1
|
Chen M, Fang Y, Ge Y, Qiu S, Dworkin L, Gong R. The redox-sensitive GSK3β is a key regulator of glomerular podocyte injury in type 2 diabetic kidney disease. Redox Biol 2024; 72:103127. [PMID: 38527400 PMCID: PMC10979123 DOI: 10.1016/j.redox.2024.103127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/06/2024] [Accepted: 03/15/2024] [Indexed: 03/27/2024] Open
Abstract
Emerging evidence suggests that GSK3β, a redox-sensitive transducer downstream of insulin signaling, acts as a convergent point for myriad pathways implicated in kidney injury, repair, and regeneration. However, its role in diabetic kidney disease remains controversial. In cultured glomerular podocytes, exposure to a milieu of type 2 diabetes elicited prominent signs of podocyte injury and degeneration, marked by loss of homeostatic marker proteins like synaptopodin, actin cytoskeleton disruption, oxidative stress, apoptosis, and stress-induced premature senescence, as shown by increased staining for senescence-associated β-galactosidase activity, amplified formation of γH2AX foci, and elevated expression of mediators of senescence signaling, like p21 and p16INK4A. These degenerative changes coincided with GSK3β hyperactivity, as evidenced by GSK3β overexpression and reduced inhibitory phosphorylation of GSK3β, and were averted by tideglusib, a highly-selective small molecule inhibitor of GSK3β. In agreement, post-hoc analysis of a publicly-available glomerular transcriptomics dataset from patients with type 2 diabetic nephropathy revealed that the curated diabetic nephropathy-related gene set was enriched in high GSK3β expression group. Mechanistically, GSK3β-modulated nuclear factor Nrf2 signaling is involved in diabetic podocytopathy, because GSK3β knockdown reinforced Nrf2 antioxidant response and suppressed oxidative stress, resulting in an improvement in podocyte injury and senescence. Conversely, ectopic expression of the constitutively active mutant of GSK3β impaired Nrf2 antioxidant response and augmented oxidative stress, culminating in an exacerbated diabetic podocyte injury and senescence. Moreover, IRS-1 was found to be a cognate substrate of GSK3β for phosphorylation at IRS-1S332, which negatively regulates IRS-1 activity. GSK3β hyperactivity promoted IRS-1 phosphorylation, denoting a desensitized insulin signaling. Consistently, in vivo in db/db mice with diabetic nephropathy, GSK3β was hyperactive in glomerular podocytes, associated with IRS-1 hyperphosphorylation, impaired Nrf2 response and premature senescence. Our finding suggests that GSK3β is likely a novel therapeutic target for treating type 2 diabetic glomerular injury.
Collapse
Affiliation(s)
- Mengxuan Chen
- Division of Nephrology, Department of Medicine, University of Toledo College of Medicine, Toledo, OH, USA
| | - Yudong Fang
- Division of Nephrology, Department of Medicine, University of Toledo College of Medicine, Toledo, OH, USA
| | - Yan Ge
- Division of Nephrology, Department of Medicine, University of Toledo College of Medicine, Toledo, OH, USA
| | - Shuhao Qiu
- Division of Nephrology, Department of Medicine, University of Toledo College of Medicine, Toledo, OH, USA
| | - Lance Dworkin
- Division of Nephrology, Department of Medicine, University of Toledo College of Medicine, Toledo, OH, USA; Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, USA
| | - Rujun Gong
- Division of Nephrology, Department of Medicine, University of Toledo College of Medicine, Toledo, OH, USA; Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, USA; Center for Diabetes and Endocrine Research, University of Toledo Medical Center, Toledo, OH, USA.
| |
Collapse
|
2
|
Cai YT, Li Z, Wang YY, Li C, Ma QY. A novel GSK3β inhibitor 5n attenuates acute kidney injury. Heliyon 2024; 10:e29159. [PMID: 38644860 PMCID: PMC11031767 DOI: 10.1016/j.heliyon.2024.e29159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 03/31/2024] [Accepted: 04/02/2024] [Indexed: 04/23/2024] Open
Abstract
Acute kidney injury (AKI) is a clinical syndrome with high morbidity and mortality caused by various factor. The specific strategies for AKI are still lacking. GSK3β is widely expressed in the kidneys. In acute models of injury, GSK3β promotes the systemic inflammatory response, increases the proinflammatory release of cytokines, induces apoptosis, and alters cell proliferation. We screened a series of 3-(4-pyridyl)-5-(4-sulfamido-phenyl)-1,2,4-oxadiazole derivatives which are recognized as new GSK3β inhibitors, and found that 5n had the least toxicity and the best cell protection. We then tested the anti-inflammatory and reno-protective effect of 5n in cisplatin-treated tubular epithelial cells. 5n had anti-inflammation effect indicated by phosphor-NF-κB detection. Finally, we found that 5n ameliorated renal injury and inflammation in cisplatin-induced AKI mouse model. Silencing GSK3β inhibited cell injury and inflammation induced by cisplatin. We found that GSK3β interacted with PP2Ac to modulate the activity of NF-κB. In conclusion, 5n, the novel GSK3β inhibitor, protects against AKI via PP2Ac-dependent mechanisms which may provide a potential strategy for the treatment of AKI in clinic.
Collapse
Affiliation(s)
- Yu-ting Cai
- Department of Nephrology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Zeng Li
- The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Yue-yue Wang
- The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Chao Li
- The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Qiu-ying Ma
- Department of pharmacy, 1. The First Affiliated Hospital of Anhui Medical University, 2. Anhui Public Health Clinical Center, No. 100 Huaihai Road, Hefei, Anhui, 230012, China
| |
Collapse
|
3
|
Li Y, Li X, Yang Y, Li F, Chen Q, Zhao Z, Zhang N, Li H. Hepatocyte growth factor attenuates high glucose-disturbed mitochondrial dynamics in podocytes by decreasing ARF6-dependent DRP1 translocation. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119623. [PMID: 37913847 DOI: 10.1016/j.bbamcr.2023.119623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 10/23/2023] [Accepted: 10/24/2023] [Indexed: 11/03/2023]
Abstract
Diabetic nephropathy (DN), one of the most common complications of Diabetes Mellitus, is the leading cause of end-stage renal diseases worldwide. Our previous study proved that hepatocyte growth factor (HGF) alleviated renal damages in mice with type 1 Diabetes Mellitus by suppressing overproduction of reactive oxygen species (ROS) in podocytes, while the further mechanism of how HGF lessens ROS production had not been clarified yet. ADP-ribosylation factor 6 (ARF6), the member of the small GTPases superfamilies, is widely spread among epithelial cells and can be activated by the HGF/c-Met signaling. Thus, this study was aimed to explore whether HGF could function on mitochondrial homeostasis, the main resource of ROS, in podocytes exposed to diabetic conditions via ARF6 activation. Our in vivo data showed that HGF markedly ameliorated the pathological damages in kidneys of db/db mice, especially the sharp decline of podocyte number, which was mostly blocked by the ARF6 inhibitor SecinH3. Correspondingly, our in vitro data revealed that HGF protected against high glucose-induced podocyte injuries by increasing ARF6 activity. Besides, this ARF6-dependent beneficial effect of HGF on podocytes was accompanied by improved mitochondrial dynamics and declined DRP1 translocation from cytosol to mitochondria. Collectively, our findings confirm the ability of HGF maintaining mitochondrial homeostasis in diabetic podocytes via decreasing ARF6-dependent DRP1 translocation and shed light on the novel mechanism of HGF treatment for DN.
Collapse
Affiliation(s)
- Yankun Li
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Xue Li
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yuling Yang
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Fengxia Li
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Qi Chen
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Zhonghua Zhao
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Nong Zhang
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Hui Li
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
| |
Collapse
|
4
|
Wang X, Yang J, Wang W, Li Y, Yang Y. Decreasing REDD1 expression protects against high glucose-induced apoptosis, oxidative stress and inflammatory injury in podocytes through regulation of the AKT/GSK-3β/Nrf2 pathway. Immunopharmacol Immunotoxicol 2023; 45:527-538. [PMID: 36883011 DOI: 10.1080/08923973.2023.2183351] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 02/10/2023] [Indexed: 02/24/2023]
Abstract
OBJECTIVE Our goal in this work was to investigate the possible role and mechanism of regulated in development and DNA damage response 1 (REDD1) in mediating high glucose (HG)-induced podocyte injury in vitro. MATERIALS AND METHODS Mouse podocytes were stimulated with HG to establish HG injury model. Protein expression was examined by Western blotting. Cell viability was measured by cell counting kit-8 assay. Cell apoptosis was assessed by annexin V-FITC/propidium iodide and TUNEL apoptotic assays. Levels of reactive oxygen species (ROS), malondialdehyde (MDA), superoxide dismutase (SOD), and glutathione peroxidase (GPx) were quantified by commercial kits. Concentrations of tumor necrosis factor (TNF)-α, interleukin (IL)-6, and IL-1β were measured by ELISA. RESULTS A marked increase in REDD1 expression was observed in podocytes stimulated with HG. Reduced REDD1 expression strikingly restrained HG-induced increases in apoptosis, oxidative stress, and inflammation response in cultured podocytes. Decreasing REDD1 expression enhanced nuclear factor erythroid 2-related factor 2 (Nrf2) activation in HG-exposed podocytes via regulation of the AKT/glycogen synthase kinase-3 beta (GSK-3β) pathway. Inhibition of AKT or reactivation of GSK-3β prominently abolished Nrf2 activation induced by decreasing REDD1 expression. Pharmacological repression of Nrf2 markedly reversed the protective effects of decreasing REDD1 expression in HG-injured podocytes. CONCLUSION Our data demonstrate that decreasing REDD1 expression protects cultured podocytes from HG-induced injuries by potentiating Nrf2 signaling through regulation of the AKT/GSK-3β pathway. Our work underscores the potential role of REDD1-mediated podocyte injury during the development of diabetic kidney disease.
Collapse
Affiliation(s)
- Xiaojing Wang
- The First Clinical Medical College of Shanxi Medical University, Taiyuan, China
- Department of Endocrinology, Shanxi Yuncheng Central Hospital, Yuncheng, China
| | - Jing Yang
- The First Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Wenxing Wang
- Department of Endocrinology, Shanxi Yuncheng Central Hospital, Yuncheng, China
| | - Yun Li
- Department of Endocrinology, Shanxi Yuncheng Central Hospital, Yuncheng, China
| | - Yue Yang
- Department of Endocrinology, Shanxi Yuncheng Central Hospital, Yuncheng, China
| |
Collapse
|
5
|
Zeng Y, Xiong C, Chen Y, Yang C, Li Q. Effects and mechanism of Rictor interference in podocyte injury induced by high glucose. Exp Ther Med 2023; 26:473. [PMID: 37753299 PMCID: PMC10518650 DOI: 10.3892/etm.2023.12172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 07/07/2023] [Indexed: 09/28/2023] Open
Abstract
Rapamycin-insensitive companion of mTOR (Rictor) is a critical effector of mTOR protein complex 2 (mTORC2). The aim of the present study was to investigate the effect of Rictor in the mTORC2 signaling pathway in high glucose (HG)-induced diabetic podocyte injury by silencing the expression of Rictor. In the present study, mouse podocytes were treated with glucose (150 mM) and mannitol (200 mM), the Rictor gene was silenced using small interfering RNA (siRNA). Apoptosis was detected by flow cytometry, whereas podocyte cytoskeletal protein expression was detected by western blotting (WB) and immunofluorescence staining. The results demonstrated that, compared with that in the control group, the podocyte apoptotic rate was significantly increased in the mannitol group (negative group) and the groups that were treated with glucose (model groups). The podocyte apoptotic rate in the model + Rictor siRNA group was significantly decreased compared with that in the negative, model and the model glucose + siRNA negative control (NC) groups. WB indicated that the protein expression levels of podocalyxin and synaptopodin were reduced in the model and model + siRNA NC groups compared with those in the normal control and negative groups. Additionally, the protein expression levels of α-smooth muscle actin (α-SMA) and P-AKT/AKT were increased in the model and model + siRNA NC groups compared with the those in control and negative groups. Compared with those the model and model + siRNA NC groups, the protein expression levels of podocalyxin and synaptopodin were increased, whilst those of the α-SMA and P-AKT/AKT proteins were decreased, in the model + Rictor siRNA group. Results from immunofluorescence analysis were basically consistent with those of WB. Therefore, results of the present study suggest that silencing of the Rictor gene may reduce the damage to podocytes induced by HG, such that the Rictor/mTORC2 signaling pathway may be involved in the remodeling of podocyte actin cytoskeletal in diabetes.
Collapse
Affiliation(s)
- Yan Zeng
- Department of Nephrology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Changbin Xiong
- Department of Nephrology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yinxiang Chen
- Department of Nephrology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Chunyun Yang
- Department of Nephrology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Qiuyue Li
- Department of Nephrology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
6
|
Urinary Markers of Tubular Injury and Renal Fibrosis in Patients with Type 2 Diabetes and Different Phenotypes of Chronic Kidney Disease. Life (Basel) 2023; 13:life13020343. [PMID: 36836700 PMCID: PMC9961033 DOI: 10.3390/life13020343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/21/2023] [Accepted: 01/22/2023] [Indexed: 01/31/2023] Open
Abstract
This study assessed the urinary excretion of markers and mediators of tubular injury and renal fibrosis in patients with type 2 diabetes (T2D) and non-albuminuric and albuminuric patterns of chronic kidney disease (CKD). One hundred and forty patients with long-term T2D and different patterns of CKD and twenty non-diabetic individuals were included. Urinary retinol-binding protein 4 (RBP-4), glutathione-S-transferase α1 and π (GST-α1 and GST-π), transforming growth factor β (TGF-β), type I and type IV collagen (Col1 and Col4), bone morphogenic protein 7 (BMP-7), and hepatocyte growth factor (HGF) were assessed by ELISA. Patients with T2D demonstrated increased urinary excretion of RBP-4, GST-π, Col4, BMP-7, and HGF (all p < 0.05 vs. control). The excretion of RBP-4, GST-π, Col1, and Col4 was increased in patients with elevated albumin-to-creatinine ratio (UACR; all p < 0.05 vs. control), while BMP-7 and HGF were increased innormoalbuminuric patients also (p < 0.05). Urinary RBP-4, GST-α1, Col1, Col4, and HGF correlated positively with UACR; meanwhile, no correlations with glomerular filtration rate were found. The results demonstrate that elevated urinary excretions of the markers of tubular injury (RBP-4, GST-π) and renal fibrosis (Col1, Col4), as well as HGF, an antifibrotic regulator, are associated with the albuminuric pattern of CKD in subjects with T2D.
Collapse
|
7
|
Audzeyenka I, Rachubik P, Typiak M, Kulesza T, Topolewska A, Rogacka D, Angielski S, Saleem MA, Piwkowska A. Hyperglycemia alters mitochondrial respiration efficiency and mitophagy in human podocytes. Exp Cell Res 2021; 407:112758. [PMID: 34437881 DOI: 10.1016/j.yexcr.2021.112758] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 07/15/2021] [Accepted: 07/26/2021] [Indexed: 12/19/2022]
Abstract
Podocytes constitute the outer layer of the renal glomerular filtration barrier. Their energy requirements strongly depend on efficient oxidative respiration, which is tightly connected with mitochondrial dynamics. We hypothesized that hyperglycemia modulates energy metabolism in glomeruli and podocytes and contributes to the development of diabetic kidney disease. We found that oxygen consumption rates were severely reduced in glomeruli from diabetic rats and in human podocytes that were cultured in high glucose concentration (30 mM; HG). In these models, all of the mitochondrial respiratory parameters, including basal and maximal respiration, ATP production, and spare respiratory capacity, were significantly decreased. Podocytes that were treated with HG showed a fragmented mitochondrial network, together with a decrease in expression of the mitochondrial fusion markers MFN1, MFN2, and OPA1, and an increase in the activity of the fission marker DRP1. We showed that markers of mitochondrial biogenesis, such as PGC-1α and TFAM, decreased in HG-treated podocytes. Moreover, PINK1/parkin-dependent mitophagy was inhibited in these cells. These results provide evidence that hyperglycemia impairs mitochondrial dynamics and turnover, which may underlie the remarkable deterioration of mitochondrial respiration parameters in glomeruli and podocytes.
Collapse
Affiliation(s)
- Irena Audzeyenka
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Wita Stwosza St. 63, 80-308, Gdansk, Poland; Faculty of Chemistry, University of Gdansk, Wita Stwosza St. 63, 80-308, Gdansk, Poland.
| | - Patrycja Rachubik
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Wita Stwosza St. 63, 80-308, Gdansk, Poland
| | - Marlena Typiak
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Wita Stwosza St. 63, 80-308, Gdansk, Poland
| | - Tomasz Kulesza
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Wita Stwosza St. 63, 80-308, Gdansk, Poland
| | - Anna Topolewska
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Wita Stwosza St. 63, 80-308, Gdansk, Poland; Faculty of Chemistry, University of Gdansk, Wita Stwosza St. 63, 80-308, Gdansk, Poland
| | - Dorota Rogacka
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Wita Stwosza St. 63, 80-308, Gdansk, Poland; Faculty of Chemistry, University of Gdansk, Wita Stwosza St. 63, 80-308, Gdansk, Poland
| | - Stefan Angielski
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Wita Stwosza St. 63, 80-308, Gdansk, Poland
| | - Moin A Saleem
- Bristol Renal, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, United Kingdom
| | - Agnieszka Piwkowska
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Wita Stwosza St. 63, 80-308, Gdansk, Poland; Faculty of Chemistry, University of Gdansk, Wita Stwosza St. 63, 80-308, Gdansk, Poland
| |
Collapse
|
8
|
Li L, An JN, Lee J, Shin DJ, Zhu SM, Kim JH, Kim DK, Ryu DR, Kim S, Lee JP. Hepatocyte growth factor and soluble cMet levels in plasma are prognostic biomarkers of mortality in patients with severe acute kidney injury. Kidney Res Clin Pract 2021; 40:596-610. [PMID: 34510856 PMCID: PMC8685369 DOI: 10.23876/j.krcp.20.258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 06/07/2021] [Indexed: 11/21/2022] Open
Abstract
Background Hepatocyte growth factor (HGF)/cMet pathway is necessary for repair and regeneration following acute kidney injury (AKI). We evaluated the clinical potential of plasma HGF and soluble cMet as prognostic biomarkers for severe AKI requiring continuous renal replacement therapy (CRRT). Methods One hundred thirty-six patients with severe AKI who participated in the VENUS (volume management under body composition monitoring in critically ill patients on CRRT) trial between 2017 and 2019 were enrolled in this study. We investigated associations between plasma HGF and cMet concentrations and all-cause mortality. Results Plasma HGF and soluble cMet levels were positively correlated. Patients were divided into three groups based on their HGF and soluble cMet concentrations. The day D 0, D2, and D7 highest concentration HGF groups had significantly higher in-hospital mortality after adjusting for sex, body mass index, Acute Physiology and Chronic Health Evaluation II, and age-adjusted Charlson comorbidity index score, especially on D7 (hazard ratio, 4.26; 95% confidence interval, 1.71–10.62; p = 0.002). D7 soluble cMet level was also associated with mortality. Receiver operating characteristic curve analysis indicated that D7 HGF and soluble cMet levels were best at predicting mortality. Addition of plasma HGF and soluble cMet to conventional prognostic indices significantly improved the predictive value for mortality on D7. However, plasma HGF and soluble cMet were not associated with fluid status. Conclusion Plasma HGF and soluble cMet levels were significant predictors of the outcomes of severe AKI patients undergoing CRRT. There was no correlation between plasma HGF and soluble cMet levels and fluid balance.
Collapse
Affiliation(s)
- Lilin Li
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Intensive Care Unit, Yanbian University Hospital, Jilin, China
| | - Jung Nam An
- Department of Internal Medicine, Hallym Sacred Heart Hospital, Anyang, Republic of Korea
| | - Jeonghwan Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Internal Medicine-Nephrology, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Dong Jin Shin
- Preliminary Medicine Courses, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Shi Mao Zhu
- Department of Internal Medicine-Nephrology, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Jin Hyuk Kim
- Department of Internal Medicine-Nephrology, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Dong Ki Kim
- Department of Internal Medicine-Nephrology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Dong-Ryeol Ryu
- Department of Internal Medicine-Nephrology, Ewha Womans University School of Medicine, Seoul, Republic of Korea
| | - Sejoong Kim
- Department of Internal Medicine-Nephrology, Seoul National University Bundang Hospital, Republic of Korea
| | - Jung Pyo Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Internal Medicine-Nephrology, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| |
Collapse
|
9
|
Hou B, Li Y, Li X, Zhang C, Zhao Z, Chen Q, Zhang N, Li H. HGF protected against diabetic nephropathy via autophagy-lysosome pathway in podocyte by modulating PI3K/Akt-GSK3β-TFEB axis. Cell Signal 2020; 75:109744. [PMID: 32827692 DOI: 10.1016/j.cellsig.2020.109744] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 08/17/2020] [Accepted: 08/17/2020] [Indexed: 01/08/2023]
Abstract
Podocyte loss is a detrimental feature and major cause of proteinuria in diabetic nephropathy (DN). Our previous study revealed that hepatocyte growth factor (HGF) prevented high glucose-induced podocyte injury via enhancing autophagy. In the current study, we aimed to assess the role of HGF on podocyte homeostasis in DN and clarify its mechanisms further. Diabetic mice treated with HGF had markedly reduced ratio of kidney weight to body weight, urinary albumin excretion, podocyte loss and matrix expansion compared with that in the non-treated counterpart. Simultaneously, HGF-treated diabetic mice exhibited increased autophagy activity as indicated by the decreased accumulation of sequestosome 1 (SQSTM1/ p62) and increased microtubule-associated proteins 1 light chains 3 (LC3) II/LC3I ratio. These beneficial effects of HGF were blocked by HGF/c-Met inhibitor Crizotinib or phosphatidylinositide 3-kinases (PI3K) inhibitor LY294002. Moreover, HGF treatment obviously prevented inactivation of the protein kinase B (Akt)-glycogen synthase kinase 3 beta (GSK3β)-transcription factor EB (TFEB) axis in high glucose-stimulated podocytes, which was associated with improved lysosome function and autophagy. Accordingly, adenovirus vector encoding constitutively active GSK3β (Ad-GSK3β-S9A) offset whereas small interfering RNA against GSK3β (GSK3β siRNA) recapitulated salutary effects of HGF on lysosome number and autophagy in podocytes. These results suggested that HGF protected against diabetic nephropathy through restoring podocyte autophagy, which at least partially involved PI3K/Akt-GSK3β-TFEB axis-mediated lysosomal function improvement.
Collapse
Affiliation(s)
- Bo Hou
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yankun Li
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Xue Li
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Congying Zhang
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Zhonghua Zhao
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Qi Chen
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Nong Zhang
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Hui Li
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
| |
Collapse
|
10
|
Russell KL, Gorgulho CM, Allen A, Vakaki M, Wang Y, Facciabene A, Lee D, Roy P, Buchser WJ, Appleman LJ, Maranchie J, Storkus WJ, Lotze MT. Inhibiting Autophagy in Renal Cell Cancer and the Associated Tumor Endothelium. ACTA ACUST UNITED AC 2020; 25:165-177. [PMID: 31135523 PMCID: PMC10395074 DOI: 10.1097/ppo.0000000000000374] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The clear cell subtype of kidney cancer encompasses most renal cell carcinoma cases and is associated with the loss of von Hippel-Lindau gene function or expression. Subsequent loss or mutation of the other allele influences cellular stress responses involving nutrient and hypoxia sensing. Autophagy is an important regulatory process promoting the disposal of unnecessary or degraded cellular components, tightly linked to almost all cellular processes. Organelles and proteins that become damaged or that are no longer needed in the cell are sequestered and digested in autophagosomes upon fusing with lysosomes, or alternatively, released via vesicular exocytosis. Tumor development tends to disrupt the regulation of the balance between this process and apoptosis, permitting prolonged cell survival and increased replication. Completed trials of autophagic inhibitors using hydroxychloroquine in combination with other anticancer agents including rapalogues and high-dose interleukin 2 have now been reported. The complex nature of autophagy and the unique biology of clear cell renal cell carcinoma warrant further understanding to better develop the next generation of relevant anticancer agents.
Collapse
Affiliation(s)
| | | | - Abigail Allen
- Bioengineering, University of Pittsburgh, Pittsburgh, PA
| | | | | | - Andrea Facciabene
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA
| | | | - Partha Roy
- Bioengineering, University of Pittsburgh, Pittsburgh, PA
| | | | | | | | | | | |
Collapse
|
11
|
Ischemic Postconditioning Alleviates Intestinal Ischemia-Reperfusion Injury by Enhancing Autophagy and Suppressing Oxidative Stress through the Akt/GSK-3 β/Nrf2 Pathway in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6954764. [PMID: 32256957 PMCID: PMC7102478 DOI: 10.1155/2020/6954764] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 12/26/2019] [Accepted: 01/29/2020] [Indexed: 02/06/2023]
Abstract
Aims Ischemic postconditioning (IPO) has a strong protective effect against intestinal ischemia-reperfusion (IIR) injury that is partly related to autophagy. However, the precise mechanisms involved are unknown. Methods C57BL/6J mice were subjected to unilateral IIR with or without IPO. After 45 min ischemia and 120 min reperfusion, intestinal tissues and blood were collected for examination. HE staining and Chiu's score were used to evaluate pathologic injury. We test markers of intestinal barrier function and oxidative stress. Finally, we used WB to detect the expression of key proteins of autophagy and the Akt/GSK-3β/Nrf2 pathway. Results IPO significantly attenuated IIR injury. Expression levels of LC3 II/I, Beclin-1, and p62 were altered during IIR, indicating that IPO enhanced autophagy. IPO also activated Akt, inhibited GSK-3β/Nrf2 pathway. Conclusion Our study indicates that IPO can ameliorate IIR injury by evoking autophagy, activating Akt, inactivating GSK-3β, and activating Nrf2. These findings may provide novel insights for the alleviation of IIR injury.β/Nrf2 pathway.
Collapse
|
12
|
Syndecan-1 Shedding Inhibition to Protect Against Ischemic Acute Kidney Injury Through HGF Target Signaling Pathway. Transplantation 2019; 102:e331-e344. [PMID: 29557914 DOI: 10.1097/tp.0000000000002170] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND The hepatocyte growth factor (HGF) target pathway plays pivotal renoprotective roles after acute kidney injury. Syndecan-1 (SDC-1) serves as the coreceptor for HGF. Shedding of SDC-1 is involved in various pathological processes. Thus, we hypothesized that ischemia/reperfusion injury induced SDC-1 shedding, and inhibiting SDC-1 shedding would protect against kidney injury by potentiating activation of the HGF receptor mesenchymal epithelial transition factor (c-Met). METHODS Expression of SDC-1 and its sheddases were observed in kidneys of sham and ischemia/reperfusion (I/R) mice. To inhibit SDC-1 shedding, mice were injected with the sheddase inhibitor GM6001 before I/R surgery, and then, renal inflammation, tubular apoptosis, and activation of the c-Met/AKT/glycogen synthase kinase-3β (GSK-3β) pathway were analyzed. In vitro, human proximal tubular cell lines were pretreated with GM6001 under hypoxia/reperfusion conditions. The apoptosis and viability of cells and expression of c-Met/AKT/GSK-3β pathway components were evaluated. The relationship was further confirmed by treatment with SU11274, a specific inhibitor of phospho-c-Met. RESULTS Shedding of SDC-1 was induced after ischemia/reperfusion injury both in vivo and in vitro. GM6001 pretreatment suppressed SDC-1 shedding, alleviated renal inflammation and tubular apoptosis, and upregulated phosphorylation of the c-Met/AKT/GSK-3β pathway. In vitro, pretreatment with GM6001 also decreased hypoxia/reperfusion-induced cell apoptosis and promoted activation of the c-Met pathway. In addition, the cytoprotective role of GM6001 was attenuated by suppressing c-Met phosphorylation with SU11274. CONCLUSIONS Our findings suggest that inhibiting I/R-induced SDC-1 shedding protected against ischemic acute kidney injury by potentiating the c-Met/AKT/GSK-3β pathway.
Collapse
|
13
|
Li Q, Zeng Y, Jiang Q, Wu C, Zhou J. Role of mTOR signaling in the regulation of high glucose-induced podocyte injury. Exp Ther Med 2019; 17:2495-2502. [PMID: 30906437 PMCID: PMC6425130 DOI: 10.3892/etm.2019.7236] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 12/12/2018] [Indexed: 02/06/2023] Open
Abstract
Podocyte injury, which promotes progressive nephropathy, is considered a key factor in the progression of diabetic nephropathy. The mammalian target of rapamycin (mTOR) signaling cascade controls cell growth, survival and metabolism. The present study investigated the role of mTOR signaling in regulating high glucose (HG)-induced podocyte injury. MTT assay and flow cytometry assay results indicated that HG significantly increased podocyte viability and apoptosis. HG effects on podocytes were suppressed by mTOR complex 1 (mTORC1) inhibitor, rapamycin, and further suppressed by dual mTORC1 and mTORC2 inhibitor, KU0063794, when compared with podocytes that received mannitol treatment. In addition, western blot analysis revealed that the expression levels of Thr-389-phosphorylated p70S6 kinase (p-p70S6K) and phosphorylated Akt (p-Akt) were significantly increased by HG when compared with mannitol treatment. Notably, rapamycin significantly inhibited HG-induced p-p70S6K expression, but did not significantly impact p-Akt expression. However, KU0063794 significantly inhibited the HG-induced p-p70S6K and p-Akt expression levels. Furthermore, the expression of ezrin was significantly reduced by HG when compared with mannitol treatment; however, α-smooth muscle actin (α-SMA) expression was significantly increased. Immunofluorescence analysis on ezrin and α-SMA supported the results of western blot analysis. KU0063794, but not rapamycin, suppressed the effect of HG on the expression levels of ezrin and α-SMA. Thus, it was suggested that the increased activation of mTOR signaling mediated HG-induced podocyte injury. In addition, the present findings suggest that the mTORC1 and mTORC2 signaling pathways may be responsible for the cell viability and apoptosis, and that the mTORC2 pathway could be primarily responsible for the regulation of cytoskeleton-associated proteins.
Collapse
Affiliation(s)
- Qiuyue Li
- Nephrology Department, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yan Zeng
- Nephrology Department, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Qing Jiang
- Nephrology Department, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Cong Wu
- Nephrology Department, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Jing Zhou
- Nephrology Department, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
14
|
A Glimpse of the Mechanisms Related to Renal Fibrosis in Diabetic Nephropathy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1165:49-79. [PMID: 31399961 DOI: 10.1007/978-981-13-8871-2_4] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Diabetic nephropathy (DN) is a common kidney disease in people with diabetes, which is also a serious microvascular complication of diabetes and the main cause of end-stage renal disease (ESRD) in developed and developing countries. Renal fibrosis is a finally pathological change in DN. Nevertheless, the relevant mechanism of cause to renal fibrosis in DN is still complex. In this review, we summarized that the role of cell growth factors, epithelial-mesenchymal transition (EMT) in the renal fibrosis of DN, we also highlighted the miRNA and inflammatory cells, such as macrophage, T lymphocyte, and mastocyte modulate the progression of DN. In addition, there are certain other mechanisms that may yet be conclusively defined. Recent studies demonstrated that some of the new signaling pathways or molecules, such as Notch, Wnt, mTOR, Epac-Rap-1 pathway, may play a pivotal role in the modulation of ECM accumulation and renal fibrosis in DN. This review aims to elucidate the mechanism of renal fibrosis in DN and has provided new insights into possible therapeutic interventions to inhibit renal fibrosis and delay the development of DN.
Collapse
|
15
|
Keshk WA, Zahran SM. Mechanistic role of cAMP and hepatocyte growth factor signaling in thioacetamide-induced nephrotoxicity: Unraveling the role of platelet rich plasma. Biomed Pharmacother 2018; 109:1078-1084. [PMID: 30551358 DOI: 10.1016/j.biopha.2018.10.121] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 10/16/2018] [Accepted: 10/20/2018] [Indexed: 12/16/2022] Open
Abstract
Chronic kidney diseases occur as result of exposure to wide range of deleterious agents as environmental pollutants, toxins and drug. Currently, there is no effective protective therapy against renal damage, fibrosis and its sequel of end stage renal disease. Platelet-rich plasma (PRP) has a progressively gained consideration in wound healing, repair/regeneration of damaged tissues and conservation of organ function. However, its impact on thioacetamide (TAA) induced chronic renal damage has not been elucidated yet. So, the present study was carried out to evaluate the possible protective and regenerative effect of PRP against TAA induced renal damage and their potential underlying mechanism. PRP treatment improved redox state, renal function disturbed histologicl features; decreased monocyte chemo-attractant protein-1 (MCP-1) level; increased Peroxisome proliferator-activated receptor gamma co-activator-1α (PGC-1α) marker of mitochondrial biogenesis and metabolism; cyclic adenosine monophosphate (cAMP); hepatocyte growth factor (HGF) and autophagy protein beclin-1 level. In addition, PRP treatment decreased apoptosis and fibrosis as evidenced by decreased active caspase3 and α-SMA expression and immunoreactivity, respectively. In conclusion, PRP could potentially protect against TTA-induced chronic kidney damage by alleviating oxidative stress, improving, mitochondrial biogenesis, autophagy, disruption of the inflammatory, apoptotic and fibrotic response induced by TTA.
Collapse
Affiliation(s)
- Walaa Arafa Keshk
- Medical Biochemistry Department, Faculty of Medicine, Tanta University, El-Geish Street, Tanta, El-Gharbia, Egypt.
| | - Samer Mahmoud Zahran
- Biochemistry Department, Faculty of Pharmacy and Drug Manufacturing, Pharos University, Alexandria, Egypt
| |
Collapse
|
16
|
Aristizabal Prada ET, Spöttl G, Maurer J, Lauseker M, Koziolek EJ, Schrader J, Grossman A, Pacak K, Beuschlein F, Auernhammer CJ, Nölting S. The role of GSK3 and its reversal with GSK3 antagonism in everolimus resistance. Endocr Relat Cancer 2018; 25:893-908. [PMID: 29895527 PMCID: PMC7439002 DOI: 10.1530/erc-18-0159] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 06/11/2018] [Indexed: 12/11/2022]
Abstract
Pancreatic neuroendocrine tumors (panNETs) are often inoperable at diagnosis. The mTORC1 inhibitor everolimus has been approved for the treatment of advanced NETs. However, the regular development of resistance to everolimus limits its clinical efficacy. We established two independent everolimus-resistant panNET (BON1) cell lines (BON1 RR1, BON1 RR2) to find potential mechanisms of resistance. After 24 weeks of permanent exposure to 10 nM everolimus, BON1 RR1 and BON1 RR2 showed stable resistance with cellular survival rates of 96.70% (IC50 = 5200 nM) and 92.30% (IC50 = 2500 nM), respectively. The control cell line showed sensitivity to 10 nM everolimus with cellular survival declining to 54.70% (IC50 = 34 nM). Both resistant cell lines did not regain sensitivity over time and showed persistent stable resistance after a drug holiday of 13 weeks. The mechanisms of resistance in our cell line model included morphological adaptations, G1 cell cycle arrest associated with reduced CDK1(cdc2) expression and decreased autophagy. Cellular migration potential was increased and indirectly linked to c-Met activation. GSK3 was over-activated in association with reduced baseline IRS-1 protein levels. Specific GSK3 inhibition strongly decreased BON1 RR1/RR2 cell survival. The combination of everolimus with the PI3Kα inhibitor BYL719 re-established everolimus sensitivity through GSK3 inhibition and restoration of autophagy. We suggest that GSK3 over-activation combined with decreased baseline IRS-1 protein levels and decreased autophagy may be a crucial feature of everolimus resistance, and hence, a possible therapeutic target.
Collapse
Affiliation(s)
- Elke Tatjana Aristizabal Prada
- Medizinische Klinik und Poliklinik IVInterdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM), Klinikum der Universität München (KUM), Ludwig-Maximilians-University, Munich, Germany
| | - Gerald Spöttl
- Medizinische Klinik und Poliklinik IVInterdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM), Klinikum der Universität München (KUM), Ludwig-Maximilians-University, Munich, Germany
| | - Julian Maurer
- Medizinische Klinik und Poliklinik IVInterdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM), Klinikum der Universität München (KUM), Ludwig-Maximilians-University, Munich, Germany
| | - Michael Lauseker
- Institute for Medical Information SciencesBiometry, and Epidemiology, Campus Grosshadern, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Eva Jolanthe Koziolek
- Department of Nuclear MedicineUniversity Medical Center Charité, Berlin, Germany
- German Cancer Consortium (DKTK)Heidelberg, Germany
- German Cancer Research Center (DKFZ)Heidelberg, Germany
| | - Jörg Schrader
- I. Medizinische Klinik und PoliklinikUniversitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Ashley Grossman
- Oxford Centre for DiabetesEndocrinology and Metabolism, University of Oxford, Oxford, UK
- Royal Free Hospital ENETS Centre of ExcellenceLondon, UK
| | - Karel Pacak
- Eunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of Health, Bethesda, Maryland, USA
| | - Felix Beuschlein
- Medizinische Klinik und Poliklinik IVInterdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM), Klinikum der Universität München (KUM), Ludwig-Maximilians-University, Munich, Germany
- Klinik für EndokrinologieDiabetologie und Klinische Ernährung, Universitätsspital Zürich, Zurich, Switzerland
| | - Christoph Joseph Auernhammer
- Medizinische Klinik und Poliklinik IVInterdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM), Klinikum der Universität München (KUM), Ludwig-Maximilians-University, Munich, Germany
| | - Svenja Nölting
- Medizinische Klinik und Poliklinik IVInterdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM), Klinikum der Universität München (KUM), Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
17
|
Yuan Y, Li X, Li M. Overexpression of miR‑17‑5p protects against high glucose‑induced endothelial cell injury by targeting E2F1‑mediated suppression of autophagy and promotion of apoptosis. Int J Mol Med 2018; 42:1559-1568. [PMID: 29786752 DOI: 10.3892/ijmm.2018.3697] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 05/03/2018] [Indexed: 11/05/2022] Open
Abstract
E2 promoter binding factor 1 (E2F1) has been reported to have an important regulatory role in cell survival during hyperglycemic conditions; however, the mechanisms remain to be fully elucidated. Bioinformatics analyses have suggested that microRNA (miR)‑17‑5p targets the 3'untranslated region (3'UTR) of E2F1. The aim of the present study was to characterize the protective effect of miR‑17‑5p/E2F1 on human umbilical vein endothelial cells (HUVECs) under high glucose (HG) conditions, to confirm the regulatory effect of miR‑17‑5p on E2F1/AMP‑activated protein kinase α2 (AMPKα2)‑mediated apoptosis and E2F1/mammalian target of rapamycin complex 1 (mTORC1)‑mediated autophagy. Bifluorescein experiments were performed to characterize the interaction between miR‑17‑5p and E2F1. The Cell Counting Kit‑8 assay, flow cytometry, immunofluorescence, and reverse transcription‑quantitative polymerase chain reaction and western blot analyses were used to detect cell viability, apoptosis, autophagy, and relative mRNA and protein expression, respectively. The results showed that HG induced the downregulation of miR‑17‑5p and upregulation of E2F1 during HUVEC injury. The downregulation of E2F1 inhibited HG‑induced HUVEC dysfunction by suppressing mTORC1‑mediated inhibition of autophagy and AMPKα2‑mediated promotion of apoptosis. The results suggested that inhibiting the expression of E2F1 protected against HG‑induced HUVEC injury via the activation of autophagy. The overexpression of miR‑17‑5p inhibited E2F1‑mediated HUVEC injury under HG conditions, which was reversed following transfection with an E2F1‑overexpression vector. The bifluorescein experiments showed that miR‑17‑5p targeted the 3'UTR of E2F1. Taken together, the results suggested that the expression of miR‑17‑5p inhibited HG‑induced endothelial cell injury by targeting E2F1.
Collapse
Affiliation(s)
- Yifeng Yuan
- Department of Interventional and Vascular Surgery, Tenth People's Hospital of Tongji University, Shanghai 200072, P.R. China
| | - Xue Li
- Department of Interventional and Vascular Surgery, Tenth People's Hospital of Tongji University, Shanghai 200072, P.R. China
| | - Maoquan Li
- Department of Interventional and Vascular Surgery, Tenth People's Hospital of Tongji University, Shanghai 200072, P.R. China
| |
Collapse
|
18
|
Zhao X, Chen Y, Tan X, Zhang L, Zhang H, Li Z, Liu S, Li R, Lin T, Liao R, Zhang Q, Dong W, Shi W, Liang X. Advanced glycation end-products suppress autophagic flux in podocytes by activating mammalian target of rapamycin and inhibiting nuclear translocation of transcription factor EB. J Pathol 2018; 245:235-248. [PMID: 29570219 PMCID: PMC5969319 DOI: 10.1002/path.5077] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 02/12/2018] [Accepted: 03/14/2018] [Indexed: 12/24/2022]
Abstract
Insufficient autophagy in podocytes is related to podocyte injury in diabetic nephropathy (DN). Advanced glycation end‐products (AGEs) are major factors of podocyte injury in DN. However, the role and mechanism of AGEs in autophagic dysfunction remain unknown. We investigated autophagic flux in AGE‐stimulated cultured podocytes using multiple assays: western blotting, reverse transcription–quantitative PCR, immunofluorescence staining, and electron microscopy. We also utilized chloroquine and a fluorescent probe to monitor the formation and turnover of autophagosomes. Mice of the db/db strain were used to model diabetes mellitus (DM) with high levels of AGEs. To mimic DM with normal levels of AGEs as a control, we treated db/db mice with pyridoxamine to block AGE formation. AGEs impaired autophagic flux in the cultured podocytes. Compared with db/db mice with normal AGEs but high glucose levels, db/db mice with high AGEs and high glucose levels exhibited lower autophagic activity. Aberrant autophagic flux was related to hyperactive mammalian target of rapamycin (mTOR), a major suppressor of autophagy. Pharmacologic inhibition of mTOR activity restored impaired autophagy. AGEs inhibited the nuclear translocation and activity of the pro‐autophagic transcription factor EB (TFEB) and thus suppressed transcription of its several autophagic target genes. Conversely, TFEB overexpression prevented AGE‐induced autophagy insufficiency. Attenuating mTOR activity recovered TFEB nuclear translocation under AGE stimulation. Co‐immunoprecipitation assays further demonstrated the interaction between mTOR and TFEB in AGE‐stimulated podocytes and in glomeruli from db/db mice. In conclusion, AGEs play a crucial part in suppressing podocyte autophagy under DM conditions. AGEs inhibited the formation and turnover of autophagosomes in podocytes by activating mTOR and inhibiting the nuclear translocation of TFEB. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Xingchen Zhao
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, PR China.,Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Institute of Geriatrics, Guangzhou, Guangdong, PR China
| | - Yuanhan Chen
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, PR China.,Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Institute of Geriatrics, Guangzhou, Guangdong, PR China
| | - Xiaofan Tan
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Institute of Geriatrics, Guangzhou, Guangdong, PR China.,Division of Nephrology, Zhongshan City People's Hospital, Zhongshan Hospital of Sun Yat-sen University, Zhongshan, Guangdong, PR China
| | - Li Zhang
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Institute of Geriatrics, Guangzhou, Guangdong, PR China
| | - Hong Zhang
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Institute of Geriatrics, Guangzhou, Guangdong, PR China
| | - Zhilian Li
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Institute of Geriatrics, Guangzhou, Guangdong, PR China
| | - Shuangxin Liu
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Institute of Geriatrics, Guangzhou, Guangdong, PR China
| | - Ruizhao Li
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Institute of Geriatrics, Guangzhou, Guangdong, PR China
| | - Ting Lin
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Institute of Geriatrics, Guangzhou, Guangdong, PR China
| | - Ruyi Liao
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Institute of Geriatrics, Guangzhou, Guangdong, PR China
| | - Qianmei Zhang
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Institute of Geriatrics, Guangzhou, Guangdong, PR China
| | - Wei Dong
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Institute of Geriatrics, Guangzhou, Guangdong, PR China
| | - Wei Shi
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, PR China.,Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Institute of Geriatrics, Guangzhou, Guangdong, PR China
| | - Xinling Liang
- Division of Nephrology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangdong Provincial Institute of Geriatrics, Guangzhou, Guangdong, PR China
| |
Collapse
|
19
|
Effect of 1,25(OH)2D3 on high glucose‑induced autophagy inhibition in peritoneum. Mol Med Rep 2017; 16:7080-7085. [PMID: 28901396 DOI: 10.3892/mmr.2017.7408] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 07/14/2017] [Indexed: 11/05/2022] Open
Abstract
High glucose (HG) may damage the structure and function of the peritoneal membrane, and is considered to be one of the most important factors that leads to peritoneal fibrosis and ultrafiltration failure. Recently, 1,25(OH)2D3, the active form of vitamin D, was demonstrated to protect against epithelial‑mesenchymal transition and fibrosis in peritoneal mesothelium and other organs. Accumulating evidence has suggested that autophagy serves a protective role in certain diseases by regulating cell survival. The present study examined whether 1,25(OH)2D3 has an effect on autophagy in peritoneal mesothelial cells. The protein level of Beclin, anti‑ubiquitin‑binding protein p62 (p62), microtubule‑associated proteins 1A/1B light chain 3B (LC3-II), mechanistic target of rapamycin (mTOR) and phosphorylated mTOR were evaluated by western blot analysis. Autophagosomes were detected under transmission electron microscopy. It was revealed that exposure to HG inhibited autophagy in peritoneal mesothelial cells. However, 1,25(OH)2D3 alleviated autophagy inhibition induced by HG in human peritoneal mesothelial cells, which activated expression of autophagy‑associated genes encoding Beclin‑1 and LC3-II downregulated the expression of p62 via mTOR signaling pathway. In a mouse model of HG‑treated peritoneal mesothelium, autophagy inhibition was observed in peritoneum, 1,25(OH)2D3 attenuated HG‑induced autophagy inhibition in peritoneal mesothelium via the mTOR signaling pathway. These findings suggested that 1,25(OH)2D3 may be a potential therapy for peritoneal injury.
Collapse
|
20
|
Su CL, Tseng CL, Ramesh C, Liu HS, Huang CYF, Yao CF. Using gene expression database to uncover biology functions of 1,4-disubstituted 1,2,3-triazole analogues synthesized via a copper (I)-catalyzed reaction. Eur J Med Chem 2017; 132:90-107. [PMID: 28342400 DOI: 10.1016/j.ejmech.2017.03.034] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 03/14/2017] [Accepted: 03/15/2017] [Indexed: 12/18/2022]
Abstract
We have synthesized bioactive 1,4-disubstituted 1,2,3-triazole analogues containing 2H-1,4-benzoxazin-3-(4H)-one derivatives via 1,3-dipolar cycloaddition in the presence of CuI. All the reactions proceeded smoothly and afforded its desired products in excellent yields. Among these analogues, 3y exhibited a better cytotoxic effect on human hepatocellular carcinoma (HCC) Hep 3B cells and displayed less cytotoxicity on normal human umbilical vein endothelial cells, compared with Sorafenib, a targeted therapy for advanced HCC. 3y also induced stronger apoptosis and autophagy. Addition of curcumin enhanced 3y-induced cytotoxicity by further induction of autophagy. Using gene expression signatures of 3y to query Connectivity Map, a glycogen synthase kinase-3 inhibitor (AR-A014418) was predicted to display similar molecular action of 3y. Experiments further demonstrate that AR-A014418 acted like 3y, and vice versa. Overall, our data suggest the chemotherapeutic potential of 3y on HCC.
Collapse
Affiliation(s)
- Chun-Li Su
- Department of Human Development and Family Studies, National Taiwan Normal University, Taipei 106, Taiwan.
| | - Chia-Ling Tseng
- Department of Human Development and Family Studies, National Taiwan Normal University, Taipei 106, Taiwan
| | - Chintakunta Ramesh
- Department of Chemistry, National Taiwan Normal University, Taipei 116, Taiwan
| | - Hsiao-Sheng Liu
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; Center of Infectious Disease and Signaling Research Center, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Chi-Ying F Huang
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei 112, Taiwan; Department of Biochemistry, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Ching-Fa Yao
- Department of Chemistry, National Taiwan Normal University, Taipei 116, Taiwan.
| |
Collapse
|
21
|
Sun J, Li ZP, Zhang RQ, Zhang HM. Repression of miR-217 protects against high glucose-induced podocyte injury and insulin resistance by restoring PTEN-mediated autophagy pathway. Biochem Biophys Res Commun 2016; 483:318-324. [PMID: 28017719 DOI: 10.1016/j.bbrc.2016.12.145] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 12/21/2016] [Indexed: 12/20/2022]
Abstract
Podocyte damage is the initial hallmark of diabetic nephropathy (DN), leading to the increasing morbidity and mortality in diabetic patients. Recent researches have corroborated the critical roles of miRNAs in the pathological progression of DN. Here, elevation of miR-217 was verified in high glucose (HG)-stimulated podocytes. Moreover, blocking miR-217 expression antagonized HG-induced cell injury by attenuating the adverse role of HG on cell viability and inhibiting ROS levels and cell apoptosis. Simultaneously, miR-217 repression restored HG-disrupted insulin resistance by elevating glucose uptake and nephrin expression, an essential component for insulin-induced glucose uptake. Mechanism assay substantiated the defective autophagy in HG-treated podocytes, which was resumed by miR-217 cessation. Importantly, suppressing autophagy pathway with 3-MA alleviated the protective roles of miR-217 down-regulation in podocyte injury and insulin resistance. Luciferase reporter analysis confirmed that PTEN was a target of miR-217 in podocytes. Additionally, blocking PTEN expression restrained autophagy restoration in miR-217-decreased cells. Furthermore, PTEN down-regulation attenuated the beneficial role of miR-217 suppression in HG-induced injury and insulin resistance. Together, this study manifests that miR-217inhibition can protectively antagonize HG-induced podocyte damage and insulin resistance by restoring the defective autophagy pathway via targeting PTEN, representing a novel and promising therapeutic target against diabetic nephropathy.
Collapse
Affiliation(s)
- Juan Sun
- School of Nursing, Xinxiang Medical University, Xinxiang, Henan, 453003, PR China
| | - Zhao Pin Li
- School of International Education, Xin Xiang Medical University, Xinxiang, Henan, 453003, PR China
| | - Rui Qin Zhang
- School of Nursing, Xinxiang Medical University, Xinxiang, Henan, 453003, PR China
| | - Hui Min Zhang
- School of Nursing, Xinxiang Medical University, Xinxiang, Henan, 453003, PR China.
| |
Collapse
|