1
|
Wang X, Gu Z, Huang Y, Wang J, Tang S, Yang X, Wang J. MicroRNA-668 alleviates renal fibrosis through PPARα/PGC-1α pathway. Eur J Med Res 2024; 29:631. [PMID: 39732711 DOI: 10.1186/s40001-024-02248-x] [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: 10/15/2024] [Accepted: 12/21/2024] [Indexed: 12/30/2024] Open
Abstract
BACKGROUND The involvement of microRNA-668 (miR-668) in the onset and progression of renal fibrosis remains unclear. To this end, we aimed to explore the relevant mechanism of miR-668 in renal fibrosis. METHODS C57BL/6 J male mice were randomly divided into sham-operated, unilateral ureteral obstruction (UUO), and UUO-fenofibrate groups. Based on transfection and drug intervention, HK-2 cells were divided into blank control, TGF-β1, TGF-β1 + fenofibrate (PPARα agonist), mimics-NC, miR-668, mimics-NC + TGF-β1, miR-668 + TGF-β1, miR-668 + TGF-β1 + fenofibrate, miR-668 + TGF-β1 + GW6471 (PPARα inhibitor), mimics-NC + TGF-β1 + fenofibrate, and mimics-NC + TGF-β1 + GW6471 groups. The pathological changes in the renal tissues were observed by hematoxylin-eosin (HE) and Masson staining. The expression of PPARα, PGC-1α, miR-668, E-cadherin, Collagen III (Col III), and α-SMA in the renal tissues or HK-2 cells was detected by western blot, immunohistochemical analyses or real-time quantitative polymerase chain reaction. The regulatory effect of miR-668 on PPARα was verified by dual-luciferase reporter assay. RESULTS The expression of PPARα and PGC-1α decreased in UUO mice and TGF-β1-induced HK-2 cells, which was improved by fenofibrate. Compared to the non-transfected group, in TGF-β1-stimulated HK-2 cells, the expression of E-cadherin, PPARα and PGC-1α increased and the expression of Col III and α-SMA decreased in the miR-668-transfected group. The dual-luciferase reporter assay indicated the regulatory effect of hsa-mir-668-3p on PPARα. CONCLUSION MiR-668 can target PPARα and positively regulate the PPARα/PGC-1α pathway to alleviate renal fibrosis.
Collapse
Affiliation(s)
- Xinran Wang
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, China
- The Critical Kidney Disease Research Center of Central South University, Changsha, China
| | - Zhoupeng Gu
- The Public Hospital Management Office, Zhuzhou, China
| | - Yan Huang
- Department of Rheumatology and Immunology, The Xiangya Changde Hospital, Central South University, Changde, China
| | - Jingyan Wang
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Shiqi Tang
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, China
- The Critical Kidney Disease Research Center of Central South University, Changsha, China
| | - Xinyu Yang
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, China
- The Critical Kidney Disease Research Center of Central South University, Changsha, China
| | - Jianwen Wang
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, China.
- The Critical Kidney Disease Research Center of Central South University, Changsha, China.
| |
Collapse
|
2
|
Masenga SK, Desta S, Hatcher M, Kirabo A, Lee DL. How PPAR-alpha mediated inflammation may affect the pathophysiology of chronic kidney disease. Curr Res Physiol 2024; 8:100133. [PMID: 39665027 PMCID: PMC11629568 DOI: 10.1016/j.crphys.2024.100133] [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: 06/06/2024] [Revised: 10/03/2024] [Accepted: 11/12/2024] [Indexed: 12/13/2024] Open
Abstract
Chronic kidney disease (CKD) is a major risk factor for death in adults. Inflammation plays a role in the pathogenesis of CKD, but the mechanisms are poorly understood. Peroxisome proliferator-activated receptor alpha (PPAR-α) is a nuclear receptor and one of the three members (PPARα, PPARβ/δ, and PPARγ) of the PPARs that plays an important role in ameliorating pathological processes that accelerate acute and chronic kidney disease. Although other PPARs members are well studied, the role of PPAR-α is not well described and its role in inflammation-mediated chronic disease is not clear. Herein, we review the role of PPAR-α in chronic kidney disease with implications for the immune system.
Collapse
Affiliation(s)
- Sepiso K. Masenga
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Livingstone Campus, Zambia
- Vanderbilt Institute for Global Health, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Selam Desta
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Physiology and Biophysics, College of Medicine, Howard University, Washington, DC, USA
| | - Mark Hatcher
- Department of Physiology and Biophysics, College of Medicine, Howard University, Washington, DC, USA
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Global Health, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Dexter L. Lee
- Department of Physiology and Biophysics, College of Medicine, Howard University, Washington, DC, USA
| |
Collapse
|
3
|
Feng L, Lin Z, Tang Z, Zhu L, Xu S, Tan X, Wang X, Mai J, Tan Q. Emodin improves renal fibrosis in chronic kidney disease by regulating mitochondrial homeostasis through the mediation of peroxisome proliferator-activated receptor-gamma coactivator-1 alpha (PGC-1α). Eur J Histochem 2024; 68:3917. [PMID: 38742403 PMCID: PMC11128849 DOI: 10.4081/ejh.2024.3917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 03/27/2024] [Indexed: 05/16/2024] Open
Abstract
Chronic kidney disease (CKD) is a leading public health issue associated with high morbidity worldwide. However, there are only a few effective therapeutic strategies for CKD. Emodin, an anthraquinone compound from rhubarb, can inhibit fibrosis in tissues and cells. Our study aims to investigate the antifibrotic effect of emodin and the underlying molecular mechanism. A unilateral ureteral obstruction (UUO)-induced rat model was established to evaluate the effect of emodin on renal fibrosis development. Hematoxylin and eosin staining, Masson's trichrome staining, and immunohistochemistry staining were performed to analyze histopathological changes and fibrotic features after emodin treatment. Subsequently, a transforming growth factor-beta 1 (TGF-β1)-induced cell model was used to assess the inhibition of emodin on cell fibrosis in vitro. Furthermore, Western blot analysis and real-time quantitative reverse transcription-polymerase chain reaction were performed to validate the regulatory mechanism of emodin on renal fibrosis progression. As a result, emodin significantly improved histopathological abnormalities in rats with UUO. The expression of fibrosis biomarkers and mitochondrial biogenesis-related proteins also decreased after emodin treatment. Moreover, emodin blocked TGF-β1-induced fibrotic phenotype, lipid accumulation, and mitochondrial homeostasis in NRK-52E cells. Conversely, peroxisome proliferator-activated receptor-gamma coactivator-1 alpha (PGC-1α) silencing significantly reversed these features in emodin-treated cells. Collectively, emodin plays an important role in regulating PGC-1α-mediated mitochondria function and energy homeostasis. This indicates that emodin exhibits great inhibition against renal fibrosis and acts as a promising inhibitor of CKD.
Collapse
Affiliation(s)
- Liuchang Feng
- Department of Nephrology, Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen.
| | - Zaoqiang Lin
- Department of Nephrology, Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen.
| | - Zeyong Tang
- Department of Nephrology, Guangzhou University of Chinese Medicine, Guangzhou.
| | - Lin Zhu
- Department of Nephrology, Shenzhen Hospital; Department of Nephrology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Shenzhen.
| | - Shu Xu
- Department of Oncology, Shenzhen Hospital, University of Chinese Academy of Sciences, Shenzhen.
| | - Xi Tan
- Medicopsychology, Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen.
| | - Xinyuan Wang
- Medicopsychology, School of Life Sciences, Beijing University of Chinese Medicine, Beijing.
| | - Jianling Mai
- Department of Hemodialysis, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou.
| | - Qinxiang Tan
- Department of Nephrology, Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen.
| |
Collapse
|
4
|
Wrońska A, Kieżun J, Kmieć Z. High-Dose Fenofibrate Stimulates Multiple Cellular Stress Pathways in the Kidney of Old Rats. Int J Mol Sci 2024; 25:3038. [PMID: 38474282 DOI: 10.3390/ijms25053038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 03/14/2024] Open
Abstract
We investigated the age-related effects of the lipid-lowering drug fenofibrate on renal stress-associated effectors. Young and old rats were fed standard chow with 0.1% or 0.5% fenofibrate. The kidney cortex tissue structure showed typical aging-related changes. In old rats, 0.1% fenofibrate reduced the thickening of basement membranes, but 0.5% fenofibrate exacerbated interstitial fibrosis. The PCR array for stress and toxicity-related targets showed that 0.1% fenofibrate mildly downregulated, whereas 0.5% upregulated multiple genes. In young rats, 0.1% fenofibrate increased some antioxidant genes' expression and decreased the immunoreactivity of oxidative stress marker 4-HNE. However, the activation of cellular antioxidant defenses was impaired in old rats. Fenofibrate modulated the expression of factors involved in hypoxia and osmotic stress signaling similarly in both age groups. Inflammatory response genes were variably modulated in the young rats, whereas old animals presented elevated expression of proinflammatory genes and TNFα immunoreactivity after 0.5% fenofibrate. In old rats, 0.1% fenofibrate more prominently than in young animals induced phospho-AMPK and PGC1α levels, and upregulated fatty acid oxidation genes. Our results show divergent effects of fenofibrate in young and old rat kidneys. The activation of multiple stress-associated effectors by high-dose fenofibrate in the aged kidney warrants caution when applying fenofibrate therapy to the elderly.
Collapse
Affiliation(s)
- Agata Wrońska
- Department of Histology, Faculty of Medicine, Medical University of Gdańsk, 80-210 Gdańsk, Poland
| | - Jacek Kieżun
- Department of Human Histology and Embryology, School of Medicine, University of Warmia and Mazury in Olsztyn, 10-082 Olsztyn, Poland
| | - Zbigniew Kmieć
- Department of Histology, Faculty of Medicine, Medical University of Gdańsk, 80-210 Gdańsk, Poland
| |
Collapse
|
5
|
Lee KM, Hwang YJ, Jung GS. Alantolactone Attenuates Renal Fibrosis via Inhibition of Transforming Growth Factor β/Smad3 Signaling Pathway. Diabetes Metab J 2024; 48:72-82. [PMID: 38173367 PMCID: PMC10850280 DOI: 10.4093/dmj.2022.0231] [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: 07/08/2022] [Accepted: 01/25/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGRUOUND Renal fibrosis is characterized by the accumulation of extracellular matrix proteins and interstitial fibrosis. Alantolactone is known to exert anticancer, anti-inflammatory, antimicrobial and antifungal effects; however, its effects on renal fibrosis remains unknown. Here, we investigated whether alantolactone attenuates renal fibrosis in mice unilateral ureteral obstruction (UUO) and evaluated the effect of alantolactone on transforming growth factor (TGF) signaling pathway in renal cells. METHODS To evaluate the therapeutic effect of alantolactone, cell counting kit-8 (CCK-8) assay, histological staining, Western blot analysis, and real-time quantitative polymerase chain reaction were performed in UUO kidneys in vivo and in TGF-β-treated renal cells in vitro. RESULTS Alantolactone (0.25 to 4 µM) did not affect the viability of renal cells. Mice orally administered 5 mg/kg of alantolactone daily for 15 days did not show mortality or liver toxicity. Alantolactone decreased UUO-induced blood urea nitrogen and serum creatinine levels. In addition, it significantly alleviated renal tubulointerstitial damage and fibrosis and decreased collagen type I, fibronectin, and α-smooth muscle actin (α-SMA) expression in UUO kidneys. In NRK-49F cells, alantolactone inhibited TGF-βstimulated expression of fibronectin, collagen type I, plasminogen activator inhibitor-1 (PAI-1), and α-SMA. In HK-2 cells, alantolactone inhibited TGF-β-stimulated expression of collagen type I and PAI-1. Alantolactone inhibited UUO-induced phosphorylation of Smad3 in UUO kidneys. In addition, it not only decreased TGF-β secretion but also Smad3 phosphorylation and translocation to nucleus in both kidney cell lines. CONCLUSION Alantolactone improves renal fibrosis by inhibiting the TGF-β/Smad3 signaling pathway in obstructive nephropathy. Thus, alantolactone is a potential therapeutic agent for chronic kidney disease.
Collapse
Affiliation(s)
- Kyeong-Min Lee
- Division of Biotechnology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea
| | - Yeo Jin Hwang
- Division of Biotechnology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea
| | - Gwon-Soo Jung
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Korea
| |
Collapse
|
6
|
Alhirmizi IAO, Uysal F, Arslan SO, Özünlü SAÇ, Koç A, Parlar A, Bayram KK. Fenofibrate Attenuates Asthma Features in an Ovalbumin-induced Mouse Model Via Suppressing NF-κB Binding Activity. Respir Physiol Neurobiol 2023:104083. [PMID: 37295485 DOI: 10.1016/j.resp.2023.104083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/07/2023] [Accepted: 05/20/2023] [Indexed: 06/12/2023]
Abstract
BACKGROUND/AIM Asthma is a chronic inflammatory disease of the airways with a high prevalence. Asthma has a complex pathophysiology and about 5-10% of patients are not fully responsive to the currently available treatments. The aim of this study is to investigate the involvement of NF-κB in the effects of fenofibrate on a mouse model of allergic asthma. MATERIALS AND METHODS A total of 49 BALB/c mice were randomly distributed into 7 groups (n=7). Allergic asthma model was created by administering i.p. injections of ovalbumin on days 0, 14 and 21, followed by provocation with inhaled ovalbumin on days 28, 29 and 30. Fenofibrate was orally given in 3 different doses; 1, 10 and 30mg/kg through days 21 to 30 of the experiment. On day 31, pulmonary function test using whole body plethysmography was performed. The mice were sacrificed 24hours later. Blood samples were obtained, and serum of each sample was separated for IgE determination. Bronchoalveolar lavage fluid (BALF) and lung tissues were collected to measure IL-5 and IL-13 levels. Nuclear extracts of lung tissues were employed to assess nuclear factor kappa B (NF-κB) p65 binding activity. RESULTS Enhanced Pause (Penh) values were significantly increased in ovalbumin-sensitized and challenged mice (p<0.01). Administration of fenofibrate (10 and 30mg/kg) resulted in improved pulmonary function as shown by significantly lower Penh values (p<0.01). Interleukin (IL) -5 and IL-13 levels in BALF and lung tissues and immunoglobulin E (IgE) levels in serum were significantly elevated in the allergic mice. IL-5 levels in the lung tissues of mice treated with 1mg/kg fenofibrate (FEN1) group were significantly reduced (p<0.01). BALF and lung tissue IL-5 and IL-13 levels in mice treated with 10 and 30mg/kg fenofibrate, FEN10 and FEN30, respectively, were significantly diminished when compared to the ovalbumin-treated (OVA) group, whereas treatment with 1mg/kg fenofibrate resulted in insignificant changes. IgE levels in the serum of FEN30 group mice have shown a prominent reduction (p<0.01). NF-κB p65 binding activity was higher in mice sensitized and challenged with ovalbumin (p<0.01). NF-κB p65 binding activity was significantly reduced in allergic mice treated with 30mg/kg (p<0.01) fenofibrate. CONCLUSIONS In this study, we showed that administration of 10 and 30mg/kg fenofibrate effectively attenuated airway hyperresponsiveness and inflammation in a mouse model of allergic asthma, possibly through inhibition of NF-κB binding activity.
Collapse
Affiliation(s)
| | - Fatma Uysal
- Medical Pharmacology Department, Faculty of Medicine, Ankara Yıldırım Beyazıt University, Ankara, Türkiye
| | - Seyfullah Oktay Arslan
- Medical Pharmacology Department, Faculty of Medicine, Ankara Yıldırım Beyazıt University, Ankara, Türkiye.
| | - Saliha Ayşenur Çam Özünlü
- Medical Pharmacology, Ankara, Türkiye Department, Faculty of Medicine, Ankara Yıldırım Beyazıt University, Ankara, Türkiye
| | - Ayşegül Koç
- Ankara Yıldırım Beyazıt University, Ankara, Türkiye
| | - Ali Parlar
- Medical Pharmacology Department, Faculty of Medicine, University of Adıyaman, Adıyaman, Türkiye
| | - Keziban Korkmaz Bayram
- Medical Genetic Department, Faculty of Medicine, Ankara Yıldırım Beyazıt University, Ankara, Türkiye
| |
Collapse
|
7
|
Paw M, Wnuk D, Madeja Z, Michalik M. PPARδ Agonist GW501516 Suppresses the TGF-β-Induced Profibrotic Response of Human Bronchial Fibroblasts from Asthmatic Patients. Int J Mol Sci 2023; 24:ijms24097721. [PMID: 37175437 PMCID: PMC10178673 DOI: 10.3390/ijms24097721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/30/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
The airway wall remodeling observed in asthma is associated with subepithelial fibrosis and enhanced activation of human bronchial fibroblasts (HBFs) in the fibroblast to myofibroblast transition (FMT), induced mainly by transforming growth factor-β (TGF-β). The relationships between asthma severity, obesity, and hyperlipidemia suggest the involvement of peroxisome proliferator-activated receptors (PPARs) in the remodeling of asthmatic bronchi. In this study, we investigated the effect of PPARδ ligands (GW501516 as an agonist, and GSK0660 as an antagonist) on the FMT potential of HBFs derived from asthmatic patients cultured in vitro. This report shows, for the first time, the inhibitory effect of a PPARδ agonist on the number of myofibroblasts and the expression of myofibroblast-related markers-α-smooth muscle actin, collagen 1, tenascin C, and connexin 43-in asthma-related TGF-β-treated HBF populations. We suggest that actin cytoskeleton reorganization and Smad2 transcriptional activity altered by GW501516 lead to the attenuation of the FMT in HBF populations derived from asthmatics. In conclusion, our data demonstrate that a PPARδ agonist stimulates antifibrotic effects in an in vitro model of bronchial subepithelial fibrosis. This suggests its potential role in the development of a possible novel therapeutic approach for the treatment of subepithelial fibrosis during asthma.
Collapse
Affiliation(s)
- Milena Paw
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland
| | - Dawid Wnuk
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland
| | - Zbigniew Madeja
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland
| | - Marta Michalik
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland
| |
Collapse
|
8
|
Li X, Sun W, Lu J, He Y, Chen Y, Ren W, Cui L, Liu Z, Wang C, Wang X, Ma L, Cheng X, Han L, Li H, Zhang H, Yuan X, Ji X, Ji A, Merriman TR, Li C. Effects of fenofibrate therapy on renal function in primary gout patients. Rheumatology (Oxford) 2021; 60:5020-5027. [PMID: 33704429 PMCID: PMC8566261 DOI: 10.1093/rheumatology/keab231] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 02/23/2021] [Indexed: 12/27/2022] Open
Abstract
Objective To investigate the incidence and potential risk factors for development of fenofibrate-associated nephrotoxicity in gout patients. Methods A total of 983 gout patients on fenofibrate treatment who visited the dedicated Gout Clinic at the Affiliated Hospital of Qingdao University between September 2016 and June 2020 were retrospectively enrolled from the electronic records system. Fenofibrate-associated nephrotoxicity was defined as an increase in serum creatinine (SCr) ≥0.3 mg/dl within 6 months of fenofibrate initiation. The change trend of SCr and uric acid levels during the treatment period were assessed by a generalised additive mixed model (GAMM). Multivariate analysis was performed for risk factors affecting elevated SCr. Results A total of 100 (10.2%) patients experienced an increase in SCr ≥0.3 mg/dl within 6 months after fenofibrate initiation. The median change of SCr in the whole cohort was 0.11 mg/dl [interquartile range (IQR) 0.03–0.20], whereas it was 0.36 (0.33–0.45) in the fenofibrate-associated nephrotoxicity group. In a multivariable regression model, chronic kidney disease (CKD) [odds ratio (OR) 2.39 (95% CI 1.48, 3.86)] and tophus [OR 2.29 (95% CI 1.39, 3.78)] were identified to be risk predictors, independent of measured covariates, of fenofibrate-associated nephrotoxicity. During the treatment period, although SCr temporarily increased, serum urate and triglyceride concentrations decreased using the interaction analysis of GAMM. Of those with fenofibrate withdrawal records, the SCr increase in 65% of patients was reversed after an average of 49 days off the drug. Conclusions This observational study implied that fenofibrate-associated nephrotoxicity occurs frequently in gout patients, especially in patients with tophi or CKD. The potential renal risks of fenofibrate usage in gout needs additional research.
Collapse
Affiliation(s)
- Xinde Li
- Department of Endocrinology and Metabolism, Shandong Provincial Key Laboratory of Metabolic Diseases and Qingdao Key Laboratory of Gout, the Affiliated Hospital of Qingdao University
| | - Wenyan Sun
- Department of Endocrinology and Metabolism, Shandong Provincial Key Laboratory of Metabolic Diseases and Qingdao Key Laboratory of Gout, the Affiliated Hospital of Qingdao University.,Institute of Metabolic Diseases, Qingdao University, Qingdao, China
| | - Jie Lu
- Department of Endocrinology and Metabolism, Shandong Provincial Key Laboratory of Metabolic Diseases and Qingdao Key Laboratory of Gout, the Affiliated Hospital of Qingdao University.,Institute of Metabolic Diseases, Qingdao University, Qingdao, China
| | - Yuwei He
- Department of Endocrinology and Metabolism, Shandong Provincial Key Laboratory of Metabolic Diseases and Qingdao Key Laboratory of Gout, the Affiliated Hospital of Qingdao University
| | - Ying Chen
- Department of Endocrinology and Metabolism, Shandong Provincial Key Laboratory of Metabolic Diseases and Qingdao Key Laboratory of Gout, the Affiliated Hospital of Qingdao University
| | - Wei Ren
- Department of Endocrinology and Metabolism, Shandong Provincial Key Laboratory of Metabolic Diseases and Qingdao Key Laboratory of Gout, the Affiliated Hospital of Qingdao University
| | - Lingling Cui
- Department of Endocrinology and Metabolism, Shandong Provincial Key Laboratory of Metabolic Diseases and Qingdao Key Laboratory of Gout, the Affiliated Hospital of Qingdao University
| | - Zhen Liu
- Department of Endocrinology and Metabolism, Shandong Provincial Key Laboratory of Metabolic Diseases and Qingdao Key Laboratory of Gout, the Affiliated Hospital of Qingdao University
| | - Can Wang
- Department of Endocrinology and Metabolism, Shandong Provincial Key Laboratory of Metabolic Diseases and Qingdao Key Laboratory of Gout, the Affiliated Hospital of Qingdao University
| | - Xuefeng Wang
- Department of Endocrinology and Metabolism, Shandong Provincial Key Laboratory of Metabolic Diseases and Qingdao Key Laboratory of Gout, the Affiliated Hospital of Qingdao University
| | - Lidan Ma
- Department of Endocrinology and Metabolism, Shandong Provincial Key Laboratory of Metabolic Diseases and Qingdao Key Laboratory of Gout, the Affiliated Hospital of Qingdao University
| | - Xiaoyu Cheng
- Department of Endocrinology and Metabolism, Shandong Provincial Key Laboratory of Metabolic Diseases and Qingdao Key Laboratory of Gout, the Affiliated Hospital of Qingdao University
| | - Lin Han
- Department of Endocrinology and Metabolism, Shandong Provincial Key Laboratory of Metabolic Diseases and Qingdao Key Laboratory of Gout, the Affiliated Hospital of Qingdao University
| | - Hailong Li
- Department of Endocrinology and Metabolism, Shandong Provincial Key Laboratory of Metabolic Diseases and Qingdao Key Laboratory of Gout, the Affiliated Hospital of Qingdao University.,Institute of Metabolic Diseases, Qingdao University, Qingdao, China
| | - Hui Zhang
- Department of Endocrinology and Metabolism, Shandong Provincial Key Laboratory of Metabolic Diseases and Qingdao Key Laboratory of Gout, the Affiliated Hospital of Qingdao University.,Institute of Metabolic Diseases, Qingdao University, Qingdao, China
| | - Xuan Yuan
- Department of Endocrinology and Metabolism, Shandong Provincial Key Laboratory of Metabolic Diseases and Qingdao Key Laboratory of Gout, the Affiliated Hospital of Qingdao University.,Institute of Metabolic Diseases, Qingdao University, Qingdao, China
| | - Xiaopeng Ji
- Department of Endocrinology and Metabolism, Shandong Provincial Key Laboratory of Metabolic Diseases and Qingdao Key Laboratory of Gout, the Affiliated Hospital of Qingdao University
| | - Aichang Ji
- Department of Endocrinology and Metabolism, Shandong Provincial Key Laboratory of Metabolic Diseases and Qingdao Key Laboratory of Gout, the Affiliated Hospital of Qingdao University
| | - Tony R Merriman
- Institute of Metabolic Diseases, Qingdao University, Qingdao, China.,Division of Clinical Immunology and Rheumatology, University of Alabama Birmingham, Birmingham, Alabama, USA.,Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Changgui Li
- Department of Endocrinology and Metabolism, Shandong Provincial Key Laboratory of Metabolic Diseases and Qingdao Key Laboratory of Gout, the Affiliated Hospital of Qingdao University.,Institute of Metabolic Diseases, Qingdao University, Qingdao, China
| |
Collapse
|
9
|
Hwang S, Chung KW. Targeting fatty acid metabolism for fibrotic disorders. Arch Pharm Res 2021; 44:839-856. [PMID: 34664210 DOI: 10.1007/s12272-021-01352-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/11/2021] [Indexed: 02/06/2023]
Abstract
Fibrosis is defined by abnormal accumulation of extracellular matrix, which can affect virtually every organ system under diseased conditions. Fibrotic tissue remodeling often leads to organ dysfunction and is highly associated with increased morbidity and mortality. The disease burden caused by fibrosis is substantial, and the medical need for effective antifibrotic therapies is essential. Significant progress has been made in understanding the molecular mechanism and pathobiology of fibrosis, such as transforming growth factor-β (TGF-β)-mediated signaling pathways. However, owing to the complex and dynamic properties of fibrotic disorders, there are currently no therapeutic options that can prevent or reverse fibrosis. Recent studies have revealed that alterations in fatty acid metabolic processes are common mechanisms and core pathways that play a central role in different fibrotic disorders. Excessive lipid accumulation or defective fatty acid oxidation is associated with increased lipotoxicity, which directly contributes to the development of fibrosis. Genetic alterations or pharmacologic targeting of fatty acid metabolic processes have great potential for the inhibition of fibrosis development. Furthermore, mechanistic studies have revealed active interactions between altered metabolic processes and fibrosis development. Several well-known fibrotic factors change the lipid metabolic processes, while altered metabolic processes actively participate in fibrosis development. This review summarizes the recent evidence linking fatty acid metabolism and fibrosis, and provides new insights into the pathogenesis of fibrotic diseases for the development of drugs for fibrosis prevention and treatment.
Collapse
Affiliation(s)
- Seonghwan Hwang
- College of Pharmacy, Pusan National University, Busan, 46214, Republic of Korea
| | - Ki Wung Chung
- College of Pharmacy, Pusan National University, Busan, 46214, Republic of Korea.
| |
Collapse
|
10
|
Li Z, Lu S, Li X. The role of metabolic reprogramming in tubular epithelial cells during the progression of acute kidney injury. Cell Mol Life Sci 2021; 78:5731-5741. [PMID: 34185125 PMCID: PMC11073237 DOI: 10.1007/s00018-021-03892-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/01/2021] [Accepted: 06/25/2021] [Indexed: 12/18/2022]
Abstract
Acute kidney injury (AKI) is one of the most common clinical syndromes. AKI is associated with significant morbidity and subsequent chronic kidney disease (CKD) development. Thus, it is urgent to develop a strategy to hinder AKI progression. Renal tubules are responsible for the reabsorption and secretion of various solutes and the damage to this part of the nephron is a key mediator of AKI. As we know, many common renal insults primarily target the highly metabolically active proximal tubular cells (PTCs). PTCs are the most energy-demanding cells in the kidney. The ATP that they use is mostly produced in their mitochondria by fatty acid β-oxidation (FAO). But, when PTCs face various biological stresses, FAO will shut down for a time that outlives injury. Recent studies have suggested that surviving PTCs can adapt to FAO disruption by increasing glycolysis when facing metabolic constraints, although PTCs do not perform glycolysis in a normal physiological state. Enhanced glycolysis in a short period compensates for impaired energy production and exerts partial renal-protective effects, but its long-term effect on renal function and AKI progression is not promising. Deranged FAO and enhanced glycolysis may contribute to the AKI to CKD transition through different molecular biological mechanisms. In this review, we concentrate on the recent pathological findings of AKI with regards to the metabolic reprogramming in PTCs, confirming that targeting metabolic reprogramming represents a potentially effective therapeutic strategy for the progression of AKI.
Collapse
Affiliation(s)
- Zhenzhen Li
- Medicial Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Shan Lu
- Emergency Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xiaobing Li
- College of Basic Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, China
| |
Collapse
|
11
|
Munro MJL, Hulsebosch SE, Marks SL, Gilor C. Efficacy of a micronized, nanocrystal fenofibrate formulation in treatment of hyperlipidemia in dogs. J Vet Intern Med 2021; 35:1733-1742. [PMID: 34096101 PMCID: PMC8295657 DOI: 10.1111/jvim.16190] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 05/13/2021] [Accepted: 05/18/2021] [Indexed: 12/15/2022] Open
Abstract
Background Safe, effective, and readily available drug therapies are required for the management of hyperlipidemia and its associated complications in dogs. Objectives To investigate the efficacy of a micronized, nanocrystal formulation of fenofibrate (Tricor) in the treatment of hyperlipidemia in dogs. Animals Ten client‐owned dogs with primary (n = 7) and secondary (n = 3) hyperlipidemia. All dogs had hypertriglyceridemia at baseline; 3 dogs also had hypercholesterolemia. Methods Prospective dose‐escalation study. Dogs were treated with fenofibrate orally once daily in up to 3 cycles of 21 days each. Fenofibrate dose was increased at the end of each cycle if hypertriglyceridemia persisted and adverse effects were not documented. Complete blood count, biochemistry, and urine protein:creatinine ratio were collected serially. Baseline (T0) parameters were compared to time of maximal reduction in serum triglyceride concentrations (T1) and reported as median (range). Results Triglycerides normalized in all dogs (T0 = 662 mg/dL [189‐2391]; T1 = 113 mg/dL [81‐132]; P = .002). Fenofibrate dose at T1 = 6.4 mg/kg PO q24h (range, 2.2‐13.5). T1 was achieved at 3 (n = 4), 6 (n = 4), and 9 (n = 2) weeks. Serum cholesterol concentrations decreased in 9 of 10 dogs. Quiet demeanor and firm stools in 1 dog were the only reported adverse reactions. Fenofibrate administration resulted in a significant reduction in median alkaline phosphatase activity (P = .049). Conclusions and Clinical Importance Over 21 to 63 days, TriCor was effective in the management of primary and secondary hyperlipidemia in dogs.
Collapse
Affiliation(s)
- Matthew J L Munro
- Veterinary Medical Teaching Hospital, School of Veterinary Medicine, University of California-Davis, 1 Garrod Drive, Davis, California 95616, USA.,Department of Veterinary Clinical Sciences, The Melbourne Veterinary School, University of Melbourne, 250 Princes Highway, Werribee, Victoria 3030, Australia
| | - Sean E Hulsebosch
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California-Davis, 1 Shields Ave., Davis, California 95616, USA
| | - Stanley L Marks
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California-Davis, 1 Shields Ave., Davis, California 95616, USA
| | - Chen Gilor
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California-Davis, 1 Shields Ave., Davis, California 95616, USA.,Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, 2560 SE 16th Ave., Gainesville, Florida 32610, USA
| |
Collapse
|
12
|
Tuleta I, Frangogiannis NG. Diabetic fibrosis. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166044. [PMID: 33378699 PMCID: PMC7867637 DOI: 10.1016/j.bbadis.2020.166044] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 11/25/2020] [Accepted: 12/07/2020] [Indexed: 12/13/2022]
Abstract
Diabetes-associated morbidity and mortality is predominantly due to complications of the disease that may cause debilitating conditions, such as heart and renal failure, hepatic insufficiency, retinopathy or peripheral neuropathy. Fibrosis, the excessive and inappropriate deposition of extracellular matrix in various tissues, is commonly found in patients with advanced type 1 or type 2 diabetes, and may contribute to organ dysfunction. Hyperglycemia, lipotoxic injury and insulin resistance activate a fibrotic response, not only through direct stimulation of matrix synthesis by fibroblasts, but also by promoting a fibrogenic phenotype in immune and vascular cells, and possibly also by triggering epithelial and endothelial cell conversion to a fibroblast-like phenotype. High glucose stimulates several fibrogenic pathways, triggering reactive oxygen species generation, stimulating neurohumoral responses, activating growth factor cascades (such as TGF-β/Smad3 and PDGFs), inducing pro-inflammatory cytokines and chemokines, generating advanced glycation end-products (AGEs) and stimulating the AGE-RAGE axis, and upregulating fibrogenic matricellular proteins. Although diabetes-activated fibrogenic signaling has common characteristics in various tissues, some organs, such as the heart, kidney and liver develop more pronounced and clinically significant fibrosis. This review manuscript summarizes current knowledge on the cellular and molecular pathways involved in diabetic fibrosis, discussing the fundamental links between metabolic perturbations and fibrogenic activation, the basis for organ-specific differences, and the promises and challenges of anti-fibrotic therapies for diabetic patients.
Collapse
Affiliation(s)
- Izabela Tuleta
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY, USA
| | - Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
13
|
Sembach FE, Østergaard MV, Vrang N, Feldt-Rasmussen B, Fosgerau K, Jelsing J, Fink LN. Rodent models of diabetic kidney disease: human translatability and preclinical validity. Drug Discov Today 2021; 26:200-217. [DOI: 10.1016/j.drudis.2020.05.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 04/27/2020] [Accepted: 05/05/2020] [Indexed: 02/06/2023]
|
14
|
Libby AE, Jones B, Lopez-Santiago I, Rowland E, Levi M. Nuclear receptors in the kidney during health and disease. Mol Aspects Med 2020; 78:100935. [PMID: 33272705 DOI: 10.1016/j.mam.2020.100935] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/24/2020] [Accepted: 11/16/2020] [Indexed: 02/06/2023]
Abstract
Over the last 30 years, nuclear receptors (NRs) have been increasingly recognized as key modulators of systemic homeostasis and as contributing factors in many diseases. In the kidney, NRs play numerous important roles in maintaining homeostasis-many of which continue to be unraveled. As "master regulators", these important transcription factors integrate and coordinate many renal processes such as circadian responses, lipid metabolism, fatty acid oxidation, glucose handling, and inflammatory responses. The use of recently-developed genetic tools and small molecule modulators have allowed for detailed studies of how renal NRs contribute to kidney homeostasis. Importantly, while NRs are intimately involved in proper kidney function, they are also implicated in a variety of renal diseases such as diabetes, acute kidney injury, and other conditions such as aging. In the last 10 years, our understanding of renal disease etiology and progression has been greatly shaped by knowledge regarding how NRs are dysregulated in these conditions. Importantly, NRs have also become attractive therapeutic targets for attenuation of renal diseases, and their modulation for this purpose has been the subject of intense investigation. Here, we review the role in health and disease of six key renal NRs including the peroxisome proliferator-activated receptors (PPAR), estrogen-related receptors (ERR), the farnesoid X receptors (FXR), estrogen receptors (ER), liver X receptors (LXR), and vitamin D receptors (VDR) with an emphasis on recent findings over the last decade. These NRs have generated a wealth of data over the last 10 years that demonstrate their crucial role in maintaining normal renal homeostasis as well as their capacity to modulate disease progression.
Collapse
Affiliation(s)
- Andrew E Libby
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, 3900 Reservoir Rd, Washington, DC, 20007, USA.
| | - Bryce Jones
- Department of Pharmacology and Physiology, Georgetown University, 3900 Reservoir Rd, Washington, DC, 20007, USA.
| | - Isabel Lopez-Santiago
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, 3900 Reservoir Rd, Washington, DC, 20007, USA.
| | - Emma Rowland
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, 3900 Reservoir Rd, Washington, DC, 20007, USA.
| | - Moshe Levi
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, 3900 Reservoir Rd, Washington, DC, 20007, USA.
| |
Collapse
|
15
|
Kaur J, Kaur T, Sharma AK, Kaur J, Yadav HN, Pathak D, Singh AP. Fenofibrate attenuates ischemia reperfusion-induced acute kidney injury and associated liver dysfunction in rats. Drug Dev Res 2020; 82:412-421. [PMID: 33226649 DOI: 10.1002/ddr.21764] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 11/10/2020] [Accepted: 11/10/2020] [Indexed: 01/09/2023]
Abstract
Ischemia/reperfusion (I/R) is one of the common reasons for acute kidney injury (AKI) and we need to develop effective therapies for treating AKI. We investigated the role of fenofibrate against I/R-induced AKI and associated hepatic dysfunction in rats. In male wistar albino rats, renal pedicle occlusion for 40 min and 24 h reperfusion resulted in AKI. I/R-induced AKI was demonstrated by measuring serum creatinine, creatinine clearance, urea, uric acid, potassium, fractional excretion of sodium and urinary microproteins. Oxidative stress in rat kidneys was quantified by assaying superoxide anion generation, thiobarbituric acid reactive substances, and reduced glutathione levels. AKI-induced hepatic damage was quantified by assaying serum aminotransferases, alkaline phosphatase and bilirubin levels. Moreover, serum cholesterol, high density lipoprotein and triglycerides were quantified. Hematoxylin-eosin staining of renal and hepatic tissues was done and the kidney and liver injury scores were determined. Immunohistology of endothelial nitric oxide synthase (eNOS) was done in rat kidneys. Fenofibrate was administered for 1 week before subjecting rats to AKI. In separate group, the nitric oxide synthase inhibitor, L-nitroarginine methyl ester (L-NAME) was administered prior to fenofibrate treatment. In I/R group, significant alteration in the serum/urine parameters indicated AKI and hepatic dysfunction along with marked increase in kidney and liver injury scores. Treatment with fenofibrate attenuated AKI and associated hepatic dysfunction. Moreover, I/R-induced decrease in renal eNOS expression was abrogated by fenofibrate. Pre-treatment with L-NAME abolished fenofibrate mediated reno- and hepato-protective effects. In conclusion, fenofibrate attenuates I/R-induced AKI and associated hepatic dysfunction putatively through modulation of eNOS expression.
Collapse
Affiliation(s)
- Jashanpreet Kaur
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, India
| | - Tajpreet Kaur
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, India.,Department of Pharmacology, Khalsa College of Pharmacy, Amritsar (INDIA), India
| | - Ashwani Kumar Sharma
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, India
| | - Japneet Kaur
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, India
| | - Harlokesh Narayan Yadav
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India.,Department of Veterinary Anatomy, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, India
| | | | - Amrit Pal Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, India
| |
Collapse
|
16
|
Yang S, Ma C, Wu H, Zhang H, Yuan F, Yang G, Yang Q, Jia L, Liang Z, Kang L. Tectorigenin attenuates diabetic nephropathy by improving vascular endothelium dysfunction through activating AdipoR1/2 pathway. Pharmacol Res 2020; 153:104678. [PMID: 32014572 DOI: 10.1016/j.phrs.2020.104678] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 01/17/2020] [Accepted: 01/30/2020] [Indexed: 01/10/2023]
Abstract
Diabetic nephropathy (DN), a kind of microvascular complication, is a primary cause of end-stage renal disease worldwide. However, therapeutic drugs for DN treatment are still in lack. The glomerular endothelium is essential to maintain selective permeability of glomerular filtration barrier and glomerular vasculature function. Growing evidences show that endothelial dysfunction or injury is the initial stage of vascular damage in DN, which can be induced by hyperglycemia, lipotoxicity, and inflammation. Therefore, to improve the function of vascular endothelium in kidney is a key point for treatment of DN. As a plant isoflavone, tectorigenin (TEC) has attracted considerable attention due to its anti-proliferative and anti-inflammatory functions. However, whether TEC could inhibit the DN development remains unknown. In this study, we examined the effects of TEC on DN development in db/db mice, a type of genetic defect diabetic mice that can spontaneously develop into severe renal dysfunction. Intriguingly, TEC treatment restored diabetes-induced glucose and lipid metabolic disorder; and improved the deterioration of renal function, particularly the renal endothelium function in db/db mice. Additionally, TEC inhibited the renal inflammation via reducing macrophages infiltration and M1 polarization. Moreover, TEC inhibited lipopolysaccharide (LPS)-induced endothelial injury and M1 polarization in vitro. Mechanistically, TEC partially restored the reduction in expression of adiponectin receptor 1/2 (AdipoR1/2), pi-LKB1, pi-AMPKα, and PPARα in vitro and in vivo. Noteworthy, these beneficial pharmacological activities mediated by TEC were significantly attenuated after AdipoR1/2 knockdown by siRNA, indicating that AdipoR1/2 plays a critical role in protection against DN. Collectively, these results suggested that TEC have a potently effect for retarding type 2 diabetes-associated DN.
Collapse
Affiliation(s)
- Shu Yang
- Department of Endocrinology, The 2nd Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, China
| | - Chuanrui Ma
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
| | - Han Wu
- Department of Endocrinology, The 2nd Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, China; Department of Endocrinology, Key Laboratory of Endocrinology, National Health Commission, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Hao Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
| | - Fengyi Yuan
- Department of Endocrinology, The 2nd Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, China
| | - Guangyan Yang
- Department of Endocrinology, The 2nd Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, China
| | - Qi Yang
- Department of Endocrinology, The 2nd Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, China
| | - Lijing Jia
- Department of Endocrinology, The 2nd Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, China.
| | - Zhen Liang
- Department of Endocrinology, The 2nd Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, China.
| | - Lin Kang
- Department of Endocrinology, The 2nd Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, China.
| |
Collapse
|
17
|
Kidney Injury Molecule-1 Is Upregulated in Renal Lipotoxicity and Mediates Palmitate-Induced Tubular Cell Injury and Inflammatory Response. Int J Mol Sci 2019; 20:ijms20143406. [PMID: 31373312 PMCID: PMC6679556 DOI: 10.3390/ijms20143406] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/05/2019] [Accepted: 07/10/2019] [Indexed: 12/20/2022] Open
Abstract
Diabetic nephropathy is increasingly recognized as a major contributor to kidney failure in patients with obesity and type 2 diabetes. This study was designed to identify the molecular mediators of kidney injury associated with metabolic syndrome with or without hyperglycemia. We compared renal gene expression profiles in Zucker lean (ZL), Zucker obese (ZO), and Zucker diabetic (ZD) rats using cDNA microarray with quantitative verification of selected transcripts by real-time PCR. Compared to the 20-week-old ZL control (glucose: 110 ± 8 mg/dL), both prediabetic ZO (glucose: 157 ± 11 mg/dL) and diabetic ZD (glucose: 481 ± 37 mg/dL) rats displayed hyperlipidemia and kidney injury with a high degree of proteinuria. cDNA microarray identified 25 inflammation and injury-related transcriptomes whose expression levels were similarly increased in ZO and ZD kidneys. Among them, kidney injury molecule-1 (KIM-1) was found to be the most highly upregulated in both ZO and ZD kidneys. Immunofluorescence staining of kidney sections revealed a strong correlation between lipid overload and KIM-1 upregulation in proximal tubules of ZO and ZD rats. In cultured primary renal tubular epithelial cells (TECs), administration of saturated fatty acid palmitate resulted in an upregulation of KIM-1, osteopontin, and CD44, which was greatly attenuated by U0126, an inhibitor of extracellular signal-regulated kinase (ERK)1/2. Moreover, knockdown of KIM-1 by siRNA interference inhibited palmitate-induced cleaved caspase-3, osteopontin, and CD44 proteins in primary TECs. Our results indicate that KIM-1 expression is upregulated in renal lipotoxicity and may play an important role in fatty acid-induced inflammation and tubular cell damage in obesity and diabetic kidney disease.
Collapse
|
18
|
Souza CS, de Sousa Oliveira BS, Viana GN, Correia TML, de Bragança AC, Canale D, Oliveira MV, de Magalhães ACM, Volpini RA, de Brito Amaral LS, de Jesus Soares T. Preventive effect of exercise training on diabetic kidney disease in ovariectomized rats with type 1 diabetes. Exp Biol Med (Maywood) 2019; 244:758-769. [PMID: 31042072 PMCID: PMC6567583 DOI: 10.1177/1535370219843830] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 03/18/2019] [Indexed: 12/25/2022] Open
Abstract
IMPACT STATEMENT To date, no studies have been found evaluating the effects of physical exercise on renal function and structure changes in ovariectomized rats with type 1 diabetes. Therefore, this work emerges with an important tool for strengthening and expanding innovative research on exercise with potential for the prevention of renal diseases in ovariectomized diabetic rats, and future development of studies that seek to increase scientific knowledge about the beneficial effects of physical exercise on renal diseases in humans.
Collapse
Affiliation(s)
- Cláudia Silva Souza
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Programa de Pós-Graduação em Biociências, Vitória da Conquista, Bahia 45029-094, Brasil
| | - Bianca Silva de Sousa Oliveira
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Programa de Pós-Graduação em Biociências, Vitória da Conquista, Bahia 45029-094, Brasil
| | - Geovanildo Nascimento Viana
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Programa de Pós-Graduação em Biociências, Vitória da Conquista, Bahia 45029-094, Brasil
| | - Thiago Macêdo Lopes Correia
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Programa de Pós-Graduação em Biociências, Vitória da Conquista, Bahia 45029-094, Brasil
| | - Ana Carolina de Bragança
- Departamento de Nefrologia, Laboratório de Pesquisa Básica-LIM12, Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-903, Brazil
| | - Daniele Canale
- Departamento de Nefrologia, Laboratório de Pesquisa Básica-LIM12, Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-903, Brazil
| | - Márcio Vasconcelos Oliveira
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Programa de Pós-Graduação em Biociências, Vitória da Conquista, Bahia 45029-094, Brasil
| | - Amélia Cristina Mendes de Magalhães
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Programa de Pós-Graduação em Biociências, Vitória da Conquista, Bahia 45029-094, Brasil
| | - Rildo Aparecido Volpini
- Departamento de Nefrologia, Laboratório de Pesquisa Básica-LIM12, Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-903, Brazil
| | - Liliany Souza de Brito Amaral
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Programa de Pós-Graduação em Biociências, Vitória da Conquista, Bahia 45029-094, Brasil
| | - Telma de Jesus Soares
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Programa de Pós-Graduação em Biociências, Vitória da Conquista, Bahia 45029-094, Brasil
| |
Collapse
|
19
|
Molecular targets of fenofibrate in the cardiovascular-renal axis: A unifying perspective of its pleiotropic benefits. Pharmacol Res 2019; 144:132-141. [PMID: 30970278 DOI: 10.1016/j.phrs.2019.03.025] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 03/28/2019] [Accepted: 03/29/2019] [Indexed: 12/17/2022]
|
20
|
Guan J, Zhao M, He C, Li X, Li Y, Sun J, Wang W, Cui YL, Zhang Q, Li BY, Qiao GF. Anti-Hypertensive Action of Fenofibrate via UCP2 Upregulation Mediated by PPAR Activation in Baroreflex Afferent Pathway. Neurosci Bull 2019; 35:15-24. [PMID: 30173356 PMCID: PMC6357279 DOI: 10.1007/s12264-018-0271-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 05/29/2018] [Indexed: 12/31/2022] Open
Abstract
Fenofibrate, an agonist for peroxisome proliferator-activated receptor alpha (PPAR-α), lowers blood pressure, but whether this action is mediated via baroreflex afferents has not been elucidated. In this study, the distribution of PPAR-α and PPAR-γ was assessed in the nodose ganglion (NG) and the nucleus of the solitary tract (NTS). Hypertension induced by drinking high fructose (HFD) was reduced, along with complete restoration of impaired baroreceptor sensitivity, by chronic treatment with fenofibrate. The molecular data also showed that both PPAR-α and PPAR-γ were dramatically up-regulated in the NG and NTS of the HFD group. Expression of the downstream signaling molecule of PPAR-α, the mitochondrial uncoupling protein 2 (UCP2), was up-regulated in the baroreflex afferent pathway under similar experimental conditions, along with amelioration of reduced superoxide dismutase activity and increased superoxide in HFD rats. These results suggest that chronic treatment with fenofibrate plays a crucial role in the neural control of blood pressure by improving baroreflex afferent function due at least partially to PPAR-mediated up-regulation of UCP2 expression and reduction of oxidative stress.
Collapse
Affiliation(s)
- Jian Guan
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Miao Zhao
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Chao He
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Xue Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Ying Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Jie Sun
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Wei Wang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Ya-Li Cui
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Qing Zhang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Bai-Yan Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China.
| | - Guo-Fen Qiao
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
21
|
Paw M, Wnuk D, Kądziołka D, Sęk A, Lasota S, Czyż J, Madeja Z, Michalik M. Fenofibrate Reduces the Asthma-Related Fibroblast-To-Myofibroblast Transition by TGF-Β/Smad2/3 Signaling Attenuation and Connexin 43-Dependent Phenotype Destabilization. Int J Mol Sci 2018; 19:ijms19092571. [PMID: 30158495 PMCID: PMC6163263 DOI: 10.3390/ijms19092571] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 08/21/2018] [Accepted: 08/27/2018] [Indexed: 12/26/2022] Open
Abstract
The activation of human bronchial fibroblasts by transforming growth factor-β1 (TGF-β1) leads to the formation of highly contractile myofibroblasts in the process of the fibroblast–myofibroblast transition (FMT). This process is crucial for subepithelial fibrosis and bronchial wall remodeling in asthma. However, this process evades current therapeutic asthma treatment strategies. Since our previous studies showed the attenuation of the TGF-β1-induced FMT in response to lipid-lowering agents (e.g., statins), we were interested to see whether a corresponding effect could be obtained upon administration of hypolipidemic agents. In this study, we investigated the effect of fenofibrate on FMT efficiency in populations of bronchial fibroblasts derived from asthmatic patients. Fenofibrate exerted a dose-dependent inhibitory effect on the FMT, even though it did not efficiently affect the expression of α-smooth muscle actin (α-SMA; marker of myofibroblasts); however, it considerably reduced its incorporation into stress fibers through connexin 43 regulation. This effect was accompanied by disturbances in the actin cytoskeleton architecture, impairments in the maturation of focal adhesions, and the fenofibrate-induced deactivation of TGF-β1/Smad2/3 signaling. These data suggest that fenofibrate interferes with myofibroblastic differentiation during asthma-related subepithelial fibrosis. The data indicate the potential application of fenofibrate in the therapy and prevention of bronchial remodeling during the asthmatic process.
Collapse
Affiliation(s)
- Milena Paw
- Faculty of Biophysics, Biochemistry and Biotechnology, Department of Cell Biology, Jagiellonian University, Gronostajowa 7, 30-378 Kraków, Poland.
| | - Dawid Wnuk
- Faculty of Biophysics, Biochemistry and Biotechnology, Department of Cell Biology, Jagiellonian University, Gronostajowa 7, 30-378 Kraków, Poland.
| | - Dominika Kądziołka
- Faculty of Biophysics, Biochemistry and Biotechnology, Department of Cell Biology, Jagiellonian University, Gronostajowa 7, 30-378 Kraków, Poland.
| | - Aleksandra Sęk
- Faculty of Biophysics, Biochemistry and Biotechnology, Department of Cell Biology, Jagiellonian University, Gronostajowa 7, 30-378 Kraków, Poland.
- Nencki Institute of Experimental Biology, Laboratory of Intracellular Ion Channels, 02-093 Warsaw, Poland.
| | - Sławomir Lasota
- Faculty of Biophysics, Biochemistry and Biotechnology, Department of Cell Biology, Jagiellonian University, Gronostajowa 7, 30-378 Kraków, Poland.
| | - Jarosław Czyż
- Faculty of Biophysics, Biochemistry and Biotechnology, Department of Cell Biology, Jagiellonian University, Gronostajowa 7, 30-378 Kraków, Poland.
| | - Zbigniew Madeja
- Faculty of Biophysics, Biochemistry and Biotechnology, Department of Cell Biology, Jagiellonian University, Gronostajowa 7, 30-378 Kraków, Poland.
| | - Marta Michalik
- Faculty of Biophysics, Biochemistry and Biotechnology, Department of Cell Biology, Jagiellonian University, Gronostajowa 7, 30-378 Kraków, Poland.
| |
Collapse
|
22
|
Monteillet L, Gjorgjieva M, Silva M, Verzieux V, Imikirene L, Duchampt A, Guillou H, Mithieux G, Rajas F. Intracellular lipids are an independent cause of liver injury and chronic kidney disease in non alcoholic fatty liver disease-like context. Mol Metab 2018; 16:100-115. [PMID: 30100243 PMCID: PMC6157648 DOI: 10.1016/j.molmet.2018.07.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 07/19/2018] [Accepted: 07/23/2018] [Indexed: 12/15/2022] Open
Abstract
Objective Ectopic lipid accumulation in the liver and kidneys is a hallmark of metabolic diseases leading to non-alcoholic fatty liver disease (NAFLD) and chronic kidney disease (CKD). Moreover, recent data have highlighted a strong correlation between NAFLD and CKD incidences. In this study, we use two mouse models of hepatic steatosis or CKD, each initiated independently of the other upon the suppression of glucose production specifically in the liver or kidneys, to elucidate the mechanisms underlying the development of CKD in the context of NAFLD-like pathology. Methods Mice with a deletion of G6pc, encoding glucose-6 phosphatase catalytic subunit, specifically in the liver (L.G6pc−/− mice) or the kidneys (K.G6pc−/− mice), were fed with either a standard diet or a high fat/high sucrose (HF/HS) diet during 9 months. These mice represent two original models of a rare metabolic disease named Glycogen Storage Disease Type Ia (GSDIa) that is characterized by both NAFLD-like pathology and CKD. Two other groups of L.G6pc−/− and K.G6pc−/− mice were fed a standard diet for 6 months and then treated with fenofibrate for 3 months. Lipid and glucose metabolisms were characterized, and NAFLD-like and CKD damages were evaluated. Results Lipid depot exacerbation upon high-calorie diet strongly accelerated hepatic and renal pathologies induced by the G6pc-deficiency. In L.G6pc−/− mice, HF/HS diet increased liver injuries, characterized by higher levels of plasmatic transaminases and increased hepatic tumor incidence. In K.G6pc−/− mice, HF/HS diet increased urinary albumin and lipocalin 2 excretion and aggravated renal fibrosis. In both cases, the worsening of NAFLD-like injuries and CKD was independent of glycogen content. Furthermore, fenofibrate, via the activation of lipid oxidation significantly decreased the hepatic or renal lipid accumulations and prevented liver or kidney damages in L.G6pc−/− and K.G6pc−/− mice, respectively. Finally, we show that L.G6pc−/− mice and K.G6pc−/− mice developed NAFLD-like pathology and CKD independently. Conclusions This study highlights the crucial role that lipids play in the independent development of both NAFLD and CKD and demonstrates the importance of lipid-lowering treatments in various metabolic diseases featured by lipid load, from the “rare” GSDIa to the “epidemic” morbid obesity or type 2 diabetes. Exacerbating lipid accumulation aggravates liver/kidney injury in GSDI. Fenofibrate-mediated PPARα activation induces hepatic and renal lipid turnover. Increased lipid turnover prevents glycogen synthesis and accumulation. PPARα–mediated metabolic reprograming prevents hepatic and renal GSDI complications. NAFLD and CKD develop independently.
Collapse
Affiliation(s)
- Laure Monteillet
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France.
| | - Monika Gjorgjieva
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France.
| | - Marine Silva
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France.
| | - Vincent Verzieux
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France.
| | - Linda Imikirene
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France.
| | - Adeline Duchampt
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France.
| | - Hervé Guillou
- Toxalim, Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, 31027, France.
| | - Gilles Mithieux
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France.
| | - Fabienne Rajas
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France.
| |
Collapse
|
23
|
Cobbs A, Ballou K, Chen X, George J, Zhao X. Saturated fatty acids bound to albumin enhance osteopontin expression and cleavage in renal proximal tubular cells. INTERNATIONAL JOURNAL OF PHYSIOLOGY, PATHOPHYSIOLOGY AND PHARMACOLOGY 2018; 10:29-38. [PMID: 29593848 PMCID: PMC5871627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 02/25/2018] [Indexed: 06/08/2023]
Abstract
Osteopontin (OPN) is one of the proinflammatory cytokines upregulated in the kidneys of diabetic animals and patients with nephropathy. An increase in urinary albumin and albumin-bound fatty acids (FA) presents a proinflammatory environment to the proximal tubules in proteinuric kidney diseases including diabetic nephropathy. This study was designed to examine if FA overload could stimulate OPN expression and cleavage in renal tubule epithelial cells. OPN gene and protein expression was examined in the kidney of Zucker diabetic (ZD) rats and cultured proximal tubular cells exposed to either bovine serum albumin (BSA) or BSA conjugated with palmitic acid (PA), the most abundant saturated plasma FA. Real-time PCR analysis confirmed an upregulation of renal cortical OPN gene correlated with albuminuria and nephropathy progression in ZD rats at the age of 7-20 weeks. Immunofluorescence staining of kidney sections revealed a massive induction of OPN protein in albumin-overloaded proximal tubules of ZD rats. A significant increase in both intact and cleaved OPN proteins was further demonstrated in the diabetic kidney and urine samples, which was attenuated by antiproteinuric treatment with losartan, an angiotensin II receptor blocker. When exposed to fatty acid-free BSA, NRK-52E cells exhibited an increase in protein levels of full-length and cleaved OPN. Moreover, the increase in OPN fragments was greatly enhanced in the presence of PA (250-500 µM). Together, our results support a stimulatory effect of albumin and conjugated FA on OPN expression and cleavage in renal tubule epithelial cells. Thus, besides lowering albuminuria/proteinuria, mitigating circulating FAs may be an effective intervention for preventing and slowing down the progression of nephropathy associated with obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Alyssa Cobbs
- Department of Physiology, Morehouse School of MedicineAtlanta 30310, GA, USA
| | - Kristopher Ballou
- Department of Physiology, Morehouse School of MedicineAtlanta 30310, GA, USA
| | - Xiaoming Chen
- Department of Physiology, Morehouse School of MedicineAtlanta 30310, GA, USA
| | - Jasmine George
- Department of Physiology, Morehouse School of MedicineAtlanta 30310, GA, USA
| | - Xueying Zhao
- Department of Physiology, Morehouse School of MedicineAtlanta 30310, GA, USA
| |
Collapse
|
24
|
Gao Q, Sarkar A, Chen Y, Xu B, Zhu X, Yuan Y, Guan T. Overexpression of heart-type fatty acid binding protein enhances fatty acid-induced podocyte injury. Exp Ther Med 2018; 15:2054-2061. [PMID: 29434805 PMCID: PMC5776647 DOI: 10.3892/etm.2017.5643] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 08/17/2017] [Indexed: 12/13/2022] Open
Abstract
Deregulated lipid metabolism is a characteristic of metabolic diseases including type 2 diabetes and obesity, and likely contributes to podocyte injury and end-stage kidney disease. Heart-type fatty acid binding protein (H-FABP) was reported to be associated with lipid metabolism. The present study investigated whether H-FABP contributes to podocyte homeostasis. Podocytes were transfected by lentiviral vector to construct a cell line which stably overexpressed H-FABP. Small interfering RNA capable of effectively silencing H-FABP was introduced into podocytes to construct a cell line with H-FABP knockdown. Certain groups were treated with palmitic acid (PA) and the fat metabolism, as well as inflammatory and oxidative stress markers were measured. PA accelerated lipid metabolism derangement, inflammatory reaction and oxidative stress in podocytes. Overexpression of H-FABP enhanced the PA-induced disequilibrium in podocytes. The mRNA and protein expression levels of acyl-coenzyme A oxidase 3 and monocyte chemotactic protein 1, and the protein expression levels of 8-hydroxy-2'-deoxyguanosine and 4-hydroxynonenal were upregulated in the H-FABP overexpression group, while the mRNA and protein expression of peroxisome proliferator activated receptor α was downregulated. Knockdown of H-FABP inhibited the PA-induced injury and lipid metabolism derangement, as well as the inflammatory reaction and oxidative stress in podocytes. These results indicated that overexpression of H-FABP enhances fatty acid-induced podocyte injury, while H-FABP inhibition may represent a potential therapeutic strategy for the prevention of lipid metabolism-associated podocyte injury.
Collapse
Affiliation(s)
- Qing Gao
- Department of Nephrology, Zhongshan Hospital, Xiamen University, Xiamen, Fujian 361004, P.R. China
| | - Alhossain Sarkar
- Department of Nephrology, Zhongshan Hospital, Xiamen University, Xiamen, Fujian 361004, P.R. China
| | - Yizhi Chen
- Department of Nephrology, Hainan Branch of Chinese PLA General Hospital, Sanya, Hainan 572013, P.R. China
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing 100086, P.R. China
| | - Bo Xu
- Department of Nephrology, Zhongshan Hospital, Xiamen University, Xiamen, Fujian 361004, P.R. China
| | - Xiaojuan Zhu
- Department of Nephrology, Zhongshan Hospital, Xiamen University, Xiamen, Fujian 361004, P.R. China
| | - Yang Yuan
- Department of Nephrology, Zhongshan Hospital, Xiamen University, Xiamen, Fujian 361004, P.R. China
| | - Tianjun Guan
- Department of Nephrology, Zhongshan Hospital, Xiamen University, Xiamen, Fujian 361004, P.R. China
| |
Collapse
|
25
|
Abstract
AKI is associated with high morbidity and mortality, and it predisposes to the development and progression of CKD. Novel strategies that minimize AKI and halt the progression of CKD are urgently needed. Normal kidney function involves numerous different cell types, such as tubular epithelial cells, endothelial cells, and podocytes, working in concert. This delicate balance involves many energy-intensive processes. Fatty acids are the preferred energy substrates for the kidney, and defects in fatty acid oxidation and mitochondrial dysfunction are universally involved in diverse causes of AKI and CKD. This review provides an overview of ATP production and energy demands in the kidney and summarizes preclinical and clinical evidence of mitochondrial dysfunction in AKI and CKD. New therapeutic strategies targeting mitochondria protection and cellular bioenergetics are presented, with emphasis on those that have been evaluated in animal models of AKI and CKD. Targeting mitochondrial function and cellular bioenergetics upstream of cellular damage may offer advantages compared with targeting downstream inflammatory and fibrosis processes.
Collapse
Affiliation(s)
- Hazel H Szeto
- Mitochondrial Therapeutics Consulting, New York, New York
| |
Collapse
|
26
|
Abstract
Fibrosis is a major player in cardiovascular disease, both as a contributor to the development of disease, as well as a post-injury response that drives progression. Despite the identification of many mechanisms responsible for cardiovascular fibrosis, to date no treatments have emerged that have effectively reduced the excess deposition of extracellular matrix associated with fibrotic conditions. Novel treatments have recently been identified that hold promise as potential therapeutic agents for cardiovascular diseases associated with fibrosis, as well as other fibrotic conditions. The purpose of this review is to provide an overview of emerging antifibrotic agents that have shown encouraging results in preclinical or early clinical studies, but have not yet been approved for use in human disease. One of these agents is bone morphogenetic protein-7 (BMP7), which has beneficial effects in multiple models of fibrotic disease. Another approach discussed involves altering the levels of micro-RNA (miR) species, including miR-29 and miR-101, which regulate the expression of fibrosis-related gene targets. Further, the antifibrotic potential of agonists of the peroxisome proliferator-activated receptors will be discussed. Finally, evidence will be reviewed in support of the polypeptide hormone relaxin. Relaxin is long known for its extracellular remodeling properties in pregnancy, and is rapidly emerging as an effective antifibrotic agent in a number of organ systems. Moreover, relaxin has potent vascular and renal effects that make it a particularly attractive approach for the treatment of cardiovascular diseases. In each case, the mechanism of action and the applicability to various fibrotic diseases will be discussed.
Collapse
Affiliation(s)
- Benita L McVicker
- Research Service, VA Nebraska-Western Iowa Health Care System, OmahaNE, United States.,Division of Gastroenterology and Hepatology, University of Nebraska Medical Center, OmahaNE, United States
| | - Robert G Bennett
- Research Service, VA Nebraska-Western Iowa Health Care System, OmahaNE, United States.,The Division of Diabetes, Endocrinology, and Metabolism, Department of Internal Medicine, University of Nebraska Medical Center, OmahaNE, United States.,Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, OmahaNE, United States
| |
Collapse
|
27
|
Zhu H, Gui Q, Hui X, Wang X, Jiang J, Ding L, Sun X, Wang Y, Chen H. TGF-β1/Smad3 Signaling Pathway Suppresses Cell Apoptosis in Cerebral Ischemic Stroke Rats. Med Sci Monit 2017; 23:366-376. [PMID: 28110342 PMCID: PMC5282965 DOI: 10.12659/msm.899195] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND We desired to observe the changes of transforming growth factor-β1/drosophila mothers against decapentaplegic protein (TGF-β1/Smad3) signaling pathway in the hippocampus region of cerebral ischemic stroke rats so that the effects of this pathway on nerve cells can be investigated. MATERIAL AND METHODS The ischemic stroke models were built by middle cerebral artery occlusion (MCAO) in vivo and oxygen-glucose deprivation (OGD) in vitro. TGF-β1 and TGF-β1 inhibitors were injected into rat models while TGF-β1, TGF-β1 siRNA, Smad3, and Smad3 siRNA were transfected into cells. Infarct sizes were measured using triphenyltetrazolium chloride (TTC) staining, while the apoptosis rate of cells were calculated by Annexin V-fluorescein isothiocyanate/propidium iodide (Annexin V-FITC/PI) staining. Levels of TGF-β1, Smad3, and Bcl-2 were examined by real-time polymerase chain reaction (RT-PCR), immunohistochemical, and Western blot analysis. RESULTS The expressions of TGF-β1/Smad3 signal pathway were significantly increased in both model rats and BV2 cells, whereas the expression of Bcl-2 was down-regulated (P<0.05). The TGF-β1/Smad3 signal pathway exhibited protective effects, including the down-regulation of infarction size in cerebral tissues and the down-regulation of apoptosis rate of BV2 cells by increasing the expression of Bcl-2 (P<0.05). In addition, these effects could be antagonized by the corresponding inhibitors and siRNA (P<0.05). CONCLUSIONS The TGF-β1/Smad3 signaling pathway was up-regulated once cerebral ischemic stroke was simulated. TGF-β1 may activate the expression of Bcl-2 via Smad3 to suppress the apoptosis of neurons.
Collapse
Affiliation(s)
- Haiping Zhu
- Department of Neurosurgery, The First People's Hospital of Changshou City, Changshou, Jiangsu, China (mainland)
| | - Qunfeng Gui
- Department of Neurosurgery, Yancheng Third People's Hospital, The affiliated Yancheng Hospital of Southeast University Medical College, Yancheng, Jiangsu, China (mainland)
| | - Xiaobo Hui
- Department of Neurosurgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China (mainland)
| | - Xiaodong Wang
- Department of Neurosurgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China (mainland)
| | - Jian Jiang
- Department of Neurosurgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China (mainland)
| | - Lianshu Ding
- Department of Neurosurgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China (mainland)
| | - Xiaoyang Sun
- Department of Neurosurgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China (mainland)
| | - Yanping Wang
- Department of Neurosurgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China (mainland)
| | - Huaqun Chen
- Department of Neurosurgery, Yancheng Third People's Hospital, The affiliated Yancheng Hospital of Southeast University Medical College, Yancheng, Jiangsu, China (mainland)
| |
Collapse
|
28
|
Al-Rasheed NM, Al-Rasheed NM, Al-Amin MA, Hasan IH, Al-Ajmi HN, Mohammad RA, Attia HA. Fenofibrate attenuates diabetic nephropathy in experimental diabetic rat's model via suppression of augmented TGF-β1/Smad3 signaling pathway. Arch Physiol Biochem 2016; 122:186-194. [PMID: 26959841 DOI: 10.3109/13813455.2016.1164186] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
CONTEXT Fibrates, the ligands of peroxisome profileferator-activated receptor-α have been shown to have a renal protective action in diabetic nephropathy (DN). OBJECTIVE This study aimed to elucidate the effect of fenofibrate on renal transforming growth factor-β1 (TGF-β1) and Smad3 in Streptozotocin (STZ)-induced DN. METHODS Diabetes was induced in rats by a single intraperitoneal injection of streptozotocin (55 mg/kg). Diabetic rats were given fenofibrate (100 mg/kg, p.o.). After 12 weeks, diabetic nephropathy biomarkers were assessed. The mRNA expression of collage I and III, TGF-β1 and Smad3 and were detected by RT-PCR. RESULTS Fenofibrate reduced significantly serum creatinine, kidney/body weight ratio, serum albumin excretion Collage I & III, TGF-β1 and Smad3 mRNA expression. CONCLUSIONS Our results give further insights into the mechanisms underlying the protective role of fenofibrate in DN, suggesting that interference with TGF-β1/Smad3 signaling pathway may be a useful therapeutic approach to prevent DN.
Collapse
Affiliation(s)
- Nouf Mohamed Al-Rasheed
- a Pharmacology and Toxicology Department , College of Pharmacy, King Saud University , Riyadh , KSA
| | - Nawal Mohamed Al-Rasheed
- a Pharmacology and Toxicology Department , College of Pharmacy, King Saud University , Riyadh , KSA
- b Pharmacology Department , College of Pharmacy, Princess Nora Bint Abdul Rahman University , KSA
| | - Maha Abdelrahman Al-Amin
- a Pharmacology and Toxicology Department , College of Pharmacy, King Saud University , Riyadh , KSA
| | - Iman Huesein Hasan
- a Pharmacology and Toxicology Department , College of Pharmacy, King Saud University , Riyadh , KSA
| | - Hanaa Najeeb Al-Ajmi
- a Pharmacology and Toxicology Department , College of Pharmacy, King Saud University , Riyadh , KSA
| | - Raeesa Ahmed Mohammad
- c Anatomy Department , Faculty of Medicine, King Saud University , Riyadh , KSA , and
| | - Hala Aboulfotooh Attia
- a Pharmacology and Toxicology Department , College of Pharmacy, King Saud University , Riyadh , KSA
- d Biochemistry Department , College of Pharmacy, Mansours University , Mansoura , Egypt
| |
Collapse
|
29
|
Effect of astragalus injection on renal tubular epithelial transdifferentiation in type 2 diabetic mice. Altern Ther Health Med 2016; 16:222. [PMID: 27422712 PMCID: PMC4947258 DOI: 10.1186/s12906-016-1208-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 06/15/2016] [Indexed: 12/02/2022]
Abstract
Background Astragalus injection is used by practitioners of traditional Chinese medicine to treat diabetic nephropathy (DN). The current study was conducted to determine the effect of astragalus on tubular epithelial transdifferentiation during the progression of DN in KKAy mice, as well as to investigate the molecular mechanism underlying this effect. Methods Diabetic, 14-week-old, male KKAy mice were randomly divided into a model group and an astragalus treatment group, while age-matched male C57BL/6 J mice were selected as controls. The treatment group received daily intraperitoneal injections of astragalus (0.03 mL/10 g per day), while the model group received injections of an equal volume of saline. Mice were euthanized after 24 weeks. Serum samples were obtained from the animals in each group for blood glucose measurement. Kidney tissue samples were used for morphometric studies. The mRNA and protein expression levels of transforming growth factor beta 1 (TGF-β1), transforming growth factor beta receptor 1 (TGFβ-R1), alpha smooth muscle actin (α-SMA), and E-cadherin were evaluated using real-time polymerase chain reaction (PCR) and western blotting. Results Astragalus significantly reduced blood glucose levels; inhibited morphological changes in the kidneys of KKAy mice; reduced mRNA and protein expression levels of TGF-β1, TGFβ-R1, and α-SMA; and increased E-cadherin expression. Conclusions Tubular epithelial transdifferentiation plays an important role in the development of DN in diabetic mice. Administration of astragalus likely prevents or mitigates DN by suppressing tubular epithelial transdifferentiation, protecting KKAy mice from renal damage.
Collapse
|
30
|
Obesity-related glomerulopathy: clinical and pathologic characteristics and pathogenesis. Nat Rev Nephrol 2016; 12:453-71. [PMID: 27263398 DOI: 10.1038/nrneph.2016.75] [Citation(s) in RCA: 472] [Impact Index Per Article: 52.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The prevalence of obesity-related glomerulopathy is increasing in parallel with the worldwide obesity epidemic. Glomerular hypertrophy and adaptive focal segmental glomerulosclerosis define the condition pathologically. The glomerulus enlarges in response to obesity-induced increases in glomerular filtration rate, renal plasma flow, filtration fraction and tubular sodium reabsorption. Normal insulin/phosphatidylinositol 3-kinase/Akt and mTOR signalling are critical for podocyte hypertrophy and adaptation. Adipokines and ectopic lipid accumulation in the kidney promote insulin resistance of podocytes and maladaptive responses to cope with the mechanical forces of renal hyperfiltration. Although most patients have stable or slowly progressive proteinuria, up to one-third develop progressive renal failure and end-stage renal disease. Renin-angiotensin-aldosterone blockade is effective in the short-term but weight loss by hypocaloric diet or bariatric surgery has induced more consistent and dramatic antiproteinuric effects and reversal of hyperfiltration. Altered fatty acid and cholesterol metabolism are increasingly recognized as key mediators of renal lipid accumulation, inflammation, oxidative stress and fibrosis. Newer therapies directed to lipid metabolism, including SREBP antagonists, PPARα agonists, FXR and TGR5 agonists, and LXR agonists, hold therapeutic promise.
Collapse
|
31
|
Yaribeygi H, Mohammadi MT. Evaluation of PPAR-α Agonist effect on Kidney Performance Through Increment of Nitric Oxide During Hyperglycemia-Induced Nephropathy in Rat. RAZAVI INTERNATIONAL JOURNAL OF MEDICINE 2016. [DOI: 10.17795/rijm37670] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
32
|
Gasparics Á, Rosivall L, Krizbai IA, Sebe A. When the endothelium scores an own goal: endothelial cells actively augment metastatic extravasation through endothelial-mesenchymal transition. Am J Physiol Heart Circ Physiol 2016; 310:H1055-63. [PMID: 26993222 DOI: 10.1152/ajpheart.00042.2016] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 03/14/2016] [Indexed: 01/05/2023]
Abstract
Endothelial-mesenchymal transition (EndMT) is an important mechanism during organ development and in certain pathological conditions. For example, EndMT contributes to myofibroblast formation during organ fibrosis, and it has been identified as an important source of cancer-associated fibroblasts, facilitating tumor progression. Recently, EndMT was proposed to modulate endothelial function during intravasation and extravasation of metastatic tumor cells. Evidence suggests that endothelial cells are not passive actors during transendothelial migration (TEM) of cancer cells, as there are profound changes in endothelial junctional protein expression, signaling, permeability, and contractility. This review describes these alterations in endothelial characteristics during TEM of metastatic tumor cells and discusses them in the context of EndMT. EndMT could play an important role during metastatic intravasation and extravasation, a novel hypothesis that may lead to new therapeutic approaches to tackle metastatic disease.
Collapse
Affiliation(s)
- Ákos Gasparics
- Department of Pathophysiology, Semmelweis University, Budapest, Hungary
| | - László Rosivall
- Department of Pathophysiology, Semmelweis University, Budapest, Hungary; Pediatrics and Nephrology Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
| | - István A Krizbai
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary; Institute of Life Sciences, Vasile Goldis Western University of Arad, Arad, Romania; and
| | - Attila Sebe
- Department of Pathophysiology, Semmelweis University, Budapest, Hungary; Division of Medical Biotechnology, Paul Ehrlich Institute, Langen, Germany
| |
Collapse
|
33
|
Mou X, Zhou DY, Zhou DY, Ma JR, Liu YH, Chen HP, Hu YB, Shou CM, Chen JW, Liu WH, Ma GL. Serum TGF-β1 as a Biomarker for Type 2 Diabetic Nephropathy: A Meta-Analysis of Randomized Controlled Trials. PLoS One 2016; 11:e0149513. [PMID: 26901047 PMCID: PMC4764759 DOI: 10.1371/journal.pone.0149513] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 02/01/2016] [Indexed: 01/27/2023] Open
Abstract
Background Abnormal expression of serum TGF-β1 was found in patients with diabetic nephropathy. However, the association of TGF-β1 with the risk of diabetic nephropathy remains unknown. The present study was undertaken to investigate whether such an association exists. Methods We searched the Chinese VIP, Wangfang, China National Knowledge Infrastructure, PubMed, Embase, and Google Scholar databases for relevant studies and extracted all eligible data. Stata12 software was used for statistical analysis. Results Nine reports met our criteria and were used for data extraction. There were 264 patients and 227 healthy controls from qualified reports in this meta-analysis. The results suggested that serum TGF-β1 levels were significantly up-regulated in patients with diabetic nephropathy; the instrumental variable was 3.94 (95% confidence interval 3.20–4.68, p<0.01). Conclusions Meta-analysis suggested that elevated serum TGF-β level in patients with diabetes is associated with a high risk of nephropathy. Further studies are required to validate these observations.
Collapse
Affiliation(s)
- Xin Mou
- Endocrinology Department, Hangzhou Red Cross Hospital, Hangzhou, 310003, China
| | - Di-Yi Zhou
- Endocrinology Department, Hangzhou Red Cross Hospital, Hangzhou, 310003, China
| | - Dan-Yang Zhou
- Endocrinology Department, Hangzhou Red Cross Hospital, Hangzhou, 310003, China
| | - Jing-Ru Ma
- Zhejiang Chinese Medical University, City Road 548, Binjiang District, Hangzhou City, Zhejiang Province, 310053, China
| | - Ying-Hui Liu
- Endocrinology Department, Hangzhou Red Cross Hospital, Hangzhou, 310003, China
| | - Hui-Ping Chen
- Endocrinology Department, Hangzhou Red Cross Hospital, Hangzhou, 310003, China
| | - Yong-Bin Hu
- Endocrinology Department, Hangzhou Red Cross Hospital, Hangzhou, 310003, China
| | - Cheng-Min Shou
- Endocrinology Department, Hangzhou Red Cross Hospital, Hangzhou, 310003, China
| | - Jia-Wei Chen
- Endocrinology Department, Hangzhou Red Cross Hospital, Hangzhou, 310003, China
| | - Wen-Hong Liu
- Zhejiang Chinese Medical University, City Road 548, Binjiang District, Hangzhou City, Zhejiang Province, 310053, China
| | - Guo-Ling Ma
- Endocrinology Department, Hangzhou Red Cross Hospital, Hangzhou, 310003, China
- * E-mail:
| |
Collapse
|
34
|
Helmy MM, Helmy MW, El-Mas MM. Additive Renoprotection by Pioglitazone and Fenofibrate against Inflammatory, Oxidative and Apoptotic Manifestations of Cisplatin Nephrotoxicity: Modulation by PPARs. PLoS One 2015; 10:e0142303. [PMID: 26536032 PMCID: PMC4633146 DOI: 10.1371/journal.pone.0142303] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 10/19/2015] [Indexed: 01/06/2023] Open
Abstract
Nephrotoxicity is a major side effect for the antineoplastic drug cisplatin. Here, we employed pharmacological, biochemical, and molecular studies to investigate the role of peroxisome proliferator-activated receptors (PPARs) in cisplatin nephrotoxicity. Rats were treated with a single i.p. dose of cisplatin (5 mg/kg) alone or combined with pioglitazone (PPARγ agonist), fenofibrate (PPARα agonist), pioglitazone plus fenofibrate, or thalidomide (Tumor necrosis factor-α inhibitor; TNF-α). Cisplatin nephrotoxicity was evidenced by rises in renal indices of functional (blood urea nitrogen, BUN, and creatinine), inflammatory (TNF-α, interleukin 6, IL-6), oxidative (increased malondialdehyde, MDA, and decreased superoxide dismutase, SOD and nitric oxide metabolites, NOx), apoptotic (caspase 3), and histological (glomerular atrophy, acute tubular necrosis and vacuolation) profiles. Cisplatin effects were partly abolished upon concurrent exposure to pioglitazone, fenofibrate, or thalidomide; more renoprotection was observed in rats treated with pioglitazaone plus fenofibrate. Immunostaining showed that renal expressions of PPARα and PPARγ were reduced by cisplatin and restored to vehicle-treated values after simultaneous treatment with pioglitazone or fenofibrate. Fenofibrate or pioglitazone renoprotection remained unaltered after concurrent blockade of PPARα (GW6471) and PPARγ (GW9662), respectively. To complement the rat studies, we also report that in human embryonic kidney cells (HEK293 cells), increases caused by cisplatin in inflammatory, apoptotic, and oxidative biomarkers were (i) partly improved after exposure to pioglitazone, fenofibrate, or thalidomide, and (ii) completely disappeared in cells treated with a combination of all three drugs. These data establish that the combined use of pioglitazone and fenofibrate additively improved manifestations of cisplatin nephrotoxicity through perhaps GW6471/GW9662-insensitive mechanisms.
Collapse
Affiliation(s)
- Mai M Helmy
- Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Maged W Helmy
- Pharmacology and Toxicology, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt
| | - Mahmoud M El-Mas
- Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
35
|
Zhao X, Jiang C, Olufade R, Liu D, Emmett N. Kidney Injury Molecule-1 Enhances Endocytosis of Albumin in Renal Proximal Tubular Cells. J Cell Physiol 2015; 231:896-907. [PMID: 26332568 DOI: 10.1002/jcp.25181] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 08/31/2015] [Indexed: 01/25/2023]
Abstract
Receptor-mediated endocytosis plays an important role in albumin reabsorption by renal proximal tubule epithelial cells. Kidney injury molecule-1 (KIM-1) is a scavenger receptor that is upregulated on the apical membrane of proximal tubules in proteinuric kidney disease. In this study, we examined the cellular localization and functional role of KIM-1 in cultured renal tubule epithelial cells (TECs). Confocal immunofluorescence microscopy reveals intracellular and cell surface localization of KIM-1 in primary renal TECs. Albumin stimulation resulted in a redistribution of KIM-1 and tight junction protein zonula occludens-1 in primary TEC monolayer. An increase in albumin internalization was observed in both primary TECs expressing endogenous KIM-1 and rat kidney cell line (NRK-52E) overexpressing exogenous KIM-1. KIM-1-induced albumin accumulation was abolished by its specific antibody. Moreover, endocytosed KIM-1 and its cargo proteins were delivered from endosomes to lysosomes for degradation in a clathrin-dependent pathway. Supportive evidence includes (1) detection of KIM-1 in Rab5-positive early endosomes, Rab7-positive late endosomes/multivesicular bodies, and LAMP1-positive lysosomes, (2) colocalization of KIM-1 and clathrin in the intracellular vesicles, and (3) blockade of KIM-1-mediated albumin internalization by chlorpromazine, an inhibitor of clathrin-dependent endocytosis. KIM-1 expression was upregulated by albumin but downregulated by transforming growth factor-β1. Taken together, our data indicate that KIM-1 increases albumin endocytosis in renal tubule epithelial cells, at least partially via a clathrin-dependent mechanism. J. Cell. Physiol. 231: 896-907, 2016. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Xueying Zhao
- Department of Physiology, Morehouse School of Medicine, Atlanta, Georgia
| | - Chen Jiang
- Department of Physiology, Morehouse School of Medicine, Atlanta, Georgia
| | - Rebecca Olufade
- Department of Physiology, Morehouse School of Medicine, Atlanta, Georgia
| | - Dong Liu
- Department of Physiology, Morehouse School of Medicine, Atlanta, Georgia
| | - Nerimiah Emmett
- Department of Physiology, Morehouse School of Medicine, Atlanta, Georgia
| |
Collapse
|
36
|
Zuo N, Zheng X, Liu H, Ma X. Fenofibrate, a PPARα agonist, protect proximal tubular cells from albumin-bound fatty acids induced apoptosis via the activation of NF-kB. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:10653-10661. [PMID: 26617775 PMCID: PMC4637590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 08/25/2015] [Indexed: 06/05/2023]
Abstract
Albumin-bound fatty acids is the main cause of renal damage, PPARα is responsible in the metabolism of fatty acids. Previous study found that PPARα played a protective role in fatty acids overload associated tubular injury. The aim of the present study is to investigate whether fenofibrate, a PPARα ligands, could contribute to the renoprotective action in fatty acids overload proximal tubule epithelial cells. We observed in HK-2 cells that fenofibrate significantly inhibited fatty acids bound albumin (FA-BSA) induced up-regulation of MCP-1 and IL-8. Treatment with fenofibrate attenuated renal oxidative stress induced by FA-BSA as evidenced by decreased MDA level, increased SOD activity and catalase, GPx-1 expression. FA-BSA induced apoptosis of HK-2 cells were also obviously prevented by fenofibrate. Furthermore, fenofibrate significantly increased the expression of PPARα mRNA and protein in FA-BSA treated cells. Finally, the activation of NF-kB induced by FA-BSA was markedly suppressed by fenofibrate. Taken together, our study describes a renoprotective role of fenofibrate in fatty acids associated tubular toxicity, and the transcriptional activation of PPARα and suppression of NF-kB were at least partially involved.
Collapse
Affiliation(s)
- Nan Zuo
- Department of Nephrology, The First Affiliated Hospital of China Medical UniversityShenyang 110001, People’s Republic of China
| | - Xiaoyu Zheng
- Department of Nephrology, The First Affiliated Hospital of China Medical UniversityShenyang 110001, People’s Republic of China
| | - Hanzhe Liu
- Department of Nephrology, The First Affiliated Hospital of China Medical UniversityShenyang 110001, People’s Republic of China
| | - Xiaoli Ma
- Department of Ophthalmology, The First Affiliated Hospital of China Medical UniversityShenyang 110001, People’s Republic of China
| |
Collapse
|
37
|
Luo H, Wang X, Wang J, Chen C, Wang N, Xu Z, Chen S, Zeng C. Chronic NF-κB blockade improves renal angiotensin II type 1 receptor functions and reduces blood pressure in Zucker diabetic rats. Cardiovasc Diabetol 2015; 14:76. [PMID: 26055622 PMCID: PMC4465496 DOI: 10.1186/s12933-015-0239-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 06/02/2015] [Indexed: 02/07/2023] Open
Abstract
Background Both angiotensin II type 1 receptor (AT1R) and nuclear factor-kappa B (NF-κB) play significant roles in the pathogenesis of hypertension and type 2 diabetes. However, the role of NF-κB in perpetuating renal AT1 receptors dysfunction remains unclear. The aim of the present study to determine whether blockade of NF-κB, could reverse the exaggerated renal AT1R function, reduce inflammatory state and oxidative stress, lower blood pressure in Zucker diabetic fatty (ZDF) rats. Methods Pyrrolidine dithiocarbamate (PDTC), a NF-κB inhibitor (150 mg/kg in drinking water)or vehicle was administered orally to 12-weeks-old ZDF rats and their respective control lean Zucker (LZ) rats for 4 weeks. Blood pressure was measured weekly by tail-cuff method. AT1R functions were determined by measuring diuretic and natriuretic responses to AT1R antagonist (candesartan; 10 μg/kg/min iv). The mRNA and protein levels of NF-κB, oxidative stress maker and AT1R were determined using quantitative real-time PCR and Western blotting, respectively. The NF-κB-DNA binding activity in renal cortex was measured by Electrophoretic mobility shift assay (EMSA). Results As compared with LZ rats, ZDF rats had higher blood pressure, impaired natriuresis and diuresis, accompanied with higher levels of oxidative stress and inflammation. Furthermore, AT1R expression was higher in renal cortex from ZDF rats; candesartan induced natriresis and diuresis, which was augmented in ZDF rats. Treatment with PDTC lowered blood pressure and improved diuretic and natriuretic effects in ZDF rats; meanwhile, the increased oxidative stress and inflammation were reduced; the increased AT1R expression and augmented candesartan-mediated natriuresis and diuresis were recoverd in ZDF rats. Our further study investigated the mechanisms of PDTC on AT1R receptor expression. It resulted that PDTC inhibited NF-κB translocation from cytosol to nucleus, inhibited binding of NF-κB with AT1R promoter, therefore, reduced AT1R expression and function. Conclusions Our present study indicates blockade of NF-κB, via inhibition of binding of NF-κB with AT1R promoter, reduces renal AT1R expression and function, improves oxidative stress and inflammatory/anti-inflammatory balance, therefore, lowers blood pressure and recovers renal function in ZDF rats. Electronic supplementary material The online version of this article (doi:10.1186/s12933-015-0239-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hao Luo
- The Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, PR China. .,Chongqing Institute of Cardiology, Chongqing, PR China.
| | - Xinquan Wang
- The Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, PR China. .,Chongqing Institute of Cardiology, Chongqing, PR China.
| | - Jialiang Wang
- The Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, PR China. .,Chongqing Institute of Cardiology, Chongqing, PR China.
| | - Caiyu Chen
- The Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, PR China. .,Chongqing Institute of Cardiology, Chongqing, PR China.
| | - Na Wang
- The Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, PR China. .,Chongqing Institute of Cardiology, Chongqing, PR China.
| | - Zaicheng Xu
- The Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, PR China. .,Chongqing Institute of Cardiology, Chongqing, PR China.
| | - Shuo Chen
- The Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, PR China. .,Chongqing Institute of Cardiology, Chongqing, PR China.
| | - Chunyu Zeng
- The Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, PR China. .,Chongqing Institute of Cardiology, Chongqing, PR China.
| |
Collapse
|
38
|
El-Haggar SM, Mostafa TM. Comparative clinical study between the effect of fenofibrate alone and its combination with pentoxifylline on biochemical parameters and liver stiffness in patients with non-alcoholic fatty liver disease. Hepatol Int 2015; 9:471-9. [PMID: 25956613 DOI: 10.1007/s12072-015-9633-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 04/12/2015] [Indexed: 12/16/2022]
Abstract
BACKGROUND Non-alcoholic fatty liver disease is a common health problem associated with increased liver and vascular specific complications. AIM The purpose of this study was to assess and compare the effect of fenofibrate alone or in combination with pentoxifylline on the measured biochemical parameters, inflammatory pathway and liver stiffness in patients with non-alcoholic fatty liver disease. METHODS The study design was randomized controlled trial. From July 2013 to June 2014, we recruited 90 non-alcoholic fatty liver patients from the Internal Medicine Department at Tanta University Hospital, Egypt. They were classified randomly into two groups to receive fenofibrate 300 mg daily or fenofibrate 300 mg daily plus pentoxifylline 1200 mg/day in three divided doses for 24 weeks. Fasting blood sample was obtained before and 24 weeks after treatment for biochemical analysis of liver and lipid panels, tumor necrosis factor-alpha, hyaluronic acid, transforming growth factor beta 1, fasting plasma insulin and fasting glucose. Liver stiffness measurement was carried out using fibro-scan. Data were statistically analyzed by paired and unpaired Student's t test. RESULTS The data obtained suggests that adding pentoxifylline to fenofibrate does not provide a beneficial effect on lipid panel, but has a beneficial effect on indirect biochemical markers of hepatic fibrosis, a direct marker linked to matrix deposition (hyaluronic acid), a cytokine/growth factor linked to liver fibrosis (transforming growth factor beta 1), the inflammatory pathway, insulin resistance and liver stiffness as compared to fenofibrate alone. CONCLUSION The combination pentoxifylline plus fenofibrate may represent a new therapeutic strategy for non-alcoholic fatty liver disease as it resulted in more beneficial effects on direct and indirect markers of liver fibrosis, liver stiffness, insulin resistance and inflammatory pathway implicated in NAFLD.
Collapse
|
39
|
Abstract
Type 2 diabetes is a pandemic disease, and its prevalence is increasing mainly due to an increase in obesity and life expectancy. Diabetic complications and their comorbidities constitute the most important economic cost of the disease and represent a significant economic burden for the healthcare systems of developed countries. Despite improving standards of care, people with diabetes remain at risk of the development and progression of microvascular diabetic complications. Therefore, the identification of novel therapeutic approaches is necessary. The aim of this article is to provide an overview of the clinical benefits of fenofibrate on microvascular diabetic complications, with special emphasis on diabetic retinopathy. In addition, the potential mechanisms of action will be briefly discussed.
Collapse
Affiliation(s)
- Rafael Simó
- Diabetes and Metabolism Research Unit. Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain,
| | | | | |
Collapse
|
40
|
Abstract
Objective: To review the mechanisms of anti-cancer activity of fenofibrate (FF) and other Peroxisome Proliferator Activator Receptor α (PPARα) agonists based on evidences reported in the published literature.Methods: We extensively reviewed the literature concerning FF as an off target anti-cancer drug. Controversies regarding conflicting findings were also addressed.Results: The main mechanism involved in anti-cancer activity is anti-angiogenesis through down-regulation of Vascular Endothelial Growth Factor (VEGF), Vascular Endothelial Growth Factor Receptor (VEGFR) and Hypoxia Inducible factor-1 α (HIF-1α), inhibition of endothelial cell migration, up-regulation of endostatin and thrombospondin-1, but there are many other contributing mechanisms like apoptosis and cell cycle arrest, down-regulation of Nuclear Factor Kappa B (NF-kB) and Protein kinase B (Akt) and decrease of cellular energy by impairing mitochondrial function. Growth impairment is related to down-regulation of Phospho-Inositol 3 Kinase (PI3K)/Akt axis and down-regulation of the p38 map kinase (MAPK) cascade. A possible role should be assigned to FF stimulated over-expression of Tribbles Homolog-3 (TRIB3) which inhibits Akt phosphorylation. Important anti-cancer and anti-metastatic activities are due to down-regulation of MCP-1 (monocyte chemotactic protein-1), decreased Metalloprotease-9 (MMP-9) production, weak down-regulation of adhesion molecules like E selectin, intercellular adhesion molecules (ICAM) and Vascular Endothelial Adhesion Molecules (VCAM), and decreased secretion of chemokines like Interleukin-6 (IL-6), and down-regulation of cyclin D-1. There is no direct link between FF activity in lipid metabolism and anticancer activity, except for the fact that many anticancer actions are dependent from PPARα agonism. FF exhibits also PPARα independent anti-cancer activities.Conclusions: There are strong evidences indicating that FF can disrupt growth-related activities in many different cancers, due to anti-angiogenesis and anti-inflammatory effects. Therefore FF may be useful as a complementary adjunct treatment of cancer, particularly included in anti-angiogenic protocols like those currently increasingly used in glioblastoma. There are sound reasons to initiate well planned phase II clinical trials for FF as a complementary adjunct treatment of cancer.
Collapse
|
41
|
Balakumar P, Varatharajan R, Nyo YH, Renushia R, Raaginey D, Oh AN, Akhtar SS, Rupeshkumar M, Sundram K, Dhanaraj SA. Fenofibrate and dipyridamole treatments in low-doses either alone or in combination blunted the development of nephropathy in diabetic rats. Pharmacol Res 2014; 90:36-47. [PMID: 25263930 DOI: 10.1016/j.phrs.2014.08.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 08/20/2014] [Accepted: 08/26/2014] [Indexed: 12/19/2022]
Abstract
Low-doses of fenofibrate and dipyridamole have pleiotropic renoprotective actions in diabetic rats. This study investigated their combined effect relative to their individual treatments and lisinopril in rats with diabetic nephropathy. Streptozotocin (55mg/kg, i.p., once)-administered diabetic rats were allowed for 10 weeks to develop nephropathy. Diabetic rats after 10 weeks developed nephropathy with discernible renal structural and functional changes as assessed in terms of increase in kidney weight to body weight ratio (KW/BW), and elevations of serum creatinine, urea and uric acid, which accompanied with elevated serum triglycerides and decreased high-density lipoproteins. Hematoxylin-eosin, periodic acid Schiff and Masson trichrome staining confirmed renal pathological changes in diabetic rats that included glomerular capsular wall distortion, mesangial cell expansion, glomerular microvascular condensation, tubular damage and degeneration and fibrosis. Low-dose fenofibrate (30mg/kg, p.o., 4 weeks) and low-dose dipyridamole (20mg/kg, p.o., 4 weeks) treatment either alone or in combination considerably reduced renal structural and functional abnormalities in diabetic rats, but without affecting the elevated glucose level. Fenofibrate, but not dipyridamole, significantly prevented the lipid alteration and importantly the uric acid elevation in diabetic rats. Lisinopril (5mg/kg, p.o., 4 weeks, reference compound), prevented the hyperglycemia, lipid alteration and development of diabetic nephropathy. Lipid alteration and uric acid elevation, besides hyperglycemia, could play key roles in the development of nephropathy. Low-doses of fenofibrate and dipyridamole treatment either alone or in combination markedly prevented the diabetes-induced nephropathy. Their combination was as effective as to their individual treatment, but not superior in preventing the development of diabetic nephropathy.
Collapse
Affiliation(s)
- Pitchai Balakumar
- Pharmacology Unit, Faculty of Pharmacy, AIMST University, Semeling, 08100 Bedong, Malaysia.
| | - Rajavel Varatharajan
- Pharmacology Unit, Faculty of Pharmacy, AIMST University, Semeling, 08100 Bedong, Malaysia
| | - Ying Hui Nyo
- Pharmacology Unit, Faculty of Pharmacy, AIMST University, Semeling, 08100 Bedong, Malaysia
| | - Raja Renushia
- Pharmacology Unit, Faculty of Pharmacy, AIMST University, Semeling, 08100 Bedong, Malaysia
| | - Devarajan Raaginey
- Pharmacology Unit, Faculty of Pharmacy, AIMST University, Semeling, 08100 Bedong, Malaysia
| | - Ann Nah Oh
- Pharmacology Unit, Faculty of Pharmacy, AIMST University, Semeling, 08100 Bedong, Malaysia
| | - Shaikh Sohrab Akhtar
- Pharmacology Unit, Faculty of Pharmacy, AIMST University, Semeling, 08100 Bedong, Malaysia
| | - Mani Rupeshkumar
- Pharmacology Unit, Faculty of Pharmacy, AIMST University, Semeling, 08100 Bedong, Malaysia
| | - Karupiah Sundram
- Pharmaceutical Chemistry Unit, Faculty of Pharmacy, AIMST University, Semeling, 08100 Bedong, Malaysia
| | - Sokkalingam A Dhanaraj
- Pharmaceutical Technology Unit, Faculty of Pharmacy, AIMST University, Semeling, 08100 Bedong, Malaysia
| |
Collapse
|
42
|
Effects of CP-900691, a novel peroxisome proliferator-activated receptor α, agonist on diabetic nephropathy in the BTBR ob/ob mouse. J Transl Med 2014; 94:851-62. [PMID: 24955894 PMCID: PMC4404155 DOI: 10.1038/labinvest.2014.80] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 05/02/2014] [Accepted: 05/20/2014] [Indexed: 12/14/2022] Open
Abstract
Piperidine-based peroxisome proliferator-activated receptor-α agonists are agents that are efficacious in improving lipid, glycemic, and inflammatory indicators in diabetes and obesity. This study sought to determine whether CP-900691 ((S)-3-[3-(1-carboxy-1-methyl-ethoxy)-phenyl]-piperidine-1-carboxylic acid 4-trifluoromethyl-benzyl ester; CP), a member of this novel class of agents, by decreasing plasma triglycerides, could prevent diabetic nephropathy in the Black and Tan, BRachyuric (BTBR) ob/ob mouse model of type 2 diabetes mellitus. Four-week old female BTBR WT and BTBR ob/ob mice received either regular chow or one containing CP (3 mg/kg per day) for 14 weeks. CP elevated plasma high-density lipoprotein, albuminuria, and urinary excretion of 8-epi PGF(2α), a product of the nonenzymatic metabolism of arachidonic acid and whose production is elevated in oxidative stress, in BTBR WT mice. In BTBR ob/ob mice, CP reduced plasma triglycerides and non-esterified fatty acids, fasting blood glucose, body weight, and plasma interleukin-6, while concomitantly improving insulin resistance. Despite these beneficial metabolic effects, CP had no effect on elevated plasma insulin, 8-epi PGF(2α) excretion, and albuminuria, and surprisingly, did not ameliorate the development of diabetic nephropathy, having no effect on the accumulation of renal macrophages, glomerular hypertrophy, and increased mesangial matrix expansion. In addition, CP did not increase plasma high-density lipoprotein in BTBR ob/ob mice, while paradoxically increasing total cholesterol levels. These findings indicate that 8-epi PGF(2α), possibly along with hyperinsulinemia and inflammatory and dysfunctional lipoproteins, is integral to the development of diabetic nephropathy and should be considered as a potential target of therapy in the treatment of diabetic nephropathy.
Collapse
|
43
|
Peng T, Wang J, Zhen J, Hu Z, Yang X. Effect of benazepril on the transdifferentiation of renal tubular epithelial cells from diabetic rats. Biomed Rep 2014; 2:490-494. [PMID: 24944793 DOI: 10.3892/br.2014.277] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Accepted: 03/11/2014] [Indexed: 01/09/2023] Open
Abstract
The aim of this study was to investigate the effect of benazepril on the transdifferentiation of renal tubular epithelial cells from diabetic rats. Thirty male Sprague-Dawley rats were included in the present study. Eight of the 30 rats were randomly selected and served as the normal control group (N group), while the remaining 22 rats, injected with streptozotocin (STZ), comprised the diabetic rat model. Rats with diabetes were randomly divided into the diabetic (DM group) and benazepril (B group) groups. The total course was conducted over 12 weeks. Blood glucose, body weight, kidney/body weight, 24-h urinary protein, serum creatinine and blood urea nitrogen were measured at the start and end of the study. We observed the tubulointerstitial pathological changes, and applied immunohistochemistry and western blotting to detect the expression of α-smooth muscle actin (α-SMA) in renal tissue. The levels of blood glucose, kidney/body weight, 24-h urinary protein, serum creatinine, blood urea nitrogen and tubulointerstitial damage index (TII) in the DM group were significantly higher than that in the N group (p<0.01). Except for blood glucose and kidney/body weight, the remaining indices were lower in the B group compared with those in the DM group (p<0.01). Immunohistochemical staining results revealed the expression of α-SMA in renal tubular epithelial cells to be significantly higher in the DM and B groups compared with the control (N) group (p<0.01). Western blot analysis revealed that the expression of α-SMA in diabetic renal tissue increased 3.27-fold compared with that of the N group, while the expression of α-SMA in the B group decreased 45% compared with that in the DM group. In conclusion, benazepril significantly reduced the expression of α-SMA in renal tubular epithelial cells obtained from diabetic rats, inhibited the transdifferentiation of renal tubular epithelial cells and played an important role in kidney protection.
Collapse
Affiliation(s)
- Tao Peng
- Department of Nephrology, Shandong University, Qilu Hospital, Jinan, Shandong 250012, P.R. China
| | - Jie Wang
- Department of Nephrology, Shandong University, Qilu Hospital, Jinan, Shandong 250012, P.R. China
| | - Junhui Zhen
- Department of Pathology, Shandong University, Qilu Hospital, Jinan, Shandong 250012, P.R. China
| | - Zhao Hu
- Department of Nephrology, Shandong University, Qilu Hospital, Jinan, Shandong 250012, P.R. China
| | - Xiangdong Yang
- Department of Nephrology, Shandong University, Qilu Hospital, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
44
|
Nie Y, Li S, Yi Y, Su W, Chai X, Jia D, Wang Q. Effects of astragalus injection on the TGFβ/Smad pathway in the kidney in type 2 diabetic mice. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 14:148. [PMID: 24885228 PMCID: PMC4023174 DOI: 10.1186/1472-6882-14-148] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Accepted: 04/29/2014] [Indexed: 11/13/2022]
Abstract
Background In traditional Chinese medicine, astragalus injection is used to treat diabetic nephropathy (DN). The current study was conducted to determine the effects of astragalus injection on DN by assessing potential modulation of the transforming growth factor beta TGFβ/Smad signaling pathway. Methods Diabetic, male KKAy mice, aged 14 weeks were randomly divided into a model group and an astragalus treatment group, while age-matched male C57BL/6J mice were selected as controls. The treatment group received daily intraperitoneal injections of astragalus (0.03 ml/10 g.d), while the model group received injections of an equivalent volume of saline. Mice were euthanized after 24 weeks. Serum samples were obtained from animals in each group, and blood glucose, creatinine, and urea nitrogen levels were measured. Tissue samples from the kidney were used for morphometric studies. The expression of TGFβ1, TGFβR-Ι, Smad3, and Smad7 were evaluated using reverse transcription-polymerase chain reaction (RT-PCR), and western blot analysis. Results Mice in the model group became obese, and suffered complications, including hyperglycemia, polyuria, and proteinuria. Astragalus treatment significantly reduced albuminuria, improved renal function, and ameliorated changes in renal histopathology. Moreover, administration of astragalus injection increased Smad7 expression, and inhibited the expression of TGFβR-Ι, Smad3 and its phosphorylation, and decreased the mRNA level of TGFβ1. Conclusions The TGFβ/Smad signaling pathway plays an important role in the development of DN. Administration of astragalus injection could prevent or mitigate DN by rebalancing TGFβ/Smad signaling, and could play a protective role in DN-induced renal damage in KKAy mice.
Collapse
|
45
|
Zhao X, Zhang Y, Leander M, Li L, Wang G, Emmett N. Altered expression profile of renal α(1D)-adrenergic receptor in diabetes and its modulation by PPAR agonists. J Diabetes Res 2014; 2014:725634. [PMID: 24772448 PMCID: PMC3977090 DOI: 10.1155/2014/725634] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 02/14/2014] [Accepted: 02/17/2014] [Indexed: 01/05/2023] Open
Abstract
Alpha(1D)-adrenergic receptor (α(1D)-AR) plays important roles in regulating physiological and pathological responses mediated by catecholamines, particularly in the cardiovascular and urinary systems. The present study was designed to investigate the expression profile of α(1D)-AR in the diabetic kidneys and its modulation by activation of peroxisome proliferator-activated receptors (PPARs). 12-week-old Zucker lean (ZL) and Zucker diabetic fatty (ZD) rats were treated with fenofibrate or rosiglitazone for 8-10 weeks. Gene microarray, real-time PCR, and confocal immunofluorescence microscopy were performed to assess mRNA and protein expression of α(1D)-AR in rat kidney tissue. Using microarray, we found that α(1D)-AR gene was dramatically upregulated in 22-week-old ZD rats compared to ZL controls. Quantitative PCR analysis verified a 16-fold increase in α(1D)-AR mRNA in renal cortex from ZD animals compared to normal controls. Chronic treatment with fenofibrate or rosiglitazone reduced renal cortical α(1D)-AR gene. Immunofluorescence staining confirmed that α(1D)-AR protein was induced in the glomeruli and tubules of diabetic rats. Moreover, dual immunostaining for α(1D)-AR and kidney injury molecule-1 indicated that α(1D)-AR was expressed in dedifferentiated proximal tubules of diabetic Zucker rats. Taken together, our results show that α(1D)-AR expression is upregulated in the diabetic kidneys. PPAR activation suppressed renal expression of α(1D)-AR in diabetic nephropathy.
Collapse
MESH Headings
- Animals
- Cell Dedifferentiation/drug effects
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Diabetic Nephropathies/prevention & control
- Fenofibrate/therapeutic use
- Gene Expression Profiling
- Hypoglycemic Agents/therapeutic use
- Kidney/drug effects
- Kidney/metabolism
- Kidney/pathology
- Kidney Glomerulus/drug effects
- Kidney Glomerulus/metabolism
- Kidney Glomerulus/pathology
- Kidney Tubules/drug effects
- Kidney Tubules/metabolism
- Kidney Tubules/pathology
- Male
- Obesity/complications
- PPAR alpha/agonists
- PPAR alpha/metabolism
- PPAR gamma/agonists
- PPAR gamma/metabolism
- Peroxisome Proliferator-Activated Receptors/agonists
- Peroxisome Proliferator-Activated Receptors/genetics
- Peroxisome Proliferator-Activated Receptors/metabolism
- Rats
- Rats, Zucker
- Receptors, Adrenergic, alpha-1/chemistry
- Receptors, Adrenergic, alpha-1/genetics
- Receptors, Adrenergic, alpha-1/metabolism
- Rosiglitazone
- Thiazolidinediones/therapeutic use
- Up-Regulation/drug effects
Collapse
Affiliation(s)
- Xueying Zhao
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Yuanyuan Zhang
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Michelle Leander
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Lingyun Li
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Guoshen Wang
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Nerimiah Emmett
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| |
Collapse
|
46
|
Hu B, Wu Z, Hergert P, Henke CA, Bitterman PB, Phan SH. Regulation of myofibroblast differentiation by poly(ADP-ribose) polymerase 1. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 182:71-83. [PMID: 23260200 DOI: 10.1016/j.ajpath.2012.09.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Revised: 08/21/2012] [Accepted: 09/13/2012] [Indexed: 02/08/2023]
Abstract
Poly(ADP-ribosyl)ation (PARylation) is a post-translational protein modification effected by enzymes belonging to the poly(ADP-ribose) polymerase (PARP) superfamily, mainly by PARP-1. The key acceptors of poly(ADP-ribose) include PARP-1 itself, histones, DNA repair proteins, and transcription factors. Because many of these factors are involved in the regulation of myofibroblast differentiation, we examined the role of PARylation on myofibroblast differentiation. Overexpression of PARP-1 with an expression plasmid activated expression of the α-SMA gene (Acta2), a marker of myofibroblast differentiation in lung fibroblasts. Suppression of PARP-1 activity or gene expression with PARP-1 inhibitors or siRNA, respectively, had the opposite effect on these cells. PARP-1-deficient cells also had reduced α-SMA gene expression. DNA pyrosequencing identified hypermethylated regions of the α-SMA gene in PARP-1-deficient cells, relative to wild-type cells. Interestingly, and of potential relevance to human idiopathic pulmonary fibrosis, PARP activity in lung fibroblasts isolated from idiopathic pulmonary fibrosis patients was significantly higher than that in cells isolated from control subjects. Furthermore, PARP-1-deficient mice exhibited reduced pulmonary fibrosis in response to bleomycin-induced lung injury, relative to wild-type controls. These results suggest that PARylation is important for myofibroblast differentiation and the pathogenesis of pulmonary fibrosis.
Collapse
Affiliation(s)
- Biao Hu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109-2200, USA
| | | | | | | | | | | |
Collapse
|
47
|
Wang W, Lin Q, Lin R, Zhang J, Ren F, Zhang J, Ji M, Li Y. PPARα agonist fenofibrate attenuates TNF-α-induced CD40 expression in 3T3-L1 adipocytes via the SIRT1-dependent signaling pathway. Exp Cell Res 2013; 319:1523-33. [DOI: 10.1016/j.yexcr.2013.04.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Revised: 03/14/2013] [Accepted: 04/09/2013] [Indexed: 11/30/2022]
|
48
|
Kostapanos MS, Florentin M, Elisaf MS. Fenofibrate and the kidney: an overview. Eur J Clin Invest 2013; 43:522-531. [PMID: 23480615 DOI: 10.1111/eci.12068] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Accepted: 02/11/2013] [Indexed: 12/18/2022]
Abstract
BACKGROUND Fenofibrate has been used for the management of atherogenic dyslipidaemia for many years. Reports of fenofibrate-associated increases in serum creatinine (SCr) levels raised concerns regarding deleterious effects on renal function. DESIGN In this narrative review, we discuss available literature on the effect of fenofibrate on the kidney. RESULTS Most clinical studies showed a rapid (within weeks) raising effect of fenofibrate on SCr levels. This was often accompanied by declined estimated glomerular filtration rate. Risk predictors of this adverse effect might include increased age, impaired renal function and high-dose treatment. Also, the concomitant use of medications affecting renal hemodynamics (e.g. angiotensin-converting enzyme-inhibitors (ACEi) and angiotensin receptor blockers) may predispose to fenofibrate-associated increased SCr levels. Interestingly, SCr increases by fenofibrate were transient and reversible even without treatment discontinuation. Furthermore, fenofibrate was associated with a slower progression of renal function impairment and albuminuria in a long-term basis. Also, fenofibrate might be protective against pathological changes in diabetic nephropathy and hypertensive glomerulosclerosis. In this context, it is uncertain whether fenofibrate-associated increase in SCr levels mirrors true renal function deterioration. Several theories have been expressed. The most dominant one involved the inhibition of renal vasodilatory prostaglandins reducing renal plasma flow and glomerular pressure. Increased creatinine secretion or reduced creatinine clearance by fenofibrate was also suggested. These hypotheses should be settled by further studies. CONCLUSIONS Fenofibrate may not be a nephrotoxic drug. However, a close monitoring of SCr levels is relevant especially in high-risk patients. Increases in SCr levels ≥30% can impose treatment discontinuation.
Collapse
Affiliation(s)
- Michael S Kostapanos
- Department of Internal Medicine, Medical School, University of Ioannina, Ioannina, Greece
| | | | | |
Collapse
|
49
|
Kim JE, Lee MH, Nam DH, Song HK, Kang YS, Lee JE, Kim HW, Cha JJ, Hyun YY, Han SY, Han KH, Han JY, Cha DR. Celastrol, an NF-κB inhibitor, improves insulin resistance and attenuates renal injury in db/db mice. PLoS One 2013; 8:e62068. [PMID: 23637966 PMCID: PMC3637455 DOI: 10.1371/journal.pone.0062068] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 03/18/2013] [Indexed: 12/12/2022] Open
Abstract
The NF-κB pathway plays an important role in chronic inflammatory and autoimmune diseases. Recently, NF-κB has also been suggested as an important mechanism linking obesity, inflammation, and metabolic disorders. However, there is no current evidence regarding the mechanism of action of NF-κB inhibition in insulin resistance and diabetic nephropathy in type 2 diabetic animal models. We investigated the effects of the NF-κB inhibitor celastrol in db/db mice. The treatment with celastrol for 2 months significantly lowered fasting plasma glucose (FPG), HbA1C and homeostasis model assessment index (HOMA-IR) levels. Celastrol also exhibited significant decreases in body weight, kidney/body weight and adiposity. Celastrol reduced insulin resistance and lipid abnormalities and led to higher plasma adiponectin levels. Celastrol treatment also significantly mitigated lipid accumulation and oxidative stress in organs including the kidney, liver and adipose tissue. The treated group also exhibited significantly lower creatinine levels and urinary albumin excretion was markedly reduced. Celastrol treatment significantly lowered mesangial expansion and suppressed type IV collagen, PAI-1 and TGFβ1 expressions in renal tissues. Celastrol also improved abnormal lipid metabolism, oxidative stress and proinflammatory cytokine activity in the kidney. In cultured podocytes, celastrol treatment abolished saturated fatty acid-induced proinflammatory cytokine synthesis. Taken together, celastrol treatment not only improved insulin resistance, glycemic control and oxidative stress, but also improved renal functional and structural changes through both metabolic and anti-inflammatory effects in the kidney. These results suggest that targeted therapy for NF-κB may be a useful new therapeutic approach for the management of type II diabetes and diabetic nephropathy.
Collapse
Affiliation(s)
- Jung Eun Kim
- Department of Internal Medicine, Division of Nephrology, Korea University, Ansan City, Kyungki-Do, Korea
| | - Mi Hwa Lee
- Department of Internal Medicine, Division of Nephrology, Korea University, Ansan City, Kyungki-Do, Korea
| | - Deok Hwa Nam
- Department of Internal Medicine, Division of Nephrology, Korea University, Ansan City, Kyungki-Do, Korea
| | - Hye Kyoung Song
- Department of Internal Medicine, Division of Nephrology, Korea University, Ansan City, Kyungki-Do, Korea
| | - Young Sun Kang
- Department of Internal Medicine, Division of Nephrology, Korea University, Ansan City, Kyungki-Do, Korea
| | - Ji Eun Lee
- Department of Internal Medicine, Division of Nephrology, Wonkwang University, Gunpo City, Kyungki-Do, Korea
| | - Hyun Wook Kim
- Department of Internal Medicine, Division of Nephrology, Wonkwang University, Gunpo City, Kyungki-Do, Korea
| | - Jin Joo Cha
- Department of Internal Medicine, Division of Nephrology, Korea University, Ansan City, Kyungki-Do, Korea
| | - Young Youl Hyun
- Department of Internal Medicine, Division of Nephrology, Sungkyunkwan University, Seoul, Korea
| | - Sang Youb Han
- Department of Internal Medicine, Division of Nephrology, Inje University, Goyang City, Kyungki-Do, Korea
| | - Kum Hyun Han
- Department of Internal Medicine, Division of Nephrology, Inje University, Goyang City, Kyungki-Do, Korea
| | - Jee Young Han
- Department of Pathology, Inha University, Incheon City, Kyungki-Do, Korea
| | - Dae Ryong Cha
- Department of Internal Medicine, Division of Nephrology, Korea University, Ansan City, Kyungki-Do, Korea
| |
Collapse
|
50
|
Bénardeau A, Verry P, Atzpodien EA, Funk JM, Meyer M, Mizrahi J, Winter M, Wright MB, Uhles S, Sebokova E. Effects of the dual PPAR-α/γ agonist aleglitazar on glycaemic control and organ protection in the Zucker diabetic fatty rat. Diabetes Obes Metab 2013; 15:164-74. [PMID: 22958363 DOI: 10.1111/dom.12006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 06/12/2012] [Accepted: 09/02/2012] [Indexed: 12/27/2022]
Abstract
AIMS To evaluate the effects of aleglitazar, a dual peroxisome proliferator-activated receptor-α/γ agonist, on the development of diabetes-related organ dysfunction, in relation to glycaemic and lipid changes, in Zucker diabetic fatty (ZDF) rats. METHODS Six-week-old, male ZDF rats received aleglitazar 0.3 mg/kg/day or vehicle as food admix for 13 weeks (n = 10 per group). Age-matched male Zucker lean rats served as non-diabetic controls. Plasma and renal markers were measured at several time points. Histopathology and quantitative immunohistochemistry were performed at 13 weeks. RESULTS Glycated haemoglobin (5.4 vs. 9.2%) and blood glucose (8.3 ± 0.3 vs. 26.1 ± 1.0 mmol/l) were significantly reduced at 12 weeks with aleglitazar versus vehicle-treated ZDF rats (both p < 0.01), while aleglitazar preserved near-normal plasma insulin levels. Aleglitazar prevented the development of hypertriglyceridaemia (1.4 ± 0.1 vs. 8.5 ± 0.9 mmol/l) and reduced plasma non-esterified fatty acids (0.09 ± 0.02 vs. 0.26 ± 0.04 mmol/l) relative to vehicle-treated animals (both p < 0.01). Urinary glucose and protein concentrations were significantly reduced at 13 weeks with aleglitazar versus vehicle-treated rats (both p < 0.01). Consistent with its effect on glycaemic control, aleglitazar protected β-cell morphology, as evidenced by preservation of islet integrity, and reduction of β-cell apoptosis and islet fibrosis. Aleglitazar prevented renal glomerular hypertrophy, podocyte degeneration, glomerulosclerosis, tubulo-interstitial lesions and development of cataracts. CONCLUSIONS Aleglitazar strongly improved glycaemic and lipid parameters while protecting key tissues, including the pancreas, kidneys and eyes, against diabetes-associated structural and functional changes in the ZDF rat.
Collapse
Affiliation(s)
- A Bénardeau
- pRED, Pharma Research & Early Development, DTA Cardiovascular & Metabolism, F. Hoffmann-La Roche AG, Basel, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|