101
|
Muneer A, Shamsher Khan RM. Endoplasmic Reticulum Stress: Implications for Neuropsychiatric Disorders. Chonnam Med J 2019; 55:8-19. [PMID: 30740335 PMCID: PMC6351318 DOI: 10.4068/cmj.2019.55.1.8] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/11/2018] [Accepted: 11/09/2018] [Indexed: 11/06/2022] Open
Abstract
The Endoplasmic reticulum (ER), an indispensable sub-cellular component of the eukaryotic cell carries out essential functions, is critical to the survival of the organism. The chaperone proteins and the folding enzymes which are multi-domain ER effectors carry out 3-dimensional conformation of nascent polypeptides and check misfolded protein aggregation, easing the exit of functional proteins from the ER. Diverse conditions, for instance redox imbalance, alterations in ionic calcium levels, and inflammatory signaling can perturb the functioning of the ER, leading to a build-up of unfolded or misfolded proteins in the lumen. This results in ER stress, and aiming to reinstate protein homeostasis, a well conserved reaction called the unfolded protein response (UPR) is elicited. Equally, in protracted cellular stress or inadequate compensatory reaction, UPR pathway leads to cell loss. Dysfunctional ER mechanisms are responsible for neuronal degeneration in numerous human diseases, for instance Alzheimer's, Parkinson's and Huntington's diseases. In addition, mounting proof indicates that ER stress is incriminated in psychiatric diseases like major depressive disorder, bipolar disorder, and schizophrenia. Accumulating evidence suggests that pharmacological agents regulating the working of ER may have a role in diminishing advancing neuronal dysfunction in neuropsychiatric disorders. Here, new findings are examined which link the foremost mechanisms connecting ER stress and cell homeostasis. Furthermore, a supposed new pathogenic model of major neuropsychiatry disorders is provided, with ER stress proposed as the pivotal step in disease development.
Collapse
Affiliation(s)
- Ather Muneer
- Islamic International Medical College, Riphah International University, Rawalpindi, Pakistan
| | | |
Collapse
|
102
|
Jao TM, Nangaku M, Wu CH, Sugahara M, Saito H, Maekawa H, Ishimoto Y, Aoe M, Inoue T, Tanaka T, Staels B, Mori K, Inagi R. ATF6α downregulation of PPARα promotes lipotoxicity-induced tubulointerstitial fibrosis. Kidney Int 2019; 95:577-589. [PMID: 30639234 DOI: 10.1016/j.kint.2018.09.023] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 08/28/2018] [Accepted: 09/20/2018] [Indexed: 01/09/2023]
Abstract
Tubulointerstitial fibrosis is a strong predictor of progression in patients with chronic kidney disease, and is often accompanied by lipid accumulation in renal tubules. However, the molecular mechanisms modulating the relationship between lipotoxicity and tubulointerstitial fibrosis remain obscure. ATF6α, a transcription factor of the unfolded protein response, is reported to be an upstream regulator of fatty acid metabolism. Owing to their high energy demand, proximal tubular cells (PTCs) use fatty acids as their main energy source. We therefore hypothesized that ATF6α regulates PTC fatty acid metabolism, contributing to lipotoxicity-induced tubulointerstitial fibrosis. Overexpression of activated ATF6α transcriptionally downregulated peroxisome proliferator-activated receptor-α (PPARα), the master regulator of lipid metabolism, leading to reduced activity of fatty acid β-oxidation and cytosolic accumulation of lipid droplets in a human PTC line (HK-2). ATF6α-induced lipid accumulation caused mitochondrial dysfunction, enhanced apoptosis, and increased expression of connective tissue growth factor (CTGF), as well as reduced cell viability. Atf6α-/- mice had sustained expression of PPARα and less tubular lipid accumulation following unilateral ischemia-reperfusion injury (uIRI), resulting in the amelioration of apoptosis; reduced expression of CTGF, α-smooth muscle actin, and collagen I; and less tubulointerstitial fibrosis. Administration of fenofibrate, a PPARα agonist, reduced lipid accumulation and tubulointerstitial fibrosis in the uIRI model. Taken together, these findings suggest that ATF6α deranges fatty acid metabolism in PTCs, which leads to lipotoxicity-mediated apoptosis and CTGF upregulation, both of which promote tubulointerstitial fibrosis.
Collapse
Affiliation(s)
- Tzu-Ming Jao
- Division of Chronic Kidney Disease Pathophysiology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Masaomi Nangaku
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Chia-Hsien Wu
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Mai Sugahara
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Hisako Saito
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Hiroshi Maekawa
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Yu Ishimoto
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Mari Aoe
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Tsuyoshi Inoue
- Division of Chronic Kidney Disease Pathophysiology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Tetsuhiro Tanaka
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Bart Staels
- Evaluation et Gestion Informatique de la Diversité Génétique, Université de Lille, Centre Hospitalier Régional Universitaire de Lille, Institut Pasteur de Lille, Institut National de la Santé et de la Recherche Médicale Unite Mixte de Recherche 1011, Lille, France
| | - Kazutoshi Mori
- Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto, Japan
| | - Reiko Inagi
- Division of Chronic Kidney Disease Pathophysiology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan.
| |
Collapse
|
103
|
Liu W, Tian X, Wu T, Liu L, Guo Y, Wang C. PDE5A Suppresses Proteasome Activity Leading to Insulin Resistance in C2C12 Myotubes. Int J Endocrinol 2019; 2019:3054820. [PMID: 30774657 PMCID: PMC6350610 DOI: 10.1155/2019/3054820] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 09/30/2018] [Accepted: 11/12/2018] [Indexed: 01/20/2023] Open
Abstract
OBJECTIVE The involvement of phosphodiesterase type 5 (PDE5) in the development of insulin resistance has been reported recently. However, the underlying molecular mechanism remains unclear. The present study aims at investigating the potential impacts of PDE5A on insulin signaling in C2C12 skeletal muscle myotubes and uncover the related mechanism. METHODS C2C12 myoblasts were differentiated into myotubes. Western blot was performed to detect the levels of proteins and phosphorylated proteins. Glucose uptake was determined by a colorimetric kit. The overexpression or knockdown of specific protein was carried out by infecting the myotubes with adenoviruses carrying cDNA or shRNA corresponding to the targeted protein, respectively. RESULTS PDE5A was demonstrated to negatively regulate insulin signaling, evidenced by the opposite effects on the suppression or enhancement of the insulin-stimulated Akt phosphorylation and 2-deoxy-D-glucose (2-DG) uptake in C2C12 myotubes, when PDE5A was overexpressed or knockdown, respectively. Interestingly, PDE5A overexpression led to significantly enhanced, while its knockdown resulted in markedly reduced, endoplasmic reticulum (ER) stress. Inhibition of ER stress improved PDE5A overexpression-induced insulin resistance. In addition, PDE5A was found to suppress proteasome activity. Inhibition of PDE5 by its selective inhibitor icariin restored PDE5A overexpression-reduced proteasome activity and mitigated PDE5A overexpression-induced ER stress. Consistently, icariin administration also markedly attenuated the detrimental impacts of PDE5A overexpression on insulin signaling. CONCLUSIONS These results suggest that PDE5A suppresses proteasome activity, which results in ER stress and subsequent insulin resistance in C2C12 myotubes.
Collapse
Affiliation(s)
- Wei Liu
- Department of Otorhinolaryngology, Head and Neck Surgery, The Second People's Hospital of Jingzhou City, Jingzhou, 434000 Hubei, China
| | - Xiaojun Tian
- Department of Critical Care Medicine, The Second People's Hospital of Jingzhou City, Jingzhou, 434000 Hubei, China
| | - Ti Wu
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, 430071 Hubei, China
| | - Le Liu
- Department of Pathology and Pathophysiology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071 Hubei, China
| | - Yanghongyun Guo
- Department of Pathology and Pathophysiology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071 Hubei, China
| | - Changhua Wang
- Department of Pathology and Pathophysiology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071 Hubei, China
| |
Collapse
|
104
|
Katuri A, Bryant JL, Patel D, Patel V, Andhavarapu S, Asemu G, Davis H, Makar TK. HIVAN associated tubular pathology with reference to ER stress, mitochondrial changes, and autophagy. Exp Mol Pathol 2018; 106:139-148. [PMID: 30605635 DOI: 10.1016/j.yexmp.2018.12.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 11/25/2018] [Accepted: 12/29/2018] [Indexed: 12/31/2022]
Abstract
Human immunodeficiency virus associated nephropathy (HIVAN) is a unique form of a renal parenchymal disorder. This disease and its characteristics can be accredited to incorporation of DNA and mRNA of human immunodeficiency virus type 1 into the renal parenchymal cells. A proper understanding of the intricacies of HIVAN and the underlying mechanisms associated with renal function and disorders is vital for the potential development of a reliable treatment for HIVAN. Specifically, the renal tubule segment of the kidney is characterized by its transport capabilities and its ability to reabsorb water and salts into the blood. However, the segment is also known for certain disorders, such as renal tubular epithelial cell infection and microcyst formation, which are also closely linked to HIVAN. Furthermore, certain organelles, like the endoplasmic reticulum (ER), mitochondria, and lysosome, are vital for certain underlying mechanisms in kidney cells. A paradigm of the importance of said organelles can be seen in documented cases of HIVAN where the renal disorder results increased ER stress due to HIV viral propagation. This balance can be restored through the synthesis of secretory proteins, but, in return, the secretion requires more energy; therefore, there is a noticeable increase in mitochondrial stress. The increased ER changes and mitochondrial stress will greatly upregulate the process of autophagy, which involves the cell's lysosomes. In conjunction, we found that ER stress and mitochondrial changes are associated in the Tg26 animal model of HIVAN. The aim of our review is to consolidate current knowledge of important mechanisms in HIVAN, specifically related to the renal tubules' association with ER stress, mitochondrial changes and autophagy. Although the specific regulatory mechanism detailing the cross-talk between the various organelles is unknown in HIVAN, the continued research in this field may potentially shed light on a possible improved treatment for HIVAN.
Collapse
Affiliation(s)
- Akhil Katuri
- Department of Neurology, University of Maryland, Baltimore, MD 21201, United States
| | - Joseph L Bryant
- Animal Model Division, Institute of Human Virology, Baltimore, MD 21201, United States
| | - Dhruvil Patel
- Department of Neurology, University of Maryland, Baltimore, MD 21201, United States
| | - Vivek Patel
- Department of Neurology, University of Maryland, Baltimore, MD 21201, United States
| | - Sanketh Andhavarapu
- Department of Neurology, University of Maryland, Baltimore, MD 21201, United States
| | - Girma Asemu
- Animal Model Division, Institute of Human Virology, Baltimore, MD 21201, United States
| | - Harry Davis
- Animal Model Division, Institute of Human Virology, Baltimore, MD 21201, United States
| | - Tapas K Makar
- Department of Neurology, University of Maryland, Baltimore, MD 21201, United States; VA Medical Center, Baltimore, MD 21201, United States.
| |
Collapse
|
105
|
Chatterjee J, Langhnoja J, Pillai PP, Mustak MS. Neuroprotective effect of quercetin against radiation-induced endoplasmic reticulum stress in neurons. J Biochem Mol Toxicol 2018; 33:e22242. [DOI: 10.1002/jbt.22242] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 08/16/2018] [Accepted: 09/07/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Jit Chatterjee
- Department of Applied Zoology; Mangalore University, Mangalagangotri; Mangalore India
| | - Jaldeep Langhnoja
- Division of Neurobiology, Department of Zoology; Maharaja Sayajirao University of Baroda; Pratapgunj, Vadodara, Gujarat India
| | - Prakash P Pillai
- Division of Neurobiology, Department of Zoology; Maharaja Sayajirao University of Baroda; Pratapgunj, Vadodara, Gujarat India
| | - Mohammed S Mustak
- Department of Applied Zoology; Mangalore University, Mangalagangotri; Mangalore India
| |
Collapse
|
106
|
Danilova T, Lindahl M. Emerging Roles for Mesencephalic Astrocyte-Derived Neurotrophic Factor (MANF) in Pancreatic Beta Cells and Diabetes. Front Physiol 2018; 9:1457. [PMID: 30386256 PMCID: PMC6198132 DOI: 10.3389/fphys.2018.01457] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 09/26/2018] [Indexed: 12/31/2022] Open
Abstract
Mesencephalic astrocyte-derived neurotrophic factor (MANF) was originally identified as a secreted trophic factor for dopamine neurons in vitro. It protects and restores damaged cells in rodent models of Parkinson's disease, brain and heart ischemia, spinocerebellar ataxia and retina in vivo. However, its exact mechanism of action is not known. MANF is widely expressed in most human and mouse organs with high levels in secretory tissues. Intracellularly, MANF localizes to the endoplasmic reticulum (ER) and ER stress increases it's expression in cells and tissues. Furthermore, increased MANF levels has been detected in the sera of young children with newly diagnosed Type 1 (T1D) diabetes and Type 2 (T2D) diabetic patients. ER stress is caused by the accumulation of misfolded and aggregated proteins in the ER. It activates a cellular defense mechanism, the unfolded protein response (UPR), a signaling cascade trying to restore ER homeostasis. However, if prolonged, unresolved ER stress leads to apoptosis. Unresolved ER stress contributes to the progressive death of pancreatic insulin-producing beta cells in both T1D and T2D. Diabetes mellitus is characterized by hyperglycemia, caused by the inability of the beta cells to maintain sufficient levels of circulating insulin. The current medications, insulin and antidiabetic drugs, alleviate diabetic symptoms but cannot reconstitute physiological insulin secretion which increases the risk of devastating vascular complications of the disease. Thus, one of the main strategies in improving current diabetes therapy is to define and validate novel approaches to protect beta cells from stress as well as activate their regeneration. Embryonic deletion of the Manf gene in mice led to gradual postnatal development of insulin-deficient diabetes caused by reduced beta cell proliferation and increased beta cell death due to increased and sustained ER stress. In vitro, recombinant MANF partly protected mouse and human beta cells from ER stress-induced beta cell death and potentiated mouse and human beta cell proliferation. Importantly, in vivo overexpression of MANF in the pancreas of T1D mice led to increased beta cell proliferation and decreased beta cell death, suggesting that MANF could be a new therapeutic candidate for beta cell protection and regeneration in diabetes.
Collapse
Affiliation(s)
- Tatiana Danilova
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Maria Lindahl
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| |
Collapse
|
107
|
Shu S, Zhu J, Liu Z, Tang C, Cai J, Dong Z. Endoplasmic reticulum stress is activated in post-ischemic kidneys to promote chronic kidney disease. EBioMedicine 2018; 37:269-280. [PMID: 30314894 PMCID: PMC6286638 DOI: 10.1016/j.ebiom.2018.10.006] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 09/22/2018] [Accepted: 10/02/2018] [Indexed: 02/06/2023] Open
Abstract
Background Acute kidney injury (AKI) may lead to the development of chronic kidney disease (CKD), i.e. AKI-CKD transition, but the underlying mechanism remains largely unclear. Endoplasmic reticulum (ER) stress is characterized by the accumulation of unfolded or misfolded proteins in ER resulting in a cellular stress response. The role of ER stress in AKI-CKD transition remains unknown. Methods In this study, we examined ER stress in the mouse model of AKI-CKD transition after unilateral renal ischemia-reperfusion injury (uIR). To determine the role of ER stress in AKI-CKD transition, we tested the effects of two chemical chaperones: Tauroursodeoxycholic acid (TUDCA) and 4-phenylbutyric acid (4-PBA). Findings uIR led to the induction of ER stress in kidneys, as indicated by increased expression of UPR molecules CHOP (C/EBP homologous protein) and BiP(binding immunoglobulin protein; also called GRP78–78 kDa glucoseregulated protein). Given at 3 days after uIR, both TUDCA and 4-PBA blocked ER stress in post-ischemic kidneys. Notably, both chemicals promoted renal recovery and suppressed tubulointerstitial injury as manifested by the reduction of tubular atrophy, renal fibrosis and myofibroblast activation. Inhibition of ER stress further attenuated renal tubular epithelial cell apoptosis, inflammation and autophagy in post-ischemic kidneys. Interpretation These findings suggest that ER stress contributes critically to the development of chronic kidney pathologies and CKD following AKI, and inhibition of ER stress may represent a potential therapeutic strategy to impede AKI-CKD transition.
Collapse
Affiliation(s)
- Shaoqun Shu
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, Hunan, China
| | - Jiefu Zhu
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, Hunan, China; Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Zhiwen Liu
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, Hunan, China
| | - Chengyuan Tang
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, Hunan, China
| | - Juan Cai
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, Hunan, China
| | - Zheng Dong
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, Hunan, China; Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, GA, USA.
| |
Collapse
|
108
|
van Raaij S, Masereeuw R, Swinkels D, van Swelm R. Inhibition of Nrf2 alters cell stress induced by chronic iron exposure in human proximal tubular epithelial cells. Toxicol Lett 2018; 295:179-186. [DOI: 10.1016/j.toxlet.2018.06.1218] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 06/18/2018] [Accepted: 06/26/2018] [Indexed: 01/19/2023]
|
109
|
Min SY, Ha DS, Ha TS. Puromycin aminonucleoside triggers apoptosis in podocytes by inducing endoplasmic reticulum stress. Kidney Res Clin Pract 2018; 37:210-221. [PMID: 30254845 PMCID: PMC6147198 DOI: 10.23876/j.krcp.2018.37.3.210] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 05/14/2018] [Accepted: 06/26/2018] [Indexed: 01/13/2023] Open
Abstract
Background Puromycin aminonucleoside (PAN) is a known podocytotoxin. PAN-induced nephrosis is a widely used animal model for studying human idiopathic nephrotic syndrome. Abnormal protein accumulation associated with podocyte-specific endoplasmic reticulum (ER) stress damages cells structurally and functionally, which in turn induces apoptosis and severe proteinuria. In the present study, we investigated the effect of PAN on ER stress and apoptosis in podocytes in vitro. Methods Mouse podocytes were cultured and treated with various concentrations of PAN. ER stress markers were then evaluated by western blotting, and apoptosis was evaluated by fluorescence-activated cell sorting (FACS) and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assays. Results PAN treatment increased ER stress markers such as activating transcription factor (ATF) 6α and caspase-12 in a dose-dependent manner at 12 and 24 hours, respectively. These markers were reduced by chemical chaperones, such as sodium 4-phenylbutyric acid and tauroursodeoxycholic acid. PAN treatment also increased 78 kD glucose-regulated protein (GRP78)/binding immunoglobulin protein (BiP) at the earlier stage of 12 hours. PAN significantly induced podocyte apoptosis in concentration- and time-dependent manners, as seen using FACS and TUNEL assays. This result was improved by Nox4 siRNA, ATF6 siRNA, and chemical chaperones. LY294002, a PI3-kinase inhibitor, significantly boosted ER stress and apoptosis. PAN-induced ER stress increased oxidative stress and subsequently induced apoptosis, and could be mitigated by inhibition of PI3-kinase signaling. Conclusion Our findings suggest that PAN induces ER stress in podocytes mainly through the GRP78/BiP, ATF6α, and caspase-12 pathways, which trigger apoptosis via induction of oxidative stress. This stress is mitigated by inhibiting PI3-kinase signaling.
Collapse
Affiliation(s)
- Seo-Yun Min
- Department of Pediatrics, Chungbuk National University College of Medicine, Cheongju, Korea
| | - Dong-Soo Ha
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Tae-Sun Ha
- Department of Pediatrics, Chungbuk National University College of Medicine, Cheongju, Korea
| |
Collapse
|
110
|
Nougarède A, Tesnière C, Ylanko J, Rimokh R, Gillet G, Andrews DW. Improved IRE1 and PERK Pathway Sensors for Multiplex Endoplasmic Reticulum Stress Assay Reveal Stress Response to Nuclear Dyes Used for Image Segmentation. Assay Drug Dev Technol 2018; 16:350-360. [PMID: 30088945 DOI: 10.1089/adt.2018.862] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In response to a variety of insults the unfolded protein response (UPR) is a major cell program quickly engaged to promote either cell survival or if stress levels cannot be relieved, apoptosis. UPR relies on three major pathways, named from the endoplasmic reticulum (ER) resident proteins IRE1α, PERK, and ATF6 that mediate response. Current tools to measure the activation of these ER stress response pathways in mammalian cells are cumbersome and not compatible with high-throughput imaging. In this study, we present IRE1α and PERK sensors with improved sensitivity, based on the canonical events of xbp1 splicing and ATF4 translation at ORF3. These sensors can be integrated into host cell genomes through lentiviral transduction, opening the way for use in a wide array of immortalized or primary mammalian cells. We demonstrate that high-throughput single-cell analysis offers unprecedented kinetic details compared with endpoint measurement of IRE1α and PERK activity. Finally, we point out the limitations of dye-based nuclear segmentation for live cell imaging applications, as we show that these dyes induce UPR and can strongly affect both the kinetic and dynamic responses of IRE1α and PERK pathways.
Collapse
Affiliation(s)
- Adrien Nougarède
- 1 Biological Sciences Platform, Sunnybrook Research Institute , Sunnybrook Health Science Centre, Toronto, Ontario, Canada
| | - Chloé Tesnière
- 2 Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon , Lyon, France
| | - Jarkko Ylanko
- 1 Biological Sciences Platform, Sunnybrook Research Institute , Sunnybrook Health Science Centre, Toronto, Ontario, Canada
| | - Ruth Rimokh
- 2 Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon , Lyon, France
| | - Germain Gillet
- 2 Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon , Lyon, France .,3 Laboratoire d'anatomie et Cytologie Pathologiques, Hospices Civils de Lyon, Centre Hospitalier Lyon Sud , Pierre Bénite, France
| | - David W Andrews
- 1 Biological Sciences Platform, Sunnybrook Research Institute , Sunnybrook Health Science Centre, Toronto, Ontario, Canada .,4 Department of Medical Biophysics, University of Toronto , Toronto, Ontario, Canada .,5 Department of Biochemistry, University of Toronto , Toronto, Ontario, Canada
| |
Collapse
|
111
|
Gewin LS. Renal fibrosis: Primacy of the proximal tubule. Matrix Biol 2018; 68-69:248-262. [PMID: 29425694 PMCID: PMC6015527 DOI: 10.1016/j.matbio.2018.02.006] [Citation(s) in RCA: 153] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 02/02/2018] [Accepted: 02/03/2018] [Indexed: 12/20/2022]
Abstract
Tubulointerstitial fibrosis (TIF) is the hallmark of chronic kidney disease and best predictor of renal survival. Many different cell types contribute to TIF progression including tubular epithelial cells, myofibroblasts, endothelia, and inflammatory cells. Previously, most of the attention has centered on myofibroblasts given their central importance in extracellular matrix production. However, emerging data focuses on how the response of the proximal tubule, a specialized epithelial segment vulnerable to injury, plays a central role in TIF progression. Several proximal tubular responses such as de-differentiation, cell cycle changes, autophagy, and metabolic changes may be adaptive initially, but can lead to maladaptive responses that promote TIF both through autocrine and paracrine effects. This review discusses the current paradigm of TIF progression and the increasingly important role of the proximal tubule in promoting TIF both in tubulointerstitial and glomerular injuries. A better understanding and appreciation of the role of the proximal tubule in TIF has important implications for therapeutic strategies to halt chronic kidney disease progression.
Collapse
Affiliation(s)
- Leslie S Gewin
- The Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, United States.
| |
Collapse
|
112
|
Gallazzini M, Pallet N. Endoplasmic reticulum stress and kidney dysfunction. Biol Cell 2018; 110:205-216. [DOI: 10.1111/boc.201800019] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 06/14/2018] [Accepted: 06/28/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Morgan Gallazzini
- INSERM U1151 - CNRS UMR 8253; Institut Necker Enfants Malades; Paris France
- INSERM U1147; Centre Universitaire des Saints Pères; Paris France
| | - Nicolas Pallet
- INSERM U1151 - CNRS UMR 8253; Institut Necker Enfants Malades; Paris France
- INSERM U1147; Centre Universitaire des Saints Pères; Paris France
- Université Paris Descartes; Paris France
- Service de Néphrologie; Hôpital Européen Georges Pompidou; Paris
- Service de Biochimie; Hôpital Européen Gorges Pompidou; Paris France
| |
Collapse
|
113
|
Yan M, Shu S, Chunyuan G, Tang C, Dong Z. Endoplasmic reticulum stress in ischemic and nephrotoxic acute kidney injury. Ann Med 2018; 50:381-390. [PMID: 29895209 PMCID: PMC6333465 DOI: 10.1080/07853890.2018.1489142] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 03/02/2018] [Accepted: 03/19/2018] [Indexed: 12/13/2022] Open
Abstract
Acute kidney injury (AKI) is a medical condition characterized by kidney damage with a rapid decline of renal function, which is associated with high mortality and morbidity. Recent research has further established an intimate relationship between AKI and chronic kidney disease. Perturbations of kidney cells in AKI result in the accumulation of unfolded and misfolded proteins in the endoplasmic reticulum (ER), leading to unfolded protein response (UPR) or ER stress. In this review, we analyze the role and regulation of ER stress in AKI triggered by renal ischemia-reperfusion and cisplatin nephrotoxicity. The balance between the two major components of UPR, the adaptive pathway and the apoptotic pathway, plays a critical role in determining the cell fate in ER stress. The adaptive pathway is evoked to attenuate translation, induce chaperones, maintain protein homeostasis and promote cell survival. Prolonged ER stress activates the apoptotic pathway, resulting in the elimination of dysfunctional cells. Therefore, regulating ER stress in kidney cells may provide a therapeutic target in AKI. KEY MESSAGES Perturbations of kidney cells in acute kidney injury result in the accumulation of unfolded and misfolded proteins in ER, leading to unfolded protein response (UPR) or ER stress. The balance between the adaptive pathway and the apoptotic pathway of UPR plays a critical role in determining the cell fate in ER stress. Modulation of ER stress in kidney cells may provide a therapeutic strategy for acute kidney injury.
Collapse
Affiliation(s)
- Mingjuan Yan
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Nephrology, The First people’s Hospital of Changde City, Changde, Hunan, China
| | - Shaoqun Shu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guo Chunyuan
- Department of Nephrology, The First people’s Hospital of Changde City, Changde, Hunan, China
| | - Chengyuan Tang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zheng Dong
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia, U.S.A
| |
Collapse
|
114
|
Tsvetkov P, Adler J, Myers N, Biran A, Reuven N, Shaul Y. Oncogenic addiction to high 26S proteasome level. Cell Death Dis 2018; 9:773. [PMID: 29991718 PMCID: PMC6039477 DOI: 10.1038/s41419-018-0806-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 05/15/2018] [Accepted: 05/17/2018] [Indexed: 12/15/2022]
Abstract
Proteasomes are large intracellular complexes responsible for the degradation of cellular proteins. The altered protein homeostasis of cancer cells results in increased dependency on proteasome function. The cellular proteasome composition comprises the 20S catalytic complex that is frequently capped with the 19S regulatory particle in forming the 26S proteasome. Proteasome inhibitors target the catalytic barrel (20S) and thus this inhibition does not allow the deconvolution of the distinct roles of 20S versus 26S proteasomes in cancer progression. We examined the degree of dependency of cancer cells specifically to the level of the 26S proteasome complex. Oncogenic transformation of human and mouse immortalized cells with mutant Ras induced a strong posttranscriptional increase of the 26S proteasome subunits, giving rise to high 26S complex levels. Depletion of a single subunit of the 19S RP was sufficient to reduce the 26S proteasome level and lower the cellular 26S/20S ratio. Under this condition the viability of the Ras-transformed MCF10A cells was severely compromised. This observation led us to hypothesize that cancer cell survival is dependent on maximal utilization of its 26S proteasomes. We validated this possibility in a large number of cancer cell lines and found that partial reduction of the 26S proteasome level impairs viability in all cancer cells examined and was not correlated with cell doubling time or reduction efficiency. Interstingly, normal human fibroblasts are refractory to the same type of 26S proteasome reduction. The suppression of 26S proteasomes in cancer cells activated the UPR and caspase-3 and cells stained positive with Annexin V. In addition, suppression of the 26S proteasome resulted in cellular proteasome redistribution, cytoplasm shrinkage, and nuclear deformation, the hallmarks of apoptosis. The observed tumor cell-specific addiction to the 26S proteasome levels sets the stage for future strategies in exploiting this dependency in cancer therapy.
Collapse
Affiliation(s)
- Peter Tsvetkov
- Department of Molecular Genetics, Weizmann Institute of Science, 76100, Rehovot, Israel.,Broad Institute of MIT and Harvard, 415 Main St., Cambridge, MA, 02142, USA
| | - Julia Adler
- Department of Molecular Genetics, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Nadav Myers
- Department of Molecular Genetics, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Assaf Biran
- Department of Molecular Genetics, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Nina Reuven
- Department of Molecular Genetics, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Yosef Shaul
- Department of Molecular Genetics, Weizmann Institute of Science, 76100, Rehovot, Israel.
| |
Collapse
|
115
|
Anusornvongchai T, Nangaku M, Jao TM, Wu CH, Ishimoto Y, Maekawa H, Tanaka T, Shimizu A, Yamamoto M, Suzuki N, Sassa R, Inagi R. Palmitate deranges erythropoietin production via transcription factor ATF4 activation of unfolded protein response. Kidney Int 2018; 94:536-550. [PMID: 29887316 DOI: 10.1016/j.kint.2018.03.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Revised: 02/02/2018] [Accepted: 03/02/2018] [Indexed: 01/22/2023]
Abstract
Lipotoxicity plays an important role in the progression of chronic kidney damage via various mechanisms, such as endoplasmic reticulum stress. Several studies proposed renal lipotoxicity in glomerular and tubular cells but the effect of lipid on renal erythropoietin (EPO)-producing (REP) cells in the interstitium has not been elucidated. Since renal anemia is caused by derangement of EPO production in REP cells, we evaluated the effect of palmitate, a representative long-chain saturated fatty acid, on EPO production and the endoplasmic reticulum stress pathway. EPO production was suppressed by palmitate (palmitate-conjugated bovine serum albumin [BSA]) or a high palmitate diet, but not oleic acid-conjugated BSA or a high oleic acid diet, especially under cobalt-induced pseudo-hypoxia both in vitro and in vivo. Importantly, suppression of EPO production was not induced by a decrease in transcription factor HIF activity, while it was significantly associated with endoplasmic reticulum stress, particularly transcription factor ATF4 activation, which suppresses 3'-enhancer activity of the EPO gene. ATF4 knockdown by siRNA significantly attenuated the suppressive effect of palmitate on EPO production. Studies utilizing inherited super-anemic mice (ISAM) mated with EPO-Cre mice (ISAM-REC mice) for lineage-labeling of REP cells showed that ATF4 activation by palmitate suppressed EPO production in REP cells. Laser capture microdissection confirmed ATF4 activation in the interstitial area of ISAM-REC mice treated with palmitate-conjugated BSA. Thus, endoplasmic reticulum stress induced by palmitate suppressed EPO expression by REP cells in a manner independent of HIF activation. The link between endoplasmic reticulum stress, dyslipidemia, and hypoxia may contribute to development and progression of anemia in CKD.
Collapse
Affiliation(s)
- Thitinun Anusornvongchai
- Division of CKD Pathophysiology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan; Department of Internal Medicine, Lerdsin General Hospital, Department of Medical Services, Bangkok, Thailand
| | - Masaomi Nangaku
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Tzu-Ming Jao
- Division of CKD Pathophysiology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Chia-Hsien Wu
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Yu Ishimoto
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Hiroshi Maekawa
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Tetsuhiro Tanaka
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Akira Shimizu
- Department of Analytic Human Pathology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Masayuki Yamamoto
- Division of Interdisciplinary Medical Science, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Norio Suzuki
- Division of Oxygen Biology, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | | | - Reiko Inagi
- Division of CKD Pathophysiology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan.
| |
Collapse
|
116
|
Yee A, Papillon J, Guillemette J, Kaufman DR, Kennedy CRJ, Cybulsky AV. Proteostasis as a therapeutic target in glomerular injury associated with mutant α-actinin-4. Am J Physiol Renal Physiol 2018; 315:F954-F966. [PMID: 29873512 DOI: 10.1152/ajprenal.00082.2018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Mutations in α-actinin-4 (actinin-4) result in hereditary focal segmental glomerulosclerosis (FSGS) in humans. Actinin-4 mutants induce podocyte injury because of dysregulation of the cytoskeleton and proteotoxicity. Injury may be associated with endoplasmic reticulum (ER) stress and polyubiquitination of proteins. We assessed if the chemical chaperone 4-phenylbutyrate (4-PBA) can ameliorate the proteotoxicity of an actinin-4 mutant. Actinin-4 K255E, which causes FSGS in humans (K256E in the mouse), showed enhanced ubiquitination, accelerated degradation, aggregate formation, and enhanced association with filamentous (F)-actin in glomerular epithelial cells (GECs). The mutant disrupted ER function and stimulated autophagy. 4-PBA reduced actinin-4 K256E aggregation and its tight association with F-actin. Transgenic mice that express actinin-4 K256E in podocytes develop podocyte injury, proteinuria, and FSGS in association with glomerular ER stress. Treatment of these mice with 4-PBA in the drinking water over a 10-wk period significantly reduced albuminuria and ER stress. Another drug, celastrol, which enhanced expression of ER and cytosolic chaperones in GECs, tended to reduce actinin-4 aggregation but did not decrease the tight association of actinin-4 K256E with F-actin and did not reduce albuminuria in actinin-4 K256E transgenic mice. Thus, chemical chaperones, such as 4-PBA, may represent a novel therapeutic approach to certain hereditary glomerular diseases.
Collapse
Affiliation(s)
- Albert Yee
- Department of Medicine, McGill University Health Centre Research Institute, McGill University , Montreal, Quebec , Canada
| | - Joan Papillon
- Department of Medicine, McGill University Health Centre Research Institute, McGill University , Montreal, Quebec , Canada
| | - Julie Guillemette
- Department of Medicine, McGill University Health Centre Research Institute, McGill University , Montreal, Quebec , Canada
| | - Daniel R Kaufman
- Department of Medicine, McGill University Health Centre Research Institute, McGill University , Montreal, Quebec , Canada
| | - Chris R J Kennedy
- Kidney Research Centre, Department of Medicine, The Ottawa Hospital, University of Ottawa , Ottawa, Ontario , Canada
| | - Andrey V Cybulsky
- Department of Medicine, McGill University Health Centre Research Institute, McGill University , Montreal, Quebec , Canada
| |
Collapse
|
117
|
Jheng JR, Chen YS, Ao UI, Chan DC, Huang JW, Hung KY, Tarng DC, Chiang CK. The double-edged sword of endoplasmic reticulum stress in uremic sarcopenia through myogenesis perturbation. J Cachexia Sarcopenia Muscle 2018; 9:570-584. [PMID: 29380555 PMCID: PMC5989876 DOI: 10.1002/jcsm.12288] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 12/11/2017] [Accepted: 12/30/2017] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND Sarcopenia is the age-related degeneration characterized with the decline of skeletal muscle mass, strength, and function. The imbalance of protein synthesis and degradation which jeopardizes immune, hormone regulation, and muscle-motor neuron connection is the main cause of sarcopenia. There is limited knowledge regarding molecular mechanism of sarcopenia. As the endoplasmic reticulum is the control centre of the protein syntheses and degradation, we hypothesized that endoplasmic reticulum stress and unfolded protein response (UPR) play an important in the development of sarcopenia. Understanding the sarcopenia molecular mechanisms may benefit the therapeutic diagnosis and treatment in the future. METHODS Mouse myoblast C2C12 cells are exposed to designated time and concentration of indoxyl sulfate (IS), a uremic toxin of chronic kidney disease. The proliferation, differentiation, and the expression of atrogin 1 are examined. The protein and mRNA expression of IS treated-C2C12 cells are inspected to distinguish the role of ER stress and oxidative stress underlying the sarcopenia. RESULTS Indoxyl sulfate inhibits myoblast differentiation. We demonstrate that as the number of multi-nuclei myotube decreased, the differentiation markers including myoD, myoG, and myosin heavy chain are also suppressed. Indoxyl sulfate inhibits myoblast proliferation and induces the myotubular atrophy marker atrogin-1 protein expression. Indoxyl sulfate stimulates eIF2α phosphorylation and XBP1 mRNA splicing in UPR. Interestingly, the oxidative stress is related to eIF2α phosphorylation but not XBP1 mRNA splicing. The eIF2α phosphorylation triggered by IS reduces myoD, myoG, and myosin heavy chain protein expression, which represents the anti-myogenic modulation on the early differentiation event. The XBP1 mRNA splicing induced by IS, however, is considered the adaptive response to restore the myogenic differentiation. CONCLUSIONS Our studies indicated that the ER stress and UPR modulation are critical in the chronic kidney disease uremic toxin-accumulated sarcopenia model. We believe that UPR-related signals showed great potential in clinical application.
Collapse
Affiliation(s)
- Jia-Rong Jheng
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yuan-Siao Chen
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Un Iong Ao
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ding-Cheng Chan
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Geriatrics and Gerontology, National Taiwan University Hospital, Taipei, Taiwan.,Superintendent's Office, National Taiwan University Hospital, Chu-Tung Branch, Taipei, Taiwan
| | - Jenq-Wen Huang
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Kuang-Yu Hung
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Der-Cheng Tarng
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chih-Kang Chiang
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Integrated Diagnostics and Therapeutics, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
118
|
Taurine Supplementation Alleviates Puromycin Aminonucleoside Damage by Modulating Endoplasmic Reticulum Stress and Mitochondrial-Related Apoptosis in Rat Kidney. Nutrients 2018; 10:nu10060689. [PMID: 29843457 PMCID: PMC6024760 DOI: 10.3390/nu10060689] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 05/24/2018] [Accepted: 05/25/2018] [Indexed: 12/27/2022] Open
Abstract
Taurine (TAU) is a sulfur-containing beta amino acid that is not involved in protein composition and anabolism, conditionally essential in mammals provided through diet. Growing evidence supports a protective role of TAU supply in osmoregulation, calcium flux, and reduction of inflammation and oxidant damage in renal diseases like diabetes. Endoplasmic reticulum (ER) stress, due to abnormal proteostasis, is a contributor to nephrotic syndrome and related renal damage. Here, we investigated the effect of dietary TAU (1.5% in drinking water for 15 days) in an established rat model that mimics human minimal change nephrosis, consisting of a single puromycin aminonucleoside (PAN) injection (intraperitoneally 15 mg/100 g body weight), with sacrifice after eight days. TAU limited proteinuria and podocytes foot processes effacement, and balanced slit diaphragm nephrin and glomerular claudin 1 expressions. In cortical proximal tubules, TAU improved lysosomal density, ER perimeter, restored proper ER-mitochondria tethering and mitochondrial cristae, and decreased inflammation. Remarkably, TAU downregulated glomerular ER stress markers (GRP78, GRP94), pro-apoptotic C/EBP homologous protein, activated caspase 3, tubular caspase1, and mitochondrial chaperone GRP75, but maintained anti-apoptotic HSP25. In conclusion, TAU, by targeting upstream ER stress separate from mitochondria dysfunctions at crucial renal sites, might be a promising dietary supplement in the treatment of the drug-resistant nephrotic syndrome.
Collapse
|
119
|
Deshpande S, Abdollahi M, Wang M, Lanting L, Kato M, Natarajan R. Reduced Autophagy by a microRNA-mediated Signaling Cascade in Diabetes-induced Renal Glomerular Hypertrophy. Sci Rep 2018; 8:6954. [PMID: 29725042 PMCID: PMC5934412 DOI: 10.1038/s41598-018-25295-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 04/18/2018] [Indexed: 12/12/2022] Open
Abstract
Autophagy plays a key role in the pathogenesis of kidney diseases, however its role in diabetic nephropathy (DN), and particularly in kidney glomerular mesangial cells (MCs) is not very clear. Transforming Growth Factor- β1 (TGF-β), a key player in the pathogenesis of DN, regulates expression of various microRNAs (miRNAs), some of which are known to regulate the expression of autophagy genes. Here we demonstrate that miR-192, induced by TGF-β signaling, plays an important role in regulating autophagy in DN. The expression of key autophagy genes was decreased in kidneys of streptozotocin-injected type-1 and type-2 (db/db) diabetic mice and this was reversed by treatment with Locked Nucleic Acid (LNA) modified miR-192 inhibitors. Changes in autophagy gene expression were also attenuated in kidneys of diabetic miR-192-KO mice. In vitro studies using mouse glomerular mesangial cells (MMCs) also showed a decrease in autophagy gene expression with TGF-β treatment. miR-192 mimic oligonucleotides also decreased the expression of certain autophagy genes. These results demonstrate that TGF-β and miR-192 decrease autophagy in MMCs under diabetic conditions and this can be reversed by inhibition or deletion of miR-192, further supporting miR-192 as a useful therapeutic target for DN.
Collapse
Affiliation(s)
- Supriya Deshpande
- Department of Diabetes Complications and Metabolism, Diabetes Metabolism Research Institute and Beckman Research Institute of City of Hope, Duarte, California, 91010, USA
| | - Maryam Abdollahi
- Department of Diabetes Complications and Metabolism, Diabetes Metabolism Research Institute and Beckman Research Institute of City of Hope, Duarte, California, 91010, USA
| | - Mei Wang
- Department of Diabetes Complications and Metabolism, Diabetes Metabolism Research Institute and Beckman Research Institute of City of Hope, Duarte, California, 91010, USA
| | - Linda Lanting
- Department of Diabetes Complications and Metabolism, Diabetes Metabolism Research Institute and Beckman Research Institute of City of Hope, Duarte, California, 91010, USA
| | - Mitsuo Kato
- Department of Diabetes Complications and Metabolism, Diabetes Metabolism Research Institute and Beckman Research Institute of City of Hope, Duarte, California, 91010, USA.
| | - Rama Natarajan
- Department of Diabetes Complications and Metabolism, Diabetes Metabolism Research Institute and Beckman Research Institute of City of Hope, Duarte, California, 91010, USA.
| |
Collapse
|
120
|
Cornec-Le Gall E, Olson RJ, Besse W, Heyer CM, Gainullin VG, Smith JM, Audrézet MP, Hopp K, Porath B, Shi B, Baheti S, Senum SR, Arroyo J, Madsen CD, Férec C, Joly D, Jouret F, Fikri-Benbrahim O, Charasse C, Coulibaly JM, Yu AS, Khalili K, Pei Y, Somlo S, Le Meur Y, Torres VE, Harris PC. Monoallelic Mutations to DNAJB11 Cause Atypical Autosomal-Dominant Polycystic Kidney Disease. Am J Hum Genet 2018; 102:832-844. [PMID: 29706351 PMCID: PMC5986722 DOI: 10.1016/j.ajhg.2018.03.013] [Citation(s) in RCA: 212] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 03/08/2018] [Indexed: 01/05/2023] Open
Abstract
Autosomal-dominant polycystic kidney disease (ADPKD) is characterized by the progressive development of kidney cysts, often resulting in end-stage renal disease (ESRD). This disorder is genetically heterogeneous with ∼7% of families genetically unresolved. We performed whole-exome sequencing (WES) in two multiplex ADPKD-like pedigrees, and we analyzed a further 591 genetically unresolved, phenotypically similar families by targeted next-generation sequencing of 65 candidate genes. WES identified a DNAJB11 missense variant (p.Pro54Arg) in two family members presenting with non-enlarged polycystic kidneys and a frameshifting change (c.166_167insTT) in a second family with small renal and liver cysts. DNAJB11 is a co-factor of BiP, a key chaperone in the endoplasmic reticulum controlling folding, trafficking, and degradation of secreted and membrane proteins. Five additional multigenerational families carrying DNAJB11 mutations were identified by the targeted analysis. The clinical phenotype was consistent in the 23 affected members, with non-enlarged cystic kidneys that often evolved to kidney atrophy; 7 subjects reached ESRD from 59 to 89 years. The lack of kidney enlargement, histologically evident interstitial fibrosis in non-cystic parenchyma, and recurring episodes of gout (one family) suggested partial phenotypic overlap with autosomal-dominant tubulointerstitial diseases (ADTKD). Characterization of DNAJB11-null cells and kidney samples from affected individuals revealed a pathogenesis associated with maturation and trafficking defects involving the ADPKD protein, PC1, and ADTKD proteins, such as UMOD. DNAJB11-associated disease is a phenotypic hybrid of ADPKD and ADTKD, characterized by normal-sized cystic kidneys and progressive interstitial fibrosis resulting in late-onset ESRD.
Collapse
Affiliation(s)
- Emilie Cornec-Le Gall
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA; Department of Nephrology, University Hospital, European University of Brittany, Brest, Brittany 29200, France; Department of Molecular Genetics, National Institute of Health and Medical Sciences, INSERM U1078, Brest 29200, France
| | - Rory J Olson
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Whitney Besse
- Section of Nephrology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Christina M Heyer
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Jessica M Smith
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA
| | - Marie-Pierre Audrézet
- Department of Molecular Genetics, National Institute of Health and Medical Sciences, INSERM U1078, Brest 29200, France
| | - Katharina Hopp
- Division of Renal Diseases and Hypertension, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80202, USA
| | - Binu Porath
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA
| | - Beili Shi
- Division of Nephrology, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Saurabh Baheti
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN 55905, USA
| | - Sarah R Senum
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA
| | - Jennifer Arroyo
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA
| | - Charles D Madsen
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA
| | - Claude Férec
- Department of Molecular Genetics, National Institute of Health and Medical Sciences, INSERM U1078, Brest 29200, France
| | - Dominique Joly
- Service of Nephrology, Necker Hospital, Paris 75231, France
| | - François Jouret
- Division of Nephrology, University of Liège, Liège 4000, Belgium
| | | | | | | | - Alan S Yu
- Kidney Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Korosh Khalili
- Department of Medical Imaging, University Health Network, Toronto, ON M5G 2C4, Canada
| | - York Pei
- Division of Nephrology, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Stefan Somlo
- Section of Nephrology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Yannick Le Meur
- Department of Nephrology, University Hospital, European University of Brittany, Brest, Brittany 29200, France
| | - Vicente E Torres
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA
| | - Peter C Harris
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA; Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
121
|
Zhang H, Zhang W, Jiao F, Li X, Zhang H, Wang L, Gong Z. The Nephroprotective Effect of MS-275 on Lipopolysaccharide (LPS)-Induced Acute Kidney Injury by Inhibiting Reactive Oxygen Species (ROS)-Oxidative Stress and Endoplasmic Reticulum Stress. Med Sci Monit 2018; 24:2620-2630. [PMID: 29704392 PMCID: PMC5944402 DOI: 10.12659/msm.906362] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background Histone deacetylase (HDAC) inhibitors can attenuate acute kidney injury (AKI)-mediated damage and reduce fibrosis in kidney disease models. The aim of the present study was to investigate the effects of the HDAC inhibitor MS-275 on lipopolysaccharide (LPS)-induced AKI and the associated mechanisms. Material/Methods A LPS-induced model in 6–8 weeks-old mice was established by intraperitoneal injection of LPS (10 mg/kg), with pre-treatment of MS-275 (2 mg/kg/day) administered intraperitoneally for five days. In addition, HK-2 cells were exposed to LPS (1 μg/mL) at 0.1 nM, 1 nM, 10 nM, and 100 nM. For our in vitro MS-275 study, detection programs included histology, biochemical, immunohistochemistry, mRNA and protein expression as well as apoptosis. Results MS-275 ameliorated renal damage, enhanced the survival rate of the LPS-induced sepsis model, decreased the expressions of TNF-α, IL-1β, IL-6, COX-2, and NF-κBp65 nucleus translocation, suppressed the HDAC activity which was enhanced in septic AKI mice, and enhanced the acetylation of histone H3 and H4. Reactive oxygen species (ROS) production was enhanced in the kidney of LPS mice compared to control mice, while MS-275 suppressed the production of ROS in kidney tissue. In the in vitro studies, MS-275 reduced the LPS-induced apoptosis of HK-2 cells, inhibited ROS and MDA production, increased the production GSH and SOD activity, decreased the expressions of CHOP, GRP78, caspase3, and capase12, which was related to endoplasmic reticulum stress in LPS stimulated HK-2 cells. Conclusions MS-275 pre-treatment improved renal function and ameliorated histological alterations, inflammation, and ROS production in LPS-induced AKI mice and may act through inhibiting ROS-oxidative stress and endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Haiyue Zhang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei, China (mainland)
| | - Wenbin Zhang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei, China (mainland)
| | - Fangzhou Jiao
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei, China (mainland)
| | - Xun Li
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei, China (mainland)
| | - Hong Zhang
- Department of Pharmaceutical, Renmin Hospital of Wuhan University, Wuhan, Hubei, China (mainland)
| | - Luwen Wang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei, China (mainland)
| | - Zuojiong Gong
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei, China (mainland)
| |
Collapse
|
122
|
Mito-TEMPO Alleviates Renal Fibrosis by Reducing Inflammation, Mitochondrial Dysfunction, and Endoplasmic Reticulum Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:5828120. [PMID: 29765500 PMCID: PMC5889907 DOI: 10.1155/2018/5828120] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/01/2018] [Accepted: 01/15/2018] [Indexed: 12/14/2022]
Abstract
Background Renal fibrosis is a common pathological symptom of chronic kidney disease (CKD). Many studies support that mitochondrial dysfunction and endoplasmic reticulum (ER) stress are implicated in the pathogenesis of CKD. In our study, we investigated the benefits and underlying mechanisms of Mito-TEMPO on renal fibrosis in 5/6 nephrectomy mice. Methods Mice were randomly divided into five groups as follows: control group, CKD group, CKD + Mito-TEMPO (1 mg·kg-1·day-1) group, CKD + Mito-TEMPO (3 mg·kg-1·day-1) group, and Mito-TEMPO group (3 mg·kg-1·day-1). Renal fibrosis was evaluated by PAS, Masson staining, immunohistochemistry, and real-time PCR. Oxidative stress markers such as SOD2 activity and MDA level in serum and isolated mitochondria from renal tissue were measured by assay kits. Mitochondrial superoxide production was evaluated by MitoSOX staining and Western blot. Mitochondrial dysfunction was assessed by electron microscopy and real-time PCR. ER stress-associated protein was measured by Western blot. Results Impaired renal function and renal fibrosis were significantly improved by Mito-TEMPO treatment. Furthermore, inflammation cytokines, profibrotic factors, oxidative stress markers, mitochondrial dysfunction, and ER stress were all increased in the CKD group. However, these effects were significantly ameliorated in the Mito-TEMPO treatment group. Conclusions Mito-TEMPO ameliorates renal fibrosis by alleviating mitochondrial dysfunction and endoplasmic reticulum stress possibly through the Sirt3-SOD2 pathway, which sheds new light on prevention of renal fibrosis in chronic kidney disease.
Collapse
|
123
|
Renal Proteomic Responses to Severe Sepsis and Surgical Trauma: Dynamic Analysis of Porcine Tissue Biopsies. Shock 2018; 46:453-64. [PMID: 27070328 DOI: 10.1097/shk.0000000000000613] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Although the burden of septic acute kidney injury continues to increase, the molecular pathogenesis remains largely obscure. The aim of this exploratory study was a discovery-driven analysis of dynamic kidney tissue protein expression changes applied for the first time in a classic large mammal model of sepsis. To achieve this goal, analyses of protein expression alterations were performed in serial samples of kidney cortical biopsies (before, 12 and 22 h of sepsis) in mechanically ventilated pigs challenged with continuous infusion of pseudomonas aeruginosa and compared with sham-operated control data. Global protein expression was analyzed using two-dimensional gel electrophoresis and mass spectrometry-based proteomics. Normodynamic sepsis was associated with 43% reduction in glomerular filtration. The exposure to surgical stress per se altered the renal protein expression profile, while sepsis induced distinct and highly dynamic proteome evolution shifting the balance toward cellular distress phenotype. We identified 20 proteins whose expression changes discriminated effects of sepsis from those induced by surgery. The data implicate endoplasmic reticulum stress, oxidative stress, mitochondrial energy metabolism, immune/inflammatory signaling, and tubular transport as major activated pathways. Thus, by coupling the power of sequential tissue proteomics with whole-animal physiological studies, our study helped to establish a first global overview of critical renal proteomic events occurring during surgical trauma and early sepsis in a porcine model. The study supports the notion that multiple potentially subtle and even transient changes in several proteins which are members of key functional interrelated systems appear to play a role in septic acute kidney injury.
Collapse
|
124
|
Fohlen B, Tavernier Q, Huynh TM, Caradeuc C, Le Corre D, Bertho G, Cholley B, Pallet N. Real-Time and Non-invasive Monitoring of the Activation of the IRE1α-XBP1 Pathway in Individuals with Hemodynamic Impairment. EBioMedicine 2017; 27:284-292. [PMID: 29276149 PMCID: PMC5828547 DOI: 10.1016/j.ebiom.2017.12.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 12/07/2017] [Accepted: 12/19/2017] [Indexed: 02/06/2023] Open
Abstract
Many stressors that are encountered upon kidney injury are likely to trigger endoplasmic reticulum (ER) stress, subsequently activating transcriptional, translational and metabolic reprogramming. Monitoring early cellular adaptive responses engaged after hemodynamic impairment yields may represent a clinically relevant approach. However, a non-invasive method for detecting the ER stress response has not been developed. We combined a metabolomic approach with genetic marker analyses using urine from individuals undergoing scheduled cardiac surgery under cardiopulmonary bypass to investigate the feasibility and significance of monitoring the ER stress response in the kidney. We developed an original method based on fragment analysis that measures urinary levels of the spliced X-box binding protein 1 (sXBP1) mRNA as a proxy of inositol-requiring enzyme 1α (IRE1α) activity because sXBP1 is absolutely sensitive and specific for ER stress. The early engagement of the ER stress response after ischemic stress is critical for protecting against tissue damage, and individuals who mount a robust adaptive response are protected against AKI. The clinical consequences of our findings are of considerable importance because ER stress is involved in numerous conditions that lead to AKI and chronic kidney disease; in addition, the detection of ER stress is straightforward and immediately available in routine practice. Endoplasmic Reticulum (ER) stress is involved in the pathophysiology of numerous kidney diseases. We developed a method based on fragment analysis that measures urinary levels of XBP1 mRNA to detect renal ER stress. ER stress occurs early after cardiopulmonary bypass, a procedure leading to acute kidney injury. The detection of renal ER stress in this context can predict the occurrence of acute kidney injury.
The better care of patients with kidney disease requires the identification of biomarkers of ongoing tissue injury to provide therapies to slow disease progression. In this study, we have developed for the first time a non-invasive (urinary) biomarker of a protective cellular process called Endoplasmic Reticulum stress that occurs early in the kidney after ischemic injury. Renal ischemic injury follows cardiac surgery and could lead to acute kidney injury. Our results indicate that the early detection of individuals who do not activate Endoplasmic Reticulum stress could help to identify individuals who will develop acute kidney injury.
Collapse
Affiliation(s)
- Baptiste Fohlen
- Department d'Anésthésie et de Réanimation Cardiovasculaire, Hôpital Européen Georges Pompidou, Paris, France; Assistance Publique-Hôpitaux de Paris (APHP), Paris, France; Université Paris Descartes, Paris, France
| | - Quentin Tavernier
- Université Paris Descartes, Paris, France; Institut National de la Santé et la Recherche Médicale (INSERM) U1147, Paris, France
| | - Thi-Mum Huynh
- Department d'Anésthésie et de Réanimation Cardiovasculaire, Hôpital Européen Georges Pompidou, Paris, France; Assistance Publique-Hôpitaux de Paris (APHP), Paris, France; Université Paris Descartes, Paris, France
| | - Cédric Caradeuc
- Université Paris Descartes, Paris, France; Centre National pour le Recherche Scientifique (CNRS) U8601, Paris, France
| | - Delphine Le Corre
- Université Paris Descartes, Paris, France; Institut National de la Santé et la Recherche Médicale (INSERM) U1147, Paris, France
| | - Gildas Bertho
- Université Paris Descartes, Paris, France; Centre National pour le Recherche Scientifique (CNRS) U8601, Paris, France
| | - Bernard Cholley
- Department d'Anésthésie et de Réanimation Cardiovasculaire, Hôpital Européen Georges Pompidou, Paris, France; Assistance Publique-Hôpitaux de Paris (APHP), Paris, France; Université Paris Descartes, Paris, France
| | - Nicolas Pallet
- Assistance Publique-Hôpitaux de Paris (APHP), Paris, France; Université Paris Descartes, Paris, France; Institut National de la Santé et la Recherche Médicale (INSERM) U1147, Paris, France; Service de Néphrologie, Hôpital Européen Georges Pompidou, Paris, France; Service de Biochimie, Hôpital Européen Georges Pompidou, Paris, France.
| |
Collapse
|
125
|
Chemical chaperon 4-phenylbutyrate protects against the endoplasmic reticulum stress-mediated renal fibrosis in vivo and in vitro. Oncotarget 2017; 7:22116-27. [PMID: 26959118 PMCID: PMC5008348 DOI: 10.18632/oncotarget.7904] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 02/21/2016] [Indexed: 12/16/2022] Open
Abstract
Renal tubulointerstitial fibrosis is the common and final pathologic change of kidney in end-stage renal disease. Interesting, endoplasmic reticulum (ER) stress is known to contribute to the pathophysiological mechanisms during the development of renal fibrosis. Here, we investigated the effects of chemical chaperon sodium 4-phenylbutyrate (4-PBA) on renal fibrosis in vivo and in vitro. In a rat unilateral ureteral obstruction (UUO) model, 4-PBA mimicked endogenous ER chaperon in the kidneys and significantly reduced glucose regulated protein 78 (GRP78), CCAAT/enhancer binding protein (C/EBP) homologous protein (CHOP), activating transcription factor 4 (ATF4), and phosphorylated JNK protein expressions as well as restored spliced X-box-binding protein 1 (XBP1) expressions in the kidneys of UUO rats. 4-PBA also attenuated the increases of α-smooth muscle actin (α-SMA), connective tissue growth factor (CTGF) protein expressions, tubulointerstitial fibrosis, and apoptosis in the kidneys of UUO rats. Moreover, transforming growth factor (TGF)-β markedly increased ER stress-associated molecules, profibrotic factors, and apoptotic markers in the renal tubular cells (NRK-52E), all of which could be significantly counteracted by 4-PBA treatment. 4-PBA also diminished TGF-β-increased CTGF promoter activity and CTGF mRNA expression in NRK-52E cells. Taken together, our results indicated that 4-PBA acts as an ER chaperone to ameliorate ER stress-induced renal tubular cell apoptosis and renal fibrosis.
Collapse
|
126
|
Huang Y, Sun Y, Cao Y, Sun H, Li M, You H, Su D, Li Y, Liang X. HRD1 prevents apoptosis in renal tubular epithelial cells by mediating eIF2α ubiquitylation and degradation. Cell Death Dis 2017; 8:3202. [PMID: 29233968 PMCID: PMC5870601 DOI: 10.1038/s41419-017-0002-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 09/09/2017] [Accepted: 09/21/2017] [Indexed: 12/30/2022]
Abstract
Apoptosis of renal tubular epithelial cells is a key feature of the pathogenicity associated with tubulointerstitial fibrosis and other kidney diseases. One factor that regulates important cellular processes like apoptosis and cell proliferation is HRD1, an E3 ubiquitin ligase that acts by promoting ubiquitylation and degradation of its target protein. However, the detailed mechanisms by which HRD1 acts as a regulator of apoptosis in renal tubular epithelial cells have not been established. In our previous liquid chromatography-tandem mass spectrometry (LC-MS/MS) study (Mol Endocrinol. 2016;30:600–613), we demonstrated that one substrate of HRD1 was eIF2α, a critical protein in the PERK-eIF2α-ATF4-CHOP signaling pathway of endoplasmic reticulum (ER) stress. Here, we show that eIF2α expression was increased and HRD1 expression decreased when apoptosis was induced in HKC-8 cells by palmitic acid (PA) or high glucose (HG). HRD1 expression was also lower in kidney tissues from mice with diabetic nephropathy (DN) than in control mice. Forced expression of HRD1 also inhibited apoptosis in HKC-8 cells, while HRD1 overexpression decreased the expression of phosphorylated eIF2α and eIF2α. Further analysis indicated that HRD1 interacted with eIF2α and promoted its ubiquitylation and degradation by the proteasome. Moreover, the HRD1 protection of PA-treated HKC-8 cells was blunted by transfection with Myc-eIF2α. Thus, eIF2α ubiquitylation by HRD1 protects tubular epithelial cells from apoptosis caused by HG and PA, indicating a novel upstream target for therapeutic prevention of renal tubulointerstitial injury.
Collapse
Affiliation(s)
- Yujie Huang
- Renal Division, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.,Department of Pathology, Jiangsu Province Hospital of TCM, Affiliated Hospital of Nanjing University of TCM, Nanjing, 210026, Jiangsu Province, China
| | - Yifei Sun
- Department of Pathophysiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Yizhi Cao
- Department of Pathophysiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Hui Sun
- Renal Division, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Min Li
- Renal Division, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.,Department of Pathophysiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Hui You
- Renal Division, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Dongming Su
- Center of Pathology and Clinical Laboratory, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211166, Jiangsu Province, China
| | - Yanjiao Li
- Department of Endocrinology, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi Province, China
| | - Xiubin Liang
- Renal Division, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211166, Jiangsu, China. .,Department of Pathophysiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
| |
Collapse
|
127
|
Kim Y, Park SJ, Manson SR, Molina CA, Kidd K, Thiessen-Philbrook H, Perry RJ, Liapis H, Kmoch S, Parikh CR, Bleyer AJ, Chen YM. Elevated urinary CRELD2 is associated with endoplasmic reticulum stress-mediated kidney disease. JCI Insight 2017; 2:92896. [PMID: 29212948 DOI: 10.1172/jci.insight.92896] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 11/01/2017] [Indexed: 02/06/2023] Open
Abstract
ER stress has emerged as a signaling platform underlying the pathogenesis of various kidney diseases. Thus, there is an urgent need to develop ER stress biomarkers in the incipient stages of ER stress-mediated kidney disease, when a kidney biopsy is not yet clinically indicated, for early therapeutic intervention. Cysteine-rich with EGF-like domains 2 (CRELD2) is a newly identified protein that is induced and secreted under ER stress. For the first time to our knowledge, we demonstrate that CRELD2 can serve as a sensitive urinary biomarker for detecting ER stress in podocytes or renal tubular cells in murine models of podocyte ER stress-induced nephrotic syndrome and tunicamycin- or ischemia-reperfusion-induced acute kidney injury (AKI), respectively. Most importantly, urinary CRELD2 elevation occurs in patients with autosomal dominant tubulointerstitial kidney disease caused by UMOD mutations, a prototypical tubular ER stress disease. In addition, in pediatric patients undergoing cardiac surgery, detectable urine levels of CRELD2 within postoperative 6 hours strongly associate with severe AKI after surgery. In conclusion, our study has identified CRELD2 as a potentially novel urinary ER stress biomarker with potential utility in early diagnosis, risk stratification, treatment response monitoring, and directing of ER-targeted therapies in selected patient subgroups in the emerging era of precision nephrology.
Collapse
Affiliation(s)
- Yeawon Kim
- Division of Nephrology, Department of Internal Medicine
| | - Sun-Ji Park
- Division of Nephrology, Department of Internal Medicine
| | - Scott R Manson
- Division of Urology, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Carlos Af Molina
- Division of Urology, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA.,Division of Urology, Department of Surgery and Anatomy, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Kendrah Kidd
- Section of Nephrology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | | | - Rebecca J Perry
- Section of Nephrology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Helen Liapis
- RTE Professor of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri, USA.,Arkana Laboratories, Little Rock, Arkansas, USA
| | - Stanislav Kmoch
- Section of Nephrology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA.,Institute for Inherited Metabolic Disorders, Charles University in Prague, Prague, Czech Republic
| | - Chirag R Parikh
- Section of Nephrology, Yale University School of Medicine, New Haven, Connecticut.,Clinical Epidemiology Research Center, Veterans Affairs Medical Center, West Haven, Connecticut, USA
| | - Anthony J Bleyer
- Section of Nephrology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | | |
Collapse
|
128
|
Amanullah A, Upadhyay A, Joshi V, Mishra R, Jana NR, Mishra A. Progressing neurobiological strategies against proteostasis failure: Challenges in neurodegeneration. Prog Neurobiol 2017; 159:1-38. [DOI: 10.1016/j.pneurobio.2017.08.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 06/01/2017] [Accepted: 08/25/2017] [Indexed: 02/07/2023]
|
129
|
Cheng YC, Chen CA, Chen HC. Endoplasmic reticulum stress-induced cell death in podocytes. Nephrology (Carlton) 2017; 22 Suppl 4:43-49. [DOI: 10.1111/nep.13145] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2017] [Indexed: 01/07/2023]
Affiliation(s)
- Yu-Chi Cheng
- Division of Nephrology, Department of Internal Medicine; Kaohsiung Medical University Hospital, Kaohsiung Medical University; Kaohsiung Taiwan
| | - Chien-An Chen
- Division of Nephrology; Sinlau Hospital; Tainan Taiwan
| | - Hung-Chun Chen
- Division of Nephrology, Department of Internal Medicine; Kaohsiung Medical University Hospital, Kaohsiung Medical University; Kaohsiung Taiwan
| |
Collapse
|
130
|
Cybulsky AV. Endoplasmic reticulum stress, the unfolded protein response and autophagy in kidney diseases. Nat Rev Nephrol 2017; 13:681-696. [DOI: 10.1038/nrneph.2017.129] [Citation(s) in RCA: 392] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
131
|
Kim Y, Park SJ, Chen YM. Mesencephalic astrocyte-derived neurotrophic factor (MANF), a new player in endoplasmic reticulum diseases: structure, biology, and therapeutic roles. Transl Res 2017; 188:1-9. [PMID: 28719799 PMCID: PMC5601018 DOI: 10.1016/j.trsl.2017.06.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Revised: 06/20/2017] [Accepted: 06/23/2017] [Indexed: 02/06/2023]
Abstract
Mesencephalic astrocyte-derived neurotrophic factor (MANF), a newly identified 18-kDa soluble protein, localizes to the luminal endoplasmic reticulum (ER), whose stress can stimulate MANF expression and secretion. In Drosophila and zebrafish, MANF regulates dopaminergic neuron development. In contrast, in mice, MANF deficiency leads to diabetes and activation of the unfolded protein response. Recent studies in rodent models have demonstrated that MANF mitigates diabetes, exerts neurotrophic function in neurodegenerative disease, protects cardiomyocytes and neurons in myocardial infarction and cerebral ischemia, respectively, and promotes immune cell phenotype switch from proinflammatory macrophages to prorepair anti-inflammatory macrophages. The cytoprotective mechanisms of MANF on ER stress are currently under active investigation. In addition, for the first time, we have discovered that MANF can potentially serve as a urinary ER stress biomarker in ER stress-mediated kidney disease. These studies have underscored the diagnostic and therapeutic importance of MANF in ER diseases.
Collapse
Affiliation(s)
- Yeawon Kim
- Division of Nephrology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Mo
| | - Sun-Ji Park
- Division of Nephrology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Mo
| | - Ying Maggie Chen
- Division of Nephrology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Mo.
| |
Collapse
|
132
|
Foreseeing the future of glomerular disease through slits: miR-NPNT axis. Kidney Int 2017; 92:782-784. [DOI: 10.1016/j.kint.2017.04.036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 03/29/2017] [Accepted: 04/04/2017] [Indexed: 01/08/2023]
|
133
|
Han Q, Zhu H, Chen X, Liu Z. Non-genetic mechanisms of diabetic nephropathy. Front Med 2017; 11:319-332. [PMID: 28871454 DOI: 10.1007/s11684-017-0569-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 06/22/2017] [Indexed: 12/12/2022]
Abstract
Diabetic nephropathy (DN) is one of the most common microvascular complications in diabetes mellitus patients and is characterized by thickened glomerular basement membrane, increased extracellular matrix formation, and podocyte loss. These phenomena lead to proteinuria and altered glomerular filtration rate, that is, the rate initially increases but progressively decreases. DN has become the leading cause of end-stage renal disease. Its prevalence shows a rapid growth trend and causes heavy social and economic burden in many countries. However, this disease is multifactorial, and its mechanism is poorly understood due to the complex pathogenesis of DN. In this review, we highlight the new molecular insights about the pathogenesis of DN from the aspects of immune inflammation response, epithelial-mesenchymal transition, apoptosis and mitochondrial damage, epigenetics, and podocyte-endothelial communication. This work offers groundwork for understanding the initiation and progression of DN, as well as provides ideas for developing new prevention and treatment measures.
Collapse
Affiliation(s)
- Qiuxia Han
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing, 100853, China
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Hanyu Zhu
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing, 100853, China.
| | - Xiangmei Chen
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing, 100853, China
| | - Zhangsuo Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
134
|
Lei Y, Wang S, Ren B, Wang J, Chen J, Lu J, Zhan S, Fu Y, Huang L, Tan J. CHOP favors endoplasmic reticulum stress-induced apoptosis in hepatocellular carcinoma cells via inhibition of autophagy. PLoS One 2017; 12:e0183680. [PMID: 28841673 PMCID: PMC5571976 DOI: 10.1371/journal.pone.0183680] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Accepted: 08/09/2017] [Indexed: 12/19/2022] Open
Abstract
C/EBP-homologous protein (CHOP) is an important component of the endoplasmic reticulum (ER) stress response. We demonstrated the induction of ER stress in response to tunicamycin stimulation, as evidenced by increased expression of chaperone proteins Grp78, Grp94, and enhanced eukaryotic initiation factor 2 subunit 1 (eIF2α) phosphorylation in hepatocellular carcinoma cells. Tunicamycin-induced ER stress resulted in apoptosis and autophagy simultaneously. While inhibition of autophagy mediated by 3-methyladenine pretreatment or direct knockdown of LC3B promoted cell apoptosis, activation of autophagy with rapamycin decreased tunicamycin- induced apoptosis in HCC cells. Furthermore, CHOP was shown to be significantly upregulated upon treatment with tunicamycin in HCC cells. Specific knockdown of CHOP not only enhanced tunicamycin-induced autophagy, but also significantly attenuated ER stress-induced apoptosis in HCC cells. Accordingly, simultaneous inhibition of autophagy in HCC cells with CHOP-knockdown could partially resensitize ER stress-induced apoptosis. Taken together, our data indicate that CHOP may favor ER stress-induced apoptosis in HCC cells via inhibition of autophagy in vitro.
Collapse
Affiliation(s)
- Yan Lei
- Department of Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou General Hospital, Xiamen University, Fuzhou, Fujian, China
| | - Shuiliang Wang
- Department of Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou General Hospital, Xiamen University, Fuzhou, Fujian, China
| | - Bingshuang Ren
- Department of Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou General Hospital, Xiamen University, Fuzhou, Fujian, China
| | - Jin Wang
- Department of Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou General Hospital, Xiamen University, Fuzhou, Fujian, China
| | - Jin Chen
- Department of Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou General Hospital, Xiamen University, Fuzhou, Fujian, China
| | - Jun Lu
- Department of Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou General Hospital, Xiamen University, Fuzhou, Fujian, China
| | - Shihuai Zhan
- Department of Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou General Hospital, Xiamen University, Fuzhou, Fujian, China
| | - Yunfeng Fu
- Department of Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou General Hospital, Xiamen University, Fuzhou, Fujian, China
| | - Lianghu Huang
- Department of Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou General Hospital, Xiamen University, Fuzhou, Fujian, China
- * E-mail: (LH); (JT)
| | - Jianming Tan
- Department of Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou General Hospital, Xiamen University, Fuzhou, Fujian, China
- * E-mail: (LH); (JT)
| |
Collapse
|
135
|
A Comparative Study of the Predictive Values of Urinary Acute Kidney Injury Markers Angiogenin and Kidney Injury Molecule 1 for the Outcomes of Kidney Allografts. Transplant Direct 2017; 3:e204. [PMID: 28979926 PMCID: PMC5585420 DOI: 10.1097/txd.0000000000000720] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 06/28/2017] [Indexed: 11/25/2022] Open
Abstract
Supplemental digital content is available in the text. Background Whether injury-related molecules in urines of individuals with ischemia-reperfusion injury (IRI) are independent predictors of graft outcomes and provide additional information compared with usual risk factors remains to be established. Methods We explored a cohort of 244 kidney transplant recipients who systematically had a urine collection 10 days after transplantation. The injury-related markers kidney injury molecule-1 (KIM-1) and angiogenin (ANG) levels in urines were measured. We determined the prognostic values of these markers on graft outcomes. Results Urinary KIM-1 and ANG concentrations were strongly correlated to each other and were significantly and independently associated with cold ischemia time, delayed graft function, and plasma creatinine 10 days after transplantation, indicating that these markers reflect the severity of IRI. However, urinary ANG and KIM-1 were not predictive of histological changes on protocol biopsies performed 3 and 12 months after transplantation. Finally, urinary ANG and urinary KIM-1 were not associated with graft survival. Conclusions Together, our results indicate that, in a cohort of 244 kidney transplant recipients, urinary ANG and KIM-1 levels in a single measurement 10 days after transplantation reflect the severity of IRI after kidney transplantation, but are neither independent predictors of renal function, histological changes and graft survival.
Collapse
|
136
|
Astragaloside IV protects against podocyte injury via SERCA2-dependent ER stress reduction and AMPKα-regulated autophagy induction in streptozotocin-induced diabetic nephropathy. Sci Rep 2017; 7:6852. [PMID: 28761152 PMCID: PMC5537362 DOI: 10.1038/s41598-017-07061-7] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 06/26/2017] [Indexed: 12/25/2022] Open
Abstract
Aberrant endoplasmic reticulum (ER) stress and autophagy are associated with diabetic nephropathy. Here we investigated the effect of astragaloside IV (AS-IV) on the progression of diabetic nephropathy (DN) and the underlying mechanism involving ER stress and autophagy in streptozotocin (STZ)-induced diabetic mice and high glucose (HG)-incubated podocytes. The diabetic mice developed progressive albuminuria and glomerulosclerosis within 8 weeks, which were significantly ameliorated by AS-IV treatment in a dose-dependent manner. Moreover, diabetes or HG-induced podocyte apoptosis was markedly attenuated by AS-IV, paralleled by a marked remission in ER stress and a remarkable restoration in impaired autophagy, which were associated with a significant improvement in the expression of sarcoendoplasmic reticulum Ca2+ ATPase 2b (SERCA2b) and AMP-activated protein kinase α (AMPKα) phosphorylation, respectively. Knockdown of SERCA2 in podocytes induced ER stress and largely abolished the protective effect of AS-IV, but had no obvious effect on the expression of autophagy-associated proteins. On the other hand, blockade of either autophagy induction or AMPKα activation could also significantly mitigate AS-IV-induced beneficial effect. Collectively, these results suggest that AS-IV prevented the progression of DN, which is mediated at least in part by SERCA2-dependent ER stress attenuation and AMPKα-promoted autophagy induction.
Collapse
|
137
|
Kuo CY, Lin CH, Hsu T. VHL Inactivation in Precancerous Kidney Cells Induces an Inflammatory Response via ER Stress-Activated IRE1 α Signaling. Cancer Res 2017; 77:3406-3416. [PMID: 28533271 DOI: 10.1158/0008-5472.can-16-3196] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 04/06/2017] [Accepted: 05/09/2017] [Indexed: 11/16/2022]
Abstract
Mutations and epigenetic inactivation of the tumor suppressor gene von Hippel-Lindau (VHL) are major causes of clear-cell renal cell carcinoma (ccRCC) that may originate from chronic inflammation. However, the role of VHL loss of function in the development of ccRCC via inflammation remains poorly understood. VHL-mutant cells exhibit metabolic abnormalities that can cause chronic endoplasmic reticulum (ER) stress and unfolded protein response. We hypothesize that unresolved ER stress induces the inflammatory responses observed in ccRCC. ER stress markers including BiP and XBP1s were significantly increased in cultured and primary VHL loss-of-function kidney cells. In epithelial cells, the kinase activity of IRE1α was required for the induction of NF-κB and JNK and for the recruitment of macrophages. IRE1α kinase activity was also important for the development of fibrotic phenotype in conditional Vhlh knockout mice. Our results offer insights into the therapeutic potential against ccRCC development by relieving metabolic stress. Such cancer prevention strategy may be critical for high-risk cohorts such as the familial VHL disease patients. Cancer Res; 77(13); 3406-16. ©2017 AACR.
Collapse
Affiliation(s)
- Chan-Yen Kuo
- Department of Biomedical Sciences and Engineering, National Central University, Jhongli, Taiwan.,Institute of Systems Biology and Bioinformatics, National Central University, Jhongli, Taiwan
| | - Chih-Hung Lin
- Department of Biomedical Sciences and Engineering, National Central University, Jhongli, Taiwan.,Department of Internal Medicine, Cathay General Hospital, Taipei, Taiwan
| | - Tien Hsu
- Department of Biomedical Sciences and Engineering, National Central University, Jhongli, Taiwan. .,Institute of Systems Biology and Bioinformatics, National Central University, Jhongli, Taiwan.,Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| |
Collapse
|
138
|
Abstract
A broad definition of preconditioning is "the preparation for a subsequent action." Mounting evidence demonstrates that novel remote preconditioning paradigms, in which protective stimuli experienced locally can capacitate systemic tolerance and enhanced cell viability upon exposure to ensuing cellular insults, have been largely successful in the field of cardiovascular ischemia/reperfusion injury. To ensure successful protective preconditioning, some models (including the uterus) have been demonstrated to activate the unfolded protein response (UPR), which is a cellular stress response controlled at the level of the endoplasmic reticulum. However, in the context of remote preconditioning, activation of these intracellular molecular pathways must result in the extracellular transmission of adaptive signals to remote targets. In our recently published manuscript, we have described the activation of the UPR in the pregnant uterine myocyte to be associated with increased uterine myocyte quiescence and normal gestational length. We hypothesize that ubiquitous uterine gestational stresses experienced in every pregnancy, which have been demonstrated in other systems to activate the UPR, may induce a robust paracrine dissemination of a uterine secretome, for example, glucose-regulated protein 78, with preconditioning-like properties. Furthermore, we speculate that the gestational stress-induced uterine secretome acts to promote both local and systemic tolerance to the ensuing gestational insults, allowing for the maintenance of uterine quiescence. In this context, preterm labor may be the result of a pregnant uterus experiencing a stress it cannot accommodate or when it is unable to host an appropriate UPR resulting in insufficient preconditioning and a diminished local and systemic capacity to tolerate pregnancy-dependent increases in normal gestational stress. This is highly attractive from a clinical viewpoint as we ultimately aim to identify local and systemic adaptations that may serve as preconditioning stimuli for use as a strategy to restore appropriate preconditioning profiles to prolong uterine quiescence in pregnancy.
Collapse
Affiliation(s)
- Judith Ingles
- 1 Department of Physiology, Wayne State University Perinatal Initiative, School of Medicine, Wayne State University, Detroit, MI, USA.,2 Department of Obstetrics and Gynecology, Wayne State University Perinatal Initiative, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Chandrashekara N Kyathanahalli
- 1 Department of Physiology, Wayne State University Perinatal Initiative, School of Medicine, Wayne State University, Detroit, MI, USA.,2 Department of Obstetrics and Gynecology, Wayne State University Perinatal Initiative, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Pancharatnam Jeyasuria
- 1 Department of Physiology, Wayne State University Perinatal Initiative, School of Medicine, Wayne State University, Detroit, MI, USA.,2 Department of Obstetrics and Gynecology, Wayne State University Perinatal Initiative, School of Medicine, Wayne State University, Detroit, MI, USA.,3 Perinatal Research Initiative Wayne State University School of Medicine, Wane State University, Detroit, MI, USA
| | - Jennifer C Condon
- 1 Department of Physiology, Wayne State University Perinatal Initiative, School of Medicine, Wayne State University, Detroit, MI, USA.,2 Department of Obstetrics and Gynecology, Wayne State University Perinatal Initiative, School of Medicine, Wayne State University, Detroit, MI, USA.,3 Perinatal Research Initiative Wayne State University School of Medicine, Wane State University, Detroit, MI, USA
| |
Collapse
|
139
|
Dietary Metabolites and Chronic Kidney Disease. Nutrients 2017; 9:nu9040358. [PMID: 28375181 PMCID: PMC5409697 DOI: 10.3390/nu9040358] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 03/30/2017] [Accepted: 03/31/2017] [Indexed: 01/04/2023] Open
Abstract
Dietary contents and their metabolites are closely related to chronic kidney disease (CKD) progression. Advanced glycated end products (AGEs) are a type of uremic toxin produced by glycation. AGE accumulation is not only the result of elevated glucose levels or reduced renal clearance capacity, but it also promotes CKD progression. Indoxyl sulfate, another uremic toxin derived from amino acid metabolism, accumulates as CKD progresses and induces tubulointerstitial fibrosis and glomerular sclerosis. Specific types of amino acids (d-serine) or fatty acids (palmitate) are reported to be closely associated with CKD progression. Promising therapeutic targets associated with nutrition include uremic toxin absorbents and inhibitors of AGEs or the receptor for AGEs (RAGE). Probiotics and prebiotics maintain gut flora balance and also prevent CKD progression by enhancing gut barriers and reducing uremic toxin formation. Nrf2 signaling not only ameliorates oxidative stress but also reduces elevated AGE levels. Bardoxolone methyl, an Nrf2 activator and NF-κB suppressor, has been tested as a therapeutic agent, but the phase 3 clinical trial was terminated owing to the high rate of cardiovascular events. However, a phase 2 trial has been initiated in Japan, and the preliminary analysis reveals promising results without an increase in cardiovascular events.
Collapse
|
140
|
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
|
141
|
Schnaper HW. The Tubulointerstitial Pathophysiology of Progressive Kidney Disease. Adv Chronic Kidney Dis 2017; 24:107-116. [PMID: 28284376 PMCID: PMC5351778 DOI: 10.1053/j.ackd.2016.11.011] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 11/07/2016] [Accepted: 11/13/2016] [Indexed: 02/07/2023]
Abstract
Accumulating evidence suggests that the central locus for the progression of CKD is the renal proximal tubule. As injured tubular epithelial cells dedifferentiate in attempted repair, they stimulate inflammation and recruit myofibroblasts. At the same time, tissue loss stimulates remnant nephron hypertrophy. Increased tubular transport workload eventually exceeds the energy-generating capacity of the hypertrophied nephrons, leading to anerobic metabolism, acidosis, hypoxia, endoplasmic reticulum stress, and the induction of additional inflammatory and fibrogenic responses. The result is a vicious cycle of injury, misdirected repair, maladaptive responses, and more nephron loss. Therapy that might be advantageous at one phase of this progression pathway could be deleterious during other phases. Thus, interrupting this downward spiral requires narrowly targeted approaches that promote healing and adequate function without generating further entry into the progression cycle.
Collapse
Affiliation(s)
- H William Schnaper
- Division of Kidney Diseases, Department of Pediatrics, Northwestern University Feinberg School of Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL.
| |
Collapse
|
142
|
Maekawa H, Inagi R. Stress Signal Network between Hypoxia and ER Stress in Chronic Kidney Disease. Front Physiol 2017; 8:74. [PMID: 28228736 PMCID: PMC5296310 DOI: 10.3389/fphys.2017.00074] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 01/26/2017] [Indexed: 11/24/2022] Open
Abstract
Chronic kidney disease (CKD) is characterized by an irreversible decrease in kidney function and induction of various metabolic dysfunctions. Accumulated findings reveal that chronic hypoxic stress and endoplasmic reticulum (ER) stress are involved in a range of pathogenic conditions, including the progression of CKD. Because of the presence of an arteriovenous oxygen shunt, the kidney is thought to be susceptible to hypoxia. Chronic kidney hypoxia is induced by a number of pathogenic conditions, including renal ischemia, reduced peritubular capillary, and tubulointerstitial fibrosis. The ER is an organelle which helps maintain the quality of proteins through the unfolded protein response (UPR) pathway, and ER dysfunction associated with maladaptive UPR activation is named ER stress. ER stress is reported to be related to some of the effects of pathogenesis in kidney, particularly in the podocyte slit diaphragm and tubulointerstitium. Furthermore, chronic hypoxia mediates ER stress in blood vessel endothelial cells and tubulointerstitium via several mechanisms, including oxidative stress, epigenetic alteration, lipid metabolism, and the AKT pathway. In summary, a growing consensus considers that these stresses interact via complicated stress signal networks, which leads to the exacerbation of CKD (Figure 1). This stress signal network might be a target for interventions aimed at ameliorating CKD.
Collapse
Affiliation(s)
- Hiroshi Maekawa
- Division of Nephrology and Endocrinology, University of Tokyo Graduate School of Medicine Tokyo, Japan
| | - Reiko Inagi
- Division of Chronic Kidney Disease Pathophysiology, University of Tokyo Graduate School of Medicine Tokyo, Japan
| |
Collapse
|
143
|
Endoplasmic reticulum stress inhibition attenuates hypertensive chronic kidney disease through reduction in proteinuria. Sci Rep 2017; 7:41572. [PMID: 28148966 PMCID: PMC5288651 DOI: 10.1038/srep41572] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 12/21/2016] [Indexed: 12/22/2022] Open
Abstract
Endoplasmic reticulum (ER) stress is implicated in chronic kidney disease (CKD) development in patients and in animal models. Here we show that ER stress inhibition through 4-phenylbutyric acid (4-PBA) administration decreases blood pressure, albuminuria, and tubular casts in an angiotensin II/deoxycorticosterone acetate/salt murine model of CKD. Lower albuminuria in 4-PBA-treated mice was associated with higher levels of cubilin protein in renal tissue membrane fractions. 4-PBA decreased renal interstitial fibrosis, renal CD3+ T-cell and macrophage infiltration, mRNA expression of TGFβ1, Wnt signaling molecules, and ER stress-induced pro-inflammatory genes. CHOP deficient mice that underwent this model of CKD developed hypertension comparable to wild type mice, but had less albuminuria and tubular casts. CHOP deficiency resulted in higher nephrin levels and decreased glomerulosclerosis compared to wild type mice; this effect was accompanied by lower macrophage infiltration and fibrosis. Our findings portray ER stress inhibition as a means to alleviate hypertensive CKD by preserving glomerular barrier integrity and tubular function. These results demonstrate ER stress modulation as a novel target for preserving renal function in hypertensive CKD.
Collapse
|
144
|
|
145
|
Hirakawa Y, Inagi R. Glycative Stress and Its Defense Machinery Glyoxalase 1 in Renal Pathogenesis. Int J Mol Sci 2017; 18:ijms18010174. [PMID: 28106734 PMCID: PMC5297806 DOI: 10.3390/ijms18010174] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 01/05/2017] [Accepted: 01/10/2017] [Indexed: 12/13/2022] Open
Abstract
Chronic kidney disease is a major public health problem around the world. Because the kidney plays a role in reducing glycative stress, renal dysfunction results in increased glycative stress. In turn, glycative stress, especially that due to advanced glycated end products (AGEs) and their precursors such as reactive carbonyl compounds, exacerbates chronic kidney disease and is related to premature aging in chronic kidney disease, whether caused by diabetes mellitus or otherwise. Factors which hinder a sufficient reduction in glycative stress include the inhibition of anti-glycation enzymes (e.g., GLO-1), as well as pathogenically activated endoplasmic reticulum (ER) stress and hypoxia in the kidney. Promising strategies aimed at halting the vicious cycle between chronic kidney disease and increases in glycative stress include the suppression of AGE accumulation in the body and the enhancement of GLO-1 to strengthen the host defense machinery against glycative stress.
Collapse
Affiliation(s)
- Yosuke Hirakawa
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| | - Reiko Inagi
- Division of Chronic Kidney Disease (CKD) Pathophysiology, The University of Tokyo Graduate School of Medicine, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| |
Collapse
|
146
|
Guo H, Cao A, Chu S, Wang Y, Zang Y, Mao X, Wang H, Wang Y, Liu C, Zhang X, Peng W. Astragaloside IV Attenuates Podocyte Apoptosis Mediated by Endoplasmic Reticulum Stress through Upregulating Sarco/Endoplasmic Reticulum Ca 2+-ATPase 2 Expression in Diabetic Nephropathy. Front Pharmacol 2016; 7:500. [PMID: 28066247 PMCID: PMC5174081 DOI: 10.3389/fphar.2016.00500] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 12/05/2016] [Indexed: 12/18/2022] Open
Abstract
Sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) plays a central role in the pathogenesis of diabetes. This protein has been recognized as a potential target for diabetic therapy. In this study, we identified astragaloside IV (AS-IV) as a potent modulator of SERCA inhibiting renal injury in diabetic status. Increasing doses of AS-IV (2, 6, and 18 mg kg-1 day-1) were administered intragastrically to db/db mice for 8 weeks. Biochemical and histopathological approaches were conducted to evaluate the therapeutic effects of AS-IV. Cultured mouse podocytes were used to further explore the underlying mechanism in vitro. AS-IV dose-dependently increased SERCA activity and SERCA2 expression, and suppressed ER stress-mediated and mitochondria-mediated apoptosis in db/db mouse kidney. AS-IV also normalized glucose tolerance and insulin sensitivity, improved renal function, and ameliorated glomerulosclerosis and renal inflammation in db/db mice. In palmitate stimulated podocytes, AS-IV markedly improved inhibitions of SERCA activity and SERCA2 expression, restored intracellular Ca2+ homeostasis, and attenuated podocyte apoptosis in a dose-dependent manner with a concomitant abrogation of ER stress as evidenced by the downregulation of GRP78, cleaved ATF6, phospho-IRE1α and phospho-PERK, and the inactivation of both ER stress-mediated and mitochondria-mediated apoptotic pathways. Furthermore, SERCA2b knockdown eliminated the effect of AS-IV on ER stress and ER stress-mediated apoptotic pathway, whereas its overexpression exhibited an anti-apoptotic effect. Our data obtained from in vivo and in vitro studies demonstrate that AS-IV attenuates renal injury in diabetes subsequent to inhibiting ER stress-induced podocyte apoptosis through restoring SERCA activity and SERCA2 expression.
Collapse
Affiliation(s)
- Hengjiang Guo
- Laboratory of Renal Disease, Putuo Hospital, Shanghai University of Traditional Chinese Medicine Shanghai, China
| | - Aili Cao
- Laboratory of Renal Disease, Putuo Hospital, Shanghai University of Traditional Chinese Medicine Shanghai, China
| | - Shuang Chu
- Laboratory of Renal Disease, Putuo Hospital, Shanghai University of Traditional Chinese Medicine Shanghai, China
| | - Yi Wang
- Laboratory of Renal Disease, Putuo Hospital, Shanghai University of Traditional Chinese Medicine Shanghai, China
| | - Yingjun Zang
- Laboratory of Renal Disease, Putuo Hospital, Shanghai University of Traditional Chinese Medicine Shanghai, China
| | - Xiaodong Mao
- Laboratory of Renal Disease, Putuo Hospital, Shanghai University of Traditional Chinese Medicine Shanghai, China
| | - Hao Wang
- Department of Nephrology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine Shanghai, China
| | - Yunman Wang
- Department of Nephrology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine Shanghai, China
| | - Cheng Liu
- Experimental Research Center, Putuo Hospital, Shanghai University of Traditional Chinese Medicine Shanghai, China
| | - Xuemei Zhang
- Department of Pharmacology, School of Pharmacy, Fudan University Shanghai, China
| | - Wen Peng
- Laboratory of Renal Disease, Putuo Hospital, Shanghai University of Traditional Chinese MedicineShanghai, China; Department of Nephrology, Putuo Hospital, Shanghai University of Traditional Chinese MedicineShanghai, China
| |
Collapse
|
147
|
Kuwagata S, Kume S, Chin-Kanasaki M, Araki H, Araki S, Nakazawa J, Sugaya T, Koya D, Haneda M, Maegawa H, Uzu T. MicroRNA148b-3p inhibits mTORC1-dependent apoptosis in diabetes by repressing TNFR2 in proximal tubular cells. Kidney Int 2016; 90:1211-1225. [DOI: 10.1016/j.kint.2016.06.036] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 06/26/2016] [Accepted: 06/30/2016] [Indexed: 11/24/2022]
|
148
|
Kim Y, Lee H, Manson SR, Lindahl M, Evans B, Miner JH, Urano F, Chen YM. Mesencephalic Astrocyte-Derived Neurotrophic Factor as a Urine Biomarker for Endoplasmic Reticulum Stress-Related Kidney Diseases. J Am Soc Nephrol 2016; 27:2974-2982. [PMID: 26940092 PMCID: PMC5042655 DOI: 10.1681/asn.2014100986] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 01/15/2016] [Indexed: 12/14/2022] Open
Abstract
Endoplasmic reticulum (ER) stress and disrupted proteostasis contribute to the pathogenesis of a variety of glomerular and tubular diseases. Thus, it is imperative to develop noninvasive biomarkers for detecting ER stress in podocytes or tubular cells in the incipient stage of disease, when a kidney biopsy is not yet clinically indicated. Mesencephalic astrocyte-derived neurotrophic factor (MANF) localizes to the ER lumen and is secreted in response to ER stress in several cell types. Here, using mouse models of human nephrotic syndrome caused by mutant laminin β2 protein-induced podocyte ER stress and AKI triggered by tunicamycin- or ischemia-reperfusion-induced tubular ER stress, we examined MANF as a potential urine biomarker for detecting ER stress in podocytes or renal tubular cells. ER stress upregulated MANF expression in podocytes and tubular cells. Notably, urinary MANF excretion concurrent with podocyte or tubular cell ER stress preceded clinical or histologic manifestations of the corresponding disease. Thus, MANF can potentially serve as a urine diagnostic or prognostic biomarker in ER stress-related kidney diseases to help stratify disease risk, predict disease progression, monitor treatment response, and identify subgroups of patients who can be treated with ER stress modulators in a highly targeted manner.
Collapse
Affiliation(s)
- Yeawon Kim
- Division of Nephrology, Department of Internal Medicine
| | - Heedoo Lee
- Division of Nephrology, Department of Internal Medicine
| | | | - Maria Lindahl
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland; and
| | - Bradley Evans
- Proteomics and Mass Spectrometry Facility, Donald Danforth Plant Science Center, St. Louis, Missouri
| | - Jeffrey H Miner
- Division of Nephrology, Department of Internal Medicine, Departments of Cell Biology and Physiology and
| | - Fumihiko Urano
- Division of Endocrinology, Metabolism and Lipid Research, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri; Pathology and Immunology, and
| | | |
Collapse
|
149
|
Tavernier Q, Mami I, Rabant M, Karras A, Laurent-Puig P, Chevet E, Thervet E, Anglicheau D, Pallet N. Urinary Angiogenin Reflects the Magnitude of Kidney Injury at the Infrahistologic Level. J Am Soc Nephrol 2016; 28:678-690. [PMID: 27436854 DOI: 10.1681/asn.2016020218] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 06/07/2016] [Indexed: 12/26/2022] Open
Abstract
The ribonuclease angiogenin is a component of the mammalian stress response that is secreted by renal epithelial cells on activation of the inositol-requiring enzyme 1α (IRE1α)-active spliced X-box binding protein 1 (sXBP1) axis and instrumental to the adaptation to AKI associated with endoplasmic reticulum stress. To determine whether the amount of angiogenin in urine of individuals with a kidney injury reflects the magnitude of the lesions and provides information on the risk of organ failure, we examined individuals referred for a kidney injury and determined the biochemical characteristics of urinary angiogenin and its diagnostic and prognostic values. Urinary angiogenin did not correlate with the urinary concentrations of high molecular weight proteins and correlated only weakly with low molecular weight proteins, suggestive of tubular production. In a cohort of 242 kidney transplant recipients with acute allograft dysfunction, higher urinary angiogenin concentrations at the time of the biopsy associated with worse renal function and higher proteinuria but did not correlate with histologic lesions as defined in the Banff classification. Kidney transplant recipients with urinary angiogenin amounts in the highest 50% had a risk of graft failure 3.59 times as high (95% confidence interval, 1.12 to 15.94) as that of patients with amounts in the lowest 50%. Finally, the amount of urinary angiogenin reflected the activity of the IRE1α-XBP1 axis in allografts. Our approach identified urinary angiogenin as a noninvasive indicator of the extent of tissue damage, independent of the histologic lesions, and a risk predictor of kidney allograft failure.
Collapse
Affiliation(s)
- Quentin Tavernier
- Institut National de la Santé et la Recherche Médicale (INSERM) U1147, Centre Universitaire des Saints Pères, Paris, France.,Université Paris Descartes, Paris, France
| | - Iadh Mami
- Institut National de la Santé et la Recherche Médicale (INSERM) U1147, Centre Universitaire des Saints Pères, Paris, France.,Université Paris Descartes, Paris, France
| | - Marion Rabant
- Université Paris Descartes, Paris, France.,Service d'Anatomopathologie and
| | - Alexandre Karras
- Université Paris Descartes, Paris, France.,Service de Néphrologie and
| | - Pierre Laurent-Puig
- Institut National de la Santé et la Recherche Médicale (INSERM) U1147, Centre Universitaire des Saints Pères, Paris, France.,Université Paris Descartes, Paris, France.,Service de Biochimie, Hôpital Européen Georges Pompidou, Paris, France
| | - Eric Chevet
- Oncogenesis, Stress, Signaling Team, ER-440 Université de Rennes-1, Rennes, France; and
| | - Eric Thervet
- Université Paris Descartes, Paris, France.,Service de Néphrologie and
| | - Dany Anglicheau
- Université Paris Descartes, Paris, France.,Service de Néphrologie et Transplantation Rénale, Hopital Necker, Paris, France.,INSERM U1151, Institut Necker Enfants Malades, Paris, France
| | - Nicolas Pallet
- Institut National de la Santé et la Recherche Médicale (INSERM) U1147, Centre Universitaire des Saints Pères, Paris, France; .,Université Paris Descartes, Paris, France.,Service de Biochimie, Hôpital Européen Georges Pompidou, Paris, France
| |
Collapse
|
150
|
Lindahl M, Saarma M, Lindholm P. Unconventional neurotrophic factors CDNF and MANF: Structure, physiological functions and therapeutic potential. Neurobiol Dis 2016; 97:90-102. [PMID: 27425895 DOI: 10.1016/j.nbd.2016.07.009] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 06/29/2016] [Accepted: 07/13/2016] [Indexed: 12/30/2022] Open
Abstract
Cerebral dopamine neurotrophic factor (CDNF) and mesencephalic astrocyte-derived neurotrophic factor (MANF) promote the survival of midbrain dopaminergic neurons which degenerate in Parkinson's disease (PD). However, CDNF and MANF are structurally and functionally clearly distinct from the classical, target-derived neurotrophic factors (NTFs) that are solely secreted proteins. In cells, CDNF and MANF localize in the endoplasmic reticulum (ER) and evidence suggests that MANF, and possibly CDNF, is important for the maintenance of ER homeostasis. MANF expression is particularly high in secretory tissues with extensive protein production and thus a high ER protein folding load. Deletion of MANF in mice results in a diabetic phenotype and the activation of unfolded protein response (UPR) in the pancreatic islets. However, information about the intracellular and extracellular mechanisms of MANF and CDNF action is still limited. Here we will discuss the structural motifs and physiological functions of CDNF and MANF as well as their therapeutic potential for the treatment of neurodegenerative diseases and diabetes. Currently available knockout models of MANF and CDNF in mice, zebrafish and fruit fly will increase information about the biology of these interesting proteins.
Collapse
Affiliation(s)
- Maria Lindahl
- Institute of Biotechnology, P.O.Box 56, Viikinkaari 5, FI-00014, University of Helsinki, Finland
| | - Mart Saarma
- Institute of Biotechnology, P.O.Box 56, Viikinkaari 5, FI-00014, University of Helsinki, Finland
| | - Päivi Lindholm
- Institute of Biotechnology, P.O.Box 56, Viikinkaari 5, FI-00014, University of Helsinki, Finland.
| |
Collapse
|