1
|
Liu Q, Hao T, Yang B, Zhang J, Pan S, Wu C, Tang Y, Zhou Y, Zhao Z, Du J, Li Y, Mai K, Ai Q. Autophagy dysfunction links palmitic acid with macrophage inflammatory responses in large yellow croaker (Larimichthys crocea). FISH & SHELLFISH IMMUNOLOGY 2025; 163:110319. [PMID: 40209962 DOI: 10.1016/j.fsi.2025.110319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 04/07/2025] [Accepted: 04/07/2025] [Indexed: 04/12/2025]
Abstract
Autophagy is a cellular degradation process reliant on lysosome, crucial for preserving intracellular homeostasis. The key saturated fatty acid palmitic acid (PA) has been demonstrated to exert regulatory effects on autophagic activity in mammals. However, the precise impact of PA on autophagy and its role in fish remains incompletely understood. Thus, this study aimed to investigate the regulation of PA on autophagy and explore the role of autophagy in inflammatory responses triggered by PA in the head kidney macrophages of large yellow croaker. This study indicates that PA exposure can inhibit macrophage autophagy by reducing the expression of genes related to autophagy (e.g., beclin1, ulk1, and lc3), activating the negative regulator mTORC1 signaling pathway (p70S6K and S6), and hindering autophagic flux. This effect was observed to be amplified with increasing exposure time and concentration of PA. Similarly to the in vitro results, the palm oil (PO) diet significantly reduced autophagic activity in the head kidney of the croaker in vivo. Subsequent studies demonstrated that restoring autophagy led to a notable reduction in the expression of PA and PO-induced pro-inflammatory genes (il-1β, il-6, tnf-α, and cox-2), the activation of the MAPK signaling pathway (p38 and JNK), and the NLRP3 inflammasome levels, both in vitro and in vivo. In contrast, further inhibition of autophagy produced the opposite effect in vitro. In conclusion, this study demonstrates that PA exerts a dynamic inhibitory effect on autophagy in the head kidney macrophage, which in turn promotes PA-induced inflammatory responses. These findings provide valuable insights into how PA influences autophagy and inflammatory responses in fish immune cells, contributing to the theoretical framework for improving the use of vegetable oils in aquaculture.
Collapse
Affiliation(s)
- Qiangde Liu
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, PR China
| | - Tingting Hao
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, PR China
| | - Bingyuan Yang
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, PR China
| | - Jinze Zhang
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, PR China
| | - Shijie Pan
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, PR China
| | - Caixia Wu
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, PR China
| | - Yuhang Tang
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, PR China
| | - Yan Zhou
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, PR China
| | - Zengqi Zhao
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, PR China
| | - Jianlong Du
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, PR China
| | - Yueru Li
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, PR China
| | - Kangsen Mai
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, PR China
| | - Qinghui Ai
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, PR China.
| |
Collapse
|
2
|
Fan Y, He J, Shi L, Zhang M, Chen Y, Xu L, Han N, Jiang Y. Identification of potential key lipid metabolism-related genes involved in tubular injury in diabetic kidney disease by bioinformatics analysis. Acta Diabetol 2024; 61:1053-1068. [PMID: 38691241 DOI: 10.1007/s00592-024-02278-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 03/19/2024] [Indexed: 05/03/2024]
Abstract
AIMS Accumulating evidences indicate that abnormalities in tubular lipid metabolism play a crucial role in the development of diabetic kidney disease (DKD). We aim to identify novel lipid metabolism-related genes associated with tubular injury in DKD by utilizing bioinformatics approaches. METHODS Differentially expressed genes (DEGs) between control and DKD tubular tissue samples were screened from the Gene Expression Omnibus (GEO) database, and then were intersected with lipid metabolism-related genes. Hub genes were further determined by combined weighted gene correlation network analysis (WGCNA) and protein-protein interaction (PPI) network. We performed enrichment analysis, immune analysis, clustering analysis, and constructed networks between hub genes and miRNAs, transcription factors and small molecule drugs. Receiver operating characteristic (ROC) curves were employed to evaluate the diagnostic efficacy of hub genes. We validated the relationships between hub genes and DKD with external datasets and our own clinical samples. RESULTS There were 5 of 37 lipid metabolism-related DEGs identified as hub genes. Enrichment analysis demonstrated that lipid metabolism-related DEGs were enriched in pathways such as peroxisome proliferator-activated receptors (PPAR) signaling and pyruvate metabolism. Hub genes had potential regulatory relationships with a variety of miRNAs, transcription factors and small molecule drugs, and had high diagnostic efficacy. Immune infiltration analysis revealed that 13 immune cells were altered in DKD, and hub genes exhibited significant correlations with a variety of immune cells. Through clustering analysis, DKD patients could be classified into 3 immune subtypes and 2 lipid metabolism subtypes, respectively. The tubular expression of hub genes in DKD was further verified by other external datasets, and immunohistochemistry (IHC) staining showed that except ACACB, the other 4 hub genes (LPL, AHR, ME1 and ALOX5) exhibited the same results as the bioinformatics analysis. CONCLUSION Our study identified several key lipid metabolism-related genes (LPL, AHR, ME1 and ALOX5) that might be involved in tubular injury in DKD, which provide new insights and perspectives for exploring the pathogenesis and potential therapeutic targets of DKD.
Collapse
Affiliation(s)
- Yuanshuo Fan
- Department of Endocrinology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Juan He
- Department of Endocrinology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China.
| | - Lixin Shi
- Department of Endocrinology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China.
| | - Miao Zhang
- Department of Endocrinology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Ye Chen
- Department of Nephrology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Lifen Xu
- Department of Pathology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Na Han
- Department of Endocrinology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Yuecheng Jiang
- Guizhou Provincial People's Hospital, Guiyang, 550002, China
| |
Collapse
|
3
|
The genetic side of diabetic kidney disease: a review. Int Urol Nephrol 2023; 55:335-343. [PMID: 35974289 DOI: 10.1007/s11255-022-03319-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 07/24/2022] [Indexed: 01/25/2023]
Abstract
BACKGROUND Diabetic kidney disease (DKD) is one of the most common complications of diabetes, with approximately 30-40% of patients with type 1 diabetes mellitus and 20% of patients with type 2 diabetes mellitus eventually developing DKD. If DKD is not controlled in the early clinical stage and proteinuria develops, the disease will progress to end-stage renal disease. The pathogenesis of DKD remains largely unknown and is multifactorial, likely due to interactions between genetic and environmental factors. Familial clustering also supports a critical role of hereditary factors in DKD. The development of gene detection technology has promoted the exploration of DKD susceptibility genes in different cohorts of patients with diabetes. Identifying susceptibility genes can provide insights into the pathogenesis of DKD, as well as a basis for its clinical diagnosis and therapy. RESULTS Numerous candidate gene loci have been found to be associated with DKD, many of which play critical regulatory roles in the pathogenesis of this disease, including genes involved in glycol-metabolism, lipid metabolism, the renin-angiotensin-aldosterone system, inflammation and oxidative stress. In this review, we summarize the functions of several susceptibility genes involved in the development of DKD. CONCLUSION Based on our findings, we recommend that studying susceptibility gene polymorphisms can lead to a better understanding of the pathogenesis of DKD and could help prevent this disease or improve its outcomes.
Collapse
|
4
|
Kayampilly P, Roeser N, Rajendiran TM, Pennathur S, Afshinnia F. Acetyl Co-A Carboxylase Inhibition Halts Hyperglycemia Induced Upregulation of De Novo Lipogenesis in Podocytes and Proximal Tubular Cells. Metabolites 2022; 12:940. [PMID: 36295842 PMCID: PMC9610518 DOI: 10.3390/metabo12100940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 09/28/2022] [Accepted: 09/30/2022] [Indexed: 11/28/2022] Open
Abstract
The effect of glycemic stress on de novo lipogenesis (DNL) in podocytes and tubular epithelial cells is understudied. This study is aimed (A) to show the effect of glycemic stress on DNL, and (B) to assess the effect of acetyl-Co A (ACC) inhibition on halting upregulation of DNL, on the expression of other lipid regulatory genes in the DNL pathway, and on markers of fibrosis and apoptosis in podocytes and tubular epithelial cells. We used cultured mouse primary tubular epithelial cells, mouse proximal tubular (BUMPT) cells, and immortal mouse podocytes and measured their percentage of labeled 13C2-palmitate as a marker of DNL after incubation with 13C2 acetate in response to high glucose concentration (25 mM). We then tested the effect of ACC inhibition by complimentary strategies utilizing CRISPR/cas9 deletion or incubation with Acaca and Acacb GapmeRs or using a small molecule inhibitor on DNL under hyperglycemic concentration. Exposure to high glucose concentration (25 mM) compared to osmotic controlled low glucose concentration (5.5 mM) significantly increased labeled palmitate after 24 h up to 72 h in podocytes and primary tubular cells. Knocking out of the ACC coding Acaca and Acacb genes by CRISPR/cas9, downregulation of Acaca and Acacb by specific antisense LNA GapmeRs and inhibition of ACC by firsocostat similarly halted/mitigated upregulation of DNL and decreased markers of fibrosis and programmed cell death in podocytes and various tubular cells. ACC inhibition is a potential therapeutic target to mitigate or halt hyperglycemia-induced upregulation of DNL in podocytes and tubular cells.
Collapse
Affiliation(s)
- Pradeep Kayampilly
- Department of Internal Medicine-Nephrology, University of Michigan, Ann Arbor, MI 48105, USA
| | - Nancy Roeser
- Department of Internal Medicine-Nephrology, University of Michigan, Ann Arbor, MI 48105, USA
| | - Thekkelnaycke M Rajendiran
- Michigan Regional Comprehensive Metabolomics Resource Core, University of Michigan, Ann Arbor, MI 48105, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Subramaniam Pennathur
- Department of Internal Medicine-Nephrology, University of Michigan, Ann Arbor, MI 48105, USA
- Michigan Regional Comprehensive Metabolomics Resource Core, University of Michigan, Ann Arbor, MI 48105, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Farsad Afshinnia
- Department of Internal Medicine-Nephrology, University of Michigan, Ann Arbor, MI 48105, USA
| |
Collapse
|
5
|
Lin D, Qin R, Guo L. Thyroid stimulating hormone aggravates diabetic retinopathy through the mitochondrial apoptotic pathway. J Cell Physiol 2021; 237:868-880. [PMID: 34435365 DOI: 10.1002/jcp.30563] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 08/11/2021] [Accepted: 08/12/2021] [Indexed: 11/09/2022]
Abstract
Diabetic retinopathy (DR) is a common complication of diabetes mellitus. High glucose-induced mitochondrial apoptosis is involved in the loss of retinal pericytes (PCs), which is considered to be a predominant pathologic change of diabetic retinopathy (DR). A high thyroid stimulating hormone (TSH) serum level is associated with an increased prevalence of DR in diabetic patients. Here, we investigated whether TSH regulated glucose-induced PCs loss through TSH-receptor (TSHR)-dependent mitochondrial apoptosis. First, the serum TSH level was found to be an independent risk factor for DR in Type 2 diabetic study participants (odds ratio = 2.294; 95% confidence interval: 1.925-2.733; p ≤ 0.001). Second, human PCs were treated with different concentrations of glucose, with or without bovine TSH (b-TSH). Glucose induced mitochondrial apoptosis through various mechanisms, including through regulating the expression of apoptosis-related proteins and inducing mitochondrial dysfunction, which could be deteriorated by costimulation of glucose and b-TSH. Additionally, we detected functional TSHR in PCs; blocking TSHR significantly restricted TSH-induced apoptosis. Thus, the presence of functional TSHR in human retinal PCs may facilitate the effect of high TSH on high glucose-induced PCs loss through TSHR-dependent mitochondrial apoptosis.
Collapse
Affiliation(s)
- Dong Lin
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology, Beijing, China.,Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Department of Immunology, School of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ruijie Qin
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology, Beijing, China.,Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Lixin Guo
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology, Beijing, China.,Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
6
|
An Overview of Lipid Metabolism and Nonalcoholic Fatty Liver Disease. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4020249. [PMID: 32733940 PMCID: PMC7383338 DOI: 10.1155/2020/4020249] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 06/14/2020] [Accepted: 06/25/2020] [Indexed: 12/11/2022]
Abstract
The occurrence of nonalcoholic fatty liver disease (NAFLD) is associated with major abnormalities of hepatic lipid metabolism. We propose that lipid abnormalities directly or indirectly contribute to NAFLD, especially fatty acid accumulation, arachidonic acid metabolic disturbance, and ceramide overload. The effects of lipid intake and accumulation on NAFLD and NAFLD treatment are explained with theoretical and experimental details. Overall, these findings provide further understanding of lipid metabolism in NAFLD and may lead to novel therapies.
Collapse
|
7
|
Inhibitor of growth 2 regulates the high glucose-induced cell cycle arrest and epithelial-to-mesenchymal transition in renal proximal tubular cells. J Physiol Biochem 2020; 76:373-382. [PMID: 32424454 DOI: 10.1007/s13105-020-00743-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 04/23/2020] [Indexed: 02/06/2023]
Abstract
The epithelial-to-mesenchymal transition (EMT)-based tubulointerstitial fibrosis is the major pathological feature of diabetic kidney disease (DKD). While several studies have linked cell cycle dysregulation to various kidney injuries in recent years, its involvement in fibrosis of DKD is far from being clarified. ING2 (inhibitor of growth 2) is the second member of the inhibitor of growth family and participates in the regulation of many cellular processes. So far the role of ING2 in DKD remains largely unknown. In the present study, ING2 expression was detected by western blotting and immunofluorescent staining both in vitro high glucose-stimulated human proximal tubular epithelial cells (HK-2) and in vivo streptozotocin-induced diabetic mice. Cell proliferation was analyzed by CCK-8 and EdU assay, and cell cycle arrest was measured by flow cytometry. Quantitative polymerase chain reaction (qPCR) and western blotting were used to detect the EMT markers, and the p53 signaling activation was evaluated by chromatin immunoprecipitation (ChIP), qPCR, and western blotting. We found that the proliferation of the cells was reduced upon high glucose stimulation, which was accompanied by cell cycle arrest. The expression of ING2 was increased in hyperglycemia conditions both in vivo and in vitro. ING2 suppression ameliorated the reduced proliferation and cell cycle arrest induced by high glucose in HK-2 cells. Moreover, ING2 knockdown suppressed p21 expression by reducing p53 acetylation and finally alleviated the EMT progress in the high glucose-stimulated HK-2 cells. Our study demonstrated that cell cycle regulation is bound up with the kidney fibrosis in DKD, suggesting a novel function of ING2 as a potential therapeutic strategy targeting cell cycle arrest for DKD.
Collapse
|
8
|
GCN5L1 controls renal lipotoxicity through regulating acetylation of fatty acid oxidation enzymes. J Physiol Biochem 2019; 75:597-606. [PMID: 31760589 DOI: 10.1007/s13105-019-00711-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 10/31/2019] [Indexed: 01/25/2023]
Abstract
Dyslipidemia is a common risk factor of chronic kidney disease (CKD). Current notion suggests that insufficient intracellular fatty acid oxidation (FAO) and subsequently enhanced fatty acid esterification within renal resident cells, a process termed as renal lipotoxicity, is the key pathogenic event responsible for dyslipidemia-induced kidney injury. However, the detailed mechanism is not fully elucidated. Recently, accumulating data indicated that acetylation modification is an important regulating manner for both mitochondrial function and energy metabolism, while whether acetylation modification is involved in renal lipotoxicity is of little known. In the present study, the expression level of global lysine acetylation was detected by immunohistochemistry in high-fat diet mice and western blot in palmitic acid (PA) stimulated HK-2 cells. The acetylation levels of long-chain acyl-CoA dehydrogenases (LCAD) and β-hydroxyacyl-CoA dehydrogenase (β-HAD) were measured by immunoprecipitation. And a multifunction microplate reader was applied to detect FAO rate, triglyceride and acyl-CoA contents, and the enzyme activities, with cellular lipid accumulation identified by Oil Red O staining. We evidenced the acetylation levels of LCAD and β-HAD that were enhanced, which led to decreased enzymatic activities and impaired FAO rate. Furthermore, renal protein hyperacetylation induced by lipid overload was associated with increased expression of GCN5L1. And the silence of GCN5L1 in tubular epithelial cells resulted in deacetylation and activation of LCAD and β-HAD. Finally, excess lipids induced lipotoxicity and epithelial-mesenchymal transition (EMT) were ameliorated by GCN5L1 suppression, suggesting GCN5L1-mediated mitochondrial LCAD and β-HAD acetylation might be a key pathogenic event underlying excess lipids induced FAO impairment.
Collapse
|
9
|
Wei L, Xiao Y, Li L, Xiong X, Han Y, Zhu X, Sun L. The Susceptibility Genes in Diabetic Nephropathy. KIDNEY DISEASES 2018; 4:226-237. [PMID: 30574499 DOI: 10.1159/000492633] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 08/03/2018] [Indexed: 12/22/2022]
Abstract
Background Diabetes mellitus (DM) poses a severe threat to global public health. Diabetic nephropathy (DN) is one of the most common complications of diabetes and the leading cause of end-stage renal disease (ESRD). Approximately 30-40% of DM patients in the world progress to ESRD, which emphasizes the effect of genetic factors on DN. Family clustering also supports the important role of hereditary factors in DN and ESRD. Therefore, a large number of genetic studies have been carried out to identify susceptibility genes in different diabetic cohorts. Extensive susceptibility genes of DN and ESRD have not been identified until recently. Summary and Key Messages Some of these associated genes function as pivotal regulators in the pathogenesis of DN, such as those related to glycometabolism and lipid metabolism. However, the functions of most of these genes remain unclear. In this article, we review several susceptibility genes according to their genetic functions to make it easier to determine their exact effect on DN and to provide a better understanding of the advancements from genetic studies. However, several challenges associated with investigating the genetic factors of DN still exist. For instance, it is difficult to determine whether these variants affect the expression of the protein they encode or other cytokines. More efforts should be made to determine how these genes influence the progression of DN. In addition, many results could not be replicated among races, suggesting that the association between genetic polymorphisms and DN is race-specific. Therefore, large, well-designed studies involving more relevant variables and ethnic groups and more relevant functional studies are urgently needed. These studies may be beneficial and retard the progression of DN by early intervention, especially for patients who carry certain risk alleles or genotypes.
Collapse
Affiliation(s)
- Ling Wei
- Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Ying Xiao
- Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Li Li
- Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiaofen Xiong
- Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Yachun Han
- Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Xuejing Zhu
- Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Lin Sun
- Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
10
|
Liu X, Zhang Y, Shi M, Wang Y, Zhang F, Yan R, Liu L, Xiao Y, Guo B. Notch1 regulates PTEN expression to exacerbate renal tubulointerstitial fibrosis in diabetic nephropathy by inhibiting autophagy via interactions with Hes1. Biochem Biophys Res Commun 2018; 497:1110-1116. [PMID: 29496446 DOI: 10.1016/j.bbrc.2018.02.187] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 02/25/2018] [Indexed: 02/07/2023]
Abstract
Diabetic nephropathy (DN) is a serious clinical microvascular complication of diabetes mellitus. DN is characterized by the accumulation of extracellular matrix, resulting in progressive fibrosis leading to the loss of renal function. Notch1 and phosphatase and tensin homolog deleted on chromosome ten (PTEN) signaling have been associated with fibrosis. Autophagy serves as an essential regulator of tubular cellular homeostasis. However, how these molecules control the balance between fibrosis and autophagy, the main homeostatic mechanism regulating fibrosis, is not well understood. This association was confirmed using Notch1-siRNA in vitro, which prevented the increase in Hes1 and restored PTEN expression. In contrast, transfection with pHAGE-Hes1 repressed PTEN promoter-driven luciferase activity, implying a direct relationship between Hes1 and PTEN. The expression of Notch1 and Hes1 was increased in diabetic db/db mice by western blotting; in contrast, the expression of PTEN was decreased. Importantly, the dysregulation of these signaling molecules was associated with an increase in extracellular matrix proteins (Collagen-I and III) and the inhibition of autophagy. Similar results were evident in response to high glucose concentrations in vitro in the NRK-52e cells. Therefore, the high glucose concentrations present in diabetes promote fibrosis through the Notch1 pathway via Hes1, while inhibiting the PTEN and autophagy. In conclusion, the inhibition of PTEN by Notch1/Hes1 in response to high glucose concentration inhibits autophagy, which is associated with the progression of fibrosis. Therefore, these signaling molecules may represent novel therapeutic targets in diabetic nephropathy.
Collapse
Affiliation(s)
- XingMei Liu
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, 550025, China; Laboratory of Pathogenesis Research, Drug Prevention and Treatment of Major Diseases, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - YingYing Zhang
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, 550025, China; Laboratory of Pathogenesis Research, Drug Prevention and Treatment of Major Diseases, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - MingJun Shi
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, 550025, China; Laboratory of Pathogenesis Research, Drug Prevention and Treatment of Major Diseases, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - YuanYuan Wang
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, 550025, China; Laboratory of Pathogenesis Research, Drug Prevention and Treatment of Major Diseases, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - Fan Zhang
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, 550025, China; Laboratory of Pathogenesis Research, Drug Prevention and Treatment of Major Diseases, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - Rui Yan
- Department of Nephrology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550002, China
| | - LingLing Liu
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, 550025, China; Laboratory of Pathogenesis Research, Drug Prevention and Treatment of Major Diseases, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - Ying Xiao
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, 550025, China; Laboratory of Pathogenesis Research, Drug Prevention and Treatment of Major Diseases, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - Bing Guo
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, 550025, China; Laboratory of Pathogenesis Research, Drug Prevention and Treatment of Major Diseases, Guizhou Medical University, Guiyang, Guizhou, 550025, China.
| |
Collapse
|
11
|
Inhibition of insulin resistance by PGE1 via autophagy-dependent FGF21 pathway in diabetic nephropathy. Sci Rep 2018; 8:9. [PMID: 29311680 PMCID: PMC5758726 DOI: 10.1038/s41598-017-18427-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 12/11/2017] [Indexed: 12/22/2022] Open
Abstract
Insulin resistance is a critical process in the initiation and progression of diabetic nephropathy (DN). Alprostadil (Prostaglandin E1, PGE1) had protective effects on renal function. However, it is unknown whether PGE1 inhibited insulin resistance in renal tubule epithelial cells via autophagy, which plays a protective role in DN against insulin resistance. Insulin resistance was induced by palmitic acid (PA) in human HK-2 cells, shown as the decrease of insulin-stimulated AKT phosphorylation, glucose transporter-4 (GLUT4), glucose uptake and enhanced phosphorylation of insulin receptor substrate 1(IRS-1) at site serine 307 (pIRS-1ser307) and downregulated expression of IRS-1. Along with less abundance of p62, autophagy markers LC3B and Beclin-1 significantly increased in HK-2 cells exposed to PA. Such abnormal changes were significantly reversed by PGE1, which mimicked the role of autophagy gene 7 small interfering RNA (ATG7 siRNA). Furthermore, PGE1 promoted the protein expression of autophagy-related fibroblast growth factor-21 (FGF21), which alleviated insulin resistance. Results from western blotting and immunohistochemistry indicated that PGE1 remarkably restored autophagy, insulin resistance and the FGF21 expression in rat kidney of type 2 diabetes mellitus (T2DM). Collectively, we demonstrated the potential protection of PGE1 on insulin resistance in renal tubules via autophagy-dependent FGF21 pathway in preventing the progression of DN.
Collapse
|
12
|
Han E, Shin E, Kim G, Lee JY, Lee YH, Lee BW, Kang ES, Cha BS. Combining SGLT2 Inhibition With a Thiazolidinedione Additively Attenuate the Very Early Phase of Diabetic Nephropathy Progression in Type 2 Diabetes Mellitus. Front Endocrinol (Lausanne) 2018; 9:412. [PMID: 30072956 PMCID: PMC6060671 DOI: 10.3389/fendo.2018.00412] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 07/02/2018] [Indexed: 12/19/2022] Open
Abstract
Although both sodium glucose co-transporter 2 inhibition by dapagliflozin and thiazolidinedione, pioglitazone have glucose-lowering and anti-inflammatory effects, the therapeutic efficacy of their combination on diabetic nephropathy has not been investigated. 9-week-old male db/db mice were randomly assigned to 4 groups and administrated with (1) vehicle, (2) dapagliflozin, (3) pioglitazone, or (4) dapagliflozin and pioglitazone combination. Human proximal tubule (HK-2) cells were treated with glucose or palmitate acid in the presence of medium, dapagliflozin, pioglitazone, or both. Glomerular tuft area and mesangial expansion of the kidney more reduced in the combination group compared to control and single therapy groups. Podocyte foot process width and glomerular basement membrane thickness decreased regardless of treatment, while the combination group showed the slowest renal hypertrophy progression (P < 0.05). The combination treatment decreased MCP-1, type I and IV collagen expression in the renal cortex. Only the combination treatment decreased the expression of angiotensinogen, IL-6, and TGF-β while it enhanced HK-2 cell survival (all P < 0.05). In conclusion, dapagliflozin and pioglitazone preserved renal function, and combination therapy showed the greatest benefit. These findings suggest that the combination therapy of dapagliflozin with pioglitazone is more effective than the single therapy for preventing the progression of nephropathy in patients with type 2 diabetes.
Collapse
Affiliation(s)
- Eugene Han
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Keimyung University School of Medicine, Daegu, South Korea
- Graduate School, Yonsei University College of Medicine, Seoul, South Korea
| | - Eugene Shin
- Institute of Endocrine Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Gyuri Kim
- Graduate School, Yonsei University College of Medicine, Seoul, South Korea
| | - Ji-Yeon Lee
- Graduate School, Yonsei University College of Medicine, Seoul, South Korea
- Institute of Endocrine Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Yong-ho Lee
- Graduate School, Yonsei University College of Medicine, Seoul, South Korea
- Institute of Endocrine Research, Yonsei University College of Medicine, Seoul, South Korea
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Byung-Wan Lee
- Graduate School, Yonsei University College of Medicine, Seoul, South Korea
- Institute of Endocrine Research, Yonsei University College of Medicine, Seoul, South Korea
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Eun Seok Kang
- Graduate School, Yonsei University College of Medicine, Seoul, South Korea
- Institute of Endocrine Research, Yonsei University College of Medicine, Seoul, South Korea
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Bong-Soo Cha
- Graduate School, Yonsei University College of Medicine, Seoul, South Korea
- Institute of Endocrine Research, Yonsei University College of Medicine, Seoul, South Korea
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
- *Correspondence: Bong-Soo Cha
| |
Collapse
|
13
|
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: 39] [Impact Index Per Article: 4.9] [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
|
14
|
Guo H, Wang B, Li H, Ling L, Niu J, Gu Y. Glucagon-like peptide-1 analog prevents obesity-related glomerulopathy by inhibiting excessive autophagy in podocytes. Am J Physiol Renal Physiol 2017; 314:F181-F189. [PMID: 29070572 DOI: 10.1152/ajprenal.00302.2017] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
To investigate the role of glucagon-like peptide-1 analog (GLP-1) in high-fat diet-induced obesity-related glomerulopathy (ORG). Male C57BL/6 mice fed a high-fat diet for 12 wk were treated with GLP-1 (200 μg/kg) or 0.9% saline for 4 wk. Fasting blood glucose and insulin and the expression of podocin, nephrin, phosphoinositide 3-kinase (PI3K), glucose transporter type (Glut4), and microtubule-associated protein 1A/1B-light chain 3 (LC3) were assayed. Glomerular morphology and podocyte foot structure were evaluated by periodic acid-Schiff staining and electron microscopy. Podocytes were treated with 150 nM GLP-1 and incubated with 400 μM palmitic acid (PA) for 12 h. The effect on autophagy was assessed by podocyte-specific Glut4 siRNA. Insulin resistance and autophagy were assayed by immunofluorescence and Western blotting. The high-fat diet resulted in weight gain, ectopic glomerular lipid accumulation, increased insulin resistance, and fusion of podophyte foot processes. The decreased translocation of Glut4 to the plasma membrane and excess autophagy seen in mice fed a high-fat diet and in PA-treated cultured podocytes were attenuated by GLP-1. Podocyte-specific Glut4 siRNA promoted autophagy, and rapamycin-enhanced autophagy worsened the podocyte injury caused by PA. Excess autophagy in podocytes was induced by inhibition of Glut4 translocation to the plasma membrane and was involved in the pathology of ORG. GLP-1 restored insulin sensitivity and ameliorated renal injury by decreasing the level of autophagy.
Collapse
Affiliation(s)
- Honglei Guo
- Division of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University , Shanghai , China.,Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University (Jiangsu Province Hospital), Nanjing, Jiangsu Province, China
| | - Bin Wang
- Department of Nephrology, Zhongda Hospital Southeast University, Nanjing, Jiangsu Province, China
| | - Hongmei Li
- Division of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University , Shanghai , China
| | - Lilu Ling
- Division of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University , Shanghai , China
| | - Jianying Niu
- Division of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University , Shanghai , China
| | - Yong Gu
- Division of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University , Shanghai , China.,Division of Nephrology, Huashan Hospital and Institute of Nephrology, Fudan University , Shanghai , China
| |
Collapse
|
15
|
Hou S, Zhang T, Li Y, Guo F, Jin X. Glycyrrhizic Acid Prevents Diabetic Nephropathy by Activating AMPK/SIRT1/PGC-1 α Signaling in db/db Mice. J Diabetes Res 2017; 2017:2865912. [PMID: 29238727 PMCID: PMC5697128 DOI: 10.1155/2017/2865912] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 09/01/2017] [Accepted: 09/19/2017] [Indexed: 12/14/2022] Open
Abstract
Diabetic nephropathy (DN) is a major cause of end-stage renal disease (ESRD). Glycyrrhizic acid (GA) is an effective inhibitor of reactive oxygen species (ROS) production. We investigated the role of GA in the progression of renal injury in DN. Albumin (Alb)/creatinine (crea) levels were significantly lower, and renal histopathology was attenuated in the diabetic db/db mice that were treated with GA (15 mg/kg via intraperitoneal injection) once per day for eight weeks. These changes were associated with significantly lower levels of α-smooth muscle actin (α-SMA) and transforming growth factor β1 (TGF-β1) expression. Additionally, diabetic db/db mice displayed more terminal deoxynucleotidyl transferase-mediated nick-end labeling- (TUNEL-) positive nuclei and diabetes-induced ROS production in the kidneys, and these effects were attenuated by the treatment with GA, which activated adenosine monophosphate-activated protein kinase (AMPK)/silent information regulator 1 (SIRT1)/peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) signaling in the kidneys. In summary, in diabetic db/db mice, the effect of GA on DN involved, in part, the inhibition of ROS and the activation of AMPK/SIRT1/PGC-1α signaling in the kidneys. GA, therefore, shows therapeutic potential for preventing and treating DN.
Collapse
Affiliation(s)
- Shaozhang Hou
- Department of Pathology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
- Institute of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Ting Zhang
- Department of Pathology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
- Institute of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Yuan Li
- Department of Nursing, Ningxia Medical University, Yinchuan 750004, China
| | - Fengying Guo
- Department of Pathology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
- Institute of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Xiu Jin
- Affiliated Hospital of Jining Medical College, Jining 272000, China
| |
Collapse
|
16
|
Musso G, Cassader M, Cohney S, De Michieli F, Pinach S, Saba F, Gambino R. Fatty Liver and Chronic Kidney Disease: Novel Mechanistic Insights and Therapeutic Opportunities. Diabetes Care 2016; 39:1830-45. [PMID: 27660122 DOI: 10.2337/dc15-1182] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 07/10/2016] [Indexed: 02/03/2023]
Abstract
Chronic kidney disease (CKD) is a risk factor for end-stage renal disease (ESRD) and cardiovascular disease (CVD). ESRD or CVD develop in a substantial proportion of patients with CKD receiving standard-of-care therapy, and mortality in CKD remains unchanged. These data suggest that key pathogenetic mechanisms underlying CKD progression go unaffected by current treatments. Growing evidence suggests that nonalcoholic fatty liver disease (NAFLD) and CKD share common pathogenetic mechanisms and potential therapeutic targets. Common nutritional conditions predisposing to both NAFLD and CKD include excessive fructose intake and vitamin D deficiency. Modulation of nuclear transcription factors regulating key pathways of lipid metabolism, inflammation, and fibrosis, including peroxisome proliferator-activated receptors and farnesoid X receptor, is advancing to stage III clinical development. The relevance of epigenetic regulation in the pathogenesis of NAFLD and CKD is also emerging, and modulation of microRNA21 is a promising therapeutic target. Although single antioxidant supplementation has yielded variable results, modulation of key effectors of redox regulation and molecular sensors of intracellular energy, nutrient, or oxygen status show promising preclinical results. Other emerging therapeutic approaches target key mediators of inflammation, such as chemokines; fibrogenesis, such as galectin-3; or gut dysfunction through gut microbiota manipulation and incretin-based therapies. Furthermore, NAFLD per se affects CKD through lipoprotein metabolism and hepatokine secretion, and conversely, targeting the renal tubule by sodium-glucose cotransporter 2 inhibitors can improve both CKD and NAFLD. Implications for the treatment of NAFLD and CKD are discussed in light of this new therapeutic armamentarium.
Collapse
Affiliation(s)
- Giovanni Musso
- Humanitas Gradenigo Hospital, University of Turin, Turin, Italy
| | - Maurizio Cassader
- Department of Medical Sciences, San Giovanni Battista Hospital, University of Turin, Turin, Italy
| | - Solomon Cohney
- Department of Nephrology, Western & Royal Melbourne Hospitals, Melbourne, VIC, Australia
| | - Franco De Michieli
- Department of Medical Sciences, San Giovanni Battista Hospital, University of Turin, Turin, Italy
| | - Silvia Pinach
- Department of Medical Sciences, San Giovanni Battista Hospital, University of Turin, Turin, Italy
| | - Francesca Saba
- Department of Medical Sciences, San Giovanni Battista Hospital, University of Turin, Turin, Italy
| | - Roberto Gambino
- Department of Medical Sciences, San Giovanni Battista Hospital, University of Turin, Turin, Italy
| |
Collapse
|
17
|
Wang S, Lu Y, Sun X, Wu D, Fu B, Chen Y, Deng H, Chen X. Identification of common and differential mechanisms of glomerulus and tubule senescence in 24-month-old rats by quantitative LC-MS/MS. Proteomics 2016; 16:2706-2717. [PMID: 27452873 DOI: 10.1002/pmic.201600121] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 07/05/2016] [Accepted: 07/20/2016] [Indexed: 12/20/2022]
Affiliation(s)
- Shiyu Wang
- Department of Nephrology; Chinese PLA General Hospital; Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases; Beijing P.R. China
- Department of Nephrology; The Second Hospital of Jilin University; Changchun Jilin P.R. China
| | - Yang Lu
- Department of Nephrology; Chinese PLA General Hospital; Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases; Beijing P.R. China
| | - Xuefeng Sun
- Department of Nephrology; Chinese PLA General Hospital; Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases; Beijing P.R. China
| | - Di Wu
- Department of Nephrology; Chinese PLA General Hospital; Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases; Beijing P.R. China
| | - Bo Fu
- Department of Nephrology; Chinese PLA General Hospital; Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases; Beijing P.R. China
| | - Yuling Chen
- MOE Key Laboratory of Bioinformatics; School of Life Sciences; Tsinghua University; Beijing P.R. China
| | - Haiteng Deng
- MOE Key Laboratory of Bioinformatics; School of Life Sciences; Tsinghua University; Beijing P.R. China
| | - Xiangmei Chen
- Department of Nephrology; Chinese PLA General Hospital; Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases; Beijing P.R. China
| |
Collapse
|
18
|
Xin W, Li Z, Xu Y, Yu Y, Zhou Q, Chen L, Wan Q. Autophagy protects human podocytes from high glucose-induced injury by preventing insulin resistance. Metabolism 2016; 65:1307-15. [PMID: 27506738 DOI: 10.1016/j.metabol.2016.05.015] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 05/05/2016] [Accepted: 05/24/2016] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Insulin resistance is correlated with the progress of albuminuria in diabetic patients, and podocytes are crucial for maintaining the normal function of the glomerular filtration barrier. In the present study, we aimed to investigate the high glucose-induced insulin resistance and cell injury in human podocytes and the putative role of autophagy in this process. METHODS Human podocytes were cultured in high glucose-supplemented medium and low glucose and high osmotic conditions were used for the controls. Autophagy in the podocytes was regulated using rapamycin or 3-methyladenine stimulation. Next, autophagy markers including LC3B, Beclin-1, and p62 were investigated using western blot and qPCR, and the insulin responsiveness was analyzed based on glucose uptake and by using the phosphorylation of the insulin receptor with Nephrin as a podocyte injury marker. RESULTS The basal autophagy level decreased under the high glucose conditions, which was accompanied by a decrease in the glucose uptake and phosphorylation of the insulin receptor in the human podocytes. More interestingly, the glucose uptake and the phosphorylation of the insulin receptor were decreased by 3-MA stimulation and increased by rapamycin, illustrating that the responsiveness of insulin was regulated by autophagy. The activation of autophagy by rapamycin also ameliorated cell injury in the human podocytes. CONCLUSIONS The presence or activation of autophagy was found to play a protective role in human podocytes against high glucose-induced insulin resistance and cell injury, which indicates a novel cellular mechanism and provides a potential therapeutic target for diabetic nephropathy (DN).
Collapse
Affiliation(s)
- Wei Xin
- Department of Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, China
| | - Zhaoping Li
- Institute of Nutrition and Food Hygiene, School of Public Health, Shandong University, Jinan, 250012, China
| | - Ying Xu
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, China
| | - Yue Yu
- Institute of Nutrition and Food Hygiene, School of Public Health, Shandong University, Jinan, 250012, China
| | - Qi Zhou
- School of Medicine, Shandong University, Jinan, 250012, China
| | - Liyong Chen
- Department of Nutrition, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, China.
| | - Qiang Wan
- Department of Nephrology, Qianfoshan Hospital Affiliated to Shandong University, Jinan, 250014, China.
| |
Collapse
|
19
|
Bader CA, Shandala T, Carter EA, Ivask A, Guinan T, Hickey SM, Werrett MV, Wright PJ, Simpson PV, Stagni S, Voelcker NH, Lay PA, Massi M, Plush SE, Brooks DA. A Molecular Probe for the Detection of Polar Lipids in Live Cells. PLoS One 2016; 11:e0161557. [PMID: 27551717 PMCID: PMC4994960 DOI: 10.1371/journal.pone.0161557] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 08/07/2016] [Indexed: 01/08/2023] Open
Abstract
Lipids have an important role in many aspects of cell biology, including membrane architecture/compartment formation, intracellular traffic, signalling, hormone regulation, inflammation, energy storage and metabolism. Lipid biology is therefore integrally involved in major human diseases, including metabolic disorders, neurodegenerative diseases, obesity, heart disease, immune disorders and cancers, which commonly display altered lipid transport and metabolism. However, the investigation of these important cellular processes has been limited by the availability of specific tools to visualise lipids in live cells. Here we describe the potential for ReZolve-L1™ to localise to intracellular compartments containing polar lipids, such as for example sphingomyelin and phosphatidylethanolamine. In live Drosophila fat body tissue from third instar larvae, ReZolve-L1™ interacted mainly with lipid droplets, including the core region of these organelles. The presence of polar lipids in the core of these lipid droplets was confirmed by Raman mapping and while this was consistent with the distribution of ReZolve-L1™ it did not exclude that the molecular probe might be detecting other lipid species. In response to complete starvation conditions, ReZolve-L1™ was detected mainly in Atg8-GFP autophagic compartments, and showed reduced staining in the lipid droplets of fat body cells. The induction of autophagy by Tor inhibition also increased ReZolve-L1™ detection in autophagic compartments, whereas Atg9 knock down impaired autophagosome formation and altered the distribution of ReZolve-L1™. Finally, during Drosophila metamorphosis fat body tissues showed increased ReZolve-L1™ staining in autophagic compartments at two hours post puparium formation, when compared to earlier developmental time points. We concluded that ReZolve-L1™ is a new live cell imaging tool, which can be used as an imaging reagent for the detection of polar lipids in different intracellular compartments.
Collapse
Affiliation(s)
- Christie A. Bader
- School of Pharmacy and Medical Science, University of South Australia, Adelaide, South Australia, Australia
| | - Tetyana Shandala
- School of Pharmacy and Medical Science, University of South Australia, Adelaide, South Australia, Australia
| | - Elizabeth A. Carter
- Vibrational Spectroscopy Core Facility, The University of Sydney, Sydney, New South Wales, Australia
| | - Angela Ivask
- Future Industries Institute, University of South Australia, Mawson Lakes, South Australia, Australia
| | - Taryn Guinan
- Future Industries Institute, University of South Australia, Mawson Lakes, South Australia, Australia
| | - Shane M. Hickey
- School of Pharmacy and Medical Science, University of South Australia, Adelaide, South Australia, Australia
| | - Melissa V. Werrett
- Department of Chemistry and Nanochemistry Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Phillip J. Wright
- Department of Chemistry and Nanochemistry Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Peter V. Simpson
- Department of Chemistry and Nanochemistry Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Stefano Stagni
- Department of Industrial Chemistry “Toso Montanari”, University of Bologna, Bologna, Italy
| | - Nicolas H. Voelcker
- Future Industries Institute, University of South Australia, Mawson Lakes, South Australia, Australia
| | - Peter A. Lay
- Vibrational Spectroscopy Core Facility, The University of Sydney, Sydney, New South Wales, Australia
| | - Massimiliano Massi
- Department of Chemistry and Nanochemistry Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Sally E. Plush
- School of Pharmacy and Medical Science, University of South Australia, Adelaide, South Australia, Australia
| | - Douglas A. Brooks
- School of Pharmacy and Medical Science, University of South Australia, Adelaide, South Australia, Australia
- * E-mail:
| |
Collapse
|
20
|
Abstract
Many common renal insults such as ischemia and toxic injury primarily target the tubular epithelial cells, especially the highly metabolically active proximal tubular segment. Tubular epithelial cells are particularly dependent on autophagy to maintain homeostasis and respond to stressors. The pattern of autophagy in the kidney has a unique spatial and chronologic signature. Recent evidence has shown that there is complex cross-talk between autophagy and various cell death pathways. This review specifically discusses the interplay between autophagy and cell death in the renal tubular epithelia. It is imperative to review this topic because recent discoveries have improved our mechanistic understanding of the autophagic process and have highlighted its broad clinical applications, making autophagy a major target for drug development.
Collapse
Affiliation(s)
- Andrea Havasi
- Department of Nephrology, Boston University Medical Center, Boston, MA.
| | - Zheng Dong
- Department of Nephrology, Second Xiangya Hospital of Central South University, Changsha, China; Department of Cellular Biology and Anatomy, Medical College of Georgia and Charlie Norwood VA Medical Center, Augusta, GA
| |
Collapse
|
21
|
Gatticchi L, Bellezza I, Del Sordo R, Peirce MJ, Sidoni A, Roberti R, Minelli A. The Tm7sf2 Gene Deficiency Protects Mice against Endotoxin-Induced Acute Kidney Injury. PLoS One 2015; 10:e0141885. [PMID: 26540160 PMCID: PMC4635018 DOI: 10.1371/journal.pone.0141885] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 10/14/2015] [Indexed: 12/18/2022] Open
Abstract
Cholesterol is essential for diverse cellular functions and cellular and whole-body cholesterol homeostasis is highly controlled. Cholesterol can also influence cellular susceptibility to injury. The connection between cholesterol metabolism and inflammation is exemplified by the Tm7sf2 gene, the absence of which reveals an essential role in cholesterol biosynthesis under stress conditions but also results in an inflammatory phenotype, i.e. NF-κB activation and TNFα up-regulation. Here, by using Tm7sf2+/+and Tm7sf2−/− mice, we investigated whether the Tm7sf2 gene, through its role in cholesterol biosynthesis under stress conditions, is involved in the renal failure induced by the administration of LPS. We found that the loss of Tm7sf2 gene results in significantly reduced blood urea nitrogen levels accompanied by decreased renal inflammatory response and neutral lipid accumulation. The increased expression of fatty acids catabolic enzymes reduces the need of the renal autophagy, a known crucial nutrient-sensing pathway in lipid metabolism. Moreover, we observed that the Tm7sf2 insufficiency is responsible for the inhibition of the NF-κB signalling thus dampening the inflammatory response and leading to a reduced renal damage. These results suggest a pivotal role for Tm7sf2 in renal inflammatory and lipotoxic response under endotoxemic conditions.
Collapse
Affiliation(s)
- Leonardo Gatticchi
- Department of Experimental Medicine, University of Perugia, Piazzale Gambuli, 06124 Perugia, Italy
| | - Ilaria Bellezza
- Department of Experimental Medicine, University of Perugia, Piazzale Gambuli, 06124 Perugia, Italy
| | - Rachele Del Sordo
- Department of Experimental Medicine, University of Perugia, Piazzale Gambuli, 06124 Perugia, Italy
| | - Matthew J. Peirce
- Department of Experimental Medicine, University of Perugia, Piazzale Gambuli, 06124 Perugia, Italy
| | - Angelo Sidoni
- Department of Experimental Medicine, University of Perugia, Piazzale Gambuli, 06124 Perugia, Italy
| | - Rita Roberti
- Department of Experimental Medicine, University of Perugia, Piazzale Gambuli, 06124 Perugia, Italy
| | - Alba Minelli
- Department of Experimental Medicine, University of Perugia, Piazzale Gambuli, 06124 Perugia, Italy
- * E-mail:
| |
Collapse
|