioxygenases/genetics
- Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism
- Male
- Myocardial Infarction/enzymology
- Myocardial Infarction/pathology
- Myocardial Infarction/physiopathology
- Myocardial Infarction/prevention & control
- Myocardial Reperfusion Injury/enzymology
- Myocardial Reperfusion Injury/pathology
- Myocardial Reperfusion Injury/physiopathology
- Myocardial Reperfusion Injury/prevention & control
- Myocardium/enzymology
- Myocardium/pathology
- Proteolysis
- Rats, Sprague-Dawley
- Signal Transduction/drug effects
- Ventricular Function, Left/drug effects
- Ventricular Remodeling/drug effects
Collapse
Affiliation(s)
- Jan Neckář
- Department of Pharmacology and Toxicology, Medical College of Wisconsin , Milwaukee, Wisconsin
- Department of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences , Prague , Czech Republic
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine Physiology , Prague , Czech Republic
| | - Anna Hsu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Md Abdul Hye Khan
- Department of Pharmacology and Toxicology, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Garrett J Gross
- Department of Pharmacology and Toxicology, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Kasem Nithipatikom
- Department of Pharmacology and Toxicology, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Michaela Cyprová
- Department of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences , Prague , Czech Republic
| | - Daniel Benák
- Department of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences , Prague , Czech Republic
| | - Markéta Hlaváčková
- Department of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences , Prague , Czech Republic
| | - Dita Sotáková-Kašparová
- Department of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences , Prague , Czech Republic
| | - John R Falck
- Department of Biochemistry, University of Texas Southwestern Medical Center , Dallas, Texas
| | - David Sedmera
- Department of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences , Prague , Czech Republic
- Institute of Anatomy, First Faculty of Medicine, Charles University , Prague , Czech Republic
| | - František Kolář
- Department of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences , Prague , Czech Republic
| | - John D Imig
- Department of Pharmacology and Toxicology, Medical College of Wisconsin , Milwaukee, Wisconsin
| |
Collapse
|
57
|
Kraus A, Peters DJM, Klanke B, Weidemann A, Willam C, Schley G, Kunzelmann K, Eckardt KU, Buchholz B. HIF-1α promotes cyst progression in a mouse model of autosomal dominant polycystic kidney disease. Kidney Int 2018; 94:887-899. [PMID: 30173898 DOI: 10.1016/j.kint.2018.06.008] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 06/01/2018] [Accepted: 06/07/2018] [Indexed: 11/29/2022]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is mainly caused by mutations of the PKD1 gene and characterized by growth of bilateral renal cysts. Cyst growth is accompanied by regional hypoxia and induction of hypoxia-inducible factor (HIF)-1α in cyst-lining epithelial cells. To determine the relevance of HIF-1α for cyst growth in vivo we used an inducible kidney epithelium-specific knockout mouse to delete Pkd1 at postnatal day 20 or 35 to induce polycystic kidney disease of different severity and analyzed the effects of Hif-1α co-deletion and HIF-1α stabilization using a prolyl-hydroxylase inhibitor. HIF-1α expression was enhanced in kidneys with progressive cyst growth induced by early Pkd1 deletion, but unchanged in the milder phenotype induced by later Pkd1 deletion. Hif-1α co-deletion significantly attenuated cyst growth in the severe, but not in the mild, phenotype. Application of a prolyl-hydroxylase inhibitor resulted in severe aggravation of the mild phenotype with rapid loss of renal function. HIF-1α expression was associated with induction of genes that mediate calcium-activated chloride secretion. Thus, HIF-1α does not seem to play a role in early cyst formation, but accelerates cyst growth during progressive polycystic kidney disease. This novel mechanism of cyst growth may qualify as a therapeutic target.
Collapse
Affiliation(s)
- Andre Kraus
- Department of Nephrology and Hypertension, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Dorien J M Peters
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Bernd Klanke
- Department of Nephrology and Hypertension, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Alexander Weidemann
- Department of Nephrology and Hypertension, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Carsten Willam
- Department of Nephrology and Hypertension, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Gunnar Schley
- Department of Nephrology and Hypertension, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Karl Kunzelmann
- Department of Physiology, University of Regensburg, Regensburg, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Hypertension, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Bjoern Buchholz
- Department of Nephrology and Hypertension, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
58
|
Shi W, Xu D, Gu J, Xue C, Yang B, Fu L, Song S, Liu D, Zhou W, Lv J, Sun K, Chen M, Mei C. Saikosaponin-d inhibits proliferation by up-regulating autophagy via the CaMKKβ-AMPK-mTOR pathway in ADPKD cells. Mol Cell Biochem 2018; 449:219-226. [PMID: 29675630 DOI: 10.1007/s11010-018-3358-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 04/16/2018] [Indexed: 02/03/2023]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a common heritable human disease. Recently, the role of repressed autophagy in ADPKD has drawn increasing attention. Here, we investigate the mechanism underlying the effect of Saikosaponin-d (SSd), a sarcoplasmic/endoplasmic reticulum Ca2+ ATPase pump (SERCA) inhibitor. We show that SSd suppresses proliferation in ADPKD cells by up-regulating autophagy. We found that treatment with SSd results in the accumulation of intracellular calcium, which in turn activates the CaMKKβ-AMPK signalling cascade, inhibits mTOR signalling and induces autophagy. Conversely, we also found that treatment with an autophagy inhibitor (3-methyladenine), AMPK inhibitor (Compound C), CaMKKβ inhibitor (STO-609) and intracellular calcium chelator (BAPTA/AM) could reduce autophagy puncta formation mediated by SSd. Our results demonstrated that SSd induces autophagy through the CaMKKβ-AMPK-mTOR signalling pathway in ADPKD cells, indicating that SSd might be a potential therapy for ADPKD and that SERCA might be a new target for ADPKD treatment.
Collapse
Affiliation(s)
- Weiwei Shi
- Kidney Institute of PLA, Department of Nephrology, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, People's Republic of China
| | - Dechao Xu
- Kidney Institute of PLA, Department of Nephrology, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, People's Republic of China
| | - Junhui Gu
- Kidney Institute of PLA, Department of Nephrology, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, People's Republic of China
| | - Cheng Xue
- Kidney Institute of PLA, Department of Nephrology, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, People's Republic of China
| | - Bo Yang
- Kidney Institute of PLA, Department of Nephrology, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, People's Republic of China
| | - Lili Fu
- Kidney Institute of PLA, Department of Nephrology, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, People's Republic of China
| | - Shuwei Song
- Kidney Institute of PLA, Department of Nephrology, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, People's Republic of China
| | - Dongmei Liu
- Kidney Institute of PLA, Department of Nephrology, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, People's Republic of China
| | - Wei Zhou
- Kidney Institute of PLA, Department of Nephrology, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, People's Republic of China
| | - Jiayi Lv
- Kidney Institute of PLA, Department of Nephrology, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, People's Republic of China
| | - Ke Sun
- Kidney Institute of PLA, Department of Nephrology, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, People's Republic of China
| | - Meihan Chen
- Kidney Institute of PLA, Department of Nephrology, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, People's Republic of China
| | - Changlin Mei
- Kidney Institute of PLA, Department of Nephrology, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, People's Republic of China.
| |
Collapse
|
59
|
Ow CPC, Ngo JP, Ullah MM, Hilliard LM, Evans RG. Renal hypoxia in kidney disease: Cause or consequence? Acta Physiol (Oxf) 2018; 222:e12999. [PMID: 29159875 DOI: 10.1111/apha.12999] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 11/10/2017] [Accepted: 11/15/2017] [Indexed: 02/06/2023]
Abstract
Tissue hypoxia has been proposed as an important factor in the pathophysiology of both chronic kidney disease (CKD) and acute kidney injury (AKI), initiating and propagating a vicious cycle of tubular injury, vascular rarefaction, and fibrosis and thus exacerbation of hypoxia. Here, we critically evaluate this proposition by systematically reviewing the literature relevant to the following six questions: (i) Is kidney disease always associated with tissue hypoxia? (ii) Does tissue hypoxia drive signalling cascades that lead to tissue damage and dysfunction? (iii) Does tissue hypoxia per se lead to kidney disease? (iv) Does tissue hypoxia precede pathology? (v) Does tissue hypoxia colocalize with pathology? (vi) Does prevention of tissue hypoxia prevent kidney disease? We conclude that tissue hypoxia is a common feature of both AKI and CKD. Furthermore, at least under in vitro conditions, renal tissue hypoxia drives signalling cascades that lead to tissue damage and dysfunction. Tissue hypoxia itself can lead to renal pathology, independent of other known risk factors for kidney disease. There is also some evidence that tissue hypoxia precedes renal pathology, at least in some forms of kidney disease. However, we have made relatively little progress in determining the spatial relationships between tissue hypoxia and pathological processes (i.e. colocalization) or whether therapies targeted to reduce tissue hypoxia can prevent or delay the progression of renal disease. Thus, the hypothesis that tissue hypoxia is a "common pathway" to both AKI and CKD still remains to be adequately tested.
Collapse
Affiliation(s)
- C. P. C. Ow
- Cardiovascular Disease Program Biomedicine Discovery Institute and Department of Physiology Monash University Melbourne Vic. Australia
| | - J. P. Ngo
- Cardiovascular Disease Program Biomedicine Discovery Institute and Department of Physiology Monash University Melbourne Vic. Australia
| | - M. M. Ullah
- Cardiovascular Disease Program Biomedicine Discovery Institute and Department of Physiology Monash University Melbourne Vic. Australia
| | - L. M. Hilliard
- Cardiovascular Disease Program Biomedicine Discovery Institute and Department of Physiology Monash University Melbourne Vic. Australia
| | - R. G. Evans
- Cardiovascular Disease Program Biomedicine Discovery Institute and Department of Physiology Monash University Melbourne Vic. Australia
| |
Collapse
|
60
|
Masyuk AI, Masyuk TV, Lorenzo Pisarello MJ, Ding JF, Loarca L, Huang BQ, LaRusso NF. Cholangiocyte autophagy contributes to hepatic cystogenesis in polycystic liver disease and represents a potential therapeutic target. Hepatology 2018; 67:1088-1108. [PMID: 29023824 PMCID: PMC5826832 DOI: 10.1002/hep.29577] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 09/11/2017] [Accepted: 10/03/2017] [Indexed: 12/24/2022]
Abstract
UNLABELLED Polycystic liver disease (PLD) is a group of genetic disorders with limited treatment options and significant morbidity. Hepatic cysts arise from cholangiocytes exhibiting a hyperproliferative phenotype. Considering that hyperproliferation of many cell types is associated with alterations in autophagy, we hypothesized that autophagy is altered in PLD cholangiocytes, contributes to hepatic cystogenesis, and might represent a potential therapeutic target. We employed functional pathway cluster analysis and next-generation sequencing, transmission electron microscopy, immunofluorescence confocal microscopy, and western blotting to assess autophagy in human and rodent PLD cholangiocytes. A three-dimensional culture model was used to study the effects of molecular and pharmacologic inhibition of autophagy on hepatic cystogenesis in vitro, and the polycystic kidney disease-specific rat, an animal model of PLD, to study the effects of hydroxychloroquine, a drug that interferes with the autophagy pathway, on disease progression in vivo. Assessment of the transcriptome of PLD cholangiocytes followed by functional pathway cluster analysis revealed that the autophagy-lysosomal pathway is one of the most altered pathways in PLD. Direct evaluation of autophagy in PLD cholangiocytes both in vitro and in vivo showed increased number and size of autophagosomes, lysosomes, and autolysosomes; overexpression of autophagy-related proteins (Atg5, Beclin1, Atg7, and LC3); and enhanced autophagic flux associated with activation of the cAMP-protein kinase A-cAMP response element-binding protein signaling pathway. Molecular and pharmacologic intervention in autophagy with ATG7 small interfering RNA, bafilomycin A1 , and hydroxychloroquine reduced proliferation of PLD cholangiocytes in vitro and growth of hepatic cysts in three-dimensional cultures. Hydroxychloroquine also efficiently inhibited hepatic cystogenesis in the polycystic kidney disease-specific rat. CONCLUSION Autophagy is increased in PLD cholangiocytes, contributes to hepatic cystogenesis, and represents a potential therapeutic target for disease treatment. (Hepatology 2018;67:1088-1108).
Collapse
Affiliation(s)
- Anatoliy I Masyuk
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, Rochester, MN
| | - Tatyana V Masyuk
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, Rochester, MN
| | - Maria J Lorenzo Pisarello
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, Rochester, MN
| | - Jingyi Francess Ding
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, Rochester, MN
| | - Lorena Loarca
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, Rochester, MN
| | - Bing Q Huang
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, Rochester, MN
| | - Nicholas F LaRusso
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, Rochester, MN
| |
Collapse
|
61
|
Nag S, Resnick A. Stabilization of hypoxia inducible factor by cobalt chloride can alter renal epithelial transport. Physiol Rep 2017; 5:5/24/e13531. [PMID: 29263117 PMCID: PMC5742700 DOI: 10.14814/phy2.13531] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 10/27/2017] [Accepted: 10/31/2017] [Indexed: 01/10/2023] Open
Abstract
Given the importance of the transcriptional regulator hypoxia-inducible factor-1 (HIF-1) for adaptive hypoxia responses, we examined the effect of stabilized HIF-1α on renal epithelial permeability and directed sodium transport. This study was motivated by histological analysis of cystic kidneys showing increased expression levels of HIF-1α and HIF-2α We hypothesize that compression induced localized ischemia-hypoxia of normal epithelia near a cyst leads to local stabilization of HIF-1α, leading to altered transepithelial transport that encourages cyst expansion. We found that stabilized HIF-1α alters both transcellular and paracellular transport through renal epithelial monolayers in a manner consistent with secretory behavior, indicating localized ischemia-hypoxia may lead to altered salt and water transport through kidney epithelial monolayers. A quantity of 100 μmol/L Cobalt chloride (CoCl2) was used acutely to stabilize HIF-1α in confluent cultures of mouse renal epithelia. We measured increased transepithelial permeability and decreased transepithelial resistance (TER) when HIF-1α was stabilized. Most interestingly, we measured a change in the direction of sodium current, most likely corresponding to abnormal secretory function, supporting our positive-feedback hypothesis.
Collapse
Affiliation(s)
- Subhra Nag
- Department of Biology, Geology and Environmental Sciences, Cleveland State University, Cleveland, Ohio
| | - Andrew Resnick
- Department of Biology, Geology and Environmental Sciences, Cleveland State University, Cleveland, Ohio .,Department of Physics Cleveland State University, Cleveland, Ohio.,Center for Gene Regulation in Health and Disease, Cleveland State University, Cleveland, Ohio
| |
Collapse
|
62
|
Zhu P, Sieben CJ, Xu X, Harris PC, Lin X. Autophagy activators suppress cystogenesis in an autosomal dominant polycystic kidney disease model. Hum Mol Genet 2017; 26:158-172. [PMID: 28007903 DOI: 10.1093/hmg/ddw376] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 10/27/2016] [Indexed: 01/08/2023] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is caused by mutations in either PKD1 or PKD2. It is one of the most common heritable human diseases with eventual development of renal failure; however, effective treatment is lacking. While inhibition of mechanistic target of rapamycin (mTOR) effectively slows cyst expansions in animal models, results from clinical studies are controversial, prompting further mechanistic studies of mTOR-based therapy. Here, we aim to establish autophagy, a downstream pathway of mTOR, as a new therapeutic target for PKD. We generated zebrafish mutants for pkd1 and noted cystic kidney and mTOR activation in pkd1a mutants, suggesting a conserved ADPKD model. Further assessment of the mutants revealed impaired autophagic flux, which was conserved in kidney epithelial cells derived from both Pkd1-null mice and ADPKD patients. We found that inhibition of autophagy by knocking down the core autophagy protein Atg5 promotes cystogenesis, while activation of autophagy using a specific inducer Beclin-1 peptide ameliorates cysts in the pkd1a model. Treatment with compound autophagy activators, including mTOR-dependent rapamycin as well as mTOR-independent carbamazepine and minoxidil, markedly attenuated cyst formation and restored kidney function. Finally, we showed that combination treatment with low doses of rapamycin and carbamazepine was able to attenuate cyst formation as effectively as a single treatment with a high dose of rapamycin alone. In summary, our results suggested a modifying effect of autophagy on ADPKD, established autophagy activation as a novel therapy for ADPKD, and presented zebrafish as an efficient vertebrate model for developing PKD therapeutic strategies.
Collapse
Affiliation(s)
- Ping Zhu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Cynthia J Sieben
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Xiaolei Xu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA.,Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| | - Peter C Harris
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Xueying Lin
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
63
|
New Roles of the Primary Cilium in Autophagy. BIOMED RESEARCH INTERNATIONAL 2017; 2017:4367019. [PMID: 28913352 PMCID: PMC5587941 DOI: 10.1155/2017/4367019] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 07/03/2017] [Indexed: 12/21/2022]
Abstract
The primary cilium is a nonmotile organelle that emanates from the surface of multiple cell types and receives signals from the environment to regulate intracellular signaling pathways. The presence of cilia, as well as their length, is important for proper cell function; shortened, elongated, or absent cilia are associated with pathological conditions. Interestingly, it has recently been shown that the molecular machinery involved in autophagy, the process of recycling of intracellular material to maintain cellular and tissue homeostasis, participates in ciliogenesis. Cilium-dependent signaling is necessary for autophagosome formation and, conversely, autophagy regulates both ciliogenesis and cilium length by degrading specific ciliary proteins. Here, we will discuss the relationship that exists between the two processes at the cellular and molecular level, highlighting what is known about the effects of ciliary dysfunction in the control of energy homeostasis in some ciliopathies.
Collapse
|
64
|
Abstract
Autophagy is a highly conserved, physiological, catabolic process, involving the lysosomal degradation of cytosolic components, including macromolecules (such as proteins and lipids) and cytosolic organelles. Autophagy is believed to be essential for the maintenance of cellular homeostasis, for a number of fundamental biological activities, and an important component of the complex response of cells to multiple forms of stress. Autophagy is involved in the pathogenesis of a number of clinically important disorders but, until recently, little was known about its connection to kidney diseases. However, there is now growing evidence that autophagy is specifically linked to the pathogenesis of important renal diseases such as acute kidney injury, diabetic nephropathy and polycystic kidney disease. However, an understanding of the precise role of autophagy in the course of kidney diseases is still in its infancy. The review points out areas of particular interest for future research, and also discusses the importance of such information on whether the pharmacologic agents that modulate autophagy are potentially usable as novel forms of treatment for various kidney diseases.
Collapse
Affiliation(s)
- Nicolas Pallet
- Inserm U1147, Université Paris Descartes, 45, rue des Saints Pères, 75006 Paris, France
| |
Collapse
|
65
|
da Costa NMM, de Siqueira AS, Ribeiro ALR, da Silva Kataoka MS, Jaeger RG, de Alves-Júnior SM, Smith AM, de Jesus Viana Pinheiro J. Role of HIF-1α and CASPASE-3 in cystogenesis of odontogenic cysts and tumors. Clin Oral Investig 2017; 22:141-149. [PMID: 28238093 DOI: 10.1007/s00784-017-2090-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 02/20/2017] [Indexed: 12/19/2022]
Abstract
OBJECTIVES Odontogenic cysts and tumors are the most relevant lesions that affect the gnathic bones. These lesions have in common the formation of cystic areas and this common feature may suggest involvement of similar mechanisms. The hypoxia inducible factor 1 alpha (HIF-1α), a responsive protein to hypoxia and caspase-3, an irreversible apoptosis marker, may contribute to cyst formation. Thus, this study aimed to investigate the immunoexpression of these proteins in odontogenic cysts and tumors. MATERIAL AND METHODS Twenty cases of ameloblastoma, keratocystic odontogenic tumor (KOT) (n = 20), radicular cyst (RC) (n = 18), dentigerous cyst (DC) (n = 11), calcifying cystic odontogenic tumor (n = 8), and dental follicle (DF) (n = 10) were used to investigate HIF-1α and caspase-3 expression in sequential serial cuts by immunohistochemistry. RESULTS HIF-1α was overexpressed in RC, DC, and ameloblastoma when compared with DF. The basal and sometimes the lower suprabasal layer showed no or very low expression in DC, KOT, and ameloblastoma, the last also showing strong expression in solid epithelial areas and initial cystic formation regions. Caspase-3 was found to be overexpressed in all lesions, with the highest expression in odontogenic cysts compared to tumors. HIF-1α and caspase-3 were localized in similar areas of the same lesions, especially in the epithelium surrounding cystic formations. CONCLUSIONS This study showed distinct immunoexpression of HIF-1α and caspase-3 in odontogenic cyst and tumors, with higher expression observed in odontogenic cysts. CLINICAL RELEVANCE These findings suggest a possible correlation between hypoxia, apoptosis, and cystogenesis, leading to understand the mechanisms responsible to cystic formation in odontogenic lesions.
Collapse
Affiliation(s)
- Natacha M M da Costa
- Cell Culture Laboratory, School of Dentistry, Federal University of Pará, Rua Augusto Corrêa, 01, Guamá, Belém, PA, 66075110, Brazil
| | - Adriane S de Siqueira
- School of Dentistry, Universidade Positivo, Av. Prof. Pedro Viriato Parigot de Souza, 5300, Curitiba, PR, 80740-050, Brazil
| | - André L R Ribeiro
- Department of Oral and Maxillofacial Surgery, University Center of Para-CESUPA, Belem, Brazil
| | - Maria S da Silva Kataoka
- Cell Culture Laboratory, School of Dentistry, Federal University of Pará, Rua Augusto Corrêa, 01, Guamá, Belém, PA, 66075110, Brazil
| | - Ruy G Jaeger
- Department of Cell and Developmental Biology Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 1524, Ed. Biomédicas 1, São Paulo, SP, 05508-000, Brazil
| | - Sérgio M de Alves-Júnior
- Cell Culture Laboratory, School of Dentistry, Federal University of Pará, Rua Augusto Corrêa, 01, Guamá, Belém, PA, 66075110, Brazil
| | - Andrew M Smith
- Department of Microbial Diseases, Eastman Dental Institute, University College London, London, UK.,The Rayne Building, Centre for Molecular Medicine, 5 University Street, London, WC1E 6JF, UK
| | - João de Jesus Viana Pinheiro
- Cell Culture Laboratory, School of Dentistry, Federal University of Pará, Rua Augusto Corrêa, 01, Guamá, Belém, PA, 66075110, Brazil.
| |
Collapse
|
66
|
Autophagy Protects against Palmitic Acid-Induced Apoptosis in Podocytes in vitro. Sci Rep 2017; 7:42764. [PMID: 28225005 PMCID: PMC5320537 DOI: 10.1038/srep42764] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 01/13/2017] [Indexed: 01/01/2023] Open
Abstract
Autophagy is a highly conserved degradation process that is involved in the clearance of proteins and damaged organelles to maintain intracellular homeostasis and cell integrity. Type 2 diabetes is often accompanied by dyslipidemia with elevated levels of free fatty acids (FFAs). Podocytes, as an important component of the filtration barrier, are susceptible to lipid disorders. The loss of podocytes causes proteinuria, which is involved in the pathogenesis of diabetic nephropathy. In the present study, we demonstrated that palmitic acid (PA) promoted autophagy in podocytes. We further found that PA increased the production of reactive oxygen species (ROS) in podocytes and that NAC (N-acetyl-cysteine), a potent antioxidant, significantly eliminated the excessive ROS and suppressed autophagy, indicating that the increased generation of ROS was associated with the palmitic acid-induced autophagy in podocytes. Moreover, we also found that PA stimulation decreased the mitochondrial membrane potential in podocytes and induced podocyte apoptosis, while the inhibition of autophagy by chloroquine (CQ) enhanced palmitic acid-induced apoptosis accompanied by increased ROS generation, and the stimulation of autophagy by rapamycin (Rap) remarkably suppressed palmitic acid-induced ROS generation and apoptosis. Taken together, these in vitro findings suggest that PA-induced autophagy in podocytes is mediated by ROS production and that autophagy plays a protective role against PA-induced podocyte apoptosis.
Collapse
|
67
|
Tanaka Y, Watari M, Saito T, Morishita Y, Ishibashi K. Enhanced Autophagy in Polycystic Kidneys of AQP11 Null Mice. Int J Mol Sci 2016; 17:ijms17121993. [PMID: 27916883 PMCID: PMC5187793 DOI: 10.3390/ijms17121993] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 11/14/2016] [Accepted: 11/16/2016] [Indexed: 11/28/2022] Open
Abstract
Aquaporin-11 (AQP11) is an intracellular water channel expressed at the endoplasmic reticulum (ER) of the proximal tubule. Its gene disruption in mice leads to intracellular vacuole formation at one week and the subsequent development of polycystic kidneys by three weeks. As the damaged proximal tubular cells with intracellular vacuoles form cysts later, we postulated that autophagy may play a role in the cyst formation and examined autophagy activity before and after cyst development in AQP11(−/−) kidneys. PCR analysis showed the increased expression of the transcript encoding LC3 (Map1lc3b) as well as other autophagy-related genes in AQP11(−/−) mice. Using green fluorescent protein (GFP)-LC3 transgenic mice and AQP11(−/−) mice, we found that the number of GFP-LC3–positive puncta was increased in the proximal tubule of AQP11(−/−) mice before the cyst formation. Interestingly, they were also observed in the cyst-lining epithelial cell. Further PCR analyses revealed the enhanced expression of apoptosis-related and ER stress–related caspase genes before and after the cyst formation, which may cause the enhanced autophagy. These results suggest the involvement of autophagy in the development and maintenance of kidney cysts in AQP11(−/−) mice.
Collapse
Affiliation(s)
- Yasuko Tanaka
- Department of Pathophysiology, Faculty of Pharmacy, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo 204-8588, Japan.
| | - Mayumi Watari
- Department of Pathophysiology, Faculty of Pharmacy, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo 204-8588, Japan.
| | - Tatsuya Saito
- Department of Pathophysiology, Faculty of Pharmacy, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo 204-8588, Japan.
| | - Yoshiyuki Morishita
- Department of Nephrology, Saitama Medical Center, Jichi Medical University, 1-847 Ohmiya, Saitama-City, Saitama 330-8503, Japan.
| | - Kenichi Ishibashi
- Department of Pathophysiology, Faculty of Pharmacy, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo 204-8588, Japan.
| |
Collapse
|
68
|
Autophagy in kidney disease and aging: lessons from rodent models. Kidney Int 2016; 90:950-964. [DOI: 10.1016/j.kint.2016.04.014] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Revised: 04/17/2016] [Accepted: 04/20/2016] [Indexed: 12/14/2022]
|
69
|
Kurbegovic A, Trudel M. Acute kidney injury induces hallmarks of polycystic kidney disease. Am J Physiol Renal Physiol 2016; 311:F740-F751. [DOI: 10.1152/ajprenal.00167.2016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 07/26/2016] [Indexed: 12/13/2022] Open
Abstract
Acute kidney injury (AKI) and autosomal dominant polycystic kidney disease (ADPKD) are considered separate entities that both frequently cause renal failure. Since ADPKD appears to depend on a polycystin-1 (Pc1) or Pc2 dosage mechanism, we investigated whether slow progression of cystogenesis in two Pkd1 transgenic mouse models can be accelerated with moderate ischemia-reperfusion injury (IRI). Transient unilateral left ischemic kidneys in both nontransgenic and transgenic mice reproducibly develop tubular dilatations, cysts, and typical PKD cellular defects within 3 mo post-IRI. Similar onset and severity of IRI induced-cystogenesis independently of genotype revealed that IRI is sufficient to promote renal cyst formation; however, this response was not further amplified by the transgene in Pkd1 mouse models. The IRI nontransgenic and transgenic kidneys showed from 16 days post-IRI strikingly increased and sustained Pkd1/Pc1 (>3-fold) and Pc2 (>8-fold) expression that can individually be cystogenic in mice. In parallel, long-term and important stimulation of hypoxia-inducible factor 1α expression was induced as in polycystic kidney disease. While mammalian target of rapamycin signaling is activated, stimulation of the Wnt pathway, with markedly increased active β-catenin and c-Myc expression in IRI renal epithelium, uncovered a similar regulatory cystogenic response shared by IRI and ADPKD. Our study demonstrates that long-term AKI induces cystogenesis and cross talk with ADPKD Pc1/Pc2 pathogenic signaling.
Collapse
Affiliation(s)
- Almira Kurbegovic
- Molecular Genetics and Development, Institut de Recherches Cliniques de Montréal, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Marie Trudel
- Molecular Genetics and Development, Institut de Recherches Cliniques de Montréal, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
70
|
Lin YF, Chiu IJ, Cheng FY, Lee YH, Wang YJ, Hsu YH, Chiu HW. The role of hypoxia-inducible factor-1α in zinc oxide nanoparticle-induced nephrotoxicity in vitro and in vivo. Part Fibre Toxicol 2016; 13:52. [PMID: 27678081 PMCID: PMC5037597 DOI: 10.1186/s12989-016-0163-3] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 09/20/2016] [Indexed: 12/01/2022] Open
Abstract
Background Zinc oxide nanoparticles (ZnO NPs) are used in an increasing number of products, including rubber manufacture, cosmetics, pigments, food additives, medicine, chemical fibers and electronics. However, the molecular mechanisms underlying ZnO NP nephrotoxicity remain unclear. In this study, we evaluated the potential toxicity of ZnO NPs in kidney cells in vitro and in vivo. Results We found that ZnO NPs were apparently engulfed by the HEK-293 human embryonic kidney cells and then induced reactive oxygen species (ROS) generation. Furthermore, exposure to ZnO NPs led to a reduction in cell viability and induction of apoptosis and autophagy. Interestingly, the ROS-induced hypoxia-inducible factor-1α (HIF-1α) signaling pathway was significantly increased following ZnO NPs exposure. Additionally, connective tissue growth factor (CTGF) and plasminogen activator inhibitor-1 (PAI-1), which are directly regulated by HIF-1 and are involved in the pathogenesis of kidney diseases, displayed significantly increased levels following ZnO NPs exposure in HEK-293 cells. HIF-1α knockdown resulted in significantly decreased levels of autophagy and increased cytotoxicity. Therefore, our results suggest that HIF-1α may have a protective role in adaptation to the toxicity of ZnO NPs in kidney cells. In an animal study, fluorescent ZnO NPs were clearly observed in the liver, lungs, kidneys, spleen and heart. ZnO NPs caused histopathological lesions in the kidney and increase in serum creatinine and blood urea nitrogen (BUN) which indicate possible renal possible damage. Moreover, ZnO NPs enhanced the HIF-1α signaling pathway, apoptosis and autophagy in mouse kidney tissues. Conclusions ZnO NPs may cause nephrotoxicity, and the results demonstrate the importance of considering the toxicological hazards of ZnO NP production and application, especially for medicinal use. Electronic supplementary material The online version of this article (doi:10.1186/s12989-016-0163-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yuh-Feng Lin
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, 110, Taipei, Taiwan
| | - I-Jen Chiu
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan
| | - Fong-Yu Cheng
- Institute of Oral Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Hsuan Lee
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ying-Jan Wang
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Biomedical Informatics, Asia University, Taichung, Taiwan.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Yung-Ho Hsu
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan.,Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hui-Wen Chiu
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan. .,Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, 110, Taipei, Taiwan.
| |
Collapse
|
71
|
Lebovitz CB, Robertson AG, Goya R, Jones SJ, Morin RD, Marra MA, Gorski SM. Cross-cancer profiling of molecular alterations within the human autophagy interaction network. Autophagy 2016. [PMID: 26208877 PMCID: PMC4590660 DOI: 10.1080/15548627.2015.1067362] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Aberrant activation or disruption of autophagy promotes tumorigenesis in various preclinical models of cancer, but whether the autophagy pathway is a target for recurrent molecular alteration in human cancer patient samples is unknown. To address this outstanding question, we surveyed 211 human autophagy-associated genes for tumor-related alterations to DNA sequence and RNA expression levels and examined their association with patient survival outcomes in multiple cancer types with sequence data from The Cancer Genome Atlas consortium. We found 3 (RB1CC1/FIP200, ULK4, WDR45/WIPI4) and one (ATG7) core autophagy genes to be under positive selection for somatic mutations in endometrial carcinoma and clear cell renal carcinoma, respectively, while 29 autophagy regulators and pathway interactors, including previously identified KEAP1, NFE2L2, and MTOR, were significantly mutated in 6 of the 11 cancer types examined. Gene expression analyses revealed that GABARAPL1 and MAP1LC3C/LC3C transcripts were less abundant in breast cancer and non-small cell lung cancers than in matched normal tissue controls; ATG4D transcripts were increased in lung squamous cell carcinoma, as were ATG16L2 transcripts in kidney cancer. Unsupervised clustering of autophagy-associated mRNA levels in tumors stratified patient overall survival in 3 of 9 cancer types (acute myeloid leukemia, clear cell renal carcinoma, and head and neck cancer). These analyses provide the first comprehensive resource of recurrently altered autophagy-associated genes in human tumors, and highlight cancer types and subtypes where perturbed autophagy may be relevant to patient overall survival.
Collapse
Affiliation(s)
- Chandra B Lebovitz
- a The Genome Sciences Centre; BC Cancer Agency ; Vancouver, BC Canada.,b Department of Molecular Biology and Biochemistry ; Simon Fraser University ; Burnaby , BC Canada
| | | | - Rodrigo Goya
- a The Genome Sciences Centre; BC Cancer Agency ; Vancouver, BC Canada.,c Centre for High-Throughput Biology; University of British Columbia ; Vancouver , BC Canada
| | - Steven J Jones
- a The Genome Sciences Centre; BC Cancer Agency ; Vancouver, BC Canada.,b Department of Molecular Biology and Biochemistry ; Simon Fraser University ; Burnaby , BC Canada.,d Department of Medical Genetics ; University of British Columbia ; Vancouver , BC Canada
| | - Ryan D Morin
- a The Genome Sciences Centre; BC Cancer Agency ; Vancouver, BC Canada.,b Department of Molecular Biology and Biochemistry ; Simon Fraser University ; Burnaby , BC Canada
| | - Marco A Marra
- a The Genome Sciences Centre; BC Cancer Agency ; Vancouver, BC Canada.,d Department of Medical Genetics ; University of British Columbia ; Vancouver , BC Canada
| | - Sharon M Gorski
- a The Genome Sciences Centre; BC Cancer Agency ; Vancouver, BC Canada.,b Department of Molecular Biology and Biochemistry ; Simon Fraser University ; Burnaby , BC Canada
| |
Collapse
|
72
|
Abstract
Many common renal insults such as ischemia and toxic injury primarily target the tubular epithelial cells, especially the highly metabolically active proximal tubular segment. Tubular epithelial cells are particularly dependent on autophagy to maintain homeostasis and respond to stressors. The pattern of autophagy in the kidney has a unique spatial and chronologic signature. Recent evidence has shown that there is complex cross-talk between autophagy and various cell death pathways. This review specifically discusses the interplay between autophagy and cell death in the renal tubular epithelia. It is imperative to review this topic because recent discoveries have improved our mechanistic understanding of the autophagic process and have highlighted its broad clinical applications, making autophagy a major target for drug development.
Collapse
Affiliation(s)
- Andrea Havasi
- Department of Nephrology, Boston University Medical Center, Boston, MA.
| | - Zheng Dong
- Department of Nephrology, Second Xiangya Hospital of Central South University, Changsha, China; Department of Cellular Biology and Anatomy, Medical College of Georgia and Charlie Norwood VA Medical Center, Augusta, GA
| |
Collapse
|
73
|
De Rechter S, Decuypere JP, Ivanova E, van den Heuvel LP, De Smedt H, Levtchenko E, Mekahli D. Autophagy in renal diseases. Pediatr Nephrol 2016; 31:737-52. [PMID: 26141928 DOI: 10.1007/s00467-015-3134-2] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 05/14/2015] [Accepted: 05/20/2015] [Indexed: 10/23/2022]
Abstract
Autophagy is the cell biology process in which cytoplasmic components are degraded in lysosomes to maintain cellular homeostasis and energy production. In the healthy kidney, autophagy plays an important role in the homeostasis and viability of renal cells such as podocytes and tubular epithelial cells and of immune cells. Recently, evidence is mounting that (dys)regulation of autophagy is implicated in the pathogenesis of various renal diseases, and might be an attractive target for new renoprotective therapies. In this review, we provide an overview of the role of autophagy in kidney physiology and kidney diseases.
Collapse
Affiliation(s)
- Stéphanie De Rechter
- Department of Paediatric Nephrology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium. .,Laboratory of Paediatrics, KU Leuven, Leuven, Belgium.
| | - Jean-Paul Decuypere
- Laboratory of Abdominal Transplantation, Department of Microbiology and Immunology Biomedical Sciences Group, KU Leuven, Leuven, Belgium.,Department of Abdominal Transplant Surgery, University Hospitals Leuven, Leuven, Belgium
| | | | - Lambertus P van den Heuvel
- Laboratory of Paediatrics, KU Leuven, Leuven, Belgium.,Translational Metabolic Laboratory and Department of Paediatric Nephrology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Humbert De Smedt
- Laboratory of Molecular and Cellular Signalling, KU Leuven, Leuven, Belgium
| | - Elena Levtchenko
- Department of Paediatric Nephrology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium.,Laboratory of Paediatrics, KU Leuven, Leuven, Belgium
| | - Djalila Mekahli
- Department of Paediatric Nephrology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium.,Laboratory of Paediatrics, KU Leuven, Leuven, Belgium
| |
Collapse
|
74
|
Wang S, Livingston MJ, Su Y, Dong Z. Reciprocal regulation of cilia and autophagy via the MTOR and proteasome pathways. Autophagy 2016; 11:607-16. [PMID: 25906314 DOI: 10.1080/15548627.2015.1023983] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Primary cilium is an organelle that plays significant roles in a number of cellular functions ranging from cell mechanosensation, proliferation, and differentiation to apoptosis. Autophagy is an evolutionarily conserved cellular function in biology and indispensable for cellular homeostasis. Both cilia and autophagy have been linked to different types of genetic and acquired human diseases. Their interaction has been suggested very recently, but the underlying mechanisms are still not fully understood. We examined autophagy in cells with suppressed cilia and measured cilium length in autophagy-activated or -suppressed cells. It was found that autophagy was repressed in cells with short cilia. Further investigation showed that MTOR activation was enhanced in cilia-suppressed cells and the MTOR inhibitor rapamycin could largely reverse autophagy suppression. In human kidney proximal tubular cells (HK2), autophagy induction was associated with cilium elongation. Conversely, autophagy inhibition by 3-methyladenine (3-MA) and chloroquine (CQ) as well as bafilomycin A1 (Baf) led to short cilia. Cilia were also shorter in cultured atg5-knockout (KO) cells and in atg7-KO kidney proximal tubular cells in mice. MG132, an inhibitor of the proteasome, could significantly restore cilium length in atg5-KO cells, being concomitant with the proteasome activity. Together, the results suggest that cilia and autophagy regulate reciprocally through the MTOR signaling pathway and ubiquitin-proteasome system.
Collapse
Key Words
- 3-MA, 3-methyladenine
- 70kDa, polypeptide 1
- ANKS6, ankyrin repeat and sterile α motif domain containing 6
- ATG/atg, autophagy-related
- Ac-TUBA, acetylated-tubulin α
- Baf, bafilomycin A1
- CF, confluence
- CQ, chloroquine
- DAPI, 4′-6-diamidino-2-phenylindole
- FBS, fetal bovine serum
- HK2, human kidney proximal tubular cells
- IFT, intraflagellar transport
- KAP3, kinesin family-associated protein 3
- KD, knockdown
- KIF3A/3B, kinesin family member 3A/3B
- KO, knockout
- LTA, lotus tetragonolobus agglutinin
- MAP1LC3B/LC3B, microtubule-associated protein 1 light chain 3 β
- MEF, mouse embryonic fibroblast
- MTOR
- MTOR, mechanistic target of rapamycin
- OFD1, oral-ficial-digital syndrome 1
- PBS, phosphate-buffered saline
- PKD, polycystic kidney disease
- RKRB, Krebs-Henseleit saline containing 25 mM NaHCO3
- RPS6KB1, ribosomal protein S6 kinase
- Rapa, rapamycin
- SD, standard deviation
- autophagy
- cilia
- polycystic kidney disease
- proteasome
Collapse
Affiliation(s)
- Shixuan Wang
- a Department of Cellular Biology and Anatomy ; Medical College of Georgia; Georgia Reagents University and Charlie Norwood VA Medical Center ; Augusta , GA USA
| | | | | | | |
Collapse
|
75
|
Pampliega O, Cuervo AM. Autophagy and primary cilia: dual interplay. Curr Opin Cell Biol 2016; 39:1-7. [PMID: 26826446 DOI: 10.1016/j.ceb.2016.01.008] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 01/08/2016] [Accepted: 01/13/2016] [Indexed: 12/11/2022]
Abstract
Primary cilia are microtubule-based organelles for sensing of the extracellular milieu and transducing this information into the cell through a variety of molecular signaling pathways. Functioning of the primary cilium has been recently connected to autophagy, a pathway for degradation of cellular components in lysosomes. Autophagy regulates the length of the cilia by removing proteins required for ciliogenesis, a phenomenon that is molecularly different if performed by basal autophagy or when autophagy is induced in response to various stressors. Here we review the current knowledge about the dual interaction between autophagy and ciliogenesis, and discuss the potential role that deregulated ciliary autophagy could have in pathologies with alterations in autophagy and ciliogenesis.
Collapse
Affiliation(s)
- Olatz Pampliega
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France.
| | - Ana Maria Cuervo
- Department of Developmental and Molecular Biology, Institute for Aging Studies, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| |
Collapse
|
76
|
Seeger-Nukpezah T, Geynisman DM, Nikonova AS, Benzing T, Golemis EA. The hallmarks of cancer: relevance to the pathogenesis of polycystic kidney disease. Nat Rev Nephrol 2015; 11:515-34. [PMID: 25870008 PMCID: PMC5902186 DOI: 10.1038/nrneph.2015.46] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a progressive inherited disorder in which renal tissue is gradually replaced with fluid-filled cysts, giving rise to chronic kidney disease (CKD) and progressive loss of renal function. ADPKD is also associated with liver ductal cysts, hypertension, chronic pain and extra-renal problems such as cerebral aneurysms. Intriguingly, improved understanding of the signalling and pathological derangements characteristic of ADPKD has revealed marked similarities to those of solid tumours, even though the gross presentation of tumours and the greater morbidity and mortality associated with tumour invasion and metastasis would initially suggest entirely different disease processes. The commonalities between ADPKD and cancer are provocative, particularly in the context of recent preclinical and clinical studies of ADPKD that have shown promise with drugs that were originally developed for cancer. The potential therapeutic benefit of such repurposing has led us to review in detail the pathological features of ADPKD through the lens of the defined, classic hallmarks of cancer. In addition, we have evaluated features typical of ADPKD, and determined whether evidence supports the presence of such features in cancer cells. This analysis, which places pathological processes in the context of defined signalling pathways and approved signalling inhibitors, highlights potential avenues for further research and therapeutic exploitation in both diseases.
Collapse
Affiliation(s)
- Tamina Seeger-Nukpezah
- Department I of Internal Medicine and Centre for Integrated Oncology, University of Cologne, Kerpenerstrasse 62, D-50937 Cologne, Germany
| | - Daniel M Geynisman
- Department of Medical Oncology, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | - Anna S Nikonova
- Department of Developmental Therapeutics, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | - Thomas Benzing
- Department II of Internal Medicine and Centre for Molecular Medicine Cologne, University of Cologne, Kerpenerstrasse 62, D-50937 Cologne, Germany
| | - Erica A Golemis
- Department of Developmental Therapeutics, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| |
Collapse
|
77
|
Zhang Z, Guo M, Zhao S, Xu W, Shao J, Zhang F, Wu L, Lu Y, Zheng S. The update on transcriptional regulation of autophagy in normal and pathologic cells: A novel therapeutic target. Biomed Pharmacother 2015; 74:17-29. [DOI: 10.1016/j.biopha.2015.06.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 06/15/2015] [Indexed: 02/08/2023] Open
|
78
|
Sumida K, Hoshino J, Suwabe T, Kasai T, Hayami N, Mise K, Kawada M, Imafuku A, Hiramatsu R, Hasegawa E, Yamanouchi M, Sawa N, Narui K, Takaichi K, Ubara Y. Sleep-Disordered Breathing in Patients with Polycystic Liver and Kidney Disease Referred for Transcatheter Arterial Embolization. Clin J Am Soc Nephrol 2015; 10:949-956. [PMID: 25825485 PMCID: PMC4455207 DOI: 10.2215/cjn.06930714] [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: 07/13/2014] [Accepted: 02/10/2015] [Indexed: 12/22/2022]
Abstract
BACKGROUND AND OBJECTIVES Sleep-disordered breathing (SDB) is prevalent among patients with CKD, but its prevalence among patients with symptomatic autosomal dominant polycystic kidney disease (ADPKD) and its association with total liver and kidney volume remain unclear. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS This study examined the association between height-adjusted total liver and kidney volume (htTLKV) and SDB in a cross-sectional study of 304 adult patients with symptomatic ADPKD who were hospitalized at Toranomon Hospital for transcatheter arterial embolization and who underwent pulse oximetry between April 2008 and November 2013. SDB was defined as having a 3% oxygen desaturation index of ≥15 events per hour of sleep. Logistic regression was performed with sex-specific quartiles of htTLKV as the main predictor, using patient data and comorbidities as covariates. RESULTS Overall (54.6% women, mean age 56.2±9.4 years, 83.5% on hemodialysis), 177 of 304 patients (58.2%) had SDB. SDB was strongly associated with htTLKV quartiles, demonstrating that odds ratios (ORs) and 95% confidence intervals (95% CIs) for SDB were 1.63 (0.76 to 3.48), 2.35 (1.09 to 5.06), and 4.61 (1.98 to 10.7) for htTLKV quartiles 2-4 (P for trend, P=0.003), respectively. Older age (OR, 1.81 per 10 years; 95% CI, 1.29 to 2.55), male sex (OR, 3.87; 95% CI, 1.96 to 7.66), receiving hemodialysis (OR, 3.46; 95% CI, 1.62 to 12.1), and higher body mass index (≥25 kg/m(2)) (OR, 3.03; 95% CI, 1.08 to 8.52) were also associated with SDB. CONCLUSIONS In this highly selected population of patients with symptomatic ADPKD referred for transcatheter arterial embolization, SDB was highly prevalent and independently associated with higher htTLKV.
Collapse
Affiliation(s)
- Keiichi Sumida
- Nephrology Center, Toranomon Hospital Kajigaya, Kanagawa, Japan; Nephrology Center and
| | - Junichi Hoshino
- Nephrology Center, Toranomon Hospital Kajigaya, Kanagawa, Japan; Nephrology Center and
| | - Tatsuya Suwabe
- Nephrology Center, Toranomon Hospital Kajigaya, Kanagawa, Japan; Nephrology Center and
| | - Takatoshi Kasai
- Sleep Center, Toranomon Hospital, Tokyo, Japan; and Departments of Cardiology and Cardio-Respiratory Sleep Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Noriko Hayami
- Nephrology Center, Toranomon Hospital Kajigaya, Kanagawa, Japan; Nephrology Center and
| | - Koki Mise
- Nephrology Center, Toranomon Hospital Kajigaya, Kanagawa, Japan
| | | | | | | | | | | | | | - Koji Narui
- Sleep Center, Toranomon Hospital, Tokyo, Japan; and
| | - Kenmei Takaichi
- Nephrology Center, Toranomon Hospital Kajigaya, Kanagawa, Japan; Nephrology Center and
| | - Yoshifumi Ubara
- Nephrology Center, Toranomon Hospital Kajigaya, Kanagawa, Japan; Nephrology Center and
| |
Collapse
|
79
|
Abstract
Diabetic nephropathy (DN) is the most common cause of end-stage kidney disease worldwide, and is associated with increased morbidity and mortality in patients with both type 1 and type 2 diabetes. Increasing prevalence of diabetes has made the need for effective treatment of DN critical and thereby identifying new therapeutic targets to improve clinical management. Autophagy is a highly conserved 'self-eating' pathway by which cells degrade and recycle macromolecules and organelles. Autophagy serves as an essential mechanism to maintain homeostasis of glomeruli and tubules, and plays important roles in human health and diseases. Impairment of autophagy is implicated in the pathogenesis of DN. Emerging body of evidence suggests that targeting the autophagic pathway to activate and restore autophagy activity may be renoprotective. In this review, we examine current advances in our understanding of the roles of autophagy in diabetic kidney injury, focusing on studies in renal cells in culture, human kidney tissues, and experimental animal models of diabetes. We discuss the major nutrient-sensing signal pathways and diabetes-induced altered intracellular metabolism and cellular events, including accumulation of advanced glycation end-products, increased oxidative stress, endoplasmic reticulum stress, hypoxia, and activation of the renin-angiotensin system, which modulate autophagic activity and contribute to the development of DN. We also highlight recent studies of autophagy and transforming growth factor-β in renal fibrosis, the final common response to injury that ultimately leads to end-stage kidney failure in both type 1 and type 2 diabetes. These findings suggest the possibility that autophagy can be a therapeutic target against DN.
Collapse
Affiliation(s)
- Yan Ding
- Division of Nephrology and HypertensionJoan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College, 525 East 68th Street, Box 3, New York, New York 10065, USA
| | - Mary E Choi
- Division of Nephrology and HypertensionJoan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College, 525 East 68th Street, Box 3, New York, New York 10065, USA
| |
Collapse
|
80
|
Ow CPC, Abdelkader A, Hilliard LM, Phillips JK, Evans RG. Determinants of renal tissue hypoxia in a rat model of polycystic kidney disease. Am J Physiol Regul Integr Comp Physiol 2014; 307:R1207-15. [DOI: 10.1152/ajpregu.00202.2014] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Renal tissue oxygen tension (Po2) and its determinants have not been quantified in polycystic kidney disease (PKD). Therefore, we measured kidney tissue Po2 in the Lewis rat model of PKD (LPK) and in Lewis control rats. We also determined the relative contributions of altered renal oxygen delivery and consumption to renal tissue hypoxia in LPK rats. Po2 of the superficial cortex of 11- to 13-wk-old LPK rats, measured by Clark electrode with the rat under anesthesia, was higher within the cysts (32.8 ± 4.0 mmHg) than the superficial cortical parenchyma (18.3 ± 3.5 mmHg). Po2 in the superficial cortical parenchyma of Lewis rats was 2.5-fold greater (46.0 ± 3.1 mmHg) than in LPK rats. At each depth below the cortical surface, tissue Po2 in LPK rats was approximately half that in Lewis rats. Renal blood flow was 60% less in LPK than in Lewis rats, and arterial hemoglobin concentration was 57% less, so renal oxygen delivery was 78% less. Renal venous Po2 was 38% less in LPK than Lewis rats. Sodium reabsorption was 98% less in LPK than Lewis rats, but renal oxygen consumption did not significantly differ between the two groups. Thus, in this model of PKD, kidney tissue is severely hypoxic, at least partly because of deficient renal oxygen delivery. Nevertheless, the observation of similar renal oxygen consumption, despite markedly less sodium reabsorption, in the kidneys of LPK compared with Lewis rats, indicates the presence of inappropriately high oxygen consumption in the polycystic kidney.
Collapse
Affiliation(s)
- Connie P. C. Ow
- Department of Physiology Monash University, Melbourne, Australia; and
| | - Amany Abdelkader
- Department of Physiology Monash University, Melbourne, Australia; and
| | | | | | - Roger G. Evans
- Department of Physiology Monash University, Melbourne, Australia; and
| |
Collapse
|
81
|
Fougeray S, Pallet N. Mechanisms and biological functions of autophagy in diseased and ageing kidneys. Nat Rev Nephrol 2014; 11:34-45. [PMID: 25385287 DOI: 10.1038/nrneph.2014.201] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Autophagy degrades pathogens, altered organelles and protein aggregates, and is characterized by the sequestration of cytoplasmic cargos within double-membrane-limited vesicles called autophagosomes. The process is regulated by inputs from the cellular microenvironment, and is activated in response to nutrient scarcity and immune triggers, which signal through a complex molecular network. Activation of autophagy leads to the formation of an isolation membrane, recognition of cytoplasmic cargos, expansion of the autophagosomal membrane, fusion with lysosomes and degradation of the autophagosome and its contents. Autophagy maintains cellular homeostasis during stressful conditions, dampens inflammation and shapes adaptive immunity. A growing body of evidence has implicated autophagy in kidney health, ageing and disease; it modulates tissue responses during acute kidney injuries, regulates podocyte homeostasis and protects against age-related renal disorders. The renoprotective functions of autophagy in epithelial renal cells and podocytes are mostly mediated by the clearance of altered mitochondria, which can activate inflammasomes and apoptosis, and the removal of protein aggregates, which might trigger inflammation and cell death. In translational terms, autophagy is undoubtedly an attractive target for developing new renoprotective treatments and identifying markers of kidney injury.
Collapse
Affiliation(s)
- Sophie Fougeray
- Departments of Medicine, Microbiology and Immunology, The Research Institute of the McGill University Health Center, 2155 Guy Street, Montreal, QC H3H 2R9, Canada
| | - Nicolas Pallet
- Service de Biochimie, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, 20 Rue Leblanc, 75015 Paris, France
| |
Collapse
|
82
|
Orhon I, Dupont N, Pampliega O, Cuervo AM, Codogno P. Autophagy and regulation of cilia function and assembly. Cell Death Differ 2014; 22:389-97. [PMID: 25361082 DOI: 10.1038/cdd.2014.171] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 09/05/2014] [Accepted: 09/10/2014] [Indexed: 12/21/2022] Open
Abstract
Motile and primary cilia (PC) are microtubule-based structures located at the cell surface of many cell types. Cilia govern cellular functions ranging from motility to integration of mechanical and chemical signaling from the environment. Recent studies highlight the interplay between cilia and autophagy, a conserved cellular process responsible for intracellular degradation. Signaling from the PC recruits the autophagic machinery to trigger autophagosome formation. Conversely, autophagy regulates ciliogenesis by controlling the levels of ciliary proteins. The cross talk between autophagy and ciliated structures is a novel aspect of cell biology with major implications in development, physiology and human pathologies related to defects in cilium function.
Collapse
Affiliation(s)
- I Orhon
- 1] INSERM U1151-CNRS UMR 8253, Paris, France [2] Institut Necker Enfants-Malades (INEM), Paris, France [3] Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - N Dupont
- 1] INSERM U1151-CNRS UMR 8253, Paris, France [2] Institut Necker Enfants-Malades (INEM), Paris, France [3] Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - O Pampliega
- 1] Department of Development and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA [2] Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY, USA
| | - A M Cuervo
- 1] Department of Development and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA [2] Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY, USA
| | - P Codogno
- 1] INSERM U1151-CNRS UMR 8253, Paris, France [2] Institut Necker Enfants-Malades (INEM), Paris, France [3] Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
83
|
Yeganeh B, Ghavami S, Kroeker AL, Mahood TH, Stelmack GL, Klonisch T, Coombs KM, Halayko AJ. Suppression of influenza A virus replication in human lung epithelial cells by noncytotoxic concentrations bafilomycin A1. Am J Physiol Lung Cell Mol Physiol 2014; 308:L270-86. [PMID: 25361566 DOI: 10.1152/ajplung.00011.2014] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Subcellular trafficking within host cells plays a critical role in viral life cycles, including influenza A virus (IAV). Thus targeting relevant subcellular compartments holds promise for effective intervention to control the impact of influenza infection. Bafilomycin A1 (Baf-A1), when used at relative high concentrations (≥10 nM), inhibits vacuolar ATPase (V-ATPase) and reduces endosome acidification and lysosome number, thus inhibiting IAV replication but promoting host cell cytotoxicity. We tested the hypothesis that much lower doses of Baf-A1 also have anti-IAV activity, but without toxic effects. Thus we assessed the antiviral activity of Baf-A1 at different concentrations (0.1-100 nM) in human alveolar epithelial cells (A549) infected with IAV strain A/PR/8/34 virus (H1N1). Infected and mock-infected cells pre- and cotreated with Baf-A1 were harvested 0-24 h postinfection and analyzed by immunoblotting, immunofluorescence, and confocal and electron microscopy. We found that Baf-A1 had disparate concentration-dependent effects on subcellular organelles and suppressed affected IAV replication. At concentrations ≥10 nM Baf-A1 inhibited acid lysosome formation, which resulted in greatly reduced IAV replication and release. Notably, at a very low concentration of 0.1 nM that is insufficient to reduce lysosome number, Baf-A1 retained the capacity to significantly impair IAV nuclear accumulation as well as IAV replication and release. In contrast to the effects of high concentrations of Baf-A1, very low concentrations did not exhibit cytotoxic effects or induce apoptotic cell death, based on morphological and FACS analyses. In conclusion, our results reveal that low-concentration Baf-A1 is an effective inhibitor of IAV replication, without impacting host cell viability.
Collapse
Affiliation(s)
- Behzad Yeganeh
- Department of Physiology, University of Manitoba, Winnipeg, Manitoba, Canada; Biology of Breathing Group, Manitoba Institute of Child Health, Winnipeg, Manitoba, Canada
| | - Saeid Ghavami
- Department of Physiology, University of Manitoba, Winnipeg, Manitoba, Canada; Biology of Breathing Group, Manitoba Institute of Child Health, Winnipeg, Manitoba, Canada; Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Andrea L Kroeker
- Department of Physiology, University of Manitoba, Winnipeg, Manitoba, Canada; Biology of Breathing Group, Manitoba Institute of Child Health, Winnipeg, Manitoba, Canada
| | - Thomas H Mahood
- Department of Physiology, University of Manitoba, Winnipeg, Manitoba, Canada; Biology of Breathing Group, Manitoba Institute of Child Health, Winnipeg, Manitoba, Canada
| | - Gerald L Stelmack
- Department of Physiology, University of Manitoba, Winnipeg, Manitoba, Canada; Biology of Breathing Group, Manitoba Institute of Child Health, Winnipeg, Manitoba, Canada
| | - Thomas Klonisch
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Surgery, University of Manitoba, Winnipeg, Manitoba, Canada; and
| | - Kevin M Coombs
- Department of Physiology, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Andrew J Halayko
- Department of Physiology, University of Manitoba, Winnipeg, Manitoba, Canada; Biology of Breathing Group, Manitoba Institute of Child Health, Winnipeg, Manitoba, Canada; Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
84
|
Buchholz B, Schley G, Faria D, Kroening S, Willam C, Schreiber R, Klanke B, Burzlaff N, Jantsch J, Kunzelmann K, Eckardt KU. Hypoxia-inducible factor-1α causes renal cyst expansion through calcium-activated chloride secretion. J Am Soc Nephrol 2014; 25:465-74. [PMID: 24203996 PMCID: PMC3935579 DOI: 10.1681/asn.2013030209] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Accepted: 08/21/2013] [Indexed: 01/09/2023] Open
Abstract
Polycystic kidney diseases are characterized by numerous bilateral renal cysts that continuously enlarge and, through compression of intact nephrons, lead to a decline in kidney function over time. We previously showed that cyst enlargement is accompanied by regional hypoxia, which results in the stabilization of hypoxia-inducible transcription factor-1α (HIF-1α) in the cyst epithelium. Here we demonstrate a correlation between cyst size and the expression of the HIF-1α-target gene, glucose transporter 1, and report that HIF-1α promotes renal cyst growth in two in vitro cyst models-principal-like MDCK cells (plMDCKs) within a collagen matrix and cultured embryonic mouse kidneys stimulated with forskolin. In both models, augmenting HIF-1α levels with the prolyl hydroxylase inhibitor 2-(1-chloro-4-hydroxyisoquinoline-3-carboxamido) acetate enhanced cyst growth. In addition, inhibition of HIF-1α degradation through tubule-specific knockdown of the von Hippel-Lindau tumor suppressor increased cyst size in the embryonic kidney cyst model. In contrast, inhibition of HIF-1α by chetomin and knockdown of HIF-1α both decreased cyst growth in these models. Consistent with previous reports, plMDCK cyst enlargement was driven largely by transepithelial chloride secretion, which consists, in part, of a calcium-activated chloride conductance. plMDCKs deficient for HIF-1α almost completely lacked calcium-activated chloride secretion. We conclude that regional hypoxia in renal cysts contributes to cyst growth, primarily due to HIF-1α-dependent calcium-activated chloride secretion. These findings identify the HIF system as a novel target for inhibition of cyst growth.
Collapse
Affiliation(s)
| | | | - Diana Faria
- Department of Physiology, University of Regensburg, Regensburg, Germany; and
| | | | | | - Rainer Schreiber
- Department of Physiology, University of Regensburg, Regensburg, Germany; and
| | | | - Nicolai Burzlaff
- Chemistry and Pharmacy, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Jonathan Jantsch
- Department of Clinical Microbiology, Immunology, and Hygiene, Microbiology Institute, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Karl Kunzelmann
- Department of Physiology, University of Regensburg, Regensburg, Germany; and
| | | |
Collapse
|
85
|
Ren XS, Sato Y, Harada K, Sasaki M, Furubo S, Song JY, Nakanuma Y. Activation of the PI3K/mTOR pathway is involved in cystic proliferation of cholangiocytes of the PCK rat. PLoS One 2014; 9:e87660. [PMID: 24498161 PMCID: PMC3907540 DOI: 10.1371/journal.pone.0087660] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 12/28/2013] [Indexed: 12/13/2022] Open
Abstract
The polycystic kidney (PCK) rat is an animal model of Caroli’s disease as well as autosomal recessive polycystic kidney disease (ARPKD). The signaling pathways involving the mammalian target of rapamycin (mTOR) are aberrantly activated in ARPKD. This study investigated the effects of inhibitors for the cell signaling pathways including mTOR on cholangiocyte proliferation of the PCK rat. Cultured PCK cholangiocytes were treated with rapamycin and everolimus [inhibitors of mTOR complex 1 (mTOC1)], LY294002 [an inhibitor of phosphatidylinositol 3-kinase (PI3K)] and NVP-BEZ235 (an inhibitor of PI3K and mTORC1/2), and the cell proliferative activity was determined in relation to autophagy and apoptosis. The expression of phosphorylated (p)-mTOR, p-Akt, and PI3K was increased in PCK cholangiocytes compared to normal cholangiocytes. All inhibitors significantly inhibited the cell proliferative activity of PCK cholangiocytes, where NVP-BEZ235 had the most prominent effect. NVP-BEZ235, but not rapamycin and everolimus, further inhibited biliary cyst formation in the three-dimensional cell culture system. Rapamycin and everolimus induced apoptosis in PCK cholangiocytes, whereas NVP-BEZ235 inhibited cholangiocyte apoptosis. Notably, the autophagic response was significantly induced following the treatment with NVP-BEZ235, but not rapamycin and everolimus. Inhibition of autophagy using siRNA against protein-light chain3 and 3-methyladenine significantly increased the cell proliferative activity of PCK cholangiocytes treated with NVP-BEZ235. In vivo, treatment of the PCK rat with NVP-BEZ235 attenuated cystic dilatation of the intrahepatic bile ducts, whereas renal cyst development was unaffected. These results suggest that the aberrant activation of the PI3K/mTOR pathway is involved in cystic proliferation of cholangiocytes of the PCK rat, and inhibition of the pathway can reduce cholangiocyte proliferation via the mechanism involving apoptosis and/or autophagy.
Collapse
Affiliation(s)
- Xiang Shan Ren
- Department of Human Pathology, Kanazawa University Graduate School of Medicine, Kanazawa, Japan
- Department of Pathology, Yanbian University College of Medicine, Yanji-city, China
| | - Yasunori Sato
- Department of Human Pathology, Kanazawa University Graduate School of Medicine, Kanazawa, Japan
| | - Kenichi Harada
- Department of Human Pathology, Kanazawa University Graduate School of Medicine, Kanazawa, Japan
| | - Motoko Sasaki
- Department of Human Pathology, Kanazawa University Graduate School of Medicine, Kanazawa, Japan
| | - Shinichi Furubo
- Department of Human Pathology, Kanazawa University Graduate School of Medicine, Kanazawa, Japan
| | - Jing Yu Song
- Department of Pathology, Yanbian University College of Medicine, Yanji-city, China
| | - Yasuni Nakanuma
- Department of Human Pathology, Kanazawa University Graduate School of Medicine, Kanazawa, Japan
- Department of Pathology, Shizuoka Cancer Center, Shizuoka, Japan
- * E-mail:
| |
Collapse
|
86
|
Abstract
SIGNIFICANCE Autophagy is emerging as an important pathway in many biological processes and diseases. This review summarizes the current progress on the role of autophagy in renal physiology and pathology. RECENT ADVANCES Studies from renal cells in culture, human kidney tissues, and experimental animal models implicate that autophagy regulates many critical aspects of normal and disease conditions in the kidney, such as diabetic nephropathy and other glomerular diseases, tubular injuries, kidney development and aging, cancer, and genetic diseases associated with the kidney. CRITICAL ISSUES The importance of autophagy in the kidney has just started to be elucidated. How the process of autophagy is altered in the pathogenesis of kidney diseases and how this alteration is beneficial or detrimental to kidney functions still need to be fully understood. FUTURE DIRECTIONS Investigations that uncover the precise mechanism and regulation of autophagy in various kidney diseases may lead to new strategies for therapeutic modulation.
Collapse
Affiliation(s)
- Zhibo Wang
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School , Boston, Massachusetts
| | | |
Collapse
|
87
|
Abstract
Autosomal-dominant polycystic kidney disease is the most common form of polycystic kidney disease in adults and is caused by a mutation in the polycystic kidney disease 1 or 2 genes, which encode, respectively, polycystin-1 and polycystin-2. Autophagy is present in polycystic kidneys in rat and mouse models of polycystic kidney disease. Autophagy has yet to be shown in human polycystic kidney disease kidneys. The mechanism of cyst growth has been studied extensively in vitro and in vivo. Multiple molecules and signaling pathways have been implicated in cyst growth including mammalian target of rapamycin, the renin-angiotensin-aldosterone system, vasopressin and cyclic adenosine monophosphate, epidermal growth factor and insulin-like growth factor tyrosine kinases, vascular endothelial growth factor, extracellular signal-related kinase, tumor necrosis factor-α, cyclin-dependent kinases, caspases and apoptosis, and cyclic adenosine monophosphate-activated protein kinases. Many of the agents that inhibit these signaling pathways and slow cyst growth are also autophagy inducers such as mammalian target of rapamycin inhibitors, cyclin-dependent kinase inhibitors, caspase inhibitors, tyrosine kinase inhibitors, metformin, curcumin, and triptolide. There are reasons to believe that suppression of autophagy may play a role in cyst formation and growth. This review presents the hypothesis that suppression of autophagy may play a role in cyst formation and growth, based on the following: (1) many of the agents that protect against polycystic kidney disease also induce autophagy, (2) suppression of autophagy in polycystic kidney disease 1 knockout cells, (3) a defect in autophagy in congenital polycystic kidney mice with polycystic kidney disease, (4) how suppressed autophagy may relate to apoptosis in polycystic kidney disease, and (5) conditions with defective cilia, the ciliopathies, are associated with decreased autophagy.
Collapse
Affiliation(s)
| | - Charles L Edelstein
- Division of Renal Diseases and Hypertension, University of Colorado at Denver, Aurora, CO.
| |
Collapse
|
88
|
Liu Y, Käch A, Ziegler U, Ong ACM, Wallace DP, Arcaro A, Serra AL. The role of phospholipase D in modulating the MTOR signaling pathway in polycystic kidney disease. PLoS One 2013; 8:e73173. [PMID: 24009738 PMCID: PMC3751888 DOI: 10.1371/journal.pone.0073173] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 07/17/2013] [Indexed: 12/17/2022] Open
Abstract
The mammalian target of rapamycin (mTOR) signaling pathway is aberrantly activated in polycystic kidney disease (PKD). Emerging evidence suggests that phospholipase D (PLD) and its product phosphatidic acid (PA) regulate mTOR activity. In this study, we assessed in vitro the regulatory function of PLD and PA on the mTOR signaling pathway in PKD. We found that the basal level of PLD activity was elevated in PKD cells. Targeting PLD by small molecule inhibitors reduced cell proliferation and blocked mTOR signaling, whereas exogenous PA stimulated mTOR signaling and abolished the inhibitory effect of PLD on PKD cell proliferation. We also show that blocking PLD activity enhanced the sensitivity of PKD cells to rapamycin and that combining PLD inhibitors and rapamycin synergistically inhibited PKD cell proliferation. Furthermore, we demonstrate that targeting mTOR did not induce autophagy, whereas targeting PLD induced autophagosome formation. Taken together, our findings suggest that deregulated mTOR pathway activation is mediated partly by increased PLD signaling in PKD cells. Targeting PLD isoforms with pharmacological inhibitors may represent a new therapeutic strategy in PKD.
Collapse
Affiliation(s)
- Yang Liu
- Institute of Physiology, University of Zurich, Zürich, Switzerland
| | - Andres Käch
- Center for Microscopy and Image Analysis, University of Zurich, Zürich, Switzerland
| | - Urs Ziegler
- Center for Microscopy and Image Analysis, University of Zurich, Zürich, Switzerland
| | - Albert C. M. Ong
- Academic Unit of Nephrology, Department of Infection and Immunity, University of Sheffield, Sheffield, United Kingdom
| | - Darren P. Wallace
- Department of Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Alexandre Arcaro
- Department of Clinical Research, University of Bern, Bern, Switzerland
| | - Andreas L. Serra
- Institute of Physiology, University of Zurich, Zürich, Switzerland
- * E-mail:
| |
Collapse
|
89
|
Pushpakumar SB, Kundu S, Metreveli N, Tyagi SC, Sen U. Matrix Metalloproteinase Inhibition Mitigates Renovascular Remodeling in Salt-Sensitive Hypertension. Physiol Rep 2013; 1:e00063. [PMID: 24159376 PMCID: PMC3804376 DOI: 10.1002/phy2.63] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Extracellular matrix (ECM) remodeling is the hallmark of hypertensive nephropathy. Uncontrolled proteolytic activity due to an imbalance between matrix metalloproteinases and tissue inhibitors of metalloproteinases (MMPs/TIMPs) has been implicated in renovascular fibrosis. We hypothesized that inhibition of MMPs will reduce excess ECM deposition and modulate autophagy to attenuate hypertension. Dahl salt-sensitive (Dahl/SS) and Lewis rats were fed on high salt diet and treated without or with 1.2 mg/kg b.w. of GM6001 (MMP inhibitor) by intraperitoneal injection on alternate days for 4 weeks. Blood pressure (BP), renal cortical blood flow, vascular density, collagen, elastin, and MMPs/TIMPs were measured. GM6001 treatment significantly reduced mean BP in hypertensive Dahl/SS rats. Renal resistive index (RI) was increased in hypertensive Dahl/SS rats and Doppler flowmetry showed reduced cortical perfusion. Barium angiography demonstrated a reduction in terminal branches of renal vasculature. Inhibition of MMPs by GM6001 resulted in a significant improvement in all the parameters including renal function. In hypertensive Dahl/SS rats, protein levels of MMP-9, -2, and -13 were increased including the activity of MMP-9 and -2; TIMP-1 and -2 levels were increased whereas TIMP-3 levels were similar to Lewis controls. Administration of GM6001 reduced the activity of MMPs and increased the levels of TIMP-1, -2, and -3. MMP inhibition reduced type 1 collagen deposition and increased elastin in the intrarenal vessels indicating reduced fibrosis. Autophagy markers were decreased in hypertensive Dahl/SS rats and GM6001 treatment enhanced their levels. We conclude that MMP inhibition (GM6001) reduces adverse renovascular remodeling in hypertension by modulating ECM turnover and stimulating autophagy.
Collapse
Affiliation(s)
- Sathnur B Pushpakumar
- Department of Physiology and Biophysics, University of Louisville School of Medicine Louisville, KY-40292
| | | | | | | | | |
Collapse
|
90
|
Wang S, Dong Z. Primary cilia and kidney injury: current research status and future perspectives. Am J Physiol Renal Physiol 2013; 305:F1085-98. [PMID: 23904226 DOI: 10.1152/ajprenal.00399.2013] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cilia, membrane-enclosed organelles protruding from the apical side of cells, can be divided into two classes: motile and primary cilia. During the past decades, motile cilia have been intensively studied. However, it was not until the 1990s that people began to realize the importance of primary cilia as cellular-specific sensors, particularly in kidney tubular epithelial cells. Furthermore, accumulating evidence indicates that primary cilia may be involved in the regulation of cell proliferation, differentiation, apoptosis, and planar cell polarity. Many signaling pathways, such as Wnt, Notch, Hedgehog, and mammalian target of rapamycin, have been located to the primary cilia. Thus primary cilia have been regarded as a hub that integrates signals from the extracellular environment. More importantly, dysfunction of this organelle may contribute to the pathogenesis of a large spectrum of human genetic diseases, named ciliopathies. The significance of primary cilia in acquired human diseases such as hypertension and diabetes has gradually drawn attention. Interestingly, recent reports disclosed that cilia length varies during kidney injury, and shortening of cilia enhances the sensitivity of epithelial cells to injury cues. This review briefly summarizes the current status of cilia research and explores the potential mechanisms of cilia-length changes during kidney injury as well as provides some thoughts to allure more insightful ideas and promotes the further study of primary cilia in the context of kidney injury.
Collapse
Affiliation(s)
- Shixuan Wang
- Dept. of Cellular Biology and Anatomy, Medical College of Georgia, Augusta, GA 30912.
| | | |
Collapse
|
91
|
Lee Y, Jung J, Cho KJ, Lee SK, Park JW, Oh ILH, Kim GJ. Increased SCF/c-kit by hypoxia promotes autophagy of human placental chorionic plate-derived mesenchymal stem cells via regulating the phosphorylation of mTOR. J Cell Biochem 2012; 114:79-88. [DOI: 10.1002/jcb.24303] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Accepted: 07/17/2012] [Indexed: 12/15/2022]
|
92
|
Ding A, Kalaignanasundaram P, Ricardo SD, Abdelkader A, Witting PK, Broughton BRS, Kim HB, Wyse BF, Phillips JK, Evans RG. Chronic treatment with tempol does not significantly ameliorate renal tissue hypoxia or disease progression in a rodent model of polycystic kidney disease. Clin Exp Pharmacol Physiol 2012; 39:917-29. [DOI: 10.1111/1440-1681.12013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Alice Ding
- The Australian School of Advanced Medicine; Macquarie University; Sydney; NSW; Australia
| | | | - Sharon D Ricardo
- Department of Monash Immunology & Stem Cell Laboratories; Monash University; Melbourne; Vic.; Australia
| | - Amany Abdelkader
- Department of Physiology; Monash University; Melbourne; Vic.; Australia
| | - Paul K Witting
- Discipline of Pathology; The University of Sydney; Sydney; NSW; Australia
| | - Brad RS Broughton
- Department of Pharmacology; Monash University; Melbourne; Vic.; Australia
| | - Hyun B Kim
- Discipline of Pathology; The University of Sydney; Sydney; NSW; Australia
| | - Benjamin F Wyse
- The Australian School of Advanced Medicine; Macquarie University; Sydney; NSW; Australia
| | - Jacqueline K Phillips
- The Australian School of Advanced Medicine; Macquarie University; Sydney; NSW; Australia
| | - Roger G Evans
- Department of Physiology; Monash University; Melbourne; Vic.; Australia
| |
Collapse
|
93
|
Huber TB, Edelstein CL, Hartleben B, Inoki K, Jiang M, Koya D, Kume S, Lieberthal W, Pallet N, Quiroga A, Ravichandran K, Susztak K, Yoshida S, Dong Z. Emerging role of autophagy in kidney function, diseases and aging. Autophagy 2012; 8:1009-31. [PMID: 22692002 PMCID: PMC3429540 DOI: 10.4161/auto.19821] [Citation(s) in RCA: 215] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Revised: 02/24/2012] [Accepted: 02/27/2012] [Indexed: 02/06/2023] Open
Abstract
Autophagy is a highly conserved process that degrades cellular long-lived proteins and organelles. Accumulating evidence indicates that autophagy plays a critical role in kidney maintenance, diseases and aging. Ischemic, toxic, immunological, and oxidative insults can cause an induction of autophagy in renal epithelial cells modifying the course of various kidney diseases. This review summarizes recent insights on the role of autophagy in kidney physiology and diseases alluding to possible novel intervention strategies for treating specific kidney disorders by modifying autophagy.
Collapse
Affiliation(s)
- Tobias B Huber
- Renal Division, University Hospital Freiburg; Freiburg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Choi JH, Lee HJ, Yang TH, Kim GJ. Effects of hypoxia inducible factors-1α on autophagy and invasion of trophoblasts. Clin Exp Reprod Med 2012; 39:73-80. [PMID: 22816073 PMCID: PMC3398120 DOI: 10.5653/cerm.2012.39.2.73] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2012] [Revised: 03/21/2012] [Accepted: 03/30/2012] [Indexed: 11/22/2022] Open
Abstract
Objective This study was undertaken to determine the effect of hypoxia inducible factor (HIF)-1α on the cell death, autophagy, and invasion of trophoblasts. Methods To understand the effect of HIF-1α, we inhibited HIF-1α using siRNA under normoxia and hypoxia conditions. Invasion assay and zymography were performed to determine changes in the invasion ability of HIF-1α. Western blotting and immunofluorescence were performed to determine some of the signal events involved in apoptosis and autophagy. Results There was no difference in cell death through the inhibition of HIF-1α expression by siRNA; however, the expression of LC3 and autophagosome formation increased. On the other hand, autophagy was increased, and the invasive ability of trophoblast cells decreased according to the inhibition of HIF-1α expression by siRNA. These experimental results mean that HIF-1α genes regulate the invasive ability of trophoblasts by increasing autophagy. Conclusion This study contributes important data for understanding the mechanism of early pregnancy implantation and the invasive ability of trophoblasts by defining the relationship between the roles of HIF-1α and autophagy.
Collapse
Affiliation(s)
- Jong-Ho Choi
- Department of Biomedical Science, CHA University, Seoul, Korea
| | | | | | | |
Collapse
|
95
|
Ponnusamy M, Liu N, Sellamuthu R, Zhao TC, Mao H, Zhuang S. Autophagy protects against necrotic renal epithelial cell-induced death of renal interstitial fibroblasts. Am J Physiol Renal Physiol 2012; 303:F83-91. [PMID: 22496408 DOI: 10.1152/ajprenal.00027.2012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
We recently reported that necrotic renal proximal tubular cells (RPTC) can induce the death of renal interstitial fibroblasts. Since autophagy plays either cytoprotective or cytodestructive roles depending on the experimental condition, the present study was carried out to investigate whether necrotic RPTC would induce autophagy of renal interstitial fibroblasts and, if so, whether autophagy would contribute to cell death or exert a protective effect. Exposure of necrotic RPTC supernatant (RPTC-Sup) induced autophagy in renal interstitial fibroblast cells (NRK-49F) in a time- and dose-dependent manner, and its induction was earlier than caspase-3 activation. Inhibition of autophagy with 3-methyladenine (3-MA) or knockdown of Beclin-1, a molecule involved in the initiation of autophagosome formation, with small interference RNA (siRNA) significantly enhanced necrotic RPTC-Sup-induced cell death. Necrotic RPTC-Sup induced phosphorylation of extracellular signal-regulated kinases (ERK1/2), p38, c-Jun NH(2)-terminal kinases (JNKs), and AKT. Treatment with an ERK1/2 pathway inhibitor, but not with specific inhibitors for p38, JNKs, or AKT pathways, blocked NRK-49F autophagy and cell death upon exposure to necrotic RPTC-Sup. Furthermore, knockdown of MEK1 with siRNA also reduced autophagy along with cell death in NRK-49F exposed to necrotic RPTC-Sup. In contrast, overexpression of MEK1/2 increased RPTC-Sup-induced fibroblast cell death without enhancing autophagy. Collectively, this study demonstrates that necrotic RPTC induce both autophagy and cell death and that autophagy plays a cytoprotective or prosurvival role in renal fibroblasts. Furthermore, necrotic RPTC-induced autophagy and cell death in renal fibroblasts is mediated by the activation of the MEK1-ERK1/2 signaling pathway.
Collapse
Affiliation(s)
- Murugavel Ponnusamy
- Department of Medicine, Rhode Island Hospital, Providence, Rhode Island, USA
| | | | | | | | | | | |
Collapse
|
96
|
Anderson S, Oyama TT, Lindsley JN, Schutzer WE, Beard DR, Gattone VH, Komers R. 2-Hydroxyestradiol slows progression of experimental polycystic kidney disease. Am J Physiol Renal Physiol 2011; 302:F636-45. [PMID: 22160773 DOI: 10.1152/ajprenal.00265.2011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Male gender is a risk factor for progression of polycystic kidney disease (PKD). 17β-Estradiol (E2) protects experimentally, but clinical use is limited by adverse effects. Novel E2 metabolites provide many benefits of E2 without stimulating the estrogen receptor, and thus may be safer. We hypothesized that E2 metabolites are protective in a model of PKD. Studies were performed in male control Han:SPRD rats, and in cystic males treated with orchiectomy, 2-methoxyestradiol, 2-hydroxyestradiol (2-OHE), or vehicle, from age 3 to 12 wk. Cystic rats exhibited renal functional impairment (∼50% decrease in glomerular filtration and renal plasma flow rates, P < 0.05) and substantial cyst development (20.5 ± 2.0% of cortex area). 2-OHE was the most effective in limiting cysts (6.0 ± 0.7% of cortex area, P < 0.05 vs. vehicle-treated cystic rats) and preserving function, in association with suppression of proliferation, apoptosis, and angiogenesis markers. Downregulation of p21 expression and increased expression of Akt, the mammalian target of rapamycin (mTOR), and some of its downstream effectors were significantly reversed by 2-OHE. Thus, 2-OHE limits disease progression in a cystic rodent model. Mechanisms include reduced renal cell proliferation, apoptosis, and angiogenesis. These effects may be mediated, at least in part, by preservation of p21 and suppression of Akt and mTOR. Estradiol metabolites may represent a novel, safe intervention to slow progression of PKD.
Collapse
Affiliation(s)
- Sharon Anderson
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health & Science University, Portland, OR 97239, USA.
| | | | | | | | | | | | | |
Collapse
|
97
|
Raina S, Honer M, Krämer SD, Liu Y, Wang X, Segerer S, Wüthrich RP, Serra AL. Anti-VEGF antibody treatment accelerates polycystic kidney disease. Am J Physiol Renal Physiol 2011; 301:F773-83. [PMID: 21677148 DOI: 10.1152/ajprenal.00058.2011] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Polycystic kidney growth implies expansion of the vasculature, suggesting that vascular endothelial growth factor (VEGF)-dependent processes play a critical role and that VEGF is a putative therapeutic target. Whether an anti-VEGF antibody improves renal cystic disease has not been determined. We administrated 5 mg/kg B20.4.1, an anti-VEGF-A antibody, or vehicle intraperitoneally twice weekly to 4-wk-old male normal (+/+) and cystic (Cy/+) Han:SPRD rats for 6 wk. Renal function, urinary protein excretion, organ/body weight ratios, cyst volume, tubular epithelial cell (TEC) proliferation, renal VEGF, hypoxia-inducible factor (HIF)-1α and -2α expression, renal histology, and kidney hypoxia visualized by [(18)F]fluoromisonidazole positron emission tomography were assessed. The treated compared with untreated +/+ rats had lower TEC proliferation rates, whereas Cy/+ rats receiving B20.4.1 displayed an increased proximal TEC proliferation rate, causing enhanced cyst and kidney growth. The +/+ and Cy/+ rats receiving B20.4.1 had severe renal failure and extensive glomerular damage. Proteinuria, which was highest in anti-VEGF-treated Cy/+ and lowest in untreated normal littermates, was positively correlated with renal HIF-1α and negatively correlated with VEGF expression. The untreated Cy/+ vs. +/+ rats had higher overall [(18)F]fluoromisonidazole uptake. The +/+ rats receiving B20.4.1 vs. untreated had increased [(18)F]fluoromisonidazole uptake, whereas the uptake was unchanged among treated vs. untreated Cy/+ animals. In conclusion, B20.4.1 caused an exaggerated cystic response of the proximal tubules in cystic rats and severe kidney injury that was associated with low renal VEGF and high HIF-1α levels. Anti-VEGF drug therapy may therefore not be a treatment option for polycystic kidney disease.
Collapse
Affiliation(s)
- Shagun Raina
- Zürich Center for Integrated Human Physiology, ETH Zürich, Zürich, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|