1
|
Giglio RV, Papanas N, Rizvi AA, Ciaccio M, Patti AM, Ilias I, Pantea Stoian A, Sahebkar A, Janez A, Rizzo M. An Update on the Current and Emerging Use of Thiazolidinediones for Type 2 Diabetes. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:1475. [PMID: 36295635 PMCID: PMC9609741 DOI: 10.3390/medicina58101475] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 10/09/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022]
Abstract
Guidelines have increasingly stressed the concept that adequate glycemic control is required to prevent or decrease the macro- and microvascular complications of type 2 diabetes mellitus (T2DM). PPAR-gamma agonists ("glitazones") are no longer prioritized due to their effects on heart failure. However, the association between these drugs and innovative therapies could be a valuable tool to attenuate the risk factors of the metabolic syndrome. Glitazones are used for the treatment of diabetes and associated comorbidities. There is substantial scientific evidence demonstrating the effect of glitazones at a cardiometabolic level, as well as on hematological and neurological pathologies that point to their usefulness. The use of glitazones has always been controversial both for the type of patients who must take these drugs and for the side effects associated with them. Unfortunately, the recent guidelines do not include them among the preferred drugs for the treatment of hyperglycemia and rosiglitazone is out of the market in many countries due to an adverse cardiovascular risk profile. Even though real-life studies have proven otherwise, and their pleiotropic effects have been highlighted, they have been unable to achieve primacy in the choice of antihyperglycemic drugs. It would be appropriate to demonstrate the usefulness of pioglitazone and its therapeutic benefit with further cardiovascular safety studies.
Collapse
Affiliation(s)
- Rosaria Vincenza Giglio
- Department of Biomedicine, Neuroscience, and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy
- Department of Laboratory Medicine, University Hospital “Paolo Giaccone”, 90127 Palermo, Italy
| | - Nikolaos Papanas
- Diabetes Centre, Second Department of Internal Medicine, Democritus University of Thrace, University Hospital of Alexandroupolis, 68132 Alexandroupoli, Greece
| | - Ali Abbas Rizvi
- Department of Medicine, University of Central Florida College of Medicine, Orlando, FL 32827, USA
| | - Marcello Ciaccio
- Department of Biomedicine, Neuroscience, and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy
- Department of Laboratory Medicine, University Hospital “Paolo Giaccone”, 90127 Palermo, Italy
| | - Angelo Maria Patti
- Promise Department, School of Medicine, University of Palermo, 90133 Palermo, Italy
| | - Ioannis Ilias
- Department of Endocrinology, Diabetes and Metabolism, Elena Venizelou Hospital, 11521 Athens, Greece
| | - Anca Pantea Stoian
- Faculty of Medicine, Diabetes, Nutrition and Metabolic Diseases, Carol Davila University, 050474 Bucharest, Romania
| | - Amirhossein Sahebkar
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 1696700, Iran
| | - Andrej Janez
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Centre, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Manfredi Rizzo
- Promise Department, School of Medicine, University of Palermo, 90133 Palermo, Italy
- Faculty of Medicine, Diabetes, Nutrition and Metabolic Diseases, Carol Davila University, 050474 Bucharest, Romania
| |
Collapse
|
2
|
Gao J, Gu Z. The Role of Peroxisome Proliferator-Activated Receptors in Kidney Diseases. Front Pharmacol 2022; 13:832732. [PMID: 35308207 PMCID: PMC8931476 DOI: 10.3389/fphar.2022.832732] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/14/2022] [Indexed: 12/20/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear hormone receptor superfamily of ligand-activated transcription factors. Accumulating evidence suggests that PPARs may play an important role in the pathogenesis of kidney disease. All three members of the PPAR subfamily, PPARα, PPARβ/δ, and PPARγ, have been implicated in many renal pathophysiological conditions, including acute kidney injury, diabetic nephropathy, and chronic kidney disease, among others. Emerging data suggest that PPARs may be potential therapeutic targets for renal disease. This article reviews the physiological roles of PPARs in the kidney and discusses the therapeutic utility of PPAR agonists in the treatment of kidney disease.
Collapse
Affiliation(s)
- Jianjun Gao
- Department of Nephrology, Chinese PLA Strategic Support Force Characteristic Medical Center, Beijing, China
| | - Zhaoyan Gu
- Department of Endocrinology, Second Medical Center, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
- *Correspondence: Zhaoyan Gu,
| |
Collapse
|
3
|
Jones BA, Wang XX, Myakala K, Levi M. Nuclear Receptors and Transcription Factors in Obesity-Related Kidney Disease. Semin Nephrol 2021; 41:318-330. [PMID: 34715962 PMCID: PMC10187996 DOI: 10.1016/j.semnephrol.2021.06.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Both obesity and chronic kidney disease are increasingly common causes of morbidity and mortality worldwide. Although obesity often co-exists with diabetes and hypertension, it has become clear over the past several decades that obesity is an independent cause of chronic kidney disease, termed obesity-related glomerulopathy. This review defines the attributes of obesity-related glomerulopathy and describes potential pharmacologic interventions. Interventions discussed include peroxisome proliferator-activated receptors, the farnesoid X receptor, the Takeda G-protein-coupled receptor 5, and the vitamin D receptor.
Collapse
Affiliation(s)
- Bryce A Jones
- Department of Pharmacology and Physiology, Georgetown University, Washington, DC
| | - Xiaoxin X Wang
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, DC
| | - Komuraiah Myakala
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, DC
| | - Moshe Levi
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, DC.
| |
Collapse
|
4
|
Kim KS, Hong S, Ahn HY, Park CY. Comparative Efficacy of Lobeglitazone Versus Pioglitazone on Albuminuria in Patients with Type 2 Diabetes Mellitus. Diabetes Ther 2021; 12:171-181. [PMID: 33099742 PMCID: PMC7843821 DOI: 10.1007/s13300-020-00948-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 10/10/2020] [Indexed: 10/31/2022] Open
Abstract
INTRODUCTION The aim of this analysis was to evaluate the efficacy of lobeglitazone on albuminuria at 24 weeks of follow-up in patients with type 2 diabetes mellitus (T2DM) compared with pioglitazone using data from a randomized, double-blinded phase III trial. METHODS In the phase III trial, patients who were inadequately controlled with metformin received 0.5 mg of lobeglitazone or 15 mg of pioglitazone for 24 weeks. Post hoc, exploratory analysis was used to investigate mean changes from baseline in the urine albumin-creatinine ratio (UACR) between the lobeglitazone (N = 104) and pioglitazone (N = 101) treatment groups. RESULTS After 24 weeks of treatment, UACR was slightly decreased in the lobeglitazone group (- 4.3 mg/g creatinine [Cr]) compared to baseline and slightly increased in the pioglitazone group (5.2 mg/g Cr), with no change in the estimated glomerular filtration rate in either group; this difference was not statistically significant (P = 0.476). The incidence of new-onset microalbuminuria (2.4%) and the progression of albuminuria by > 1 stage (2.9%) in the lobeglitazone group were lower than the respective values in the pioglitazone group (6.8 and 6.1%, respectively). Of the patients in the lobeglitazone group, 50% exhibited regression to normoalbuminuria, compared to 39.3% of the patients in the pioglitazone. In subjects in the lobeglitazone group with micro- and macroalbuminuria, UACR tended to be more decreased and HbA1c was more reduced compared to those with normoalbuminuria (P = 0.014). CONCLUSION Lobeglitazone had a tendency to improve albuminuria in patients with T2DM and had comparable effects on albuminuria as pioglitazone which has demonstrated beneficial effects. TRIAL REGISTRATION ClinicalTrials.gov identifier, NCT01106131.
Collapse
Affiliation(s)
- Kyung-Soo Kim
- Department of Internal Medicine, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Republic of Korea
| | - Sangmo Hong
- Department of Internal Medicine, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Republic of Korea
| | - Hong-Yup Ahn
- Department of Statistics, Dongguk University-Seoul, Seoul, Republic of Korea
| | - Cheol-Young Park
- Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
5
|
Zhou Y, Huang Y, Ji X, Wang X, Shen L, Wang Y. Pioglitazone for the Primary and Secondary Prevention of Cardiovascular and Renal Outcomes in Patients with or at High Risk of Type 2 Diabetes Mellitus: A Meta-Analysis. J Clin Endocrinol Metab 2020; 105:5673086. [PMID: 31822895 DOI: 10.1210/clinem/dgz252] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 12/09/2019] [Indexed: 12/15/2022]
Abstract
CONTEXT The goal of the meta-analysis was to evaluate the effect of pioglitazone on the primary and secondary prevention of cardiovascular diseases (CVDs) and renal adverse events in patients with or at high risk of type 2 diabetes mellitus (T2DM). DESIGN Randomized controlled trials (RCTs) comparing pioglitazone with any control were identified through PubMed, Embase, and the Cochrane Library. Cardiovascular outcomes included major adverse cardiovascular events (MACEs, defined as the composite of nonfatal myocardial infarction, nonfatal stroke, and cardiovascular death), hospitalization for heart failure, and all-cause mortality. Renal outcomes included change in urinary albumin to creatinine ratio and 24-hour urinary protein excretion. Weighted mean difference (WMD) and risk ratio (RR) with 95% confidence intervals (CIs) were pooled. RESULTS A total of 26 studies with 19 645 participants were enrolled. Pioglitazone reduced the risk of MACE (RR, 0.8 [95% CI, 0.7-0.9]), with benefit only seen in patients with a history of established CVDs (0.8 [0.7-0.9]) and not in those without (1.0 [0.7-1.3]). Regarding the individual components, pioglitazone reduced the risk of nonfatal myocardial infarction (0.8 [0.6-1.0]) and nonfatal stroke (0.8 [0.7-0.9]), which was confined to patients with a history of established CVDs, whereas no treatment effect was found on cardiovascular death (1.0 [0.7-1.2]) regardless of the presence of established CVDs. Pioglitazone increased the risk of hospitalization for heart failure (1.3 [1.1-1.6]) and had no treatment effect on all-cause mortality (1.0 [0.8-1.1]). Pioglitazone reduced albuminuria by 18.5% (WMD 18.5% [95% CI, 21.1-16.0]), with a similar benefit in patients with different renal function categories. CONCLUSIONS Pioglitazone should be considered in patients with or at high risk of T2DM for the prevention of cardiovascular endpoints, especially in those with a history of established CVD who might benefit the most. Robust reductions in progression of renal disease are seen regardless of baseline renal function degree.
Collapse
Affiliation(s)
- Yue Zhou
- Department of Endocrinology, Affiliated Hospital of Medical College Qingdao University, Qingdao, China
| | - Yajing Huang
- Department of Endocrinology, Affiliated Hospital of Medical College Qingdao University, Qingdao, China
| | - Xiaoyun Ji
- Department of Endocrinology, Affiliated Hospital of Medical College Qingdao University, Qingdao, China
| | - Xiang Wang
- Department of Endocrinology, Affiliated Hospital of Medical College Qingdao University, Qingdao, China
| | - Liyan Shen
- Department of Endocrinology, Affiliated Hospital of Medical College Qingdao University, Qingdao, China
| | - Yangang Wang
- Department of Endocrinology, Affiliated Hospital of Medical College Qingdao University, Qingdao, China
| |
Collapse
|
6
|
Wang L, Chang JH, Buckley AF, Spurney RF. Knockout of TRPC6 promotes insulin resistance and exacerbates glomerular injury in Akita mice. Kidney Int 2020; 95:321-332. [PMID: 30665571 DOI: 10.1016/j.kint.2018.09.026] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 09/22/2018] [Accepted: 09/27/2018] [Indexed: 12/17/2022]
Abstract
Gain-of-function mutations in TRPC6 cause familial focal segmental glomerulosclerosis, and TRPC6 is upregulated in glomerular diseases including diabetic kidney disease. We studied the effect of systemic TRPC6 knockout in the Akita model of type 1 diabetes. Knockout of TRPC6 inhibited albuminuria in Akita mice at 12 and 16 weeks of age, but this difference disappeared by 20 weeks. Knockout of TRPC6 also reduced tubular injury in Akita mice; however, mesangial expansion was significantly increased. Hyperglycemia and blood pressure were similar between TRPC6 knockout and wild-type Akita mice, but knockout mice were more insulin resistant. In cultured podocytes, knockout of TRPC6 inhibited expression of the calcium/calcineurin responsive gene insulin receptor substrate 2 and decreased insulin responsiveness. Insulin resistance is reported to promote diabetic kidney disease independent of blood glucose levels. While the mechanisms are not fully understood, insulin activates both Akt2 and ERK, which inhibits apoptosis signal regulated kinase 1 (ASK1)-p38-induced apoptosis. In cultured podocytes, hyperglycemia stimulated p38 signaling and induced apoptosis, which was reduced by insulin and ASK1 inhibition and enhanced by Akt or ERK inhibition. Glomerular p38 signaling was increased in TRPC6 knockout Akita mice and was associated with enhanced expression of the p38 gene target cyclooxygenase 2. These data suggest that knockout of TRPC6 in Akita mice promotes insulin resistance and exacerbates glomerular disease independent of hyperglycemia.
Collapse
Affiliation(s)
- Liming Wang
- Division of Nephrology, Department of Medicine, Duke University Health System, Durham, North Carolina, USA
| | - Jae-Hyung Chang
- Division of Nephrology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | - Anne F Buckley
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
| | - Robert F Spurney
- Division of Nephrology, Department of Medicine, Duke University Health System, Durham, North Carolina, USA; Durham VA Medical Center, Durham, North Carolina, USA.
| |
Collapse
|
7
|
Hall G, Wang L, Spurney RF. TRPC Channels in Proteinuric Kidney Diseases. Cells 2019; 9:cells9010044. [PMID: 31877991 PMCID: PMC7016871 DOI: 10.3390/cells9010044] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 12/18/2019] [Accepted: 12/19/2019] [Indexed: 12/20/2022] Open
Abstract
Over a decade ago, mutations in the gene encoding TRPC6 (transient receptor potential cation channel, subfamily C, member 6) were linked to development of familial forms of nephrosis. Since this discovery, TRPC6 has been implicated in the pathophysiology of non-genetic forms of kidney disease including focal segmental glomerulosclerosis (FSGS), diabetic nephropathy, immune-mediated kidney diseases, and renal fibrosis. On the basis of these findings, TRPC6 has become an important target for the development of therapeutic agents to treat diverse kidney diseases. Although TRPC6 has been a major focus for drug discovery, more recent studies suggest that other TRPC family members play a role in the pathogenesis of glomerular disease processes and chronic kidney disease (CKD). This review highlights the data implicating TRPC6 and other TRPC family members in both genetic and non-genetic forms of kidney disease, focusing on TRPC3, TRPC5, and TRPC6 in a cell type (glomerular podocytes) that plays a key role in proteinuric kidney diseases.
Collapse
|
8
|
Wu L, Wang Q, Guo F, Ma X, Ji H, Liu F, Zhao Y, Qin G. MicroRNA-27a Induces Mesangial Cell Injury by Targeting of PPARγ, and its In Vivo Knockdown Prevents Progression of Diabetic Nephropathy. Sci Rep 2016; 6:26072. [PMID: 27184517 PMCID: PMC4869109 DOI: 10.1038/srep26072] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 04/26/2016] [Indexed: 11/09/2022] Open
Abstract
MicroRNAs play important roles in the pathogenesis of diabetic nephropathy (DN). In this study, we found that high glucose upregulated miR-27a expression in cultured glomerular mesangial cells and in the kidney glomeruli of streptozotocin (STZ)-induced diabetic rats. miR-27a knockdown prevented high glucose-induced mesangial cell proliferation and also blocked the upregulation of extracellular matrix (ECM)-associated profibrotic genes. Reduction of cell proliferation and profibrotic gene expression by a miR-27a inhibitor depended upon the expression of peroxisome proliferator-activated receptor γ (PPARγ). Further studies showed that miR-27a negatively regulated PPARγ expression by binding to the 3'-untranslated region of rat PPARγ. An antisense oligonucleotide specific to miR-27a (antagomir-27a) significantly reduced renal miR-27a expression in STZ-induced diabetic rats and significantly increased PPARγ levels. Antagomir-27a also reduced kidney ECM accumulation and proteinuria in STZ-induced diabetic rats. These findings suggest that specific reduction of renal miR-27a decreases renal fibrosis, which may be explained in part by its regulation of PPARγ, and that targeting miR-27a may represent a novel therapeutic approach for DN.
Collapse
Affiliation(s)
- Lina Wu
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.,Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Qingzhu Wang
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Feng Guo
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xiaojun Ma
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Hongfei Ji
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.,Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Fei Liu
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yanyan Zhao
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Guijun Qin
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| |
Collapse
|
9
|
Yogarajah T, Bee YTG, Noordin R, Yin KB. Increased peroxisome proliferator-activated receptor γ expression levels in visceral adipose tissue, and serum CCL2 and interleukin-6 levels during visceral adipose tissue accumulation. Mol Med Rep 2014; 11:515-20. [PMID: 25324014 DOI: 10.3892/mmr.2014.2686] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 06/05/2014] [Indexed: 11/06/2022] Open
Abstract
This study was conducted to determine the mRNA and protein expression levels of peroxisome proliferator-activated receptors (PPARs) in visceral adipose tissue, as well as serum adipokine levels, in Sprague Dawley rats. The rats were fed either a normal (control rats) or excessive (experimental rats) intake of food for 8 or 16 weeks, then sacrificed, at which time visceral and subcutaneous adipose tissues, as well as blood samples, were collected. The mRNA and protein expression levels of PPARs in the visceral adipose tissues were determined using reverse transcription-polymerase chain reaction and Western blotting, respectively. In addition, the levels of adipokines in the serum samples were determined using commercial ELISA kits. The results revealed that at 8 weeks, the mass of subcutaneous adipose tissue was higher than that of the visceral adipose tissue in the experimental rats, but the reverse occurred at 16 weeks. Furthermore, at 16 weeks the experimental rats exhibited an upregulation of PPARγ mRNA and protein expression levels in the visceral adipose tissues, and significant increases in the serum levels of CCL2 and interleukin (IL)-6 were observed, compared with those measured at 8 weeks. In conclusion, this study demonstrated that the PPARγ expression level was likely correlated with serum levels of CCL2 and IL-6, molecules that may facilitate visceral adipose tissue accumulation. In addition, the levels of the two adipokines in the serum may be useful as surrogate biomarkers for the expression levels of PPARγ in accumulated visceral adipose tissues.
Collapse
Affiliation(s)
- Thaneswary Yogarajah
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Yvonne-Tee Get Bee
- School of Health Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Rahmah Noordin
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Khoo Boon Yin
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800 Penang, Malaysia
| |
Collapse
|
10
|
Takagi H, Yamamoto H, Iwata K, Goto SN, Umemoto T. Effects of telmisartan on proteinuria or albuminuria: A meta-analysis of randomized trials. Int J Cardiol 2013; 167:1443-9. [DOI: 10.1016/j.ijcard.2012.04.058] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2011] [Revised: 03/08/2012] [Accepted: 04/08/2012] [Indexed: 10/28/2022]
|
11
|
Sun YM, Su Y, Li J, Wang LF. Recent advances in understanding the biochemical and molecular mechanism of diabetic nephropathy. Biochem Biophys Res Commun 2013; 433:359-61. [PMID: 23541575 DOI: 10.1016/j.bbrc.2013.02.120] [Citation(s) in RCA: 171] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 02/26/2013] [Indexed: 12/13/2022]
Abstract
Diabetic nephropathy (DN) is a chronic disease characterized by proteinuria, glomerular hypertrophy, decreased glomerular filtration and renal fibrosis with loss of renal function. DN is the leading cause of end-stage renal disease, accounting for millions of deaths worldwide. Hyperglycemia is the driving force for the development of diabetic nephropathy. The exact cause of diabetic nephropathy is unknown, but various postulated mechanisms are: hyperglycemia (causing hyperfiltration and renal injury), advanced glycosylation products, activation of cytokines. In this review article, we have discussed a number of diabetes-induced metabolites such as glucose, advanced glycation end products, protein kinase C and oxidative stress and other related factors that are implicated in the pathophysiology of the DN. An understanding of the biochemical and molecular changes especially early in the DN may lead to new and effective therapies towards prevention and amelioration of DN.
Collapse
Affiliation(s)
- Yan-Ming Sun
- Department of Cardiac Care Unit, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | | | | | | |
Collapse
|
12
|
Abstract
The most problematic issue in clinical nephrology is the relentless and progressive increase in patients with ESRD (end-stage renal disease) worldwide. The impact of diabetic nephropathy on the increasing population with CKD (chronic kidney disease) and ESRD is enormous. Three major pathways showing abnormality of intracellular metabolism have been identified in the development of diabetic nephropathy: (i) the activation of polyol and PKC (protein kinase C) pathways; (ii) the formation of advanced glycation end-products; and (iii) intraglomerular hypertension induced by glomerular hyperfiltration. Upstream of these three major pathways, hyperglycaemia is the major driving force of the progression to ESRD from diabetic nephropathy. Downstream of the three pathways, microinflammation and subsequent extracellular matrix expansion are common pathways for the progression of diabetic nephropathy. In recent years, many researchers have been convinced that the inflammation pathways play central roles in the progression of diabetic nephropathy, and the identification of new inflammatory molecules may link to the development of new therapeutic strategies. Various molecules related to the inflammation pathways in diabetic nephropathy include transcription factors, pro-inflammatory cytokines, chemokines, adhesion molecules, Toll-like receptors, adipokines and nuclear receptors, which are candidates for the new molecular targets for the treatment of diabetic nephropathy. Understanding of these molecular pathways of inflammation would translate into the development of anti-inflammation therapeutic strategies.
Collapse
|
13
|
Tovar-Palacio C, Torres N, Diaz-Villaseñor A, Tovar AR. The role of nuclear receptors in the kidney in obesity and metabolic syndrome. GENES AND NUTRITION 2012; 7:483-98. [PMID: 22532116 DOI: 10.1007/s12263-012-0295-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 04/02/2012] [Indexed: 02/06/2023]
Abstract
Nuclear receptors are ligand-activated transcriptional regulators of several key aspects of renal physiology and pathophysiology. As such, nuclear receptors control a large variety of metabolic processes, including kidney lipid metabolism, drug clearance, inflammation, fibrosis, cell differentiation, and oxidative stress. Derangement of nuclear receptor regulation, that is, mainly due to obesity may induce metabolic syndrome, may contribute to the pathogenesis and progression of chronic renal disease and may result in end-stage renal disease. This places nuclear receptors at the forefront of novel therapeutic approaches for a broad range of kidney disorders and diseases, including glomerulosclerosis, tubulointerstitial disease, renal lipotoxicity, kidney fibrosis, and hypertension. This review focuses on the importance of the transcription factors peroxisome proliferator-activated receptor alpha, peroxisome proliferator-activated receptor beta, peroxisome proliferator-activated receptor gamma, liver X receptors, farnesoid X receptor, and the pregnane X receptor/steroid and xenobiotic receptor (PXR) on the physiology and pathophysiology of renal diseases associated with obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Claudia Tovar-Palacio
- Department of Nephrology and Mineral Metabolism, National Medical Science and Nutrition Institute, Salvador Zubirán, Vasco de Quiroga No. 15, Tlalpan, 14000, Mexico, D.F., Mexico,
| | | | | | | |
Collapse
|
14
|
PPARγ as a therapeutic target in diabetic nephropathy and other renal diseases. Curr Opin Nephrol Hypertens 2012; 21:97-105. [PMID: 22143250 DOI: 10.1097/mnh.0b013e32834de526] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW Peroxisome proliferator-activated receptor gamma (PPARγ) is a ligand-activated nuclear transcription factor that regulates many important physiological processes including glucose and lipid metabolism, energy homeostasis, cell proliferation, inflammation, immunity and reproduction. The current review aims to summarize and discuss recent findings evaluating the protective effects of PPARγ against kidney diseases with a focus on diabetic nephropathy. We will also delineate the potential underlying mechanisms. RECENT FINDINGS PPARγ plays important roles in renal physiology and pathophysiology. Agonists of PPARγ exert protective effects against various kidney diseases including diabetic nephropathy, ischemic renal injury, IgA nephropathy, chemotherapy-associated kidney damage, polycystic kidney diseases and age-related kidney diseases via both systemic and renal actions. SUMMARY PPARγ agonists are effective in delaying and even preventing the progression of many renal diseases, especially diabetic nephropathy. PPARγ may represent a promising target for the treatment of renal diseases.
Collapse
|
15
|
Bolignano D, Zoccali C. Glitazones in chronic kidney disease: potential and concerns. Nutr Metab Cardiovasc Dis 2012; 22:167-175. [PMID: 22364889 DOI: 10.1016/j.numecd.2011.11.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Revised: 11/07/2011] [Accepted: 11/13/2011] [Indexed: 11/17/2022]
Abstract
AIMS Glitazones rank now among the most used hypoglycemic agents in patients with type-2 diabetes. This systematic review focuses on the cardiovascular and renal outcomes in chronic kidney disease (CKD) patients treated with these drugs. DATA SYNTHESIS Data from randomized clinical trials and a meta-analysis indicate that glitazones (particularly rosiglitazone) may increase the risk of myocardial infarction, heart failure and cardiovascular death in type-2 diabetics. Observational studies looking at survival and cardiovascular outcomes in diabetic patients with kidney failure show controversial results. Studies in experimental models and clinical studies suggest that glitazones may have favorable effects on renal disease progression, because these drugs coherently reduce urinary albumin excretion and proteinuria in diabetic and non-diabetic nephropathies. No clinical trial based on clinical end-points like kidney failure has until now tested the effect of glitazones on the evolution of chronic renal failure in these patients. CONCLUSIONS Whether the use of glitazones has a positive or a negative impact upon major cardiovascular and renal outcomes in diabetic patients remains an open, unanswered question. Specific studies are needed to assess the efficacy and safety of glitazones in a high risk population like type-2 diabetics with chronic kidney disease.
Collapse
Affiliation(s)
- D Bolignano
- CNR-IBIM, Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension of Reggio Calabria, Reggio Calabria, Italy
| | | |
Collapse
|
16
|
Bondar' IA, Klimontov VV. The role of matrix metalloproteinases and their inhibitors in the development of renal fibrosis in the patients with diabetes mellitus. ACTA ACUST UNITED AC 2012. [DOI: 10.14341/probl201258139-44] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The accumulation of components of extracellular matrix in the glomerular and interstitial compartments of the kidneys is a characteristic feature of diabetic nephropathy. The leading role in the extracellular matrix catabolism is played by matrix metalloproteinases (MMP). The activity of these enzymes is regulated by a group of inhibitors including tissue metalloproteinase inhibitors, plasminogen activator inhibitor-1, etc. Both in vivo and in vitro studies have demonstrated that a reduction of MMP activities and/or an increase of expression of MMP tissue inhibitors in the glomerular and tubular cells result in the suppression of catabolism of the components of extracellular matrix under the hyperglycemic conditions. Both circulating and urinary MMP as well as their inhibitors are considered to be new potential markers of renal fibrosis associated with diabetes mellitus. It is concluded that the directed activation of MMP and neutralization of their inhibitors provide a promising tool for the treatment of diabetic nephropathy.
Collapse
|
17
|
Lee EK, Jeong JU, Chang JW, Yang WS, Kim SB, Park SK, Park JS, Lee SK. Activation of AMP-Activated Protein Kinase Inhibits Albumin-Induced Endoplasmic Reticulum Stress and Apoptosis through Inhibition of Reactive Oxygen Species. ACTA ACUST UNITED AC 2012; 121:e38-48. [DOI: 10.1159/000342802] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Accepted: 08/14/2012] [Indexed: 11/19/2022]
|
18
|
Bilan VP, Salah EM, Bastacky S, Jones HB, Mayers RM, Zinker B, Poucher SM, Tofovic SP. Diabetic nephropathy and long-term treatment effects of rosiglitazone and enalapril in obese ZSF1 rats. J Endocrinol 2011; 210:293-308. [PMID: 21680617 DOI: 10.1530/joe-11-0122] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Diabetic nephropathy (DN) is a major cause of end-stage renal disease. Yet the pathogenic mechanisms underlying the development of DN are not fully defined, partially due to lack of suitable models that mimic the complex pathogenesis of renal disease in diabetic patients. In this study, we describe early and late renal manifestations of DN and renal responses to long-term treatments with rosiglitazone or high-dose enalapril in ZSF1 rats, a model of metabolic syndrome, diabetes, and chronic renal disease. At 8 weeks of age, obese ZSF1 rats developed metabolic syndrome and diabetes (hyperglycemia, glucosuria, hyperlipidemia, and hypertension) and early signs of renal disease (proteinuria, glomerular collagen IV deposition, tubulointerstitial inflammation, and renal hypertrophy). By 32 weeks of age, animals developed renal histopathology consistent with DN, including mesangial expansion, glomerulosclerosis, tubulointerstitial inflammation and fibrosis, tubular dilation and atrophy, and arteriolar thickening. Rosiglitazone markedly increased body weight but reduced food intake, improved glucose control, and attenuated hyperlipidemia and liver and kidney injury. In contrast, rosiglitazone markedly increased cardiac hypertrophy via a blood pressure-independent mechanism. High-dose enalapril did not improve glucose homeostasis, but normalized blood pressure, and nearly prevented diabetic renal injury. The ZSF1 model thus detects the clinical observations seen with rosiglitazone and enalapril in terms of primary and secondary endpoints of cardiac and renal effects. This and previous reports indicate that the obese ZSF1 rat meets currently accepted criteria for progressive experimental diabetic renal disease in rodents, suggesting that this may be the best available rat model for simulation of human DN.
Collapse
Affiliation(s)
- Victor P Bilan
- Division of Pulmonary, Allergy, and Critical Care Medicine, Vascular Medicine Institute, Departments of Medicine Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Wang WM, Chen H, Zhong F, Lu Y, Han L, Chen N. Inhibitory effects of rosiglitazone on lipopolysaccharide-induced inflammation in a murine model and HK-2 cells. Am J Nephrol 2011; 34:152-62. [PMID: 21734368 DOI: 10.1159/000329120] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2010] [Accepted: 05/04/2011] [Indexed: 11/19/2022]
Abstract
BACKGROUND Inflammation may play an important role in the pathogenesis of kidney disease. Agonists of the peroxisome proliferator-activated receptor-γ (PPAR-γ), such as rosiglitazone, have been recently demonstrated to regulate inflammation by modulating the production of inflammatory mediators. The purpose of this study was to examine the effects of rosiglitazone on lipopolysaccharide (LPS)-induced kidney inflammation and to explore the mechanism of its renoprotection. METHODS Mice were treated with LPS with or without pretreatment with rosiglitazone. Blood urea nitrogen (BUN), creatinine levels, the urinary albumin-to-creatinine ratio, macrophage infiltration, monocyte chemoattractant protein-1 (MCP-1) expression, PPAR-γ expression, and NF-κB and PPAR-γ activity were investigated. HK-2 cells were maintained under defined in vitro conditions, treated with either rosiglitazone and/or the PPAR-γ antagonist GW9662, and then stimulated with LPS. MCP-1, IL-8, IL-6, NF-κB activity and PPAR-γ expression were investigated. RESULTS Compared to the LPS only group, pretreatment with rosiglitazone in vivo significantly attenuated the BUN levels macrophage infiltration, MCP-1 overexpression and NF-κB activity (p < 0.05). Rosiglitazone also restored PPAR-γ expression and protein activity, which were reduced significantly in the LPS only group (p < 0.05). Furthermore, in the LPS-stimulated HK-2 cells, rosiglitazone downregulated MCP-1, IL-8 and IL-6 expression as well as NF-κB activation and increased PPAR-γ expression (p < 0.05). These effects were diminished by GW9662. CONCLUSION These results showed that pretreatment with rosiglitazone could attenuate kidney inflammation through the activation of PPAR-γ, suppression of MCP-1 overproduction and NF-κB activation. Rosiglitazone had a protective effect via a PPAR-γ-dependent pathway in LPS-treated HK-2 cells.
Collapse
Affiliation(s)
- W M Wang
- Department of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University, PR China
| | | | | | | | | | | |
Collapse
|
20
|
PPARγ and chronic kidney disease. Pediatr Nephrol 2011; 26:347-51. [PMID: 20676693 DOI: 10.1007/s00467-010-1602-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2010] [Revised: 06/23/2010] [Accepted: 06/30/2010] [Indexed: 10/19/2022]
Abstract
Peroxisome proliferator-activated receptor-γ (PPARγ) agonists, exemplified by the thiazolidinediones (TZDs), have been used extensively for their beneficial effects to improve insulin sensitivity and lipid metabolism in type 2 diabetic patients. PPARγ receptors are part of the steroid hormone nuclear receptor family and, when activated by agonist binding, can affect numerous target genes expressing PPAR response elements. Results from experimental studies and a limited number of studies in humans suggest that PPARγ agonists have manifold effects beyond those on dysmetabolic syndrome. These potentially beneficial actions are mediated via renal parenchymal and infiltrating cells and modulate fibrotic, inflammatory, immune, proliferative, reactive oxygen and mitochondrial injury pathways. Thus, the potential benefits of TZDs in chronic kidney disease impact numerous pathogenic pathways. This review will focus on evidence of the effects of TZDs in nondiabetic chronic kidney disease in experimental and human disease settings.
Collapse
|
21
|
Matsushita Y, Ogawa D, Wada J, Yamamoto N, Shikata K, Sato C, Tachibana H, Toyota N, Makino H. Activation of peroxisome proliferator-activated receptor delta inhibits streptozotocin-induced diabetic nephropathy through anti-inflammatory mechanisms in mice. Diabetes 2011; 60:960-8. [PMID: 21270242 PMCID: PMC3046857 DOI: 10.2337/db10-1361] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Activation of the nuclear hormone receptor peroxisome proliferator-activated receptor (PPAR)-δ has been shown to improve insulin resistance, adiposity, and plasma HDL levels. Several studies have reported that activation of PPARδ is atheroprotective; however, the role of PPARδ in renal function remains unclear. Here, we report the renoprotective effects of PPARδ activation in a model of streptozotocin-induced diabetic nephropathy. RESEARCH DESIGN AND METHODS Eight-week-old male C57BL/6 mice were divided into three groups: 1) nondiabetic control mice, 2) diabetic mice, and 3) diabetic mice treated with the PPARδ agonist GW0742 (1 mg/kg/day). GW0742 was administered by gavage for 8 weeks after inducing diabetes. RESULTS GW0742 decreased urinary albumin excretion without altering blood glucose levels. Macrophage infiltration, mesangial matrix accumulation, and type IV collagen deposition were substantially attenuated by GW0742. The gene expression of inflammatory mediators in the kidney cortex, such as monocyte chemoattractant protein-1 (MCP-1) and osteopontin (OPN), was also suppressed. In vitro studies demonstrated that PPARδ activation increased the expression of anti-inflammatory corepressor B-cell lymphoma-6, which subsequently suppressed MCP-1 and OPN expression. CONCLUSIONS These findings uncover a previously unrecognized mechanism for the renoprotective effects of PPARδ agonists and support the concept that PPARδ agonists may offer a novel therapeutic approach for the treatment of diabetic nephropathy.
Collapse
Affiliation(s)
- Yuichi Matsushita
- Department of Medicine and Clinical Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Daisuke Ogawa
- Department of Medicine and Clinical Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
- Department of Diabetic Nephropathy, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
- Corresponding author: Daisuke Ogawa,
| | - Jun Wada
- Department of Medicine and Clinical Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Noriko Yamamoto
- Department of Medicine and Clinical Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kenichi Shikata
- Department of Medicine and Clinical Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
- Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama, Japan
| | - Chikage Sato
- Department of Medicine and Clinical Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
- Department of Diabetic Nephropathy, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hiromi Tachibana
- Department of Medicine and Clinical Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Noriko Toyota
- Department of Medicine and Clinical Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hirofumi Makino
- Department of Medicine and Clinical Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
22
|
Lepenies J, Hewison M, Stewart PM, Quinkler M. Renal PPARγ mRNA expression increases with impairment of renal function in patients with chronic kidney disease. Nephrology (Carlton) 2011; 15:683-91. [PMID: 21040163 DOI: 10.1111/j.1440-1797.2010.01339.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
AIM Peroxisome proliferator-activated receptor gamma (PPARγ) is generally accepted as renoprotective factor in type 2 diabetes mellitus, and PPARγ agonists have been reported to reduce albuminuria. However, little is known about renal PPARγ expression in chronic kidney disease, and especially human data are scarce. We hypothesized that renal PPARγ expression is associated with extent of proteinuria, kidney function, histological diagnosis and inflammatory mediators. Therefore, we investigated PPARγ mRNA expression in human kidney biopsies. METHODS We quantified PPARγ mRNA as well as the expression of macrophage chemoattractant protein-1, transforming growth factor beta-1 and interleukin-6 in 64 human kidney biopsies from patients with chronic kidney disease and mild-to-marked proteinuria of diverse aetiology. We measured renal function, and macrophage invasion was quantified by CD68 and vascularization by CD34 immunostaining. RESULTS PPARγ mRNA expression correlated inversely with renal function. Higher blood pressure levels were associated with higher PPARγ expression levels. PPARγ mRNA expression correlated significantly (P<0.001) with macrophage chemoattractant protein-1 mRNA expression and showed a negative trend with transforming growth factor beta-1 mRNA expression. No differences in PPARγ expression were detected with regard to extent of proteinuria, histological diagnosis, macrophage invasion, interleukin-6 expression, and age or body mass index. CONCLUSIONS PPARγ expression increases with loss of renal function and may be an important factor in maintaining normal renal function serving as a key protective mechanism to renal injury.
Collapse
Affiliation(s)
- Julia Lepenies
- KfH Dialysis Center Bismarckstrasse, Clinical Endocrinology, Charite Campus Mitte, Charite University Medicine Berlin, Berlin, Germany
| | | | | | | |
Collapse
|
23
|
Welsh GI, Hale LJ, Eremina V, Jeansson M, Maezawa Y, Lennon R, Pons DA, Owen RJ, Satchell SC, Miles MJ, Caunt CJ, McArdle CA, Pavenstädt H, Tavaré JM, Herzenberg AM, Kahn CR, Mathieson PW, Quaggin SE, Saleem MA, Coward RJM. Insulin signaling to the glomerular podocyte is critical for normal kidney function. Cell Metab 2010; 12:329-340. [PMID: 20889126 PMCID: PMC4949331 DOI: 10.1016/j.cmet.2010.08.015] [Citation(s) in RCA: 352] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2010] [Revised: 05/21/2010] [Accepted: 07/07/2010] [Indexed: 12/21/2022]
Abstract
Diabetic nephropathy (DN) is the leading cause of renal failure in the world. It is characterized by albuminuria and abnormal glomerular function and is considered a hyperglycemic "microvascular" complication of diabetes, implying a primary defect in the endothelium. However, we have previously shown that human podocytes have robust responses to insulin. To determine whether insulin signaling in podocytes affects glomerular function in vivo, we generated mice with specific deletion of the insulin receptor from their podocytes. These animals develop significant albuminuria together with histological features that recapitulate DN, but in a normoglycemic environment. Examination of "normal" insulin-responsive podocytes in vivo and in vitro demonstrates that insulin signals through the MAPK and PI3K pathways via the insulin receptor and directly remodels the actin cytoskeleton of this cell. Collectively, this work reveals the critical importance of podocyte insulin sensitivity for kidney function.
Collapse
Affiliation(s)
- Gavin I Welsh
- Academic Renal Unit, School of Clinical Sciences, University of Bristol, Bristol BS8 1TH, UK
| | - Lorna J Hale
- Academic Renal Unit, School of Clinical Sciences, University of Bristol, Bristol BS8 1TH, UK
| | - Vera Eremina
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Ontario M5G 1X5, Canada
| | - Marie Jeansson
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Ontario M5G 1X5, Canada
| | - Yoshiro Maezawa
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Ontario M5G 1X5, Canada
| | - Rachel Lennon
- Academic Renal Unit, School of Clinical Sciences, University of Bristol, Bristol BS8 1TH, UK
| | - Deborah A Pons
- Academic Renal Unit, School of Clinical Sciences, University of Bristol, Bristol BS8 1TH, UK
| | - Rachel J Owen
- School of Physics, School of Clinical Sciences, University of Bristol, Bristol BS8 1TH, UK
| | - Simon C Satchell
- Academic Renal Unit, School of Clinical Sciences, University of Bristol, Bristol BS8 1TH, UK
| | - Mervyn J Miles
- School of Physics, School of Clinical Sciences, University of Bristol, Bristol BS8 1TH, UK
| | - Christopher J Caunt
- Department of Molecular Pharmacology, School of Clinical Sciences, University of Bristol, Bristol BS8 1TH, UK
| | - Craig A McArdle
- Department of Molecular Pharmacology, School of Clinical Sciences, University of Bristol, Bristol BS8 1TH, UK
| | - Hermann Pavenstädt
- Department of Internal Medicine D, Nephrology and Hypertension, University Clinics Muenster, Muenster 48149, Germany
| | - Jeremy M Tavaré
- School of Biochemistry, School of Clinical Sciences, University of Bristol, Bristol BS8 1TH, UK
| | - Andrew M Herzenberg
- Department of Pathology, University Health Network and University of Toronto, Ontario M5G 2C4, Canada
| | - C Ronald Kahn
- Joslin Diabetes Center and Harvard Medical School, Boston, MA 02215, USA
| | - Peter W Mathieson
- Academic Renal Unit, School of Clinical Sciences, University of Bristol, Bristol BS8 1TH, UK
| | - Susan E Quaggin
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Ontario M5G 1X5, Canada
| | - Moin A Saleem
- Academic Renal Unit, School of Clinical Sciences, University of Bristol, Bristol BS8 1TH, UK
| | - Richard J M Coward
- Academic Renal Unit, School of Clinical Sciences, University of Bristol, Bristol BS8 1TH, UK; Samuel Lunenfeld Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Ontario M5G 1X5, Canada.
| |
Collapse
|
24
|
Abstract
Diabetes mellitus is an established risk factor for cardiovascular disease and the leading cause of end-stage renal disease in the Western World. Thiazolidinediones (TZDs) represent a class of antidiabetic agents that exert their glucose-lowering effects by reducing insulin resistance, through stimulation of a type of nuclear receptor, called peroxisome proliferator-activated receptor-γ. Apart from improving glycemic control, TZDs were shown to exert beneficial effects on several components of the metabolic syndrome and cardiovascular risk markers. Furthermore, background and human studies have shown that TZDs reduce urinary albumin and protein excretion and interfere with most of the pathogenentic pathways involved in the development and progression of diabetic nephropathy. On the other hand, currently used TZDs have side effects, most important of which is fluid retention leading to wait gain and heart failure deterioration. With regards to cardiovascular outcomes, the anticipated benefit of TZDs was demonstrated for pioglitazone, whereas a series of previous meta-analyses linking rosiglitazone treatment with increased risk of myocardial infarction and cardiovascular death raised uncertainty around the cardiovascular safety of rosiglitazone. This article will discuss the effects of TZDs on established and emerging cardiovascular risk factors, the data on possible beneficial renal effects of these compounds, and the existing evidence from large-scale clinical trials and meta-analyses on their effects on cardiovascular outcomes, aiming to provide an overview of the cardio- and renoprotective properties of these drugs.
Collapse
Affiliation(s)
- Pantelis A Sarafidis
- Section of Nephrology and Hypertension, 1st Department of Medicine, AHEPA Hospital, Aristotle University of Thessaloniki, Greece
| | | | | |
Collapse
|
25
|
|
26
|
Nicholas SB, Liu J, Kim J, Ren Y, Collins AR, Nguyen L, Hsueh WA. Critical role for osteopontin in diabetic nephropathy. Kidney Int 2010; 77:588-600. [PMID: 20130530 DOI: 10.1038/ki.2009.518] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The profibrotic adhesion molecule, osteopontin (OPN), is upregulated in kidneys of humans and mice with diabetes. The thiazolidinedione (TZD) insulin sensitizers decrease albuminuria in diabetic nephropathy (DN) and reduce OPN expression in vascular and cardiac tissue. To examine whether OPN is a critical mediator of DN we treated db/db mice with insulin, rosiglitazone, or pioglitazone to achieve similar fasting plasma glucose levels. The urine albumin-to-creatinine ratio and glomerular OPN expression were increased in diabetic mice, but both were reduced by the TZDs more than by insulin. We administered streptozotocin to OPN-null and OPN-wild-type mice, and OPN-null mice were bred into both type 1 (Ins2(akita/+)) and 2 (db/db) diabetic mice. In each case, OPN deletion decreased albuminuria, mesangial area, and glomerular collagen IV, fibronectin and transforming growth factor (TGF)-beta in the diabetic mice compared with their respective controls. In cultured mouse mesangial cells, TZDs but not insulin decreased angiotensin II-induced OPN expression, while recombinant OPN upregulated TGF-beta, ERK/MAPK, and JNK/MAPK signaling. These studies show that OPN expression in DN mouse models enhances glomerular damage, likely through the expression of TGF-beta, while its deletion protects against disease progression, suggesting that OPN might serve as a therapeutic target.
Collapse
Affiliation(s)
- Susanne B Nicholas
- Division of Nephrology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Sarafidis PA, Stafylas PC, Georgianos PI, Saratzis AN, Lasaridis AN. Effect of thiazolidinediones on albuminuria and proteinuria in diabetes: a meta-analysis. Am J Kidney Dis 2010; 55:835-47. [PMID: 20110146 DOI: 10.1053/j.ajkd.2009.11.013] [Citation(s) in RCA: 147] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2009] [Accepted: 11/12/2009] [Indexed: 01/11/2023]
Abstract
BACKGROUND Because of the major clinical and economic burden of diabetic nephropathy, new therapeutic tools to delay its progression are needed. Recent studies suggest that thiazolidinediones have renal benefits. We aimed to evaluate the effect of thiazolidinediones on urinary albumin and protein excretion in patients with diabetes mellitus. STUDY DESIGN Systematic review and meta-analysis by searching MEDLINE/PubMed, EMBASE, and Cochrane CENTRAL databases (1991 to September 2009). SETTING & POPULATION Patients with diabetes mellitus. SELECTION CRITERIA FOR STUDIES Randomized controlled trials. INTERVENTION Thiazolidinediones (rosiglitazone and pioglitazone) compared with placebo or other antidiabetic agents. OUTCOMES Weighted (WMDs) and standardized mean differences (SMDs) for changes in urine albumin or protein excretion between the thiazolidinedione and control groups. RESULTS Of 171 originally identified articles, 15 studies (5 with rosiglitazone and 10 with pioglitazone) involving 2,860 patients were included in the analysis. In participants with baseline normo- or microalbuminuria, the WMD of proportional changes between the thiazolidinedione and control groups in urinary albumin excretion measured using time-specified collections was -64.8% (95% CI, -75.6 to -53.9) and the WMD of changes in albumin-creatinine ratio was -24.8% (95% CI, -39.6 to -10.0). Overall, in participants with normo- and microalbuminuria, thiazolidinedione treatment was associated with a significant decrease in urinary albumin excretion (SMD, -0.6 units of standard deviation [SD]; 95% CI, -0.8 to -0.4). Similarly, thiazolidinediones were associated with a significant decrease in urinary protein excretion in patients with proteinuria (SMD, -1.1 units of SD; 95% CI, -1.8 to -0.4). LIMITATIONS Significant heterogeneity across included studies in several subgroup analyses; patient-level data not available. CONCLUSIONS Treatment with thiazolidinediones significantly decreases urinary albumin and protein excretion in patients with diabetes. This finding calls for clinical trials with hard renal outcomes to elucidate the potential benefits of thiazolidinediones on diabetic nephropathy.
Collapse
Affiliation(s)
- Pantelis A Sarafidis
- Section of Nephrology and Hypertension, First Department of Medicine, AHEPA Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| | | | | | | | | |
Collapse
|
28
|
Sarafidis PA, Nilsson PM. The effects of thiazolidinediones on blood pressure levels – A systematic review. Blood Press 2009; 15:135-50. [PMID: 16864155 DOI: 10.1080/08037050600853720] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Insulin resistance has been proposed to be the underlying disorder of the so-called metabolic or insulin resistance syndrome, which represents the clustering in the same individual of several cardiovascular risk factors, such as type 2 diabetes mellitus, hypertension, abdominal obesity, elevated triglycerides and low high-density lipoprotein-cholesterol. As far as the connection of insulin resistance and compensatory hyperinsulinaemia with hypertension is concerned, a number of mechanisms possibly linking these disturbances have been described, such as activation of sympathetic nervous system, enhancement of renal sodium reabsorption, or impairment of endothelium-dependent vasodilatation. Thiazolidinediones (TZDs) constitute a class of oral antihyperglycaemic agents that act by decreasing insulin resistance, and apart from their action on glycaemic control, they have been also reported to exert beneficial effects on other parameters of the metabolic syndrome. In particular, during recent years a considerable number of animal and human studies have shown that the use of TZDs was associated with usually small but significant reductions of blood pressure (BP) levels. Since a possible beneficial action of these compounds on BP could be of particular value for patients with the metabolic syndrome, this review aimed to summarize and evaluate the literature data in the field, derived either from studies that just examined BP levels among other parameters or from studies that were specifically designed to determine the effect of a TZD on BP.
Collapse
Affiliation(s)
- Pantelis A Sarafidis
- 1st Department of Medicine, AHEPA University Hospital, Aristotle University, Thessaloniki, Greece.
| | | |
Collapse
|
29
|
Efrati S, Berman S, Chachashvili A, Cohen N, Siman-Tov Y, Averbukh Z, Weissgarten J. Rosiglitazone treatment attenuates renal tissue inflammation generated by urinary tract obstruction. Nephrology (Carlton) 2009; 14:189-97. [PMID: 19143945 DOI: 10.1111/j.1440-1797.2008.01032.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
AIM Peroxisome proliferator-activated receptor (PPAR)-gamma activation by rosiglitazone decreases manifestation of intrarenal inflammatory hallmarks. Inflammation significantly aggravates renal injury following urinary tract obstruction. The effect of rosiglitazone on renal inflammation following unilateral ureteral obstruction was investigated. METHODS Ninety-six Sprague-Dawley rats were subjected to unilateral ureteral ligation, or to sham operation. Half of each group received rosiglitazone, 5 mg/kg bodyweight per day. The animals were killed and their kidneys allocated following 1 h, 24 h or 2 weeks, for pathological examination or for intrarenal transforming growth factor (TGF)-beta, interleukin (IL)-4, IL-6, IL-10 and nitric oxide (NO) assessment by specific enzyme-linked immunosorbent assays. Apoptosis rates, extracellular matrix deposition, PPAR-gamma, alpha-smooth muscle actin (alpha-SMA) expression and macrophage infiltration were assessed by specific immunohistological stainings. RESULTS PPAR-gamma receptor expression was downregulated, and infiltration of macrophages decreased, in all rosiglitazone-treated kidneys. Rosiglitazone significantly decreased apoptosis, TGF-beta, IL-6, alpha-SMA expression and NO availability in obstructed kidneys. Synthesis of IL-10 was unaltered, while IL-4 augmented by Rosiglitazone. Rosiglitazone also affected NO and IL-4 production in sham-operated controls. CONCLUSION (i) Rosiglitazone attenuates profibrotic and pro-inflammatory responses in a rat model of ureteral obstruction-induced renal inflammation; (ii) rosiglitazone stimulates counteractive anti-inflammatory responses in the damaged kidneys; (iii) in part, rosiglitazone exerts comparable anti-inflammatory effects on obstructed kidneys and unobstructed healthy controls. Taken together, this ascertains the importance of the anti-inflammatory role of rosiglitazone treatment in amelioration of ureteral obstruction-induced renal damage.
Collapse
Affiliation(s)
- Shai Efrati
- Nephrology Division, Assaf-Harofeh Medical Center, Zerifin, Affiliated to Sackler Faculty of Medicine, Tel Aviv University, Israel.
| | | | | | | | | | | | | |
Collapse
|
30
|
Peroxisome proliferator-activated receptors in diabetic nephropathy. PPAR Res 2009; 2008:879523. [PMID: 19277201 PMCID: PMC2652581 DOI: 10.1155/2008/879523] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2008] [Accepted: 12/08/2008] [Indexed: 02/08/2023] Open
Abstract
Diabetic nephropathy is a leading cause of end-stage renal disease, which is increasing in incidence worldwide, despite intensive treatment approaches such as glycemic and blood pressure control in patients with diabetes mellitus. New therapeutic strategies are needed to prevent the onset of diabetic nephropathy. Peroxisome proliferator-activated receptors (PPARs) are ligand-activated nuclear transcription factors that play important roles in lipid and glucose homeostases. These agents might prevent the progression of diabetic nephropathy, since PPAR agonists improve dyslipidemia and insulin resistance. Furthermore, data from murine models suggest that PPAR agonists also have independent renoprotective effects by suppressing inflammation, oxidative stress, lipotoxicity, and activation of the renin-angiotensin system. This review summarizes data from clinical and experimental studies regarding the relationship between PPARs and diabetic nephropathy. The therapeutic potential of PPAR agonists in the treatment of diabetic nephropathy is also discussed.
Collapse
|
31
|
Role of PPARgamma in renoprotection in Type 2 diabetes: molecular mechanisms and therapeutic potential. Clin Sci (Lond) 2009; 116:17-26. [PMID: 19037881 DOI: 10.1042/cs20070462] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
DN (diabetic nephropathy) is a chronic disease characterized by proteinuria, glomerular hypertrophy, decreased glomerular filtration and renal fibrosis with loss of renal function. DN is the leading cause of ESRD (end-stage renal disease), accounting for millions of deaths worldwide. TZDs (thiazolidinediones) are synthetic ligands of PPARgamma (peroxisome-proliferator-activated receptor gamma), which is involved in many important physiological processes, including adipose differentiation, lipid and glucose metabolism, energy homoeostasis, cell proliferation, inflammation, reproduction and renoprotection. A large body of research over the past decade has revealed that, in addition to their insulin-sensitizing effects, TZDs play an important role in delaying and preventing the progression of chronic kidney disease in Type 2 diabetes. Although PPARgamma activation by TZDs is in general considered beneficial for the amelioration of diabetic renal complications in Type 2 diabetes, the underlying mechanism(s) remains only partially characterized. In this review, we summarize and discuss recent findings regarding the renoprotective effects of PPARgamma in Type 2 diabetes and the potential underlying mechanisms.
Collapse
|
32
|
Vingtdeux V, Dreses-Werringloer U, Zhao H, Davies P, Marambaud P. Therapeutic potential of resveratrol in Alzheimer's disease. BMC Neurosci 2008; 9 Suppl 2:S6. [PMID: 19090994 PMCID: PMC2604890 DOI: 10.1186/1471-2202-9-s2-s6] [Citation(s) in RCA: 143] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Several epidemiological studies indicate that moderate consumption of red wine is associated with a lower incidence of dementia and Alzheimer's disease. Red wine is enriched in antioxidant polyphenols with potential neuroprotective activities. Despite scepticism concerning the bioavailability of these polyphenols, in vivo data have clearly demonstrated the neuroprotective properties of the naturally occurring polyphenol resveratrol in rodent models for stress and diseases. Furthermore, recent work in cell cultures and animal models has shed light on the molecular mechanisms potentially involved in the beneficial effects of resveratrol intake against the neurodegenerative process in Alzheimer's disease.
Collapse
Affiliation(s)
- Valérie Vingtdeux
- Litwin-Zucker Research Center for the Study of Alzheimer's Disease, The Feinstein Institute for Medical Research, North Shore-LIJ, Manhasset, NY 11030, USA.
| | | | | | | | | |
Collapse
|
33
|
Thethi TK, Singh S, Fonseca V. Insulin Sensitizers and Cardiovascular Disease. Cardiovasc Endocrinol 2008. [DOI: 10.1007/978-1-59745-141-3_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
34
|
Zhang H, Saha J, Byun J, Schin M, Lorenz M, Kennedy RT, Kretzler M, Feldman EL, Pennathur S, Brosius FC. Rosiglitazone reduces renal and plasma markers of oxidative injury and reverses urinary metabolite abnormalities in the amelioration of diabetic nephropathy. Am J Physiol Renal Physiol 2008; 295:F1071-81. [PMID: 18667486 DOI: 10.1152/ajprenal.90208.2008] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Recent studies suggest that thiazolidinediones ameliorate diabetic nephropathy (DN) independently of their effect on hyperglycemia. In the current study, we confirm and extend these findings by showing that rosiglitazone treatment prevented the development of DN and reversed multiple markers of oxidative injury in DBA/2J mice made diabetic by low-dose streptozotocin. These diabetic mice developed a 14.2-fold increase in albuminuria and a 53% expansion of renal glomerular extracellular matrix after 12 wk of diabetes. These changes were largely abrogated by administration of rosiglitazone beginning 2 wk after the completion of streptozotocin injections. Rosiglitazone had no effect on glycemic control. Rosiglitazone had similar effects on insulin-treated diabetic mice after 24 wk of diabetes. Podocyte loss and glomerular fibronectin accumulation, other markers of early DN, were prevented by rosiglitazone in both 12- and 24-wk diabetic models. Surprisingly, glomerular GLUT1 levels did not increase and nephrin levels did not decrease in the diabetic animals; neither changed with rosiglitazone. Plasma and kidney markers of protein oxidation and lipid peroxidation were significantly elevated in the 24-wk diabetic animals despite insulin treatment and were reduced to near-normal levels by rosiglitazone. Finally, urinary metabolites were markedly altered by diabetes. Of 1,988 metabolite features identified by electrospray ionization time of flight mass spectrometry, levels of 56 were altered more than twofold in the urine of diabetic mice. Of these, 21 were returned to normal by rosiglitazone. Thus rosiglitazone has direct effects on the renal glomerulus to reduce reactive oxygen species accumulation to prevent type 1 diabetic mice from development of DN.
Collapse
Affiliation(s)
- Hongyu Zhang
- Department of Internal Medicine, University of Michigan, 5520 MSRB1, 1150 W. Medical Center Dr., Ann Arbor, MI 48109-5680, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Potential for peroxisome proliferator-activated receptor-?? agonists in progression: beyond metabolism. Curr Opin Nephrol Hypertens 2008; 17:282-5. [DOI: 10.1097/mnh.0b013e3282f9b1c0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
36
|
Agonists at PPAR-gamma suppress angiotensin II-induced production of plasminogen activator inhibitor-1 and extracellular matrix in rat cardiac fibroblasts. Br J Pharmacol 2008; 153:1409-19. [PMID: 18278065 DOI: 10.1038/bjp.2008.21] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND AND PURPOSE Peroxisome proliferator-activated receptor (PPAR)-gamma ligands have been shown to inhibit cardiac fibrosis. However, the underlying mechanisms are poorly understood. We investigated the regulation by PPAR-gamma ligands of angiotensin (Ang) II-induced plasminogen activator inhibitor (PAI)-1, extracellular matrix (ECM) production and cell growth in cardiac fibroblasts. EXPERIMENTAL APPROACH The effects of PPAR-gamma ligands on Ang II-induced PAI-1, ECM expression and cell growth were assessed in primary-cultured rat cardiac fibroblasts; cardiac PAI-1 and ECM production was examined in Ang II-infused rats. KEY RESULTS In growth-arrested cardiac fibroblasts, PPAR-gamma ligands rosiglitazone and 15-deoxy-Delta(12,14)-prostaglandin J2 (15d-PGJ2) dose-dependently attenuated Ang II-induced cell proliferation and expression of PAI-1, collagen type-I, collagen type-III and fibronectin. An accompanying increase in PPAR-gamma expression and activation was also observed. These suppressive effects were attenuated by the PPAR-gamma antagonists GW9662 and bisphenol A diglycidyl ether (BADGE). Moreover, rosiglitazone and 15d-PGJ2 inhibited in part the expression and phosphorylation of Ang II-induced transforming growth factor (TGF)-beta1, Smad2/3 and c-Jun NH(2)-terminal kinase (JNK). Ang II infusion in rats markedly increased left ventricular production of PAI-1, collagen and fibronectin, with a concurrent increase in the ratios of heart weight/body weight and left ventricle weight/body weight. Co-treatment with rosiglitazone significantly decreased these levels and upregulated PPAR-gamma expression. CONCLUSIONS AND IMPLICATIONS Rosiglitazone and 15d-PGJ2 suppress Ang II-induced production of PAI-1 and ECM probably via interactions between PPAR-gamma and TGF-beta1/Smad2/3 and JNK signalling pathways. It is suggested that PPAR-gamma and its ligands may have potential applications in preventing cardiac fibrosis.
Collapse
|
37
|
Ruan X, Zheng F, Guan Y. PPARs and the kidney in metabolic syndrome. Am J Physiol Renal Physiol 2008; 294:F1032-47. [PMID: 18234957 DOI: 10.1152/ajprenal.00152.2007] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The metabolic syndrome (MetS) is defined by a set of metabolic risk factors, including insulin resistance, central obesity, dyslipidemia, hyperglycemia, and hypertension for type 2 diabetes and cardiovascular disease. Although both retrospective and prospective clinical studies have revealed that MetS is associated with chronic renal disease, even with a nondiabetic cause, the cellular and molecular mechanisms in this association remain largely uncharacterized. Recently, increasing evidence suggests that peroxisome proliferator-activated receptors (PPARs), a subgroup of the nuclear hormone receptor superfamily of ligand-activated transcription factors, may play an important role in the pathogenesis of MetS. All three members of the PPAR nuclear receptor subfamily, PPARalpha, -beta/delta, and -gamma, are critical in regulating insulin sensitivity, adipogenesis, lipid metabolism, inflammation, and blood pressure. PPARs have also been implicated in many renal pathophysiological conditions, including diabetic nephropathy and glomerulosclerosis. Ligands for PPARs such as hypolipidemic PPARalpha activators, and antidiabetic thiazolidinedione PPARgamma agonists affect not only diverse aspects of MetS but also renal disease progression. Emerging data suggest that PPARs may be potential therapeutic targets for MetS and its related renal complications. This review focuses on current knowledge of the role of PPARs in MetS and discusses the potential therapeutic utility of PPAR modulators in the treatment of kidney diseases associated with MetS.
Collapse
Affiliation(s)
- Xiongzhong Ruan
- Center for Nephrology, University College of London, London, United Kingdom
| | | | | |
Collapse
|
38
|
Hong EG. Diabetic Nephropathy - Preventive effects of lithospermic acid B (LAB). KOREAN DIABETES JOURNAL 2008. [DOI: 10.4093/kdj.2008.32.1.7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Eun Gyoung Hong
- Department of Internal Medicine, College of Medicine, Hallym University, Chunchon, Korea
| |
Collapse
|
39
|
KINCAID-SMITH PRISCILLA, FAIRLEY KENNETHF, FARISH STEPHEN, BEST JAMESD, PROIETTO JOSEPH. Reduction of proteinuria by rosiglitazone in non-diabetic renal disease. Nephrology (Carlton) 2007; 13:58-62. [DOI: 10.1111/j.1440-1797.2007.00903.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
40
|
Erdogan M, Karadeniz M, Eroglu Z, Tezcanli B, Selvi N, Yilmaz C. The relationship of the peroxisome proliferator-activated receptor-gamma 2 exon 2 and exon 6 gene polymorphism in Turkish type 2 diabetic patients with and without nephropathy. Diabetes Res Clin Pract 2007; 78:355-9. [PMID: 17681394 DOI: 10.1016/j.diabres.2007.06.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2007] [Accepted: 06/16/2007] [Indexed: 10/23/2022]
Abstract
BACKGROUND Peroxisome proliferator-activated receptor gamma (PPAR gamma) has recently been shown to be associated with type 2 diabetes. We aim to investigate Turkish type 2 diabetic patients with/without diabetic nephropathy and healthy group and examined the contribution of the G/C exon 2 and T/C exon 6 of the PPAR gamma gene polymorphism to the development of diabetic nephropathy. METHODS The PPAR gamma genotypes were determined retrospectively in 43 patients with nephropathy and 48 without nephropathy and a control group of 50 healthy individuals. Genotyping of the G/C exon 2 and T/C exon 6 of the PPAR-gamma gene polymorphism for all individuals was performed by melting curve analysis of the generated amplicons after real-time online PCR. RESULTS This genotype (exon 2 and exon 6) distribution did not differ between control subjects and type 2 diabetic patients. The genotype frequencies and allele exon 2 were CC, 100%; GC, 0%; GG, 0% and C, 100%; G, 0% in diabetic patients with nephropathy versus CC, 97.9%; GC, 2.1%; GG, 0% and C 98.9%, G 1.1% in those without nephropathy. Genotype exon 6 frequencies in diabetic patients with nephropathy were (T/T) 0%, (T/C) 14%; (C/C) 86% versus (G/G) 0%; (G/C) 2.1%; (C/C) 97.9% in those without nephropathy. The PPAR gamma exon 2 and exon 6 genotype and allele frequencies were not different between diabetic patients with and without nephropathy. CONCLUSIONS PPAR gamma exon 2 and exon 6 gene polymorphism is not associated with the development of diabetic nephropathy in Turkish type 2 diabetic patients.
Collapse
Affiliation(s)
- M Erdogan
- Ege University Medical School, Department of Endocrinology and Metabolism Disease, 35100 Izmir, Turkey
| | | | | | | | | | | |
Collapse
|
41
|
Fernyhough ME, Okine E, Hausman G, Vierck JL, Dodson MV. PPARgamma and GLUT-4 expression as developmental regulators/markers for preadipocyte differentiation into an adipocyte. Domest Anim Endocrinol 2007; 33:367-78. [PMID: 17560753 DOI: 10.1016/j.domaniend.2007.05.001] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2007] [Revised: 05/07/2007] [Accepted: 05/08/2007] [Indexed: 12/25/2022]
Abstract
In this document, we have integrated knowledge about two major cellular markers found in cells of the adipocyte lineage (an adipogenic marker and a metabolic marker). This review provides information as to how differentiation of a cell (such as an adipofibroblast, fibroblast or preadipocyte) to become a viable (and new) adipocyte is under different regulation than that experienced by an immature adipocyte that is just beginning to accumulate lipid. The differentiation, prior to lipid-filling, involves PPARgamma. Subsequently, lipid-filling of the adipocyte relies on a late subset of genes and, depending on depot specificity, involves GLUT-4 or any number of other metabolic markers.
Collapse
Affiliation(s)
- M E Fernyhough
- Department of Animal Sciences, Washington State University, PO Box 646310, Pullman, WA 99164, United States
| | | | | | | | | |
Collapse
|
42
|
Efrati S, Berman S, Chachashvili A, Cohen N, Averbukh Z, Weissgarten J. Rosiglitazone treatment attenuates expression of inflammatory hallmarks in the remaining kidney following contralateral nephrectomy. Am J Nephrol 2007; 28:238-45. [PMID: 17975301 DOI: 10.1159/000110681] [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] [Received: 04/23/2007] [Accepted: 09/18/2007] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIMS Following kidney donation, living kidney donors have been reported to develop anemia and pronounced inflammation. Therapeutic strategies for ameliorating unilateral nephrectomy-induced inflammation would be beneficiary for the living donors. We applied rosiglitazone to attenuate inflammatory processes ongoing within the remaining kidney following contralateral nephrectomy. METHODS 20 Sprague-Dawley rats were subjected to left unilateral nephrectomy and 20 others to sham operation. Half of each group was treated for 2 weeks with rosiglitazone (5 mg/kg body weight). After sacrifice, intrarenal transforming growth factor (TGF)-beta, angiotensin-II (A-II), interleukin (IL)-6, IL-10, IL-4 and nitric oxide (NO) were assessed, and histologic sections stained to assess the inflammatory cell infiltration. Renal function was evaluated by creatinine, urea, cystatin C measurements. RESULTS Intrarenal IL-6, A-II and TGF-beta were significantly augmented, while NO was significantly decreased in kidneys remaining after contralateral nephrectomy. Rosiglitazone treatment abrogated augmented IL-6, A-II and TGF-beta synthesis and restored intrarenal NO availability in the remaining kidneys. Rosiglitazone also augmented anti-inflammatory IL-4 cytokine synthesis, while IL-10 production, leukocyte infiltration and renal function parameters remained unchanged. CONCLUSIONS Rosiglitazone treatment attenuates the proinflammatory responses, represented by augmented IL-6, A-II and TGF-beta production, developing in the remaining kidney following contralateral nephrectomy. In addition, by stimulating IL-4 synthesis and restoring NO availability, rosiglitazone treatment initiates counteractive anti-inflammatory responses in the remaining kidney.
Collapse
Affiliation(s)
- Shai Efrati
- Nephrology Division, Assaf Harofeh Medical Center, Affiliated to Sackler Faculty of Medicine, Tel Aviv University, Zerifin, Israel.
| | | | | | | | | | | |
Collapse
|
43
|
Efrati S, Berman S, Ilgiyeav E, Averbukh Z, Weissgarten J. PPAR-γ Activation Inhibits Angiotensin II Synthesis, Apoptosis, and Proliferation of Mesangial Cells from Spontaneously Hypertensive Rats. ACTA ACUST UNITED AC 2007; 106:e107-12. [PMID: 17622738 DOI: 10.1159/000104834] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2006] [Accepted: 02/12/2007] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIM The angiotensin II level is elevated in subjects genetically prone to develop hypertension, triggering renal hypercellularity, cytokine production, and matrix deposition. Angiotensin-converting enzyme inhibition and/or angiotensin II type 1 receptor blockade attenuate renal damage. Rosiglitazone, a peroxisome proliferator-activated receptor gamma agonist possessing antihypertensive and anti-inflammatory properties, was demonstrated to provide better renal protection than angiotensin-converting enzyme inhibitors. We studied the effects of in vivo peroxisome proliferator-activated receptor gamma activation by rosiglitazone on angiotensin II synthesis, proliferation, and apoptosis of mesangial cells of spontaneously hypertensive rats versus normotensive Sprague-Dawley rats. METHODS The animals consumed either a high-sodium diet (8% Na) or a normal-sodium diet (0.5% Na). Half of each group received rosiglitazone at 5 mg/kg/day. After 3 weeks, all rats were sacrificed and the mesangial cells isolated and cultured. Angiotensin II was assessed by radioimmunoassay, apoptosis by terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling assay, and cell proliferation by [(3)H]thymidine incorporation. RESULTS Only the spontaneously hypertensive rats which consumed the high-sodium diet developed hypertension (185 +/- 6 mm Hg vs. basal 128 +/- 5 mm Hg; p = 0.0007) which was attenuated by rosiglitazone (to 126 +/- 4 mm Hg; p = 0.34). Angiotensin II synthesis, proliferation, and apoptosis were exaggerated in mesangial cell cultures from Sprague-Dawley rats and, more so, spontaneously hypertensive rats fed the high-sodium diet, but were inhibited in cultures from rosiglitazone-treated animals. CONCLUSIONS Peroxisome proliferator-activated receptor gamma activation, in addition to lowering blood pressure, suppresses angiotensin II synthesis and downregulates angiotensin-II-mediated proliferation and apoptosis of mesangial cells. In the context of hypertension-induced renal damage, this would mean that the renoprotective role of rosiglitazone extends beyond glycemic and lipidemic control.
Collapse
Affiliation(s)
- Shai Efrati
- Nephrology Division, Department of Nephrology, Assaf Harofeh Medical Center, Zerifin, Israel.
| | | | | | | | | |
Collapse
|
44
|
Gipson DS, Gibson K, Gipson PE, Watkins S, Moxey-Mims M. Therapeutic approach to FSGS in children. Pediatr Nephrol 2007; 22:28-36. [PMID: 17109140 PMCID: PMC1784542 DOI: 10.1007/s00467-006-0310-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2006] [Revised: 07/21/2006] [Accepted: 07/27/2006] [Indexed: 11/26/2022]
Abstract
Therapy of primary focal segmental glomerulosclerosis (FSGS) in children incorporates conservative management and immunosuppression regimens to control proteinuria and preserve kidney function. In long-term cohort studies in adults and children with primary FSGS, renal survival has been directly associated with degree of proteinuria control. This educational article reviews the current therapeutic approach toward children with primary FSGS.
Collapse
Affiliation(s)
- Debbie S Gipson
- Chapel Hill School of Medicine, University of North Carolina Kidney Center, University of North Carolina, 7012 Burnett-Womack Hall, CB#7155, Chapel Hill, NC, 27599-7155, USA.
| | | | | | | | | |
Collapse
|
45
|
Pereira MG, Câmara NOS, Campaholle G, Cenedeze MA, de Paula Antunes Teixeira V, dos Reis MA, Pacheco-Silva A. Pioglitazone limits cyclosporine nephrotoxicity in rats. Int Immunopharmacol 2006; 6:1943-51. [PMID: 17161347 DOI: 10.1016/j.intimp.2006.07.024] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2006] [Accepted: 07/22/2006] [Indexed: 01/14/2023]
Abstract
Chronic cyclosporine (CsA) nephrotoxicity is a relevant factor in the pathogenesis of chronic allograft nephropathy. Pioglitazone is an agonist of PPARgamma, capable of reducing chronic inflammation. We investigated the capacity of pioglitazone in preventing renal dysfunction. Adult male Wistar rats were assigned to: Vehicle (olive oil 1 ml/kg/day), CsA (10 mg/kg/day) alone and with pioglitazone (5 or 10 mg/kg/day). The animals were sacrificed at 28 days, where blood (serum creatinine ratio, CR) and kidney samples (arteriolopathy analyses) were collected. The mRNA transcripts of TGF-beta1, PAI-1, Smad3 and 7 were evaluated by real-time PCR. As expected, CsA treatment significantly decreased renal function that peaked at day 28, compared with vehicle (CR=1.29+/-0.03 vs. 0.95+/-0.14, p<0.05). In contrast, the administration of pioglitazone 5 or 10 mg/kg combined with CsA resulted in better renal function (CR=1.09+/-0.05 and 1.14+/-0.14, respectively, p<0.05). Animals treated with CSA showed relevant arteriolopathy (49.5+/-2.86%) and pioglitazone administration significantly limited it (37.0+/-3.59% and 36.6+/-1.72%, respectively, 5 or 10 mg/kg, p<0.05). In CsA-treated animals (alone and with pioglitazone), TGF-beta1 and Smad3 increased significantly. In animals treated with CsA and pioglitazone (5 mg/kg), PAI-1 was significantly lower than CsA alone (3.96+/-0.92 vs. 7.53+/-1.38, p<0.05). Interestingly, the administration of pioglitazone 5 or 10 mg/kg was associated with an increase in Smad7 (1.79+/-0.25 and 1.75+/-0.19, respectively), compared to vehicle and to CsA-treated groups (1.08+/-0.17 and 1.17+/-0.19, respectively, p<0.05). These data provide evidence that pioglitazone acts through down regulation of pro-fibrotic cytokine PAI-1 and overexpression of the regulatory Smad7.
Collapse
Affiliation(s)
- Mauricio Galvão Pereira
- Laboratório de Imunologia Clínica e Experimental, Division of Nephrology, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, Brazil
| | | | | | | | | | | | | |
Collapse
|
46
|
Peng Y, Liu H, Liu F, Liu Y, Li J, Chen X. Troglitazone inhibits synthesis of transforming growth factor-?1 and reduces matrix production in human peritoneal mesothelial cells. Nephrology (Carlton) 2006; 11:516-23. [PMID: 17199790 DOI: 10.1111/j.1440-1797.2006.00654.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
AIM Peritoneal matrix accumulation is a characteristic of peritoneal fibrosis (PF). Continuous ambulatory peritoneal dialysis (CAPD) patients with up-regulation of transforming growth factor-beta1 (TGF-beta1) in their drained effluent show an increased risk of PF. Inhibition of TGF-beta1 expression in human peritoneal mesothelial cells (HPMC) may provide a potential treatment for PF. Peroxisome proliferator-activated receptor-gamma (PPAR-gamma) agonists are increasingly used in patients with diabetes, but their effects on extracellular matrix (ECM) turnover are unknown. The aims of this study were to investigate the effects of the PPAR-gamma agonist troglitazone on TGF-beta1 expression and matrix production in HPMC. METHODS Human peritoneal mesothelial cells were cultured from human omentum by an enzyme digestion method, grown in a medium containing 30 mmol/L D-glucose. TGF-beta1 expression and matrix production and turnover were measured in HPMC in the presence and absence of 15 micromol/L troglitazone. The mRNA expressions of TGF-beta(1), Collagen I (Col I) and fibronectin (FN) were determined by semiquantification reverse-transcriptive polymerase chain reaction (RT-PCR). The protein of TGF-beta1 was determined by ELISA and proteins of Col I, FN were determined by western blot. RESULTS The mRNA expression and protein of TGF-beta(1), Col I, FN were significantly increased in HPMC stimulated with 30 mmol/L D-glucose compared to the control group with F12 media (P < 0.01), which was reversed in the presence of troglitazone (15 micromol/L). Obvious decrease of TGF-beta(1) was found in troglitazone-treated groups as compared to groups stimulated with GS (P < 0.05). Exposure of HPMC to troglitazone reduced collagen I secretion (P < 0.05), and fibronectin secretion (P < 0.05). CONCLUSION Troglitazone reduce the expression of TGF-beta(1) in HPMC stimulated by 30 mmol/L D-glucose, and reduces ECM production. These studies suggest that the PPAR-gamma agonists may have a specific role in ameliorating the course of progressive peritoneal fibrosis under long-term peritoneal dialysis states.
Collapse
Affiliation(s)
- Youming Peng
- Department of Nephrology, Nephrology Institute of Central South University, 2nd Xiangya Hospital, Central South University, Changsha, Hunan, China
| | | | | | | | | | | |
Collapse
|
47
|
Sarafidis PA, Bakris GL. Protection of the kidney by thiazolidinediones: An assessment from bench to bedside. Kidney Int 2006; 70:1223-33. [PMID: 16883325 DOI: 10.1038/sj.ki.5001620] [Citation(s) in RCA: 164] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The global epidemic of diabetes mellitus has led to a continuous increase in the prevalence of diabetic nephropathy over the past years. Thus, diabetic nephropathy is currently the number one cause of end-stage renal disease in the Western world. It represents a major public health problem for which more effective prevention and treatment strategies are needed. Thiazolidinediones (TZDs) are a class of agents that lower blood glucose through reduction of insulin resistance in patients with type 2 diabetes. Growing evidence support the concept that TZDs have several beneficial effects on the cardiovascular system beyond their effects on glycemic control. These benefits include: blood pressure lowering, triglyceride reduction, high-density lipoprotein-cholesterol elevation, and reduction in subclinical vascular inflammation. Moreover, data from several animal and human studies support the notion that TZDs reduce urine albumin excretion and may prevent development of renal injury. The relative lack of evidence, however, demonstrating the effects of TZDs on hard renal outcomes mandates the need for well-designed trials with this particular objective. This paper summarizes all the data from clinical and experimental studies relevant to a possible renoprotective effect of TZDs and discusses actions of these compounds that may contribute toward this effect.
Collapse
Affiliation(s)
- P A Sarafidis
- Department of Preventive Medicine, Hypertension/Clinical Research Center, Rush University Medical Center, Chicago, Illinois 60612, USA
| | | |
Collapse
|
48
|
Zhang YJ, Yang X, Kong QY, Zhang YF, Chen WY, Dong XQ, Li XY, Yu XQ. Effect of 15d-PGJ2 on the expression of CD40 and RANTES induced by IFN-gamma and TNF-alpha on renal tubular epithelial cells (HK-2). Am J Nephrol 2006; 26:356-62. [PMID: 16864989 DOI: 10.1159/000094735] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2006] [Accepted: 06/21/2006] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIMS Recent evidence shows that peroxisome proliferator-activated receptor-gamma (PPAR-gamma) ameliorates a variety of inflammatory conditions. CD40 is a co-stimulatory molecule and its ligation induces production of different proinflammatory cytokines including RANTES (regulated upon activation, normal T cell expressed), which are considered as important factors in the initiation and maintenance of inflammatory response. The aim of this study was to investigate the effect of PPAR-gamma on CD40 and RANTES production on cultured human renal proximal tubular epithelial (HK-2) cells. METHODS HK-2 cells were maintained under defined in vitro conditions and treated with either PPAR-gamma agonist 15-deoxy-12,14-prostaglandin J2 (15d-PGJ2) or 15d-PGJ2 + PPAR-gamma antagonist GW9662, and then stimulated with a combination of tumor necrosis factor-alpha (TNF-alpha) and interferon-gamma (IFN-gamma). The CD40 and RANTES levels were investigated. RESULTS HK-2 cells expressed low levels of CD40 and RANTES. Activation of HK-2 cells by combined treatment of TNF-alpha and IFN-gamma results in strong synergistic effects on the expression of CD40 and the secretion of RANTES. 15d-PGJ2 significantly decreased CD40 and RANTES expression and GW9662 partly abrogated the inhibition of 15d-PGJ2 on CD40 and RANTES. CONCLUSION 15d-PGJ2 significantly decreased CD40 and RANTES expression in HK-2 cells, which were partially mediated by PPAR-gamma-dependent pathways. These results point to PPAR-gamma as a remarkable new target in the prevention of tubular inflammatory injury associated with renal disease.
Collapse
Affiliation(s)
- Ya Jie Zhang
- Department of Nephrology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PR China
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Dobrian AD. The complex role of PPARgamma in renal dysfunction in obesity: managing a Janus-faced receptor. Vascul Pharmacol 2006; 45:36-45. [PMID: 16716756 DOI: 10.1016/j.vph.2006.01.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2006] [Revised: 01/01/2006] [Accepted: 01/01/2006] [Indexed: 01/11/2023]
Abstract
Obesity is frequently accompanied by insulin resistance, type II diabetes, hypertension and atherosclerosis, a cluster of pathologies that are the major components of the metabolic syndrome. Obesity is a known cause for renal dysfunction that leads to two major renal pathologies: hypertension and glomerular and tubulointerstitial injury. Peroxizome proliferator activated receptors (PPARs) are transcription factors belonging to the nuclear hormone receptor superfamily with important functions in the regulation of metabolism. The role of PPARgamma isoforms in adipogenesis and vascular inflammation associated to obesity has been vastly studied and is well recognized, albeit not completely mechanistically understood. Also, the effect of various PPARgamma agonists on blood pressure reduction in different forms of hypertension, including obesity related hypertension has been reported, but the mechanisms involved are only beginning to be studied. Even less clear is the concurrent beneficial effect of PPARgamma agonists thiazolinendiones (TZD) on blood pressure reduction in different forms of hypertension and, at the same time, in some cases, the significant water retention leading to edema and heart failure. The occurrence of both these apparently opposite effects on the renal water and sodium handling suggests a complex role of PPARgamma in the kidney that is likely related to the metabolic state. Also, PPARgamma activation leads to a reduction in mesangial cell proliferation while stimulating apoptosis. TZD treatment reduces albuminuria in obese and diabetic humans and rodent models suggesting protective effects against renal tubuloglomerular injury. The focus of this review is to present and critically discuss the recent findings on the roles of PPARgamma in the kidney in direct relation to renal function and renal injury in obesity and obesity-initiated diabetes.
Collapse
Affiliation(s)
- Anca Dana Dobrian
- Department of Physiological Sciences, Eastern Virginia Medical School, 700W Olney Road, Lewis Hall, Room 2027, Norfolk, VA 23507, USA.
| |
Collapse
|
50
|
Goyal A, Crook ED. Thiazolidinediones and progression of renal disease in patients with diabetes. J Investig Med 2006; 54:56-61. [PMID: 16472474 DOI: 10.2310/6650.2005.05034] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
OBJECTIVE Thiazolidinediones (TZDs) are used in the treatment of type 2 diabetes mellitus (T2DM) and appear to have beneficial effects on markers of cardiovascular or renal risk that are independent of glycemic control. We examined the effects of TZDs on renal survival in a predominantly black population with T2DM. METHODS We performed a retrospective case-control study in patients with T2DM seen in our nephrology clinic in 2001 to 2002. Cases had T2DM and were on a TZD at presentation or for >or= 6 months over follow-up. Controls were matched for sex, age, duration of T2DM, and initial creatinine. Reaching end-stage renal disease (ESRD) was the primary end point. RESULTS From 387 records, 43 cases (34 blacks, 31 females) and 106 controls (96 blacks, 83 females) were identified. The baseline characteristics were similar for both groups. Both groups had moderate renal disease (estimated glomerular filtration rate approximately 40-45 mL/min). Cases had lower systolic blood pressure over follow-up (p=.02), but there was no difference in glycemic control or use of insulin. Renal survival was better among cases (age- and gender-adjusted odds ratio for reaching ESRD 0.17 [95% confidence interval 0.03-0.8]; p=.03). When adjusted for systolic blood pressure over follow-up, the tendency for improved renal survival in cases remained but was no longer significant. CONCLUSION We conclude that TZDs may protect against the progression of renal disease in T2DM. Prospective studies are required to determine the effects of TZDs on renal survival in T2DM.
Collapse
Affiliation(s)
- Abhijeet Goyal
- Department of Medicine, Wayne State University School of Medicine and the John.D.Dingell VA Medical Center, Detroit, MI, USA
| | | |
Collapse
|