1
|
Ley-Ngardigal S, Claverol S, Sobilo L, Moreau M, Hubert C, Goupil J, Poulignon A, Mahfouf W, Fatrouni H, Dard L, Juan M, Gales L, Merched A, Tokarski C, Leblanc E, Galinier A, Lacombe D, Rezvani HR, Bellvert F, Pays K, Nizard C, Amoedo ND, Bulteau AL, Rossignol R. Repression of oxidative phosphorylation by NR2F2, MTERF3 and GDF15 in human skin under high-glucose stress. Redox Biol 2025; 82:103613. [PMID: 40174478 PMCID: PMC11999475 DOI: 10.1016/j.redox.2025.103613] [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: 01/18/2025] [Revised: 03/12/2025] [Accepted: 03/24/2025] [Indexed: 04/04/2025] Open
Abstract
Lifestyle factors such as a Western diet or metabolic diseases like diabetes disrupt glucose homeostasis and induce stress responses, yet their impact on skin metabolism and structural integrity remains poorly understood. Here, we performed multiomic and bioenergetic analyses of human dermal fibroblasts (HDFs), human equivalent dermis (HED), human reconstructed skin (HRS), and skin explants from diabetic patients. We found that 12 mM glucose stress represses oxidative phosphorylation (OXPHOS) through a dual mechanism: the glucose-dependent nuclear receptor NR2F2 activates mitochondrial transcription termination factor 3 (MTERF3) while inhibiting growth-differentiation factor 15 (GDF15). Promoter assays revealed that MTERF3 is regulated by NR2F2 and MYCN, whereas GDF15 is modulated by NR2F2 and FOS. Consequently, OXPHOS proteins and mitochondrial respiration were suppressed, and MTERF3 overexpression additionally interfered with collagen biosynthesis. In contrast, GDF15 supplementation fully rescued hyperglycemia-induced bioenergetic and metabolomic alterations, suggesting a pharmacological strategy to mitigate hyperglycemic damage in the skin. Finally, silencing GDF15 or TFAM impaired fibroblast haptotaxis and skin reconstruction, underscoring the crucial role of mitochondrial energetics in dermal structure and function. Collectively, these findings identify the NR2F2-MTERF3-GDF15 axis as a key mediator of OXPHOS suppression and highlight a potential therapeutic target to preserve skin integrity under hyperglycemic stress.
Collapse
Affiliation(s)
- S Ley-Ngardigal
- INSERM U1211, 33076, Bordeaux, France; Bordeaux University, 146 rue Léo Saignat, 33076, Bordeaux, France; LVMH Recherche, Saint-Jean-de-Braye, France
| | - S Claverol
- Bordeaux University, 146 rue Léo Saignat, 33076, Bordeaux, France; Univ. Bordeaux, Bordeaux Proteome, Bordeaux, France
| | - L Sobilo
- LVMH Recherche, Saint-Jean-de-Braye, France
| | - M Moreau
- LVMH Recherche, Saint-Jean-de-Braye, France
| | - C Hubert
- INSERM U1211, 33076, Bordeaux, France; Bordeaux University, 146 rue Léo Saignat, 33076, Bordeaux, France
| | - J Goupil
- INSERM U1211, 33076, Bordeaux, France; Bordeaux University, 146 rue Léo Saignat, 33076, Bordeaux, France; CELLOMET, ADERA, 146 rue Léo Saignat, 33076, Bordeaux, France
| | - A Poulignon
- INSERM U1211, 33076, Bordeaux, France; Bordeaux University, 146 rue Léo Saignat, 33076, Bordeaux, France
| | - W Mahfouf
- Bordeaux University, 146 rue Léo Saignat, 33076, Bordeaux, France; INSERM UMR 1312, Bordeaux Institute of Oncology (BRIC), Bordeaux, France
| | - H Fatrouni
- Bordeaux University, 146 rue Léo Saignat, 33076, Bordeaux, France; INSERM UMR 1312, Bordeaux Institute of Oncology (BRIC), Bordeaux, France
| | - L Dard
- INSERM U1211, 33076, Bordeaux, France; Bordeaux University, 146 rue Léo Saignat, 33076, Bordeaux, France
| | - M Juan
- LVMH Recherche, Saint-Jean-de-Braye, France
| | - L Gales
- Metabolomics facility METATOUL, Toulouse, France
| | - A Merched
- Bordeaux University, 146 rue Léo Saignat, 33076, Bordeaux, France; INSERM UMR 1312, Bordeaux Institute of Oncology (BRIC), Bordeaux, France
| | - C Tokarski
- Bordeaux University, 146 rue Léo Saignat, 33076, Bordeaux, France; Univ. Bordeaux, Bordeaux Proteome, Bordeaux, France; Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, Pessac, F-33600, France
| | - E Leblanc
- LVMH Recherche, Saint-Jean-de-Braye, France
| | - A Galinier
- RESTORE, UMR 1301-Inserm 5070-CNRS EFS Univ. P. Sabatier, Toulouse, France
| | - D Lacombe
- INSERM U1211, 33076, Bordeaux, France; Bordeaux University, 146 rue Léo Saignat, 33076, Bordeaux, France; Medical Genetics Department, CHU Bordeaux, 33076, Bordeaux, France
| | - H R Rezvani
- Bordeaux University, 146 rue Léo Saignat, 33076, Bordeaux, France; INSERM UMR 1312, Bordeaux Institute of Oncology (BRIC), Bordeaux, France
| | - F Bellvert
- Metabolomics facility METATOUL, Toulouse, France
| | - K Pays
- LVMH Recherche, Saint-Jean-de-Braye, France
| | - C Nizard
- LVMH Recherche, Saint-Jean-de-Braye, France
| | - N Dias Amoedo
- INSERM U1211, 33076, Bordeaux, France; Bordeaux University, 146 rue Léo Saignat, 33076, Bordeaux, France; CELLOMET, ADERA, 146 rue Léo Saignat, 33076, Bordeaux, France
| | | | - R Rossignol
- INSERM U1211, 33076, Bordeaux, France; Bordeaux University, 146 rue Léo Saignat, 33076, Bordeaux, France; CELLOMET, ADERA, 146 rue Léo Saignat, 33076, Bordeaux, France.
| |
Collapse
|
2
|
Mobeen A, Joshi S, Fatima F, Bhargav A, Arif Y, Faruq M, Ramachandran S. NF-κB signaling is the major inflammatory pathway for inducing insulin resistance. 3 Biotech 2025; 15:47. [PMID: 39845928 PMCID: PMC11747027 DOI: 10.1007/s13205-024-04202-4] [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: 08/22/2024] [Accepted: 12/23/2024] [Indexed: 01/24/2025] Open
Abstract
Insulin resistance is major factor in the development of metabolic syndrome and type 2 diabetes (T2D). We extracted 430 genes from literature associated with both insulin resistance and inflammation. The highly significant pathways were Toll-like receptor signaling, PI3K-Akt signaling, cytokine-cytokine receptor interaction, pathways in cancer, TNF signaling, and NF-kappa B signaling. Among the 297 common genes in all datasets of various T2D patients' tissues including blood, muscle, liver, pancreas, and adipose tissues, 71% and 60% of these genes were differentially expressed in pancreas (GSE25724) and liver (GSE15653), respectively. A total of 169 genes contain highly conserved motifs for various transcription factors involved in immune response, thereby suggesting coordinated expression. Through co-expression analysis, we obtained three modules. The respective modules had 78, 158, and 55 genes, and TRAF2, HMGA1, and RGS5 as hub genes. Further, we used the BioNSi pathways simulation tool and identified the following five KEGG pathways perturbed in four or more tissues, namely Toll-like receptor signaling pathway, RIG-1-like receptor signaling pathway, pathways in cancer, NF-kappa B signaling pathway, and insulin resistance pathway. The genes NFKBIA and IKBKB are common to all these five pathways. In addition, using the NF-κB computational activation model, we identified that the reversal of NF-κB constitutive activation through overexpression of NFKB1 (P50 homodimer), PPARG, PIAS3 could reduce insulin resistance by almost half of its original value. To conclude, co-expression studies, gene expression network simulation, and NF-κB computational modeling substantiate the causal role of NF-κB pathway in insulin resistance. These results taken together with other published evidence suggests that the TNF-TRAF2-IKBKB-NF-κB axis could be explored as a potential target in combination with available metabolic targets in the management of insulin resistance. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-024-04202-4.
Collapse
Affiliation(s)
- Ahmed Mobeen
- CSIR Institute of Genomics & Integrative Biology, Sukhdev Vihar, New Delhi, 110025 India
| | - Sweta Joshi
- Department of Food Technology, SIST, Jamia Hamdard, New Delhi, 110062 India
| | - Firdaus Fatima
- CSIR Institute of Genomics & Integrative Biology, Sukhdev Vihar, New Delhi, 110025 India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002 India
| | - Anasuya Bhargav
- CSIR Institute of Genomics & Integrative Biology, Sukhdev Vihar, New Delhi, 110025 India
| | - Yusra Arif
- Centre of Bioinformatics, Institute of Inter Disciplinary Studies, Allahabad University, Allahabad, Uttar Pradesh 211002 India
| | - Mohammed Faruq
- CSIR Institute of Genomics & Integrative Biology, Sukhdev Vihar, New Delhi, 110025 India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002 India
| | - Srinivasan Ramachandran
- CSIR Institute of Genomics & Integrative Biology, Sukhdev Vihar, New Delhi, 110025 India
- Manav Rachna International Institute of Research and Studies, Sector 43, Delhi–Surajkund Road, Faridabad, Haryana 121004 India
| |
Collapse
|
3
|
Liu W, Gui R, Li Y, Li M, Lei Z, Jin Y, Yu Y, Li Y, Qian L, Xiong Y. Linarin Identified as a Bioactive Compound of Lycii Cortex Ameliorates Insulin Resistance and Inflammation Through the c-FOS/ARG2 Signaling Axis. Phytother Res 2025; 39:246-263. [PMID: 39523692 DOI: 10.1002/ptr.8370] [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: 01/15/2024] [Revised: 08/19/2024] [Accepted: 09/21/2024] [Indexed: 11/16/2024]
Abstract
Insulin resistance (IR) is a central pathophysiological process underlying numerous chronic metabolic disorders, including type 2 diabetes and obesity. Lycii Cortex, a widely used traditional Chinese herb, has demonstrated potential benefits in preventing and managing diabetes and IR. Whereas, the specific bioactive compounds responsible for these protective effects and their underlying mechanisms of action remain elusive. This study aimed to identify the bioactive components within Lycii Cortex that contribute to its anti-diabetic effects and to elucidate the molecular mechanisms underlying its beneficial actions on insulin resistance. Network pharmacology and molecular docking analyses were employed to identify the potential active compounds in Lycii Cortex and their corresponding target proteins. An in vitro model of IR was established using palmitic acid (PA)-treated HepG2 cells. Cell viability was assessed using the CCK-8 assay, while glucose uptake was evaluated by 2-NBDG staining and extracellular glucose measurement. To validate the in vitro findings, an in vivo model of obesity-induced IR was established using high-fat diet (HFD)-fed mice. The network pharmacology analysis preliminarily identified 13 candidate chemicals and 10 hub LyC and IR-related genes (LIRRGs). Molecular docking analysis demonstrates that Linarin as the potential active component exhibits the greatest potential to target c-FOS for preventing obesity-induced IR. Enrichment analysis suggested that Linarin-targeted pathways are correlated with inflammation. In vitro experimental validation demonstrated that Linarin was capable of protecting against PA-induced IR in HepG2 cells evidenced by improving glucose uptake ability and reducing extracellular glucose content. Additionally, we found that Linarin ablated PA-induced increase in the expression of c-FOS and inflammatory cytokines. Furthermore, in PA-treated cells, silencing c-FOS markedly improved glucose consumption, and reduced inflammation and Arginase 2 (ARG2) expression. Similarly, as exposure to PA, silencing ARG2 also ameliorated glucose uptake and inflammation, while not affecting c-FOS expression. In vivo experiments further showed that Linarin administration remarkably improved glucose tolerance and insulin sensitivity, and reduced the fat mass and body weight in HFD-induced obese mice. In this study, Linarin has been identified as the bioactive compound of Lycii Cortex to ameliorate obesity-related IR and inflammation through the c-FOS/ARG2 signaling cascade. These findings underscore the therapeutic potential of Linarin and provide valuable insights into developing novel intervention strategies for type 2 diabetes therapy.
Collapse
Affiliation(s)
- Wenxuan Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, People's Republic of China
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No. 3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, Shaanxi, People's Republic of China
| | - Runlin Gui
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, People's Republic of China
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No. 3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, Shaanxi, People's Republic of China
| | - Yang Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, People's Republic of China
| | - Man Li
- Department of Endocrinology, Xi' an No. 3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, Shaanxi, People's Republic of China
| | - Zhen Lei
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, People's Republic of China
| | - Yanyan Jin
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, People's Republic of China
| | - Yi Yu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, People's Republic of China
| | - Yujia Li
- Department of Traditional Chinese Medicine, Xi' an No. 3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, Shaanxi, People's Republic of China
| | - Lu Qian
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, People's Republic of China
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No. 3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, Shaanxi, People's Republic of China
- Xi'an Mental Health Center, Xi'an, Shaanxi, People's Republic of China
| | - Yuyan Xiong
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, People's Republic of China
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No. 3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, Shaanxi, People's Republic of China
| |
Collapse
|
4
|
He Z, Xu Y, Ma Q, Zhou C, Yang L, Lin M, Deng P, Yang Z, Gong M, Zhang H, Lu M, Li Y, Gao P, Lu Y, He M, Zhang L, Pi H, Zhang K, Qin S, Yu Z, Zhou Z, Chen C. SOX2 modulated astrocytic process plasticity is involved in arsenic-induced metabolic disorders. JOURNAL OF HAZARDOUS MATERIALS 2022; 435:128942. [PMID: 35468398 DOI: 10.1016/j.jhazmat.2022.128942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/02/2022] [Accepted: 04/13/2022] [Indexed: 06/14/2023]
Abstract
Metabolic disorders induced by arsenic exposure have attracted great public concern. However, it remains unclear whether hypothalamus-based central regulation mechanisms are involved in this process. Here, we exposed mice to 100 μg/L arsenic in drinking water and established a chronic arsenic exposure model. Our study revealed that chronic arsenic exposure caused metabolic disorders in mice including impaired glucose metabolism and decreased energy expenditure. Arsenic exposure also impaired glucose sensing and the activation of proopiomelanocortin (POMC) neurons in the hypothalamus. In particular, arsenic exposure damaged the plasticity of hypothalamic astrocytic process. Further research revealed that arsenic exposure inhibited the expression of sex-determining region Y-Box 2 (SOX2), which decreased the expression level of insulin receptors (INSRs) and the phosphorylation of AKT. The conditional deletion of astrocytic SOX2 exacerbated arsenic-induced effects on metabolic disorders, the impairment of hypothalamic astrocytic processes, and the inhibition of INSR/AKT signaling. Furthermore, the arsenic-induced impairment of astrocytic processes and inhibitory effects on INSR/AKT signaling were reversed by SOX2 overexpression in primary hypothalamic astrocytes. Together, we demonstrated here that chronic arsenic exposure caused metabolic disorders by impairing SOX2-modulated hypothalamic astrocytic process plasticity in mice. Our study provides evidence of novel central regulatory mechanisms underlying arsenic-induced metabolic disorders and emphasizes the crucial role of SOX2 in regulating the process plasticity of adult astrocytes.
Collapse
Affiliation(s)
- Zhixin He
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Yudong Xu
- Department of Environmental Medicine, School of Public Health, and Department of Emergency Medicine, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Qinlong Ma
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Chao Zhou
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China; Army 953 Hospital, Shigatse Branch of Xinqiao Hospital, Third Military Medical University, Shigatse 857099, China
| | - Lingling Yang
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Min Lin
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Ping Deng
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Zhiqi Yang
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing 400038, China
| | - Mingyue Gong
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing 400038, China
| | - Huijie Zhang
- School of Medicine, Guangxi University, Nanning 530004, Guangxi Zhuang Autonomous Region, China
| | - Muxue Lu
- School of Medicine, Guangxi University, Nanning 530004, Guangxi Zhuang Autonomous Region, China
| | - Yanqi Li
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Peng Gao
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Yonghui Lu
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Mindi He
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Lei Zhang
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Huifeng Pi
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Kuan Zhang
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing 400038, China
| | - Song Qin
- Department of Anatomy, Histology and Embryology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Zhengping Yu
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Zhou Zhou
- Department of Environmental Medicine, School of Public Health, and Department of Emergency Medicine, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China; Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing 400030, China.
| | - Chunhai Chen
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
5
|
Torimoto K, Okuno K, Kuroda R, Shanas N, Cicalese SM, Eguchi K, Elliott KJ, Kawai T, Corbett CB, Peluzzo AM, St. Paul AK, Autieri MV, Scalia R, Rizzo V, Hashimoto T, Eguchi S. Glucose consumption of vascular cell types in culture: toward optimization of experimental conditions. Am J Physiol Cell Physiol 2022; 322:C73-C85. [PMID: 34817269 PMCID: PMC8791793 DOI: 10.1152/ajpcell.00257.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
In this study, we have looked for an optimum media glucose concentration and compared glucose consumption in three vascular cell types, endothelial cells (ECs), vascular smooth muscle cells (VSMCs), and adventitial fibroblasts (AFs) with or without angiotensin II (AngII) stimulation. In a subconfluent 6-well experiment in 1 mL DMEM with a standard low (100 mg/dL), a standard high (450 mg/dL), or a mixed middle (275 mg/dL) glucose concentration, steady and significant glucose consumption was observed in all cell types. After 48-h incubation, media that contained low glucose was reduced to almost 0 mg/dL, media that contained high glucose remained significantly higher at ∼275 mg/dL, and media that contained middle glucose remained closer to physiological range. AngII treatment enhanced glucose consumption in AFs and VSMCs but not in ECs. Enhanced extracellular acidification rate by AngII was also observed in AFs. In AFs, AngII induction of target proteins at 48 h varied depending on the glucose concentration used. In low glucose media, induction of glucose regulatory protein 78 or hexokinase II was highest, whereas induction of VCAM-1 was lowest. Utilization of specific inhibitors further suggests essential roles of angiotensin II type-1 receptor and glycolysis in AngII-induced fibroblast activation. Overall, this study demonstrates a high risk of hypo- or hyperglycemic conditions when standard low or high glucose media is used with vascular cells. Moreover, these conditions may significantly alter experimental outcomes. Media glucose concentration should be monitored during any culture experiments and utilization of middle glucose media is recommended for all vascular cell types.
Collapse
Affiliation(s)
- Keiichi Torimoto
- 1Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Keisuke Okuno
- 1Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Ryohei Kuroda
- 1Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - No’Ad Shanas
- 1Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Stephanie M. Cicalese
- 1Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Kunie Eguchi
- 1Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Katherine J. Elliott
- 1Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Tatsuo Kawai
- 1Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Cali B. Corbett
- 1Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Amanda M. Peluzzo
- 1Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Amanda K. St. Paul
- 1Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Michael V. Autieri
- 1Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Rosario Scalia
- 1Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Victor Rizzo
- 1Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Tomoki Hashimoto
- 2Barrow Aneurysm and AVM Research Center, Departments of Neurosurgery and Neurobiology, Barrow Neurological Institute, Phoenix, Arizona
| | - Satoru Eguchi
- 1Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
6
|
Kumano K, Shimoda M, Hyodo T, Sakai T. The Role of Tgf-β in Growth Inhibition of Peritoneal Mesothelial Cells in High-Glucose Dialysate. Perit Dial Int 2020. [DOI: 10.1177/089686089501507s13] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Kazuo Kumano
- Kidney Center, Kitasato University Hospital, Sagamihara, Kanagawa, Japan
| | - Masuhiro Shimoda
- Kidney Center, Kitasato University Hospital, Sagamihara, Kanagawa, Japan
| | - Toru Hyodo
- Kidney Center, Kitasato University Hospital, Sagamihara, Kanagawa, Japan
| | - Tadasu Sakai
- Kidney Center, Kitasato University Hospital, Sagamihara, Kanagawa, Japan
| |
Collapse
|
7
|
Kang DH, Hong YS, Lim HJ, Choi JH, Han DS, Yoon KI. High Glucose Solution and Spent Dialysate Stimulate the Synthesis of Transforming Growth Factor-β1of Human Peritoneal Mesothelial Cells: Effect of Cytokine Costimulation. Perit Dial Int 2020. [DOI: 10.1177/089686089901900307] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
ObjectiveTo investigate the effect of high glucose and spent peritoneal dialysate on the transforming growth factor-β1(TGFβ1) synthesis of cultured human peritoneal mesothelial cells (HPMCs) and to examine the effect of costimulation with high glucose or spent dialysate, and cytokines, interleukin-1β (IL-1β), and tumor necrosis factor-α (TNFα) on TGFβ1synthesis of HPMCs.DesignHPMCs were exposed to different concentrations of glucose (30, 60, and 90 mmol/L) or spent peritoneal dialysate for 48 hours in the absence or presence of IL-1β (1 ng/mL) and TNFα (1 ng/mL). TGFβ1mRNA expression was assessed by Northern blot analysis and TGFβ1protein release by Western blot analysis and enzymelinked immunosorbent assay (ELISA).ResultsExposure of HPMCs to high glucose conditions (30, 60, and 90 mmol/L of D-glucose) induced 2.3-, 3.6-, and 4.0-fold increases in TGFβ1mRNA expression of HPMC with enhanced TGFβ1protein synthesis and secretion into the media, whereas there were no significant changes in TGFβ1synthesis with equimolar concentrations of D-mannitol. Incubation with spent dialysate also significantly increased TGFβ1mRNA expression and protein secretion compared to control media ( p < 0.05). Stimulation with IL-1β (1 ng/mL) or TNFα (1 ng/mL) resulted in a significant increase in TGFβ1mRNA expression after 48 hours: 2.7 and 2.1 times the control level, respectively. However, TNFα-induced increase in TGFβ1mRNA expression was not translated into TGFβ1protein secretion, while IL-1β stimulation induced a significant increase in TGFβ1protein secretion as well as TGFβ1mRNA expression. Combined stimulation by high glucose or spent dialysate, together with IL-1β or TNFα, showed a greater increase in TGFβ1mRNA expression and protein secretion compared to stimulation by high glucose or spent dialysate alone.ConclusionOur results clearly show that high glucose solution and spent dialysate themselves might be sufficient to stimulate the production of TGFβ1by peritoneal mesothelial cells. In peritoneal dialysis patients, this state of chronic induction of TGFβ1is further exacerbated in the presence of peritonitis because of the stimulatory effect of proinflammatory cytokines, resulting in augmented TGFβ1synthesis, thus promoting peritoneal fibrosis.
Collapse
Affiliation(s)
- Duk-Hee Kang
- Department of Internal Medicine, Yonsei University, Seoul, Korea
- Medical Research Center, College of Medicine, Ewha Women's University, Seoul, Korea
| | | | - Hyun Joung Lim
- Medical Research Center, Yonsei University, Seoul, Korea
| | - Jin-Hee Choi
- Medical Research Center, Yonsei University, Seoul, Korea
| | - Dae-Suk Han
- Medical Research Center, Yonsei University, Seoul, Korea
| | - Kyun-Il Yoon
- Department of Internal Medicine, Yonsei University, Seoul, Korea
- Medical Research Center, College of Medicine, Ewha Women's University, Seoul, Korea
| |
Collapse
|
8
|
Gupta MK, Vadde R. Identification and characterization of differentially expressed genes in Type 2 Diabetes using in silico approach. Comput Biol Chem 2019; 79:24-35. [PMID: 30708140 DOI: 10.1016/j.compbiolchem.2019.01.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 12/26/2018] [Accepted: 01/23/2019] [Indexed: 12/14/2022]
Abstract
Diabetes mellitus is clinically characterized by hyperglycemia. Though many studies have been done to understand the mechanism of Type 2 Diabetes (T2D), however, the complete network of diabetes and its associated disorders through polygenic involvement is still under debate. The present study designed to re-analyze publicly available T2D related microarray raw datasets present in GEO database and T2D genes information present in GWAS catalog for screening out differentially expressed genes (DEGs) and identify key hub genes associated with T2D. T2D related microarray data downloaded from Gene Expression Omnibus (GEO) database and re-analysis performed with in house R packages scripts for background correction, normalization and identification of DEGs in T2D. Also retrieved T2D related DEGs information from GWAS catalog. Both DEGs lists were grouped after removal of overlapping genes. These screened DEGs were utilized further for identification and characterization of key hub genes in T2D and its associated diseases using STRING, WebGestalt and Panther databases. Computational analysis reveal that out of 99 identified key hub gene candidates from 348 DEGs, only four genes (CCL2, ELMO1, VEGFA and TCF7L2) along with FOS playing key role in causing T2D and its associated disorders, like nephropathy, neuropathy, rheumatoid arthritis and cancer via p53 or Wnt signaling pathways. MIR-29, and MAZ_Q6 are identified potential target microRNA and TF along with probable drugs alprostadil, collagenase and dinoprostone for the key hub gene candidates. The results suggest that identified key DEGs may play promising roles in prevention of diabetes.
Collapse
Affiliation(s)
- Manoj Kumar Gupta
- Department of Biotechnology & Bioinformatics, Yogi Vemana University, Kadapa 516003, Andhra Pradesh, India.
| | - Ramakrishna Vadde
- Department of Biotechnology & Bioinformatics, Yogi Vemana University, Kadapa 516003, Andhra Pradesh, India.
| |
Collapse
|
9
|
Peterschmitt Y, Abdoul-Azize S, Murtaza B, Barbier M, Khan AS, Millot JL, Khan NA. Fatty Acid Lingual Application Activates Gustatory and Reward Brain Circuits in the Mouse. Nutrients 2018; 10:nu10091246. [PMID: 30200577 PMCID: PMC6163273 DOI: 10.3390/nu10091246] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 08/27/2018] [Accepted: 09/04/2018] [Indexed: 11/16/2022] Open
Abstract
The origin of spontaneous preference for dietary lipids in humans and rodents is debated, though recent compelling evidence has shown the existence of fat taste that might be considered a sixth taste quality. We investigated the implication of gustatory and reward brain circuits, triggered by linoleic acid (LA), a long-chain fatty acid. The LA was applied onto the circumvallate papillae for 30 min in conscious C57BL/6J mice, and neuronal activation was assessed using c-Fos immunohistochemistry. By using real-time reverse transcription polymerase chain reaction (RT-qPCR), we also studied the expression of mRNA encoding brain-derived neurotrophic factor (BDNF), Zif-268, and Glut-1 in some brain areas of these animals. LA induced a significant increase in c-Fos expression in the nucleus of solitary tract (NST), parabrachial nucleus (PBN), and ventroposterior medialis parvocellularis (VPMPC) of the thalamus, which are the regions known to be activated by gustatory signals. LA also triggered c-Fos expression in the central amygdala and ventral tegmental area (VTA), involved in food reward, in conjunction with emotional traits. Interestingly, we noticed a high expression of BDNF, Zif-268, and Glut-1 mRNA in the arcuate nucleus (Arc) and hippocampus (Hipp), where neuronal activation leads to memory formation. Our study demonstrates that oral lipid taste perception might trigger the activation of canonical gustatory and reward pathways.
Collapse
Affiliation(s)
- Yvan Peterschmitt
- Neurosciences Intégratives et Cliniques EA481, Université de Bourgogne Franche-Comté (UBFC), 25000 Besançon, France; (Y.P.); (M.B.)
| | - Souleymane Abdoul-Azize
- Unité Inserm U1234, Université de Rouen/IRIB, Faculté de Médecine et Pharmacie, 76183 Rouen CEDEX, France;
| | - Babar Murtaza
- Physiologie de la Nutrition & Toxicologie (NUTox), Agro-Sup, UMR U1231 INSERM/Université de Bourgogne Franche-Comté (UBFC), 6, Boulevard Gabriel, 21000 Dijon, France; (B.M.); (A.S.K.)
| | - Marie Barbier
- Neurosciences Intégratives et Cliniques EA481, Université de Bourgogne Franche-Comté (UBFC), 25000 Besançon, France; (Y.P.); (M.B.)
| | - Amira Sayed Khan
- Physiologie de la Nutrition & Toxicologie (NUTox), Agro-Sup, UMR U1231 INSERM/Université de Bourgogne Franche-Comté (UBFC), 6, Boulevard Gabriel, 21000 Dijon, France; (B.M.); (A.S.K.)
| | - Jean-Louis Millot
- Neurosciences Intégratives et Cliniques EA481, Université de Bourgogne Franche-Comté (UBFC), 25000 Besançon, France; (Y.P.); (M.B.)
| | - Naim Akhtar Khan
- Physiologie de la Nutrition & Toxicologie (NUTox), Agro-Sup, UMR U1231 INSERM/Université de Bourgogne Franche-Comté (UBFC), 6, Boulevard Gabriel, 21000 Dijon, France; (B.M.); (A.S.K.)
- Correspondence: ; Tel.: +33-38-039-6312; Fax: +33-38-039-6330
| |
Collapse
|
10
|
Association of variants in SH2B1 and RABEP1 with worsening of low-density lipoprotein and glucose parameters in patients treated with psychotropic drugs. Gene 2017; 628:8-15. [DOI: 10.1016/j.gene.2017.07.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 05/11/2017] [Accepted: 07/02/2017] [Indexed: 12/22/2022]
|
11
|
Tahara A, Tsukada J, Tomura Y, Yatsu T, Shibasaki M. Downregulation of vasopressin V1A receptors and activation of mitogen-activated protein kinase in rat mesangial cells cultured under high-glucose conditions. Clin Exp Pharmacol Physiol 2013; 39:438-46. [PMID: 22352691 DOI: 10.1111/j.1440-1681.2012.05693.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
SUMMARY In the present study we examined the effects of high extracellular glucose concentrations on vasopressin (AVP) V(1A) receptor kinetics and signal transduction in cultured rat mesangial cells. Scatchard analysis of [(3) H]-AVP binding to mesangial cell plasma membranes showed that although high glucose (30 mmol/L) decreased V(1A) receptor numbers relative to cells cultured in normal glucose (10 mmol/L), receptor affinity was not affected. This V(1A) receptor downregulation was associated with an attenuated increase in AVP-stimulated cytosolic free calcium concentrations ([Ca(2+) ](i) ). In addition, high glucose increased both the basal and AVP-stimulated activity of the classic mitogen-activated protein kinase, namely extracellular signal-regulated kinase (ERK). Furthermore, high glucose induced activation of protein kinase C (PKC) in mesangial cells that could be inhibited by coincubation with the PKC inhibitor staurosporine (10 nmol/L). Staurosporine also markedly attenuated the high glucose-induced downregulation of V(1A) receptors on mesangial cells and blocked the depressed [Ca(2+) ](i) response and increased ERK activity induced by AVP. The results indicate that high extracellular glucose downregulates V(1A) receptors on rat mesangial cells and modulates their signal transduction properties via PKC activation.
Collapse
Affiliation(s)
- Atsuo Tahara
- Drug Discovery Research, Astellas Pharma Inc., Tsukuba, Japan.
| | | | | | | | | |
Collapse
|
12
|
Li Y, Samuvel DJ, Sundararaj KP, Lopes-Virella MF, Huang Y. IL-6 and high glucose synergistically upregulate MMP-1 expression by U937 mononuclear phagocytes via ERK1/2 and JNK pathways and c-Jun. J Cell Biochem 2010; 110:248-59. [PMID: 20225236 DOI: 10.1002/jcb.22532] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Matrix metalloproteinases (MMPs) play a pivotal role in tissue remodeling and destruction in inflammation-associated diseases such as cardiovascular disease and periodontal disease. Although it is known that interleukin (IL)-6 is a key proinflamatory cytokine, it remains unclear how IL-6 regulates MMP expression by mononuclear phagocytes. Furthermore, it remains undetermined how IL-6 in combination with hyperglycemia affects MMP expression. In the present study, we investigated the regulatory effect of IL-6 alone or in combination with high glucose on MMP-1 expression by U937 mononuclear phagocytes. We found that IL-6 is a powerful stimulator for MMP-1 expression and high glucose further augmented IL-6-stimulated MMP-1 expression. We also found that high glucose, IL-6, and lipopolysaccharide act in concert to stimulate MMP-1 expression. In the studies to elucidate underlying mechanisms, the extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) pathways were found to be required for stimulation of MMP-1 by IL-6 and high glucose. We also observed that IL-6 and high glucose stimulated the expression of c-Jun, a key subunit of AP-1 known to be essential for MMP-1 transcription. The role of c-Jun in MMP-1 expression was confirmed by the finding that suppression of c-Jun expression by RNA interference significantly inhibited MMP-1 expression. Finally, we demonstrated that similarly to U937 mononuclear phagocytes, IL-6 and high glucose also stimulated MMP-1 secretion from human primary monocytes. In conclusion, this study demonstrated that IL-6 and high glucose synergistically stimulated MMP-1 expression in mononuclear phagocytes via ERK and JNK cascades and c-Jun upregulation.
Collapse
Affiliation(s)
- Yanchun Li
- Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, College of Medicine, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | | | | | |
Collapse
|
13
|
Kang YS, Song HK, Lee MH, Ko GJ, Han JY, Han SY, Han KH, Kim HK, Cha DR. Visfatin is upregulated in type-2 diabetic rats and targets renal cells. Kidney Int 2010; 78:170-81. [PMID: 20375985 DOI: 10.1038/ki.2010.98] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Visfatin (also known as pre-B cell colony-enhancing factor) is a newly discovered adipocytokine that is preferentially produced by visceral fat and regulated by cytokines promoting insulin resistance. Here we determined its renal synthesis and physiology in a genetic model of type 2 diabetes in rats. These rats had higher levels of visfatin synthesis in both glomeruli and tubulointerstitium compared to control rats. Plasma visfatin levels were significantly increased in the early stages of diabetic nephropathy and positively correlated with body weight, fasting plasma glucose, and microalbuminuria. Interestingly, visfatin synthesis was found to occur in podocytes and proximal tubular cells, as well as in adipocytes in vitro. Further, in both renal cells, visfatin synthesis was significantly increased by high glucose in the media but not by angiotensin II. Additionally, visfatin treatment induced rapid uptake of glucose and was associated with increased translocation of GLUT-1 to the cellular membrane of both renal cell types. Furthermore, visfatin induced tyrosine phosphorylation of the insulin receptor, activated downstream insulin signaling pathways such as Erk-1, Akt, and p38 MAPK, and markedly increased the levels of TGFbeta1, PAI-1, type I collagen, and MCP-1 in both renal cells. Thus, our results suggest that visfatin is produced by renal cells and has an important paracrine role in the pathogenesis of diabetic nephropathy.
Collapse
Affiliation(s)
- Young Sun Kang
- Department of Internal Medicine, Korea University, Ansan City, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Short-term modulation of extracellular signal-regulated kinase 1/2 and stress-activated protein kinase/c-Jun NH2-terminal kinase in pancreatic islets by glucose and palmitate: possible involvement of ceramide. Pancreas 2009; 38:585-92. [PMID: 19295452 DOI: 10.1097/mpa.0b013e31819fef03] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES The effect of glucose and palmitate on the phosphorylation of proteins associated with cell growth and survival (extracellular signal-regulated kinase 1/2 [ERK1/2] and stress-activated protein kinase/c-Jun NH2-terminal kinase [SAPK/JNK]) and on the expression of immediate early genes was investigated. METHODS Groups of freshly isolated rat pancreatic islets were incubated in 10-mmol/L glucose with palmitate, LY294002, or fumonisin B1 for the measurement of the phosphorylation and the content of ERK1/2, JNK/SAPK, and v-akt murine thymoma viral oncongene (AKT) (serine 473) by immunoblotting. The expressions of the immediate early genes, c-fos and c-jun, were evaluated by reverse transcription-polymerase chain reaction. RESULTS Glucose at 10 mmol/L induced ERK1/2 and AKT phosphorylations and decreased SAPK/JNK phosphorylation. Palmitate (0.1 mmol/L) abolished the glucose effect on ERK1/2, AKT, and SAPK/JNK phosphorylations. LY294002 caused a similar effect. The inhibitory effect of palmitate on glucose-induced ERK1/2 and AKT phosphorylation changes was not observed in the presence of fumonisin B1. Glucose increased c-fos and decreased c-jun expressions. Palmitate and LY294002 abolished these latter glucose effects. The presence of fumonisin B1 abolished the effect induced by palmitate on c-jun expression. CONCLUSIONS Our results suggest that short-term changes of mitogen-activated protein kinase and AKT signaling pathways and c-fos and c-jun expressions caused by glucose are abolished by palmitate through phosphatidylinositol 3-kinase inhibition via ceramide synthesis.
Collapse
|
15
|
Naito M, Shenoy A, Aoyama I, Koopmeiners JS, Komers R, Schnaper HW, Bomsztyk K. High ambient glucose augments angiotensin II-induced proinflammatory gene mRNA expression in human mesangial cells: effects of valsartan and simvastatin. Am J Nephrol 2009; 30:99-111. [PMID: 19225232 DOI: 10.1159/000203619] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2008] [Accepted: 01/16/2009] [Indexed: 12/28/2022]
Abstract
BACKGROUND Hyperglycemia may potentiate the adverse renal effects of angiotensin II (AII). In the kidney, the major target of AII action is the glomerular mesangial cell, where its hemodynamic and proinflammatory action contributes to renal injury. AII action is mediated by several types of cell receptors. Among those, the AT1 receptor has been best studied using specific AII receptor blockers (ARBs). These agents have emerged as major new modalities in the prevention and amelioration of renal disease where the ARB renoprotective anti-inflammatory properties could be more important than previously appreciated. Like the ARBs, statins may also modulate inflammatory responses that are renoprotective and complement their cholesterol-lowering effects. AIM The aim of this project was to (i) identify a repertoire of proinflammatory mesangial cell AII-inducible mRNAs; (ii) determine if the AII-induced proinflammatory mRNA responses depend on ambient glucose, and (iii) test the anti-inflammatory effectiveness of an ARB, valsartan, either alone or in combination with a statin, simvastatin. RESULTS/CONCLUSIONS Using high-density microarrays and real-time PCR we identified several AII-inducible proinflammatory mesangial genes that exhibited augmented mRNA responses in high-glucose milieu. Valsartan blocked the AII-induced mRNA expression of proinflammatory genes (i.e. MCP-1, LIF and COX-2) maintained in normal and high glucose. These observations add to the mounting evidence that ARBs have anti-inflammatory effects in the kidney, a beneficial effect that may be more important in protecting renal function in diabetic patients. While simvastatin inhibited expression of some mRNAs encoding chemokines/cytokines, it enhanced expression of mRNA encoding COX-2, a key mediator of inflammation. Thus, the non-cholesterol effects of statins on inflammatory responses appear complex.
Collapse
Affiliation(s)
- Masayo Naito
- UW Medicine Lake Union Research, University of Washington, Seattle, WA 98109, USA
| | | | | | | | | | | | | |
Collapse
|
16
|
Song HK, Lee MH, Kim BK, Park YG, Ko GJ, Kang YS, Han JY, Han SY, Han KH, Kim HK, Cha DR. Visfatin: a new player in mesangial cell physiology and diabetic nephropathy. Am J Physiol Renal Physiol 2008; 295:F1485-94. [PMID: 18768589 DOI: 10.1152/ajprenal.90231.2008] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Visfatin is an adipocytokine that improves insulin resistance and has an antidiabetic effect. However, the role of visfatin in the kidney has not yet been reported. In this experiment, the synthesis and physiological action of visfatin in cultured mesangial cells (MCs) were studied to investigate the role of visfatin in diabetic nephropathy. Visfatin was found synthesized in MCs as well as adipocytes. Visfatin synthesis was markedly increased, not by angiotensin II, but by high glucose stimuli. In addition, visfatin treatment induced a rapid uptake of glucose, peaking at 20 min after visfatin treatment in a dose-dependent manner. A small inhibiting RNA against insulin receptor significantly blocked visfatin-mediated glucose uptake. Visfatin stimuli also enhanced intracellular NAD levels, and treatment with FK866, which is a specific inhibitor of nicotinamide phosphoribosyltransferase (Nampt), significantly inhibited visfatin-induced NAD synthesis and glucose uptake. Visfatin treatment increased glucose transporter-1 (GLUT-1) protein expression in isolated cellular membranes, and pretreatment with cytochalasin B completely inhibited visfatin-induced glucose uptake. Moreover, immunofluorescent microscopy showed the migration of cytosolic GLUT-1 into cellular membranes after visfatin treatment. In accordance with these results, the activation of protein kinase B was detected after visfatin treatment. Furthermore, visfatin treatment dramatically increased the synthesis of profibrotic molecules including transforming growth factor-beta1, plasminogen activator inhibitor-1, and type I collagen, and pretreatment with cytochalasin B completely inhibited visfatin-induced upregulation of profibrotic molecules. These results suggest that visfatin is produced in MCs, which are a novel target for visfatin, and play an important role in the pathogenesis of diabetic nephropathy.
Collapse
Affiliation(s)
- Hye Kyoung Song
- Department of Internal Medicine, Korea University, Ansan City, Kyungki-Do, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Sabuda-Widemann D, Grabensee B, Schwandt C, Blume C. Mycophenolic acid inhibits the autocrine PDGF-B synthesis and PDGF-BB-induced mRNA expression of Egr-1 in rat mesangial cells. Nephrol Dial Transplant 2008; 24:52-61. [PMID: 18723570 DOI: 10.1093/ndt/gfn462] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Uncontrolled mesangial cell (MC) proliferation within the context of glomerular disease contributes to the development of glomerulosclerosis. Mesangial autocrine growth factor stimulation has been described as a pathogenic factor. We investigated the effects of mycophenolic acid (MPA), the active metabolite of the immunosuppressant mycophenolate mofetil (MMF), on proliferation factors of cultured rat MCs. MPA was tested on the expression of platelet-derived growth factor-B (PDGF-B) and its receptor beta (PDGFR-beta), the immediate early gene (IEG) c-fos and the early growth response gene-1 (Egr-1), and AP-1 activation. METHODS Growth-arrested rat MCs were stimulated with 10% fetal calf serum (FCS) or 10-25 ng/ml platelet-derived growth factor-BB (PDGF-BB) in the presence or absence of MPA (0.019-10 microM) with or without guanosine (100 microM). MC proliferation was quantified by 5-bromo-2'-deoxyuridine (BrdU) incorporation and direct cell counting. Cytotoxicity of MPA was evaluated using the MTT and LDH tests. Protein expression of PDGF-B and its receptor PDGFR-beta was quantified by western blot analysis. The effect of MPA on gene expression of PDGF-B, Egr-1 and c-fos was determined by the reverse transcriptase-polymerase chain reaction (RT-PCR). AP-1 activation was analysed by an electrophoretic mobility shift assay (EMSA). RESULTS Exposure of MCs to MPA caused a concentration-dependent inhibition of FCS-induced cell proliferation (cell number increase) with an IC50 of 0.44 +/- 0.03 microM and DNA synthesis with an IC50 of 0.52 +/- 0.02 microM without cell cytotoxicity in the therapeutic range. MPA decreased the PDGF-B protein expression and mRNA self-induction of PDGF-B but did not alter the protein expression of PDGFR-beta. MPA strongly inhibited the PDGF-BB-induced mRNA expression of Egr-1 decreasing to 7.6 +/- 2.5% after 30 min (P <or= 0.001) and to 4.7 +/- 3.1% after 1 h (P <or= 0.05), both being compared to the maximal expression induced by PDGF-BB. PDGF-BB-induced c-fos expression under MPA was unchanged after 30 min and decreased to 57 +/- 26% after 1 h (n.s.). MPA treatment did not affect PDGF-BB-induced AP-1 activity determined after 1 h and 2 h. The inhibitory MPA effect on PDGF-BB-induced PDGF-B expression was not significantly restored by guanosine (56 +/- 18% versus 32 +/- 17% after 2 h, n.s.), and MPA inhibition of PDGF-BB-induced Egr-1 expression was not reversed by exogenous guanosine. CONCLUSIONS Treatment of cultured MCs with MPA inhibits MC proliferation correlating with a downregulation of the PDGF-B gene and protein expression and a suppression of Egr-1 mRNA expression. Since exogenous guanosine was not able to reverse the inhibitory MPA effect on PDGF-B and Egr-1 expression, we conclude that the antiproliferative effect of MPA on MCs may not solely depend on dGTP depletion but on a specific interference with the autocrine PDGF-B synthesis and Egr-1 expression of MCs.
Collapse
|
18
|
Nosadini R, Carboni A, Manconi A, Angius F, Caria S, Cherchi S, Satta A, Faedda R, Obinu D, Nieddu M, Carraro A, Tonolo GC. WITHDRAWN: The decline of glomerular function is not always associated with the development of micro- and macroalbuminuria in hypertensive patients with type 2 diabetes. Diabetologia 2008. [PMID: 18607559 DOI: 10.1007/s00125-008-1079-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2008] [Accepted: 05/21/2008] [Indexed: 11/25/2022]
Affiliation(s)
- R Nosadini
- Department of Endocrinology and Metabolic Diseases, Internal Medicine Department, University of Sassari, Viale San Pietro 8, 07100, Sassari, Italy,
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Peng F, Wu D, Gao B, Ingram AJ, Zhang B, Chorneyko K, McKenzie R, Krepinsky JC. RhoA/Rho-kinase contribute to the pathogenesis of diabetic renal disease. Diabetes 2008; 57:1683-92. [PMID: 18356410 DOI: 10.2337/db07-1149] [Citation(s) in RCA: 145] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Accumulation of glomerular matrix proteins is central to the pathogenesis of diabetic nephropathy, with resident mesangial cells (MCs) known to upregulate matrix protein synthesis in response to high glucose. Because activation of the GTPase RhoA has been implicated in matrix upregulation, we studied its role in induction of the matrix protein fibronectin in diabetic MCs and in vivo in diabetic nephropathy. RESEARCH DESIGN AND METHODS Glucose (30 mmol/l)-induced RhoA/Rho-kinase, AP-1 activation, and fibronectin upregulation were assessed by immunoblotting, luciferase, electrophoretic mobility shift assay, enzyme-linked immunosorbent assay, real-time PCR, Northern blots, and immunofluorescence. Streptozotocin-induced diabetic rats were treated with the rho-kinase inhibitor fasudil, which was compared with enalapril, and functional and pathologic parameters were assessed. RESULTS Glucose led to RhoA and downstream Rho-kinase activation. Mannitol was without effect. Activity of the transcription factor AP-1, increased in diabetic MCs and kidneys, is important in the profibrotic effects of glucose, and this was dependent on Rho-kinase signaling. Upregulation of fibronectin by glucose, shown to be mediated by activator protein-1 (AP-1), was prevented by Rho-kinase inhibition. RhoA siRNA and dominant-negative RhoA also markedly attenuated fibronectin upregulation by high glucose. Applicability of these findings were tested in vivo. Fasudil prevented glomerular fibronectin upregulation, glomerular sclerosis, and proteinuria in diabetic rats, with effectiveness similar to enalapril. CONCLUSIONS High glucose activates RhoA/Rho-kinase in MCs, leading to downstream AP-1 activation and fibronectin induction. Inhibition of this pathway in vivo prevents the pathologic changes of diabetic nephropathy, supporting a potential role for inhibitors of RhoA/Rho in the treatment of diabetic renal disease.
Collapse
Affiliation(s)
- Fangfang Peng
- Division of Nephrology, McMaster University, Hamilton, Canada
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Yin DK, Yao WB, Gao XD. Chemically modified heparin inhibits mesangial cell proliferation induced by high glucose through interfering with the cell cycle. Biol Pharm Bull 2007; 30:2274-8. [PMID: 18057711 DOI: 10.1248/bpb.30.2274] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The aims of this study were to investigate whether chemically modified non-anticoagulation heparin derivate (Periodate-Oxidized/Borohydride-Reduced modified heparin (OR-heparin)) can inhibit high glucose-induced human mesangial cell proliferation and its influence on the cell cycle. OR-heparin with low anticoagulation activity inhibited high glucose-induced early proliferation in a dose-dependent manner. OR-heparin released high glucose-arrested mesangial cells at G(1) phase, and dose-dependently increased S phase. OR-heparin also inhibited high glucose-activated ERK1/2 phosphorylation, induced p27(Kip1) expression, and suppressed reactive oxygen species (ROS) accumulation in a dose-dependent manner. Our results suggest that OR-heparin releases high glucose-arrested cells on G(1) phase and inhibits high glucose-induced mesangial cell proliferation through blocking ERK1/2 phosphorylation and delaying S phase progression, which may be in correlation with OR-heparin suppressing ROS accumulation.
Collapse
Affiliation(s)
- Deng-Ke Yin
- School of Life Science and Technology, China Pharmaceutical University, 24 Tongjia Road, Nanjing City 210009, China
| | | | | |
Collapse
|
21
|
Khera TK, Martin J, Riley SG, Steadman R, Phillips AO. Glucose modulates handling of apoptotic cells by mesangial cells: involvement of TGF-beta1. J Transl Med 2007; 87:690-701. [PMID: 17530031 DOI: 10.1038/labinvest.3700555] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Glucose stimulates proapoptotic signalling pathways in mesangial cells. Studies focused on inflammatory glomerular injury have demonstrated that removal of apoptotic mesangial cells occurs by neighbouring non-apoptotic mesangial cells. The aim of this study was to define the effect of ambient glucose concentration on mesangial handling of apoptotic cells, and in addition to examine the response made by the mesangial cell. We used a co-culture model in which neutrophils aged overnight to induce apoptosis, or apoptotic mesangial cells, labelled with a fluorescent dye, were added to mesangial cells to study phagocytosis. Exposure of mesangial cells to an ambient glucose concentration of 25 mM D-glucose before addition of apoptotic cells led in an increase in mesangial cell phagocytosis. Ingestion of apoptotic cells was inhibited by blocking alpha v beta 3 integrin-vitronectin receptor or thrombospondin-1. Furthermore, glucose-dependent stimulation of phagocytosis was inhibited by a blocking antibody to TGF-beta1. Co-culture of apoptotic cells with mesangial cells stimulated synthesis of TGF-beta1 as compared to freshly isolated neutrophils. Increased TGF-beta1 synthesis was dependent on direct contact between the two cell types but was not dependent on phagocytosis of apoptotic cells, as TGF-beta1 generation was not affected by inhibition of the thrombospondin-1 pathway. We propose a model in which apoptotic cell binding but not phagocytosis stimulates enhanced mesangial cell TGF-beta1 synthesis. Furthermore phagocytosis, which involves the thrombospondin-1 pathway, is uncoupled from binding of apoptotic cells, which stimulated TGF-beta1 synthesis.
Collapse
Affiliation(s)
- Tarnjit K Khera
- Institute of Nephrology, School of Medicine, Cardiff University, Wales, UK
| | | | | | | | | |
Collapse
|
22
|
Botolin S, McCabe LR. Chronic hyperglycemia modulates osteoblast gene expression through osmotic and non-osmotic pathways. J Cell Biochem 2006; 99:411-24. [PMID: 16619259 DOI: 10.1002/jcb.20842] [Citation(s) in RCA: 179] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Insulin dependent diabetes mellitus (IDDM; type I) is a chronic disease stemming from little or no insulin production and elevated blood glucose levels. IDDM is associated with osteoporosis and increased fracture rates. The mechanisms underlying IDDM associated bone loss are not known. Previously we demonstrated that osteoblasts exhibit a response to acute (1 and 24 h) hyperglycemia and hyperosmolality. Here we examined the influence of chronic hyperglycemia (30 mM) and its associated hyperosmolality on osteoblast phenotype. Our findings demonstrate that osteoblasts respond to chronic hyperglycemia through modulated gene expression. Specifically, chronic hyperglycemia increases alkaline phosphatase activity and expression and decreases osteocalcin, MMP-13, VEGF and GAPDH expression. Of these genes, only MMP-13 mRNA levels exhibit a similar suppression in response to hyperosmotic conditions (mannitol treatment). Acute hyperglycemia for a 48-h period was also capable of inducing alkaline phosphatase and suppressing osteocalcin, MMP-13, VEGF, and GAPDH expression in differentiated osteoblasts. This suggests that acute responses in differentiated cells are maintained chronically. In addition, hyperglycemic and hyperosmotic conditions increased PPARgamma2 expression, although this increase reached significance only in 21 days chronic glucose treated cultures. Given that osteocalcin is suppressed and PPARgamma2 expression is increased in type I diabetic mouse model bones, these findings suggest that diabetes-associated hyperglycemia may modulate osteoblast gene expression, function and bone formation and thereby contribute to type I diabetic bone loss.
Collapse
MESH Headings
- Animals
- Base Sequence
- Cell Differentiation
- Chronic Disease
- Collagenases/genetics
- DNA, Complementary/genetics
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/pathology
- Gene Expression
- Glyceraldehyde-3-Phosphate Dehydrogenases/genetics
- Hyperglycemia/genetics
- Hyperglycemia/metabolism
- Hyperglycemia/pathology
- In Vitro Techniques
- Male
- Matrix Metalloproteinase 13
- Mice
- Mice, Inbred BALB C
- Osmosis
- Osteoblasts/metabolism
- Osteoblasts/pathology
- PPAR gamma/genetics
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Vascular Endothelial Growth Factor A/genetics
Collapse
Affiliation(s)
- Sergiu Botolin
- Molecular Imaging Research Center, 2201 Biomedical Physical Science Building, Department of Physiology, Michigan State University, East Lansing, Michigan 48824, USA
| | | |
Collapse
|
23
|
Leung JCK, Chan LYY, Tang SCW, Chu KM, Lai KN. Leptin induces TGF-β synthesis through functional leptin receptor expressed by human peritoneal mesothelial cell. Kidney Int 2006; 69:2078-86. [PMID: 16641931 DOI: 10.1038/sj.ki.5000409] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Marked increase in leptin concentration in spent peritoneal dialysate has been reported following continuous ambulatory peritoneal dialysis treatment. The present study was designed to determine whether functional leptin receptor is expressed by human peritoneal mesothelial cells and if so, the possible implication in dialysis. Expression of leptin receptors in cultured mesothelial cells and omental tissue was examined. The effect of leptin on the production of transforming growth factor-beta (TGF-beta) by mesothelial cells in the presence or absence of high glucose was determined using in vitro culture model of human peritoneal mesothelial cells and adipocytes. The signaling mechanism involved in leptin-induced TGF-beta synthesis by mesothelial cells was studied. Both mRNA and protein of the full-length leptin receptor are constitutively expressed in mesothelial cells. The leptin receptor expression in mesothelial cells was upregulated by glucose but not leptin. In adipocytes, glucose increased the mRNA expression and synthesis of leptin. The Janus kinase-signal transducers and activation (JAK-STAT) signal transduction pathway in mesothelial cells was activated by either exogenous or adipocytes-derived leptin. Exogenous leptin induced the release of TGF-beta by mesothelial cells. The TGF-beta synthesis induced by leptin was amplified by glucose through increased leptin receptor expression. Our novel findings reveal that functional leptin receptor is present on human peritoneal mesothelial cells. The leptin-induced TGF-beta synthesis in mesothelial cells is associated with the expression of leptin receptor and the activation of the JAK-STAT signal transduction pathway.
Collapse
Affiliation(s)
- J C K Leung
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong
| | | | | | | | | |
Collapse
|
24
|
Yeshao W, Gu J, Peng X, Nairn AC, Nadler JL. Elevated glucose activates protein synthesis in cultured cardiac myocytes. Metabolism 2005; 54:1453-60. [PMID: 16253633 DOI: 10.1016/j.metabol.2005.05.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2004] [Accepted: 05/08/2005] [Indexed: 11/25/2022]
Abstract
Diabetes mellitus results in chronic hyperglycemia, a serious metabolic disorder associated with a markedly increased risk of cardiovascular disease. However, the effects of high glucose (HG) on cardiac myocyte growth have not been fully clarified. In this study, the effect of glucose on cardiac myocyte growth was examined using leucine incorporation as an index of protein synthesis. High glucose (HG, 25 mmol/L) increased leucine incorporation (167% +/- 0.2% over normal glucose, n=4, P<.01) compared with a physiological glucose concentration (5.5 mmol/L, normal glucose). The HG-induced increase in leucine incorporation was time- and dose-dependent and was not due to osmotic changes because 25 mmol/L mannitol did not change leucine incorporation. High glucose also significantly reduced elongation factor 2 phosphorylation, an effect known to result in increased protein synthesis at the elongation step. Western blot analysis showed that HG-activated protein kinase B (PKB), also called Akt (PKB/Akt), at 18 hours. High glucose-induced leucine incorporation was attenuated with phosphatidylinositol 3-kinase (PI3K) inhibition using wortmannin and LY294002 and by rapamycin, a mammalian target of rapamycin (mTOR) inhibitor, 72%, 64%, and 65% (P<.05), respectively. High glucose also activated extracellular signal-regulated kinase 1/2 activity with peak stimulation at 5 minutes. In addition, PD98059, an inhibitor of mitogen-activated protein kinase kinase, attenuated HG-induced leucine incorporation. These data show for the first time that elevated glucose increases protein synthesis in cardiac myocytes. The increase appears to be mediated by activation of PI3K-PKB/Akt and/or PI3K-mTOR as well as extracellular signal-regulated kinase 1/2. These results provide new evidence for a direct effect of glucose independent of insulin on cardiac myocyte growth.
Collapse
Affiliation(s)
- Wen Yeshao
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Virginia Health Science Center, Charlottesville, VA 22908, USA.
| | | | | | | | | |
Collapse
|
25
|
|
26
|
Zhu ZS, Wang JM, Chen SL. Mesenteric artery remodeling and effects of imidapril and irbesartan on it in spontaneously hypertensive rats. World J Gastroenterol 2004; 10:1471-5. [PMID: 15133856 PMCID: PMC4656287 DOI: 10.3748/wjg.v10.i10.1471] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
AIM: To investigate the remodeling of mesenteric artery and the expression of TGF-β1, c-Jun in mesenteric artery and effects of imidapril and irbesartan on the remodeling in spontaneously hypertensive rats (SHR).
METHODS: Thirty SHR (male/female, 21/9), aged 13 wk, were randomly divided into 3 groups (7 male rats and 3 female rats each group): SHR group, imidapril group (imidapril 3 mg/kg·d was given in drinking water for 14 wk), and irbesartan group (irbesartan 50 mg/kg·d was given in drinking water foe 14 wk). Ten homogenous Wistar Kyoto rats, 5 males and 5 females, weighing 206 ± 49 g, were selected as normal control group (WKY group). Systolic pressure was measured on day 1, 2, 4, 6, 8, 10, 12 and 14 during the experiment and the rats were killed at the end of the experiment. Angiotensin II (Ang II) level in plasma and mesenteric arteries was measured by radioimmunoassay. The morphology of the secondary branches of mesenteric artery were examined by light microscopy and electron microscopy. Reverse transcription polymerase chain reaction (RT-PCR) was used to detect the expression of transforming growth factor TGF-β1 and c-Jun mRNA.
RESULTS: Compared with imidapril group and irbesartan group, the blood pressure was remarkably increased in SHR group. Ang II level in plasma and mesenteric arteries in SHR group was the same or lower than that in WKY group, and was higher in irbesartan group and lower in imidapril group. The remodeling of mesenteric arteries in SHR group was mostly obvious among the 4 groups. The ratio of TGF-β1 absorbed light value to GAPDH absorbed light value in the SHR group was 0.887 ± 0.019, which was significantly higher than that in WKY group, imidapril group, and irbesartan group with the ratios of 0.780 ± 0.018, 0.803 ± 0.005, and 0.847 ± 0.017, respectively (P < 0.01). Ang II level in plasma and mesenteric arteries in imidapril group was significantly lower than that in irbesartan group (P < 0.05). The c-Jun absorbed light value/GAPDH absorbed light value of mesenteric arteries in the SHR group was 0.850 ± 0.015, which was significantly higher than that in the WKY, imidapril, and irbesartan groups (0.582 ± 0.013, 0.743 ± 0.012, and 0.789 ± 0.013, respectively, P < 0.01), and was significantly lower in imidapril group than in irbesartan group (P < 0.05).
CONCLUSION: Imidapril and irbesartan can not only control blood pressure but also inhibit mesenteric arteries remodeling and mRNA expression of TGF-β1, c-Jun in SHR. Imidapril is more effective than irbesartan.
Collapse
Affiliation(s)
- Zhong-Sheng Zhu
- Department of Cardiovascular Medicine, Third Affiliated Hospital of Nanjing Medical University, Nanjing 210006, Jiangsu Province, China.
| | | | | |
Collapse
|
27
|
Weigert C, Brodbeck K, Bierhaus A, Häring HU, Schleicher ED. c-Fos-driven transcriptional activation of transforming growth factor beta-1: inhibition of high glucose-induced promoter activity by thiazolidinediones. Biochem Biophys Res Commun 2003; 304:301-7. [PMID: 12711314 DOI: 10.1016/s0006-291x(03)00599-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The peroxisome proliferator-activated receptor gamma activating compounds thiazolidinedione (TZD) have been shown to inhibit diabetes-induced glomerular transforming growth factor-beta1 (TGF-beta1) expression, thereby ameliorating diabetic nephropathy. Here we examined the hypothesis that TZDs block high glucose-induced TGF-beta1 gene activation by interaction with the activated protein kinase C-c-Fos-TGF-beta1 promoter cascade in mesangial cells. The TZD compounds troglitazone and rosiglitazone completely prevented the high glucose induction of both TGF-beta1 promoter activity and elevation in nuclear c-Fos protein levels. The scavenging properties of troglitazone were shown not to be responsible for this inhibitory action, because hydrogen peroxide-mediated stimulation of TGF-beta1 promoter activity was not blocked. TZD-treatment did not interfere with the transcriptional activity of c-Fos responsible for stimulation of the TGF-beta1 promoter. The findings suggest a molecular mechanism by which TZD-treatment reduces specifically high glucose-induced, c-Fos-mediated gene activation, since phorbol ester-induced c-Fos mRNA and protein expression and subsequent elevation of TGF-beta1 mRNA expression were not prevented by TZDs.
Collapse
Affiliation(s)
- Cora Weigert
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Pathobiochemistry, University of Tübingen, Otfried-Müller-Strasse 10, D-72076 Tübingen, Germany
| | | | | | | | | |
Collapse
|
28
|
Haneda M, Koya D, Isono M, Kikkawa R. Overview of glucose signaling in mesangial cells in diabetic nephropathy. J Am Soc Nephrol 2003; 14:1374-82. [PMID: 12707407 DOI: 10.1097/01.asn.0000064500.89551.76] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Affiliation(s)
- Masakazu Haneda
- Department of Medicine, Shiga University of Medical Science, Seta, Otsu, Shiga 520-2192, Japan.
| | | | | | | |
Collapse
|
29
|
Weigert C, Brodbeck K, Brosius FC, Huber M, Lehmann R, Friess U, Facchin S, Aulwurm S, Häring HU, Schleicher ED, Heilig CW. Evidence for a novel TGF-beta1-independent mechanism of fibronectin production in mesangial cells overexpressing glucose transporters. Diabetes 2003; 52:527-35. [PMID: 12540631 DOI: 10.2337/diabetes.52.2.527] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Recent experimental work indicates that the hyperglycemia-induced increase in mesangial matrix production, which is a hallmark in the development of diabetic nephropathy, is mediated by increased expression of GLUT1. Mesangial cells stably transfected with human GLUT1 mimic the effect of hyperglycemia on the production of the extracellular matrix proteins, particularly fibronectin, when cultured under normoglycemic conditions. Our investigation of the molecular mechanism of this effect has revealed that the enhanced fibronectin production was not mediated by the prosclerotic cytokine transforming growth factor (TGF)-beta1. We found markedly increased nuclear content in Jun proteins, leading to enhanced DNA-binding activity of activating protein 1 (AP-1). AP-1 inhibition reduced fibronectin production in a dosage-dependent manner. Moreover, inhibition of classic protein kinase C (PKC) isoforms prevented both the activation of AP-1 and the enhanced fibronectin production. In contrast to mesangial cells exposed to high glucose, no activation of the hexosamine biosynthetic, p38, or extracellular signal-related kinase 1 and 2 mitogen-activated protein kinase pathways nor any increase in TGF-beta1 synthesis could be detected, which could be explained by the absence of oxidative stress in cells transfected with the human GLUT1 gene. Our data indicate that increased glucose uptake and metabolism induce PKC-dependent AP-1 activation that is sufficient for enhanced fibronectin production, but not for increased TGF-beta1 expression.
Collapse
Affiliation(s)
- Cora Weigert
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Pathobio-Chemistry, University of Tübingen, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Lin S, Sahai A, Chugh SS, Pan X, Wallner EI, Danesh FR, Lomasney JW, Kanwar YS. High glucose stimulates synthesis of fibronectin via a novel protein kinase C, Rap1b, and B-Raf signaling pathway. J Biol Chem 2002; 277:41725-35. [PMID: 12196513 DOI: 10.1074/jbc.m203957200] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The molecular mechanism(s) by which high glucose induces fibronectin expression via G-protein activation in the kidney are largely unknown. This investigation describes the effect of high glucose (HG) on a small GTP-binding protein, Rap1b, expression and activation, and the relevance of protein kinase C (PKC) and Raf pathways in fibronectin synthesis in cultured renal glomerular mesangial cells (MCs). In vivo experiments revealed a dose-dependent increase in Rap1b expression in glomeruli of diabetic rat kidneys. Similarly, in vitro exposure of MCs to HG led to an up-regulation of Rap1b with concomitant increase in fibronectin (FN) mRNA and protein expression. The up-regulation of Rap1b mRNA was mitigated by the PKC inhibitors, calphostin C, and bisindolymaleimide, while also reducing HG- induced FN expression in non-transfected MCs. Overexpression of Rap1b by transfection with pcDNA 3.1/Rap1b in MCs resulted in the stimulation of FN synthesis; however, the PKC inhibitors had no significant effect in reducing FN expression in Rap1b-transfected MCs. Transfection of Rap1b mutants S17N (Ser --> Asn) or T61R (Thr --> Arg) in MCs inhibited the HG-induced increased FN synthesis. B-Raf and Raf-1 expression was investigated to assess whether Rap1b effects are mediated via the Raf pathway. B-Raf, and not Raf-1, expression was increased in MCs transfected with Rap1b. HG also caused activation of Rap1b, which was largely unaffected by anti-platelet-derived growth factor (PDGF) antibodies. HG-induced activation of Rap1b was specific, since Rap2b activation and expression of Rap2a and Rap2b were unaffected by HG. These findings indicate that hyperglycemia and HG cause an activation and up-regulation of Rap1b in renal glomeruli and in cultured MCs, which then stimulates FN synthesis. This effect appears to be PKC-dependent and PDGF-independent, but involves B-Raf, suggesting a novel PKC-Rap1b-B-Raf pathway responsible for HG-induced increased mesangial matrix synthesis, a hallmark of diabetic nephropathy.
Collapse
Affiliation(s)
- Sun Lin
- Department of Pathology, Northwestern University Medical School, Chicago, Illinois 60611, USA
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Amiri F, Shaw S, Wang X, Tang J, Waller JL, Eaton DC, Marrero MB. Angiotensin II activation of the JAK/STAT pathway in mesangial cells is altered by high glucose. Kidney Int 2002; 61:1605-16. [PMID: 11967010 DOI: 10.1046/j.1523-1755.2002.00311.x] [Citation(s) in RCA: 106] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND Both high glucose (HG) and angiotensin II (Ang II) causes glomerular mesangial cell (GMC) growth and increased synthesis of matrix proteins like collagen IV contributing to diabetic nephropathy. We have recently found that exposure of vascular smooth muscle cells to HG augments the Ang II activation of the growth promoting JAK/STAT pathway. We hypothesized that Ang II activation of the JAK/STAT pathway is altered by HG in GMC, and that this pathway might be linked to the Ang II-induced growth and overproduction of collagen IV in GMC in HG conditions. METHODS GMC were cultured under normal glucose (NG; 5.5 mmol/L) and HG (25 mmol/L) for 48 hours and stimulated with Ang II (0.1 micromol/L) for various times. GMC lysate was then immunoprecipitated and/or immunoblotted with SHP-1, SHP-2 and phosphospecific JAK2 and STAT antibodies. The HG and Ang II induced growth and collagen IV synthesis studies were performed in GMC transfected with JAK2 antisense or JAK2 sense. GMC growth was monitored via [3H]-thymidine incorporation, and collagen IV synthesis via ELISA. RESULTS We found that Ang II-induced JAK2, STAT1, STAT3, STAT5A/B and SHP-2 phosphorylations were enhanced by HG, whereas that of SHP-1 was reduced. Ang II-induced growth and collagen IV synthesis also were increased under HG conditions. Transfection of GMC with JAK2 antisense oligonucleotides blocked the Ang II-induced growth and collagen IV synthesis in both NG and HG conditions. CONCLUSION These results provide evidence that activation of the JAK/STAT pathway by HG or/and Ang II may be of importance in the increased GMC cell growth and collagen IV synthesis that is seen in diabetic nephropathy.
Collapse
Affiliation(s)
- Farhad Amiri
- Vascular Biology Center, Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta, Georgia 30912-2500, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Nishio T, Haneda M, Koya D, Inoki K, Maeda S, Kikkawa R. Cyclic AMP inhibits stretch-induced overexpression of fibronectin in glomerular mesangial cells. Eur J Pharmacol 2002; 437:113-22. [PMID: 11890898 DOI: 10.1016/s0014-2999(01)01559-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Glomerular hypertension is proposed to play an important role in the progression of various glomerular diseases. Glomerular mesangial cells are considered to be exposed to the stretch stress due to glomerular hypertension and are found to produce the excess amount of extracellular matrix (ECM) proteins including fibronectin when exposed to the mechanical stretch. Herein, we provide the evidence that cAMP-generating agents inhibit the stretch-induced overexpression of fibronectin through the inhibition of the stretch-induced activation of mitogen-activated protein kinases (MAPKs) in protein kinase-A-dependent manner. We also found that the mechanical stretch enhanced the binding of nuclear extracts to activator protein-1 (AP-1)-like sequences in the promoter region of rat fibronectin gene and this enhancement was also prevented by the cAMP-generating agent. These results indicate that the agents, which activate cAMP/protein kinase-A axis, may work protectively against the injury from glomerular hypertension in mesangial cells.
Collapse
Affiliation(s)
- Toshiki Nishio
- Third Department of Medicine, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| | | | | | | | | | | |
Collapse
|
33
|
Catherwood MA, Powell LA, Anderson P, McMaster D, Sharpe PC, Trimble ER. Glucose-induced oxidative stress in mesangial cells. Kidney Int 2002; 61:599-608. [PMID: 11849402 DOI: 10.1046/j.1523-1755.2002.00168.x] [Citation(s) in RCA: 128] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Hyperglycemia is a well-recognized pathogenic factor of long-term complications in diabetes mellitus. Hyperglycemia not only generates reactive oxygen species but also attenuates antioxidant mechanisms creating a state of oxidative stress. METHODS Porcine mesangial cells were cultured in high glucose (HG) for ten days to investigate the effects on the antioxidant defenses of the cell. RESULTS Mesangial cells cultured in HG conditions had significantly reduced levels of glutathione (GSH) compared with those grown in normal glucose (NG). The reduced GSH levels were accompanied by decreased gene expression of both subunits of gamma-glutamylcysteine synthetase (gamma-GCS), the rate-limiting enzyme in de novo synthesis of GSH. Elevated levels of intracellular malondialdehyde (MDA) were found in cells exposed to HG conditions. HG also caused elevated mRNA levels of the antioxidant enzymes CuZn superoxide dismutase (SOD) and MnSOD. These changes were accompanied by increased mRNA levels of extracellular matrix proteins (ECM), fibronectin (FN) and collagen IV (CIV). Addition of antioxidants to high glucose caused a significant reversal of FN and CIV gene expression; alpha-lipoic acid also up-regulated gamma-GCS gene expression and restored intracellular GSH and MDA levels. CONCLUSIONS The results demonstrate the existence of glucose-induced oxidative stress in mesangial cells as evidenced by elevated MDA and decreased GSH levels. The decreased levels of GSH are as a result of decreased mRNA expression of gamma-GCS within the cell. Antioxidants caused a significant reversal of FN and CIV gene expression, suggesting an etiological link between oxidative stress and increased ECM protein synthesis.
Collapse
Affiliation(s)
- Mark A Catherwood
- Department of Clinical Biochemistry, Queen's University of Belfast, Royal Group of Hospitals, Belfast, United Kingdom.
| | | | | | | | | | | |
Collapse
|
34
|
Abstract
Oxidative stress has been known to play an important role in the development and progression of diabetic nephropathy, but the intracellular signal transduction pathways regulated by reactive oxygen species (ROS) have not been clearly defined. High glucose (HG) induces intracellular ROS directly via glucose metabolism and auto-oxidation and indirectly through the formation of advanced glycation end products and their receptor binding. ROS mimic the stimulatory effects of HG and upregulate transforming growth factor-beta 1, plasminogen activator inhibitor-1, and extracellular matrix (ECM) proteins by glomerular mesangial cells, thus leading to mesangial expansion. ROS activate other signaling molecules, such as protein kinase C and mitogen-activated protein kinases and transcription factors, such as nuclear factor-kappa B, activator protein-1, and specificity protein 1 leading to transcription of genes encoding cytokines, growth factors, and ECM proteins. Finally, various antioxidants inhibit mesangial cell activation by HG and ameliorate features of diabetic nephropathy. These findings qualify ROS as intracellular messengers and as integral glucose-signaling molecules in glomerular mesangial cells in diabetic nephropathy. With this new concept, ROS assume a greater importance in the pathogenesis of diabetic nephropathy. Future studies elucidating other downstream-signaling molecules activated by ROS in mesangial and other renal cells will allow us to understand the final cellular responses to HG, such as proliferation, differentiation, apoptosis, and ECM accumulation. With this new information, we should be able to develop strategies for a more rational treatment of diabetic nephropathy.
Collapse
Affiliation(s)
- H Ha
- Hyonam Kidney Laboratory, Soon Chun Hyang University, 657 Hannam-dong, Yongsan-ku, Seoul 140-743, Korea
| | | |
Collapse
|
35
|
Powell LA, Nally SM, McMaster D, Catherwood MA, Trimble ER. Restoration of glutathione levels in vascular smooth muscle cells exposed to high glucose conditions. Free Radic Biol Med 2001; 31:1149-55. [PMID: 11705692 DOI: 10.1016/s0891-5849(01)00648-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Hyperglycemia-induced oxidative stress may play a key role in the pathogenesis of diabetic vascular disease. The purpose of this study was to determine the effects of glucose on levels of glutathione (a major intracellular antioxidant), the expression of gamma-glutamylcysteine synthetase (the rate-limiting enzyme in glutathione de novo synthesis), and DNA damage in human vascular smooth muscle cells in vitro. High glucose conditions and buthionine sulphoximine, an inhibitor of gamma-glutamylcysteine synthetase, reduced intracellular glutathione levels in vascular smooth muscle cells. This reduction was accompanied by a decrease in the mRNA expression of both subunits of gamma-glutamylcysteine synthetase as well as an increase in DNA damage. In high glucose conditions, incubation of the vascular smooth muscle cells with alpha-lipoic acid and L-cystine restored glutathione levels. We suggest that the decrease in GSH levels seen in high glucose conditions is mediated by the availability of cysteine (rate-limiting substrate in de novo glutathione synthesis) and the gene expression of the gamma-glutamylcysteine synthetase enzyme. Glutathione depletion is associated with an increase in DNA damage, which can be reduced when glutathione levels are restored.
Collapse
Affiliation(s)
- L A Powell
- Department of Clinical Biochemistry, The Royal Group of Hospitals, Belfast, UK.
| | | | | | | | | |
Collapse
|
36
|
Abstract
Vascular complications in diabetes mellitus are known to be associated with the activation of the protein kinase C (PKC) pathway through the de novo synthesis of diacylglycerol (DAG) from glycolytic intermediates. Specific PKC isoforms, mainly the beta- and delta-isoforms, have been shown to be persistently activated in diabetic mellitus. Multiple studies have reported that the activation of PKC leads to increased production of extracellular matrix and cytokines, enhances contractility, permeability and vascular cell proliferation, induces the activation of cytosolic phospholipase A2 and inhibits the activity of Na+-K+-ATPase. These events are not only frequently observed in diabetes mellitus but are also involved in the actions of vasoactive agents or oxidative stress. Inhibition of PKC by two different kinds of PKC inhibitors - LY333531, a selective PKC-beta-isoform inhibitor, and vitamin E, d-alpha-tocopheron - were able to prevent or reverse the various vascular dysfunctions in vitro and in vivo. Clinical studies using these compounds are now ongoing to evaluate the significance of DAG-PKC pathway activation in the development of vascular complications in diabetic patients.
Collapse
Affiliation(s)
- M Meier
- Research Division of Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
37
|
Yasuda Y, Nakamura J, Hamada Y, Nakayama M, Chaya S, Naruse K, Nakashima E, Kato K, Kamiya H, Hotta N. Role of PKC and TGF-beta receptor in glucose-induced proliferation of smooth muscle cells. Biochem Biophys Res Commun 2001; 281:71-7. [PMID: 11178962 DOI: 10.1006/bbrc.2001.4310] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The role of protein kinase C (PKC) and transforming growth factor (TGF)-beta in the proliferation of vascular smooth muscle cells (SMCs) under a high glucose condition was investigated. [3H]-thymidine incorporation under 20 mM glucose was significantly accelerated compared with that under 5.5 mM glucose, and this increase was inhibited by an anti-TGF-beta antibody or a PKC-beta specific inhibitor, LY333531. The amount of active and total TGF-beta1 in the conditioned media did not differ between 5.5 and 20 mM glucose. However, the expression of TGF-beta receptor type II under 20 mM glucose was significantly increased, but that of the TGF-beta receptor type I was not. This increased expression of the TGF-beta receptor type II was prevented by LY333531. These observations suggest that the increased expression of the TGF-beta receptor type II via PKC-beta plays an important role in the accelerated proliferation of SMCs under a high glucose condition, leading to the development of diabetic macroangiopathy.
Collapse
MESH Headings
- Activin Receptors, Type I
- Animals
- Aorta/metabolism
- Cell Division
- Cells, Cultured
- Culture Media, Conditioned/metabolism
- DNA/biosynthesis
- Dose-Response Relationship, Drug
- Enzyme Inhibitors/pharmacology
- Glucose/metabolism
- Glucose/pharmacology
- Immunoblotting
- Indoles/pharmacology
- Maleimides/pharmacology
- Muscle, Smooth/cytology
- Muscle, Smooth/metabolism
- Protein Kinase C/physiology
- Protein Serine-Threonine Kinases/metabolism
- RNA, Messenger/metabolism
- Rats
- Receptor, Transforming Growth Factor-beta Type I
- Receptor, Transforming Growth Factor-beta Type II
- Receptors, Transforming Growth Factor beta/metabolism
- Receptors, Transforming Growth Factor beta/physiology
- Reverse Transcriptase Polymerase Chain Reaction
Collapse
Affiliation(s)
- Y Yasuda
- Third Department of Internal Medicine, Nagoya University School of Medicine, Nagoya, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Isono M, Cruz MCIDELA, Chen S, Hong SW, Ziyadeh FN. Extracellular signal-regulated kinase mediates stimulation of TGF-beta1 and matrix by high glucose in mesangial cells. J Am Soc Nephrol 2000; 11:2222-2230. [PMID: 11095645 DOI: 10.1681/asn.v11122222] [Citation(s) in RCA: 96] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
High ambient glucose exerts its injurious effects on renal cells through nonenzymatic and enzymatic pathways, including altered signal transduction and upregulation of the transforming growth factor-beta (TGF-beta) system. Extracellular signal-regulated kinase (ERK), a member of the mitogen-activated protein kinase (MAPK) cascade, is activated in mesangial cells cultured in high glucose and in glomeruli of diabetic rats. However, the biologic consequences of ERK activation in the kidney have not been investigated. To clarify the role of ERK activation, mouse mesangial cells were exposed to normal (5.5 mM) or high (25 mM) glucose with or without addition of PD98059, a specific inhibitor of MAPK/ERK kinase (MEK), an upstream kinase activator of ERK. Cells that were exposed to high glucose exhibited significant increases in ERK activity, TGF-beta1 expression (total protein, mRNA levels, and promoter activity), [(3)H]-proline uptake, and alpha1(I) collagen and fibronectin mRNA levels. Treatment with PD98059 (up to 25 microM) significantly inhibited these parameters. In contrast, 25 microM PD98059 had no significant effect on any of the parameters measured in cells that were exposed to normal glucose. Overexpression of MAPK phosphatase CL 100 prevented TGF-beta1 promoter activation by high glucose, confirming the involvement of the MEK-ERK pathway in response to high glucose. The conclusion is that activation of ERK in mesangial cells is responsible for high-glucose-induced stimulation of TGF-beta1 and contributes to the increased extracellular matrix expression.
Collapse
Affiliation(s)
- Motohide Isono
- Renal-Electrolyte and Hypertension Division of the Department of Medicine and the Penn Center for the Molecular Studies of Kidney Diseases, University of Pennsylvania, Philadelphia, Pennsylvania
| | - M Carmen Iglesias-DE LA Cruz
- Renal-Electrolyte and Hypertension Division of the Department of Medicine and the Penn Center for the Molecular Studies of Kidney Diseases, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sheldon Chen
- Renal-Electrolyte and Hypertension Division of the Department of Medicine and the Penn Center for the Molecular Studies of Kidney Diseases, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Soon Won Hong
- Renal-Electrolyte and Hypertension Division of the Department of Medicine and the Penn Center for the Molecular Studies of Kidney Diseases, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Fuad N Ziyadeh
- Renal-Electrolyte and Hypertension Division of the Department of Medicine and the Penn Center for the Molecular Studies of Kidney Diseases, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
39
|
Weigert C, Sauer U, Brodbeck K, Pfeiffer A, Häring HU, Schleicher ED. AP-1 proteins mediate hyperglycemia-induced activation of the human TGF-beta1 promoter in mesangial cells. J Am Soc Nephrol 2000; 11:2007-2016. [PMID: 11053476 DOI: 10.1681/asn.v11112007] [Citation(s) in RCA: 131] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Hyperglycemia-induced overproduction of the prosclerotic cytokine transforming growth factor-beta1 (TGF-beta1) has been implicated in the pathogenesis of diabetic nephropathy. Because high glucose and phorbol esters (PMA) increase TGF-beta1 mRNA levels in mesangial cells, this study was designed to characterize these effects on the human TGF-beta1 promoter activity. With the use of luciferase reporter gene constructs containing TGF-beta1 5'-flanking sequence (from -453 to +11 bp) transfected into mesangial cells, it was found that 30 mM glucose induced a nearly twofold increase in TGF-beta1 promoter activity after 24 h of incubation in human and porcine mesangial cells. Stimulation by PMA was more effective (2.3-fold). Mutagenesis in either one of the two or both activating protein-1 (AP-1) binding sites abolished the high glucose and the PMA effect. Furthermore, addition of the AP-1 inhibitor curcumin obliterated the glucose response. Corresponding experiments revealed that the transcription factor stimulating protein 1 was not involved in mediating the glucose effect. The high glucose-induced TGF-beta1 promoter activation was also prevented by inhibitors of protein kinase C and p38 mitogen-activated proteinkinase. Electrophoretic mobility shift assays with oligonucleotides containing one of the two AP-1 binding sites showed that glucose treatment markedly enhanced the binding activity of nuclear proteins of mesangial cells, particularly to box B. Supershift assays demonstrated that JunD and c-Fos were present in the protein-DNA complexes under control and hyperglycemic conditions. The functional and structural results show that glucose regulates human TGF-beta1 gene expression through two adjacent AP-1 binding sites and gives rise to the involvement of protein kinase C and p38 mitogen-activated protein kinase in hyperglycemia-induced TGF-beta1 gene expression.
Collapse
Affiliation(s)
- Cora Weigert
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Pathobiochemistry, University of Tübingen, Tübingen, Germany
| | - Ulrich Sauer
- Institute of Pathology, University of Munich, Munich, Germany
| | - Katrin Brodbeck
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Pathobiochemistry, University of Tübingen, Tübingen, Germany
| | | | - Hans U Häring
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Pathobiochemistry, University of Tübingen, Tübingen, Germany
| | - Erwin D Schleicher
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Pathobiochemistry, University of Tübingen, Tübingen, Germany
| |
Collapse
|
40
|
Leehey DJ, Singh AK, Alavi N, Singh R. Role of angiotensin II in diabetic nephropathy. KIDNEY INTERNATIONAL. SUPPLEMENT 2000; 77:S93-8. [PMID: 10997697 DOI: 10.1046/j.1523-1755.2000.07715.x] [Citation(s) in RCA: 177] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Considerable evidence suggests that the intrarenal renin-angiotensin system plays an important role in diabetic nephropathy. Angiotensin-converting enzyme (ACE) inhibitors and angiotensin II (Ang II) receptor blockers (ARBs) can attenuate progressive glomerulosclerosis in disease models and can slow disease progression in humans. Because agents that interfere with Ang II action may decrease glomerular injury without altering glomerular pressures, it has been suggested that Ang II has direct effects on glomerular cells to induce sclerosis independent of its hemodynamic actions. To study nonhemodynamic effects of Ang II on matrix metabolism, many investigators have used cell culture systems. Glucose and Ang II have been shown to produce similar effects on renal cells in culture. For instance, incubation of mesangial cells in high-glucose media or in the presence of Ang II stimulates matrix protein synthesis and inhibits degradative enzyme (e.g., collagenase, plasmin) activity. Glucose and Ang II also can inhibit proximal tubule proteinases. Glucose increases expression of the angiotensinogen gene in proximal tubule cells and Ang II production in primary mesangial cell culture, which indicates that high glucose itself can activate the renin-angiotensin system. The effects of glucose and Ang II on mesangial matrix metabolism may be mediated by transforming growth factor-beta (TGF-beta). Exposure of mesangial cells to glucose or Ang II increases TGF-beta expression and secretion. Their effects on matrix metabolism can be blocked by anti-TGF-beta antibody or ARBs such as losartan, which also prevents the glucose-induced increment in TGF-beta secretion. Taken together, these findings support the hypothesis that the high-glucose milieu of diabetes increases Ang II production by renal, and especially, mesangial cells, which results in stimulation of TGF-beta secretion, leading to increased synthesis and decreased degradation of matrix proteins, thus producing matrix accumulation. This may be an important mechanism linking hyperglycemia and Ang II in the pathogenesis of diabetic nephropathy.
Collapse
Affiliation(s)
- D J Leehey
- Hines Veterans Affairs Hospital, Illinois 60141, USA.
| | | | | | | |
Collapse
|
41
|
Abstract
Cell cycle regulation in diabetic nephropathy. Renal hypertrophy is one of the earliest abnormalities of diabetic nephropathy. Although selected cell populations. such as tubulointerstitial fibroblasts, may undergo sustained proliferation in the diabetic environment, most renal cells such as mesangial cells are arrested in the G1-phase of the cell cycle after actively leaving G0-phase and some self-limited early proliferation. High glucose, transforming growth factor-beta (TGF-beta), angiotensin II, and probably other factors induce inhibitors of cyclin-dependent kinases (CDK) including p21Cip1 and p27KiP1. These CDK-inhibitors bind to and inactivate G1-phase cyclin/CDK complexes. The consequence is a lack in kinase activity, underphosphorylation of the retinoblastoma gene protein, and a failure to initiate the G1-S-phase transit. The half-life of CDK-inhibitors may also be increased by serine phosphorylation mediated through activated MAP kinases. Treatment of diabetic rats with angiotensin-converting enzyme inhibitors attenuates glomerular hypertrophy and abolishes the glomerular expression of the CDK-inhibitors p16INK4 and p27KiP1, thus indicating that the cell cycle arrest can be therapeutically influenced. Cell cycle proteins may also be involved in these molecular events, leading to a limited degree of tubular apoptosis, which is a feature of diabetic nephropathy. Although not definitively proven, accumulating evidence suggests that early hypertrophy of renal cells may act as pacemaker for subsequent irreversible structural changes, such as glomerulosclerosis and tubulointerstitial fibrosis. Therefore, a better understanding of altered processes of cell cycle regulation is necessary to develop novel therapeutic strategies to prevent diabetic nephropathy. The recent observation that glomerular hypertrophy and proteinuria do not develop in diabetic p21CiP1 knockout mice indicates that this approach is feasible.
Collapse
Affiliation(s)
- G Wolf
- Department of Medicine, University of Hamburg, Germany.
| |
Collapse
|
42
|
McLennan SV, Fisher E, Martell SY, Death AK, Williams PF, Lyons JG, Yue DK. Effects of glucose on matrix metalloproteinase and plasmin activities in mesangial cells: possible role in diabetic nephropathy. KIDNEY INTERNATIONAL. SUPPLEMENT 2000; 77:S81-7. [PMID: 10997695 DOI: 10.1046/j.1523-1755.2000.07713.x] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Diabetic nephropathy is characterized by an accumulation of mesangium matrix that correlates well with the loss of kidney function. High glucose concentration is known to increase the synthesis of many matrix components. Recently, we have shown that degradation of matrix also decreases in diabetes. The major enzymes responsible for matrix degradation are the matrix metalloproteinases. The physiology of these enzymes is complex and their activity is tightly regulated at many levels. At the transcriptional level matrix metalloproteinase (MMP) expression is increased by protein kinase C (PKC) agonists, and some growth factors. In contrast transforming growth factor (TGF)-beta can decrease MMP expression. Once synthesized, MMPs are secreted as inactive pro-enzymes that are activated by other MMPs or plasmin. To effect this, plasmin must be liberated from plasminogen in the pericellular environment. In turn, activated MMPs can be inhibited by binding to specific inhibitors known as tissue inhibitor of metalloproteinases (TIMP). Cell culture and animal studies have shown that high glucose (HG) decreases expression of MMPs and increases expression of TIMPs. HG can also affect MMP activation by decreasing plasmin availability and reducing expression of a membrane-bound MMP called MT1-MMP. How HG induces these changes remains to be fully elucidated. One possibility is that HG can increase TGF-beta. which may in turn alter MMP promoter activity: this area is currently being studied in our laboratory.
Collapse
Affiliation(s)
- S V McLennan
- Effects of glucose on matrix metalloproteinase and plasmin activities in mesangial cells: possible role in diabetic nephropathy.
| | | | | | | | | | | | | |
Collapse
|
43
|
Ha H, Lee HB. Reactive oxygen species as glucose signaling molecules in mesangial cells cultured under high glucose. KIDNEY INTERNATIONAL. SUPPLEMENT 2000; 77:S19-25. [PMID: 10997686 DOI: 10.1046/j.1523-1755.2000.07704.x] [Citation(s) in RCA: 209] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Oxidative stress is one of the important mediators of vascular complications in diabetes including nephropathy. High glucose (HG) generates reactive oxygen species (ROS) as a result of glucose auto-oxidation, metabolism, and formation of advanced glycosylation end products. The concept of ROS-induced tissue injury has recently been revised with the appreciation of new roles for ROS in signaling pathways and gene expression. METHODS AND RESULTS High glucose rapidly generated dichlorofluorescein-sensitive cytosolic ROS in rat and mouse mesangial cells. Neither L-glucose nor 3-O-methyl-D-glucose increased cytosolic ROS and cytochalasin B, an inhibitor of glucose transporter, effectively inhibited HG-induced ROS generation, suggesting that glucose uptake and subsequent metabolism are required in HG-induced cytosolic ROS generation. H2O2 up-regulated fibronectin mRNA expression and protein synthesis; this up-regulation was effectively inhibited by protein kinase C (PKC) inhibitor or by depletion of PKC. The HG-induced generation of ROS was, in turn, related to activation of PKC and transcription factors nuclear factor-kappaB (NF-kappaB) and activator protein-1 (AP-1) as well as to the up-regulation of transforming growth factor-beta1 (TGF-beta1), fibronectin mRNA expression and protein synthesis, because antioxidants effectively inhibited HG-induced PKC, NF-kappaB, AP-1 activation, and TGF-beta1 and fibronectin expression in mesangial cells cultured under HG. CONCLUSIONS Although signal transduction pathways linking HG, ROS, PKC, transcription factors, and extracellular matrix (ECM) protein synthesis in mesangial cells have not been fully elucidated, the current data provide evidence that ROS generated by glucose metabolism may act as integral signaling molecules under HG as in other membrane receptor signaling.
Collapse
Affiliation(s)
- H Ha
- Department of Pharmacology, Yonsei University College of Medicine and Hyonam Kidney Laboratory, Soon Chun Hyang University, Seoul, Korea
| | | |
Collapse
|
44
|
Abstract
Insulin dependent diabetes mellitus, marked by high blood glucose levels and no insulin secretion, is associated with decreased bone mass and increased fracture rates. Analysis of bone histology suggests that osteoblast phenotype and function are influenced by diabetes. To determine if elevated extracellular glucose levels could directly influence osteoblast phenotype we treated mouse osteoblasts, MC3T3-E1 cells, with 22 mM glucose and analyzed osteoblast gene expression. Collagen I mRNA levels significantly increased while osteocalcin mRNA levels decreased 24 h after the addition of glucose. Expression of other genes, actin, osteopontin, and histone H4, was unaffected. Effects on collagen I expression were seen as early as 1 h after treatment. c-Jun, an AP-1 transcription factor involved in the regulation of osteoblast gene expression and growth, was also modulated by glucose. Specifically, an increase in c-jun expression was found at 1 h and maintained for 24 h following glucose treatment. Treatment of osteoblasts with an equal concentration of mannitol completely mimicked glucose treatment effects on collagen I and c-jun expression, demonstrating that osmotic stress rather than glucose metabolism is responsible for the effects on osteoblast gene expression and phenotype. Additional studies using staurosporine and Ro-31-8220 demonstrate that protein kinase C is required for the glucose up regulation of collagen I and c-jun. Taken together, our results demonstrate that osteoblasts respond to increasing extracellular glucose concentration through an osmotic response pathway that is dependent upon protein kinase C activity and results in upregulation of c-jun and modulation of collagen I and osteocalcin expression.
Collapse
Affiliation(s)
- M Zayzafoon
- Department of Physiology, Michigan State University, East Lansing, Michigan 48824, USA
| | | | | | | |
Collapse
|
45
|
Isono M, Mogyorósi A, Han DC, Hoffman BB, Ziyadeh FN. Stimulation of TGF-beta type II receptor by high glucose in mouse mesangial cells and in diabetic kidney. Am J Physiol Renal Physiol 2000; 278:F830-8. [PMID: 10807596 DOI: 10.1152/ajprenal.2000.278.5.f830] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Transforming growth factor-beta (TGF-beta) is important in the pathogenesis of diabetic nephropathy, but little is known about the regulation of the ligand-binding TGF-beta type II signaling receptor (TbetaIIR). There were significant increases in TbetaIIR protein and mRNA levels in kidney cortex after 1-6 wk of streptozotocin-induced diabetes. Mouse mesangial cells cultured in high glucose demonstrated significantly increased TbetaIIR protein and mRNA levels compared with normal glucose. This effect was independent of stimulation of TGF-beta bioactivity by high glucose. Consistent with transcriptional activation by high glucose, the half-life ( approximately 4 h) of TbetaIIR mRNA was not affected by glucose concentration. Moreover, mouse mesangial cells transiently transfected with reporter constructs containing the first 47- or 274-bp promoter fragments of TbetaIIR demonstrated significantly increased reporter activity in high glucose. Cells grown in high glucose demonstrated increased responsiveness to a relatively small dose of exogenous TGF-beta(1) (0.5 ng/ml): [(3)H]proline incorporation and alpha(1)(IV) collagen mRNA were significantly greater in cells cultured in high than in normal glucose. Hence, the expression of TbetaIIR is increased in the diabetic kidney and in mesangial cells cultured in high glucose, primarily because of stimulation of gene transcription. TbetaIIR upregulation by high ambient glucose may contribute to the increased sensitivity of mesangial cells to the profibrogenic action of TGF-beta(1).
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Collagen/genetics
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Diabetic Nephropathies/genetics
- Diabetic Nephropathies/metabolism
- Female
- Gene Expression
- Glomerular Mesangium/drug effects
- Glomerular Mesangium/metabolism
- Glucose/pharmacology
- Mice
- Mice, Inbred C57BL
- Proline/metabolism
- Promoter Regions, Genetic
- RNA Stability
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Transforming Growth Factor beta/drug effects
- Receptors, Transforming Growth Factor beta/genetics
- Receptors, Transforming Growth Factor beta/metabolism
- Transfection
- Transforming Growth Factor beta/pharmacology
Collapse
Affiliation(s)
- M Isono
- Renal-Electrolyte and Hypertension Division, Department of Medicine, and Penn Center for the Molecular Studies of Kidney Diseases, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6144, USA
| | | | | | | | | |
Collapse
|
46
|
Shankland SJ, Wolf G. Cell cycle regulatory proteins in renal disease: role in hypertrophy, proliferation, and apoptosis. Am J Physiol Renal Physiol 2000; 278:F515-29. [PMID: 10751212 DOI: 10.1152/ajprenal.2000.278.4.f515] [Citation(s) in RCA: 122] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The response to glomerular and tubulointerstitial cell injury in most forms of renal disease includes changes in cell number (proliferation and apoptosis) and cell size (hypertrophy). These events typically precede and may be responsible for the accumulation of extracellular matrix proteins that leads to a decrease in renal function. There is increasing evidence showing that positive (cyclins and cyclin-dependent kinases) and negative (cyclin-dependent kinase inhibitors) cell cycle regulatory proteins have a critical role in regulating these fundamental cellular responses to immune and nonimmune forms of injury. Data now show that altering specific cell cycle proteins affects renal cell proliferation and improves renal function. Equally exciting is the expanding body of literature showing novel biological roles for cell cycle proteins in the regulation of cell hypertrophy and apoptosis. With increasing understanding of the role for cell cycle regulatory proteins in renal disease comes the hope for potential therapeutic interventions.
Collapse
Affiliation(s)
- S J Shankland
- Department of Medicine, Division of Nephrology, University of Washington Seattle, Washington 98195-6521, USA.
| | | |
Collapse
|
47
|
Koya D, Haneda M, Nakagawa H, Isshiki K, Sato H, Maeda S, Sugimoto T, Yasuda H, Kashiwagi A, Ways DK, King GL, Kikkawa R. Amelioration of accelerated diabetic mesangial expansion by treatment with a PKC beta inhibitor in diabetic db/db mice, a rodent model for type 2 diabetes. FASEB J 2000; 14:439-47. [PMID: 10698958 DOI: 10.1096/fasebj.14.3.439] [Citation(s) in RCA: 337] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Activation of protein kinase C (PKC) is implicated as an important mechanism by which diabetes causes vascular complications. We have recently shown that a PKC beta inhibitor ameliorates not only early diabetes-induced glomerular dysfunction such as glomerular hyperfiltration and albuminuria, but also overexpression of glomerular mRNA for transforming growth factor beta1 (TGF-beta1) and extracellular matrix (ECM) proteins in streptozotocin-induced diabetic rats, a model for type 1 diabetes. In this study, we examined the long-term effects of a PKC beta inhibitor on glomerular histology as well as on biochemical and functional abnormalities in glomeruli of db/db mice, a model for type 2 diabetes. Administration of a PKC beta inhibitor reduced urinary albumin excretion rates and inhibited glomerular PKC activation in diabetic db/db mice. Administration of a PKC beta inhibitor also prevented the mesangial expansion observed in diabetic db/db mice, possibly through attenuation of glomerular expression of TGF-beta and ECM proteins such as fibronectin and type IV collagen. These findings provide the first in vivo evidence that the long-term inhibition of PKC activation in the renal glomeruli can ameliorate glomerular pathologies in diabetic state, and thus suggest that a PKC beta inhibitor might be an useful therapeutic strategy for the treatment of diabetic nephropathy.
Collapse
Affiliation(s)
- D Koya
- Third Department of Medicine, Shiga University of Medical Science, Seta, Otsu, Shiga 520-2192, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Ingram AJ, Ly H, Thai K, Kang MJ, Scholey JW. Mesangial cell signaling cascades in response to mechanical strain and glucose. Kidney Int 1999; 56:1721-8. [PMID: 10571780 DOI: 10.1046/j.1523-1755.1999.00743.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Elevated glucose levels and glomerular hypertension (Pgc) are considered to contribute to the elaboration of matrix protein by mesangial cells (MCs) in diabetic glomeruli. MCs grown in 30 mM of glucose produce excessive matrix protein, as do MCs exposed to cyclic strain, and the combination of the two exacerbates this. Tight glucose control or reduction in Pgc clinically delays progression of diabetic nephropathy. MC c-fos is induced in response to either application of strain or high ambient glucose, inducing increases in activated protein-1 transactivational activity and extracellular matrix production. Stimuli that lead to c-fos induction pass through the three mitogen-activated protein (MAP) kinase pathways: p44/42, SAPK/JNK, and p38/HOG. We studied MAP kinase activation in MCs exposed to mechanical strain and a high glucose. METHODS MCs (passage 5 through 10) cultured for 96 hours on type 1 collagen-coated flexible-bottom plates in either 5.6 or 30 mM glucose were exposed to 5, 10, or 30 minutes of cyclic strain (60 cycles per min) by computer-driven generation of vacuums of -14 kPa, inducing 20% elongation in the diameter of the surface. Control MCs were grown on both coated rigid and flexible-bottom plates. Protein levels (by Western blot) and activity assays for all three kinase cascades were performed at baseline and after 5, 10, and 30 minutes. All experiments were performed in triplicate. RESULTS MAP kinase signaling was seen in response to stretch, and high ambient glucose affected the pattern of activation. Both p44/42 and p38HOG kinase activities showed small increases to a maximum of 2.5- to 3.5-fold greater than static MCs at 10 minutes. Activity in both kinase cascades was slightly suppressed by 30 mM glucose. In contrast, SAPK/JNK activity was present at a very low level in static MCs and increased markedly by 10 minutes of stretch. Thirty micromolars of glucose augmented this effect by a factor of six over MCs cultured in 5.6 mM glucose after 10 minutes of stretch. Neither glucose concentration nor mechanical strain had any effect on the protein expression of any of the kinases by Western blot. CONCLUSIONS MAP kinase cascade signaling is seen when physical force is applied to MCs, and glucose affects the pattern of activity. Thirty micromolars of glucose markedly increase the level of SAPK/JNK activation. This may have implications in diabetic signal transduction and matrix protein production.
Collapse
Affiliation(s)
- A J Ingram
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada.
| | | | | | | | | |
Collapse
|
49
|
Abstract
Altered growth of renal cells is one of the early abnormalities detected after the onset of diabetes. Cell culture studies whereby renal cells are exposed to high glucose concentrations have provided a considerable amount of insight into mechanisms of growth. In the glomerular compartment, there is a very early and self-limited proliferation of mesangial cells with subsequent hypertrophy, whereas proximal tubular cells primarily undergo hypertrophy. There is overwhelming evidence from in vivo and cell culture studies that induction of the transforming growth factor-beta (TGF-beta) system mediates the actions of high ambient glucose and that this system is pivotal for the hypertrophy of mesangial and tubular cells. Other factors such as hemodynamic forces, protein glycation products, and several mediators (for example, angiotensin II, endothelin-1, thromboxane, and platelet-derived growth factor) may further amplify the synthesis of TGF-beta and/or the expression of its receptors in the diabetic state. Cellular hypertrophy can be characterized by cell cycle arrest in the G1 phase. The molecular mechanism arresting mesangial cells in the G1 phase of the cell cycle is the induction of cyclin-dependent kinase (CdK) inhibitors such as p27Kip1 and p21, which bind to and inactivate cyclin-CdK complexes responsible for G1-phase exit. High-glucose-induced activation of protein kinase C and stimulated TGF-beta expression appear to be essential for stimulated expression of p27Kip1. In addition, a decreased turnover of protein caused by the inhibition of proteases contributes to hypertrophy. The development of irreversible renal changes in diabetes mellitus such as glomerulosclerosis and tubulointerstitial fibrosis is always preceded by the early hypertrophic processes in the glomerular and the tubular compartments. It may still be debated whether diabetic renal hypertrophy will inevitably lead to irreversible fibrotic changes in the absence of other factors such as altered intraglomerular hemodynamics and genetic predisposition. Nevertheless, understanding cellular growth on a molecular level may help design a novel therapeutic approach to prevent or treat diabetic nephropathy effectively.
Collapse
Affiliation(s)
- G Wolf
- Department of Medicine, University of Hamburg, Germany
| | | |
Collapse
|
50
|
Kang MJ, Wu X, Ly H, Thai K, Scholey JW. Effect of glucose on stress-activated protein kinase activity in mesangial cells and diabetic glomeruli. Kidney Int 1999; 55:2203-14. [PMID: 10354269 DOI: 10.1046/j.1523-1755.1999.00488.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND We have reported that hyperglycemia increases c-jun mRNA levels in isolated glomeruli of diabetic rats. The transcriptional activity of c-jun can be modified by phosphorylation of serine residues in the regulatory domain of the protein by stress-activated protein kinases (SAPKs), but the effect of high glucose concentrations on SAPK expression and activity is unknown. Accordingly, we studied p42/44 MAPK, p38 MAPK, and SAPK expression and activity in primary mesangial cells exposed to high glucose concentrations, as well as SAPK expression and activity in glomeruli of normal and streptozotocin-induced diabetic rats. METHODS Mesangial cells were incubated in 40 mM glucose for 30 and 60 minutes and 6, 12, 24, and 48 hours, whereas glomeruli of streptozotocin-induced diabetic rats were isolated one day and one and two weeks after the onset of hyperglycemia (blood glucose levels more than 15 mmol/liter), and were compared with age-matched normal rats. Cell lysates were subjected to Western blot analysis of SAPK and phosphorylated SAPK and an in vitro SAPK assay using recombinant c-jun. RESULTS Western blot analysis revealed that SAPK was expressed, but unphosphorylated, in unstimulated mesangial cells and whole glomerular lysates from normal rats. In accord with these observations, no SAPK activity was detected in lysates from mesangial cells or whole glomeruli from normal rats, although mesangial cell SAPK activity was readily induced in vitro by sorbitol. High glucose concentrations did not increase SAPK activity or lead to detectable phosphorylated SAPK either in vitro or in vivo. In contrast, short-term exposure to 40 mM of glucose activated both p42/44 MAPK and p38 MAPK. CONCLUSIONS We conclude that high glucose concentrations do not activate SAPK in primary cultured mesangial cells or in diabetic glomeruli during the early phase of diabetic renal hypertrophy.
Collapse
Affiliation(s)
- M J Kang
- Department of Medicine, University of Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|