当前位置: 首页 > 医学版 > 期刊论文 > 内科学 > 糖尿病学杂志 > 2006年 > 第4期 > 正文
编号:11257012
Accelerated Diabetic Nephropathy in Mice Lacking the Peroxisome ProliferatoreCActivated Receptor
     1 Division of Nephrology, Department of Internal Medicine, The Catholic University of Korea, Seoul, Korea

    2 Department of Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan

    3 Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee

    DMEM, Dulbecco’s modified Eagle’s medium; ELISA, enzyme-linked immunosorbent assay; PPAR, peroxisome proliferatoreCactivated receptor; PPRE, PPAR response element; TGF, transforming growth factor; TUNEL, terminal deoxynucleotide transferase-mediated dUTP nick-end labeling

    ABSTRACT

    Peroxisome proliferatoreCactivated receptor (PPAR), a member of the ligand-activated nuclear receptor superfamily, plays an important role in lipid metabolism and glucose homeostasis and is highly expressed in the kidney. The present studies were aimed at determining the role of PPAR in the pathogenesis of diabetic nephropathy using PPAR-knockout mice and cultured murine mesangial cells. Diabetes was induced using a low-dose streptozotocin protocol in 8-week-old male 129 SvJ PPAR-knockout and wild-type mice. Diabetic PPAR-knockout and wild-type mice developed elevated fasting blood glucose (P < 0.001) and HbA1c levels (P < 0.001). Renal functional and histopathological changes in diabetic and nondiabetic PPAR-knockout and wild-type mice were evaluated after 16 weeks of hyperglycemia. PPAR immunostaining of the cortical tubules of diabetic wild-type mice was elevated by hyperglycemia. In diabetic PPAR-knockout mice, renal disease with accompanying albuminuria, glomerular sclerosis, and mesangial area expansion was more severe than in diabetic wild-type mice (P < 0.05) and was accompanied by increased levels of serum free fatty acids and triglycerides (P < 0.01). Furthermore, they exhibited increased renal immunostaining for type IV collagen and osteopontin, which was associated with increased macrophage infiltration and glomerular apoptosis. There were no significant differences in these indexes of renal disease between nondiabetic PPAR-knockout and wild-type mice and diabetic PPAR wild-type mice. In vitro studies demonstrated that high glucose levels markedly increased the expression of type IV collagen, transforming growth factor-1, and the number of leukocytes adherent to cultured mesangial cells. Adherence of leukocytes was inhibited by the PPAR agonist fenofibrate. Taken together, PPAR deficiency appears to aggravate the severity of diabetic nephropathy through an increase in extracellular matrix formation, inflammation, and circulating free fatty acid and triglyceride concentrations. PPAR agonists may serve as useful therapeutic agents for type 1 diabetic nephropathy.

    Peroxisome proliferatoreCactivated receptor (PPAR) is a member of the nuclear hormone receptor superfamily of ligand-activated transcription factors and plays an important role in lipid metabolism (1). The role of PPAR expression in the kidney remains poorly characterized. PPAR expression predominates in the proximal tubule and the thick limb, which could contribute to dietary lipideCinduced gene expression of peroxisomal and mitochondrial -oxidation proteins in these segments (2eC4). Interestingly, the PPAR agonist clofibrate reduces blood pressure in Dahl salt-sensitive rats (5,6). PPAR has also been shown to play an important role in sustaining the balance between energy production and utilization in the kidney by regulating the expression of genes involved in fatty acid -oxidation (7). This effect may explain the observation that PPAR ligands protect the kidney from ischemia/reperfusion injury in rats. Mice lacking the PPAR gene exhibit more severe cortical necrosis and worse kidney function than wild-type control mice (7).

    PPAR is relatively abundant in tissues with a high oxidative capacity, such as the liver, kidney, and heart. Recent reports demonstrate that altered fatty acid metabolism is associated with cardiomyopathy and nephropathy (8,9). Accumulation of excess lipids in nonadipose tissues leads to cell dysfunction or cell death (8,9). This phenomenon, known as lipotoxicity, may play an important role in the pathogenesis of diabetes and heart failure in humans (8). Several mechanisms have been suggested to underlie lipotoxicity, including a direct toxic effect of fatty acids or products of their metabolism, increased production of reactive oxygen species, ATP deficiency, and fatty acideCinduced apoptosis (8,9).

    The generation of PPAR-knockout mice has helped to confirm several of the proposed actions of this nuclear receptor and has identified previously unknown actions. These mice are viable, fertile, and exhibit no gross phenotypic defects (10); however, they exhibit profound metabolic abnormalities of the liver and heart. These mice fail to exhibit peroxisome proliferation or activation of fatty acid oxidation target genes when exposed to PPAR agonists (11). They accumulate increased hepatic triglycerides in response to feeding and during fasting (11). PPAR-knockout mice develop severe hypoglycemia when fasted (12) because of impaired fatty acid oxidation and increased reliance on glucose as an energy source. In this study, we used PPAR-knockout mice to investigate the role of PPAR in diabetic nephropathy associated with type 1 diabetes.

    RESEARCH DESIGN AND METHODS

    Male PPAR-knockout and wild-type background 129SvJ mice were used for all studies. Mice were housed in a sterile environment with 12:12-h light-dark cycles and had free access to food and water. To measure 24-h albumin excretion and creatinine clearance, mice were placed in individual mouse metabolic cages (Nalgene, Rochester, NY) with access to water and food for 24 h. Eight-week-old homozygous PPAR-knockout mice (n = 12) and 129SvJ wild-type mice (n = 12) were treated with or without streptozotocin by intraperitoneal injection (50 mg · kgeC1 · dayeC1 for 5 days). After 2 weeks, fasting blood glucose was monitored using Accu-Chek meter (Roche Diagnostics, St. Louis, MO) and hyperglycemia confirmed. Mouse body weight was measured weekly. Blood glucose was measured every 2 weeks, and HbA1c (A1C) and 24-h urinary albumin and creatinine were measured every 4 weeks. Systolic blood pressure was assessed by noninvasive tail cuff system in conscious mice at the end of the study (IITC Life Science, Woodland Hills, CA). Animals were habituated to the tail cuff device before measuring the blood pressure for 5 days over 15 min to reduce variability of the measurements.

    After 16 weeks, animals were anesthetized by an intraperitoneal injection of pentobarbital sodium (55 mg/kg body wt; Nembutal; Boehringer Ingelheim, Artanrmon, NSW, Australia). The kidneys were rapidly dissected and stored in buffered formalin (10%) for subsequent immunohistochemical analyses. Blood was collected from the left ventricle and centrifuged, and plasma was stored at eC70°C for subsequent analyses. A1C was determined on red cell lysates by high-pressure liquid chromatograpy (Bio-Rad, Richmond, CA). Total cholesterol and triglyceride concentrations were measured by autoanalyzer (Hitachi 917; Tokyo, Japan). The free fatty acid levels were measured with the JCA-BM1250 automatic analyzer (JEOL, Tokyo, Japan).

    Assessment of renal function.

    At week 16, the animals were housed in metabolic cages for 24 h to collect urine for subsequent measurements of albumin concentration by an immunoassay (Bayer, Elkhart, IN). Plasma creatinine and urea concentrations were measured using autoanalyzer (Beckman Instruments, Fullerton, CA). Serum triglyceride and cholesterol levels were measured using commercial kits (nos. 33740A and 33710B and 402-20, respectively) purchased from Sigma-Aldrich.

    Light microscopic study.

    Kidney samples were collected following systemic perfusion with PBS and then fixed in 4% paraformaldehyde. Fixed tissues were embedded in paraffin. Histology was assessed following hematoxylin, eosin, and periodic acid Schiff staining. To examine the effect of PPAR deletion on glomerular area and mesangial matrix area, glomerular analysis was performed on periodic acid SchiffeCstained kidney sections. Fifteen consecutive glomerular cross sections were photographed by an examiner, blinded to the source of the tissue, using a digital camera (Olympus DP11; Olympus America, Melville, NY). Mesangial matrix area and glomerular tuft area were quantified for each glomerular cross section as previously reported (13). More than 30 glomeruli that were cut through the vascular pole were counted per kidney, and the average was used for analysis.

    Immunohistochemistry for PPAR, osteopontin, F4/80, transforming growth factor-1, type IV collagen, and caspase-3.

    We performed immunohistochemistry for PPAR, type IV collagen, transforming growth factor (TGF)-1, osteopontin, F4/80, and caspase-3. Briefly, small blocks of kidney were immediately fixed in 10% buffered formalin for 24 h before embedded in paraffin. Five micrometereCthick sections were deparaffinized, washed with PBS, and incubated with 1.5% H2O2 in methanol to block endogenous peroxidase activity. Nonspecific binding was blocked with 10% normal goat serum in PBS. Sections were incubated overnight with the anti-PPAR (1:1,500 in blocking solution), anti-osteopontin (1:1,500), antieCtype IV collagen (1:150; Biodesign International, Saco, ME), antieCTGF-1 (1:100; Santa Cruz Biotechnology, Santa Cruz, CA), anti-F4/80 (1:100; Serotek, Oxford, U.K.), and antieCcaspase-3 (1:50; SantaCruz Biotechnology) in a humidified chamber at 4°C. For immunohistochemistry, tissue sections were treated with an antigen unmasking solution consisting of 10 mmol/l Na citrate, pH 6.0, and 0.05% Tween 20. Antibodies were localized with the ABC technique (Vector Labs, Buringame, CA) and 3,3-diamninobenzidine substrate solution with nickel chloride enhancement. Sections were then dehydrated in ethanol, cleared in xylene, and mounted without counterstaining.

    All of these sections were examined in a masked manner using light microscopy (Olympus BX-50; Olympus Optical, Tokyo, Japan). For the quantification of proportional area of staining, 20 views (x400 magnification) were randomly located in the renal cortex and corticomedullary junction of each slide (Scion Image Beta 4.0.2; Scion, Frederick, MD).

    Terminal deoxynucleotide transferase-mediated dUTP nick-end labeling assay.

    Detection of apoptotic cells in formalin-fixed, paraffin-embedded tissue was performed by in situ terminal deoxynucleotide transferase-mediated dUTP nick-end labeling (TUNEL), using a commercially available kit (Apoptag Plus; Intergen Company, New York, NY). Sections were deparaffinized, digested with proteinase K (20 e蘥/ml), and incubated with hydrogen peroxyde to block endogenous peroxidase activity. After washing, slides were incubated with the TdT enzyme and then with antieCdigoxigenin-peroxidase and developed using a substrate containing diaminobenzidine. Negative controls included omission of TdT. Positive apoptotic cells exhibited an intense brown nuclear colorimetric reaction product. The number of cells positive for TUNEL reaction was determined in the whole glomeruli biopsy under x400 magnification.

    Cell culture.

    Mouse mesangial cells were obtained from primary cell culture of 129SvJ mice using sieving methods. Mouse mesangial cells cultured in Dulbecco’s modified Eagle’s medium (DMEM) containing 5 mmol/l D-glucose and 10% FBS with penicillin 100 units/ml and streptomycin 100 e蘥/ml at 37°C in atmosphere containing 5% CO2 with 95% humidity. Experiments were carried out in DMEM containing 0.2% FBS using cells between the third and sixth passages. At these passages mesangial cells showed normal morphology and function. Cell viability was measured with tryptan blue exclusion and lactate dehydrogenase assay (Sigma Chemicals, St. Louis, MO). These results showed no loss of viability after 4 days of incubation and various concentrations of medium used (data not shown).

    Effects of fenofibrate on TGF-1 and type IV collagen expression and effect of osteopontin on type IV collagen expression in cultured mouse mesangial cells.

    To examine the effects of fenofibrate on TGF-1 and type IV collagen expression in 30 mmol/l high-glucose media, mesangial cells (2 x 104) were seeded in each well of 24-well plates. After washing cells three times with 1x PBS, normal (5 mmol/l) or high-glucose (30 mmol/l) medium containing 0.1% FBS was added to the cells in the presence of or absence of fenofibrate (10 e蘭ol). Total TGF-1 and type IV collagen were measured in cell culture supernatants from mesangial cells with specific enzyme-linked immunosorbent assay (ELISA) kits (Predicta TGF-1 Kit and Genzyme; Biodesign International, Saco, MA) according to the manufactures’ instructions. The absorbance was measured at 450 nm in an ELISA reader (14).

    Leukocyte adhesion assay.

    To examine the effect of high glucose on leukocyte adhesion on mouse mesangial cells, mesangial cells were cultured for 24 h with normal (5 mmol/l) or high glucose (30 mmol/l). For adhesion assay, we also used fenofibrate (10 e蘭ol) or anti-osteopontin antibody on normal- or high-glucoseeCinduced leukocyte adhesion to mesangial cells. Leukocyte adhesion was assayed by modified methods from another study (15). Briefly, confluent mesangial cell monolayers (1 x 105) on 12-mm coverslips were coincubated for 30 min at room temperature under static condition in six-well cell culture plates with human leukocyte (1 x 105) in 1 ml of DMEM plus 1% FCS. In this assay, we used human leukocytes obtained from healthy male volunteers. To remove unattached leukocytes, each coverslip was washed by immersion three times in DMEM plus 1% FCS, then once in PBS, and fixed in 2% paraformaldehyde at 4°C. After staining with hematoxylin, the adherent leukocytes were counted in 20 selected at random high power field (x100) on each of the coverslips and the results expressed as the mean number of leukocytes for each field.

    Statistical analysis.

    Data are expressed as means ± SD. Differences between the groups were examined for statistical significance using ANOVA with Bonferroni correction (SPSS 11.5; SPSS, Chicago, IL). A P value of <0.05 was considered a statistically significant difference. In mesangial cell culture studies, the average of the six experimental measurements was used for statistical analysis.

    RESULTS

    Body weight and levels of glucose and A1C.

    The body weight of diabetic PPAR-knockout mice was less than that of nondiabetic wild-type mice throughout the entire period of experimentation (P < 0.05; Table 1). Although diabetic wild-type mice had slightly reduced body weight compared with nondiabetic wild-type mice, the difference was not significant. Diabetic PPAR-knockout and wild-type mice exhibited dramatically increased blood glucose and A1C levels throughout the entire period of experimentation (P < 0.001) compared with those of nondiabetic PPAR-knockout and wild-type mice. However, there were no significant differences in blood glucose and A1C between diabetic PPAR-knockout and diabetic wild-type mice.

    Blood chemistry and serum lipid levels.

    Na+, K+, CleC, urea nitrogen, and creatinine concentration in blood and hematocrit did not differ significantly between experimental groups (Table 1). In contrast, serum free fatty acid and triglyceride levels were significantly increased in diabetic PPAR-knockout mice compared with those of other groups (P < 0.05; Table 1). There were no differences in total cholesterol concentrations between groups.

    Kidney weight, albuminuria, creatinine, and systolic blood pressure.

    As expected, diabetic mice exhibited increased 24-h urine volumes and kidney weights compared with those of nondiabetic PPAR-knockout and wild-type mice (P < 0.05; Table 2). Interestingly, the kidney weight of diabetic PPAR-knockout mice was heavier than that of diabetic wild mice (P < 0.05; Table 2). Diabetic PPAR-knockout mice also exhibited a persistent increase in urine albumin excretion, whereas albuminuria in diabetic wild-type mice, although initially elevated, declined toward baseline after 3eC4 months of hyperglycemia (P < 0.01; Fig. 1). No significant differences in systolic blood pressure (Table 1) were observed among the groups.

    Expression of PPAR and renal histological examination.

    Immunohistochemistry revealed significantly increased PPAR expression in diabetic wild-type kidneys compared with that in nondiabetic wild-type kidneys. As expected, PPAR was not detected in kidneys of diabetic or nondiabetic PPAR-knockout mice (Fig. 2C and D). The 2.9-fold increase in renal PPAR immunostaining in the wild-type diabetic kidney compared with the nondiabetic kidney occurred primarily in the proximal tubules and thick ascending tubules (Fig. 2E). Glomerular injury in diabetic PPAR-knockout mice was characterized by mesangial extracellular matrix expansion and glomerulosclerosis (Fig. 3AeCD). There was no significant difference in glomerular area between diabetic PPAR-knockout and wild-type mice. Glomerular fractional mesangial areas were increased in diabetic PPAR-knockout mice compared with diabetic wild-type mice and nondiabetic controls (P < 0.01, Fig. 3E).

    Expression of type IV collagen, TGF-, osteopontin, and macrophage infiltration.

    Diabetes was associated with an increase in type IV collagen protein expression in the glomeruli (Fig. 4AeCD). The increase in type IV collagen in the glomeruli was more prominent in diabetic PPAR-knockout mice than in diabetic wild-type mice (P < 0.01, Fig. 4E). In the kidneys of nondiabetic PPAR-knockout and wild-type mice and diabetic wild-type mice, positive staining for TGF- was sparse and localized to the glomeruli (Fig. 4FeCI). In contrast, a prominent increase in TGF- expression was detected in diabetic PPAR-knockout kidneys (Fig. 4J).

    In the kidneys of nondiabetic PPAR-knockout and wild-type mice, sparsely positive osteopontin was observed in the interstitium and glomeruli (Fig. 4KeCN). In the kidneys of diabetic PPAR-knockout mice, osteopontin expression was significantly increased in the glomeruli compared with that of nondiabetic PPAR-knockout and wild-type mice and diabetic wild-type mice (P < 0.05; Fig. 4O). Only modest macrophage infiltration, as assessed by F4/80-positive staining, was observed in the glomeruli of nondiabetic and diabetic wild-type mice (Fig. 4PeCS). In contrast, F4/80 immunostaining was markedly increased in the glomeruli of PPAR-knockout mice. F4/80 immunostaining was strongly increased within the glomeruli of diabetic PPAR-knockout mice compared with that of nondiabetic PPAR-knockout mice (Fig. 4T).

    Expression of TUNEL and caspase 3.

    In the kidneys of nondiabetic PPAR-knockout and wild-type mice as well as in diabetic wild-type mice, a few TUNEL-positive cells were observed within the glomeruli. In the kidneys of diabetic PPAR-knockout mice, diabetes was associated with a markedly increased number of glomerular TUNEL- positive cells (P < 0.001; Fig. 5). Consistent with TUNEL staining reflecting increased apoptosis, caspase 3 immunostaining was also markedly increased in glomerular areas of diabetic PPAR knockout kidneys compared with those of other groups (P < 0.001; Fig. 6).

    Effect of PPAR agonist on glucose-induced type IV collagen and TGF-1 production in cultured mouse mesangial cells and leukocyte adhesion to mesangial cells.

    To determine whether PPAR activation directly modulates glomerular extracellular matrix production and inflammatory cell adhesion to mesangial cells, mesangial cells from a 129SvJ mouse were cultured in normal- (5 mmol/l) or high-glucose (30 mmol/l) media in the presence or absence of the PPAR agonist fenofibrate. Treatment with high glucose concentrations markedly increased type IV collagen and TGF-1 in these cells as determined by ELISA, while treatment with fenofibrate (10 e蘭ol/l) significantly suppressed the production of type IV collagen and TGF-1 (52.2 ± 8.8 vs. 39.8 ± 9.2 ng/ml, P < 0.01, Fig. 7A; 3.14 ± 0.19 vs. 2.65 ± 0.17 pg/104 cells, P < 0.05, Fig. 7B, respectively). In addition, the high-glucose treatment also increased the number of leukocytes adherent to mesangial cells (42 ± 11 vs. 119 ± 15 cells per high-power field, P < 0.05). When mesangial cells cultured in high-glucose media were pretreated with fenofibrate (10 e蘭ol/l) or an anti-osteopontin antibody, high-glucoseeCinduced leukocyte adhesion to mesangial cells was inhibited (63 ± 11 and 68 ± 15 cells per high-power field, respectively, P < 0.05, Fig. 8). In contrast, when mesangial cells were pretreated with fenofibrate and cultured in normal glucose, leukocyte adhesion to mesangial cells tended to decrease (34 ± 7 cells per high-power field), but the difference was not statistically significant.

    DISCUSSION

    We evaluated the role of PPAR activation in the development of diabetic nephropathy. PPAR immunostaining in the kidney was increased in diabetic wild-type mice compared with control mice. Genetic disruption of PPAR expression was associated with abnormal lipid metabolism, including increased circulating free fatty acid and triglyceride concentrations and exacerbation of diabetic nephropathy. More severe structural changes (glomerulosclerosis and mesangial area expansion) as well as functional changes (albuminuria) were noted in diabetic PPAR-knockout mice and were associated with an increase in profibrotic, proinflammatory, and proapoptotic pathways implicated in renal extracellular matrix accumulation. No differences in systolic blood pressure were detected.

    PPAR-knockout mice are reported to develop massive accumulations of myocardial lipids under conditions that increase fatty acid flux (16). Later, Campbell et al. (17) demonstrated that the marked increase in malonyl-CoA, a potent inhibitor of fatty acid oxidation, in the hearts of PPAR-knockout mice is due to decreased expression of malonyl-CoA carboxylase. It is well known that PPAR agonists lower triglyceride levels by increasing lipoprotein lipase gene expression via a PPAR response element (PPRE) in the LPL promoter (18) and decreasing apo C-III levels (19), thus resulting in an enhanced lipolytic activity. In our study, we found that diabetic PPAR-knockout mice exhibit profound metabolic abnormalities, including increased circulating free fatty acid and triglyceride concentrations. Taken together, these results indicate that a lack of PPAR activity in diabetic PPAR-knockout mice is associated with increased circulating free fatty acid and triglyceride concentrations, which could contribute to lipotoxicity in the kidney.

    Recently, Mishra et al. (20) demonstrated that PPAR is a diabetes-induced transcription factor that helps control the kidney’s response to lipids. Cardiac-specific PPAR overexpression increases fatty acid oxidation, elevates lipid storage droplets, and exacerbates cardiomyopathy in diabetes compared with wild-type controls (21). On the other hand, oral fibrates, PPAR agonists that lower serum triglycerides, reduced proteinuria in a subset of diabetic patients (22,23). Our study consistently showed that diabetes increased PPAR expression in diabetic wild-type mice and in vitro PPAR activation in mesangial cells by fenofibrate-attenuated extracellular matrix production associated with reduced TGF-1 levels and leukocyte adherence to mesangial cells. Therefore, the net effect of PPAR activation may be beneficial because of reduced systemic and intrarenal free fatty acid and triglyceride levels. This may underlie the milder renal damage in diabetic wild-type mice compared with diabetic PPAR-knockout mice.

    PPAR also exerts direct and indirect anti-inflammatory activities. Because PPAR enhances the activity of the -oxidation pathway, the consequent degradation of arachidonic acid and leukotrienes directly results in the termination of inflammation (24). In the vascular wall, PPARs interfere with chemo attraction, including monocyte-chemoattractant protein 1 and RANTES (regulated on activation normal T-cell expressed and secreted) and cell adhesion of monocytes, T-cells, and eosinophils (25eC27). Furthermore, PPAR activators inhibit endothelin 1 expression and cytokine-induced vascular cell adhesion molecule 1 (27). In our study, genetic deletion of PPAR, in the presence or absence of diabetes, was associated with increased F4/80 immunostaining in the glomeruli. Furthermore, in the kidneys of diabetic PPAR-knockout mice, F4/80 and osteopontin immunostaining within the glomeruli was increased over and above that of nondiabetic PPAR-knockout mice. These results indicate that both the deletion of the PPAR gene as well as diabetes might be associated with activation of the proinflammatory status. These findings demonstrate that PPAR plays a role in impeding inflammatory cell infiltration into the glomeruli in diabetes by blunting osteopontin expression. This finding is also consistent with a previous in vivo study using db/db mice (28) and with our in vitro primary mesangial cell culture study, which showed that high glucose levels resulted in leukocyte adhesion to mesangial cells and that the PPAR agonist fenofibrate blocked high-glucoseeCinduced leukocyte adhesion to mesangial cells. Therefore, it appears plausible that fenofibrate exerts renoprotective effects in addition to its effect of lowering plasma free fatty acid and triglyceride levels.

    In early diabetic nephropathy, increased amounts of type IV collagen and fibronectin in the mesangium accompany increased mesangial cell proliferation (29). In concord with this association, the glomerular lesions in PPAR-knockout mice exhibited increased type IV collagen and TGF- expression in diabetic kidney disease. The mechanisms involved in the direct anti-fibrotic effects of PPAR agonists on mesangial cells remain to be defined. Interestingly, Wilmer et al. (30) demonstrated that TGF-1 expression mediated by oxidant stress is suppressed by PPAR activation, which inhibits the TRE gene. In our study, the production of the type IV collagen and TGF-1 by mesangial cells pretreated with fenofibrate cultured under high-glucose media was also inhibited. These findings suggest that this PPAR agonist is an endogenous regulator of high-glucoseeCinduced TGF-1 expression.

    Recent studies demonstrated that saturated free fatty acids and hyperglycemia activate the intrinsic pathway of apoptosis in mesangial cells. Apoptosis of glomerular cells correlated with greater expansion of the mesangial matrix and with more severe albuminuria (31,32) associated with increased in TGF- levels in db/db mice. In contrast to nondiabetic PPAR-knockout or diabetic wild-type mice, diabetic PPAR-knockout mice developed significantly more TUNEL-positive glomerular cells. Taken together, reduction of PPAR activity in the diabetic status increases glomerular cell apoptosis and aggravates mesangial matrix expansion.

    Proteinuria in diabetes is considered to have both a hemodynamic (glomerular capillary hypertension and hyperfiltration) and a structural/cellular (alterations in basement membrane, mesangial cell matrix, and podocyte function) basis (33). In our study, diabetic PPAR-knockout mice developed profound proteinuria compared with diabetic wild-type mice. Therefore, we speculate that PPAR agonists or activity may similarly attenuate diabetic renal injury by preventing an increase in extracellular matrix formation in the mesangium and apoptosis in the glomeruli, not by the induction of hemodynamic changes. Accumulating evidence suggests that diabetic nephropathy can be viewed as an inflammatory disease triggered by disordered glucose metabolism (34,35).

    In another experiment with db/db mice, we demonstrated that PPAR activation by fenofibrate significantly increased PPAR immunostaining in the tubule and decreased the glomerular and mesangial surface areas (36). We also demonstrated that fenofibrate treatment markedly increased PPRE-luciferase activity in mesangial cells from db/db mice as determined by PPRE3X luciferase reporter analysis, which is consistent with the existence of endogenous PPAR activity in these cells. Therefore, we suggest that increased PPAR activity in the tubule may exert anti-inflammatory and anti-fibrotic effects via paracrine action resulting from increased PPRE activity in the glomeruli without significant differences in PPAR immunostaining in the glomerular areas.

    In conclusion, deficiency of PPAR in a model of type 1 diabetic nephropathy is associated with increased circulating free fatty acids and triglycerides. This likely contributes to the aggravation of proteinuria and the pathologic changes observed in this model. Activation of PPAR with fenofibrate, a ligand agonist, may effectively prevent the glomerular matrix expansion that accompanied apoptosis and inflammatory cell infiltration in the glomerulus.

    ACKNOWLEDGMENTS

    This study was supported by National Institutes of Health Grants UO1 DK61018 (to M.D.B.), DK38226 (to M.D.B.), and DK065074-01 (to Y.G.).

    We thank Dr. Dae Ryong Cha for excellent assistance. The technical assistance of Linda S. Davis and Hyung-Jung Park is also acknowledged.

    FOOTNOTES

    The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked "advertisement" in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

    REFERENCES

    Isselmann I, Green S: Activation of a member of the steroid hormone receptor superfamily by peroxisome proliferators. Nature 347:645eC650, 1990

    Guan Y, Zhang Y, David L, Breyer M: Expression of peroxisome proliferator-activated receptors in urinary tract of rabbits and humans. Am J Physiol 273:F1013eCF1022, 1997

    Braissant O, Wahli W: Differential expression of peroxisome proliferator-activated receptors--, and - during rat embryonic development. Endocrinology 139:2784eC2754, 1998

    Ouali F, Djouadi F, Merlet-Benichou C, Bastin J: Dietary lipids regulate -oxidation enzyme gene expression in the developing rat kidney. Am J Physiol 275:F777eCF784, 1998

    Roman RJ, Ma YH, Frohlich B, Markham B: Clofibrate prevents the development of hypertension in Dahl salt-sensitive rats. Hypertension 21:985eC988, 1993

    Capdevila JH, Falck JR, Harris RC: Cytochrome P 450 and arachidonic acid bioactivation: molecular and functional properties of the arachidonate monooxygenase. J Lipid Res 41:163eC181, 2000

    Portilla D, Dai G, Peters JM, Gonzalez FA, Crew MD, Proia AD: Etomoxir-induced PPAR alpha-modulated enzymes protect during acute renal failure. Am J Physiol (Renal Physiol ) 278:F667eCF675, 2000

    Schaffer JE: Lipotoxicity: when tissues overeat. Curr Opin Lipidol 14:281eC287, 2003

    Francis GA, Annicotte JS, Auwerx J: PPAR-alpha effects on the heart and other vascular tissues. Am J Physiol Heart Circ Physiol 285:H1eCH9, 2003

    Lee SS, Pineau T, Drago J, Lee EJ, Owens JW, Kroetz DL, Fernandez-Salguero PM, Westphal H, Gonzalez FJ: Targeted disruption of the alpha isoform of the peroxisome proliferator-activated receptor gene in mice results in abolishment of the pleiotropic effects of peroxisome proliferators. Mol Cell Biol 15:3012eC3022, 1995

    Peters JM, Hennuyer N, Staels B, Fruchart JC, Fievet C, Gonzalez FJ, Auwerx J: Alterations in lipoprotein metabolism in peroxisome proliferator-activated receptor alpha-deficient mice. J Biol Chem 272:27307eC27312, 1997

    Kersten S, Seydoux J, Peters JM, Gonzalez FJ, Desvergne B, Wahli W: Peroxisome proliferator-activated receptor alpha mediates the adaptive response to fasting. J Clin Invest 103:1489eC1498, 1999

    Wen M, Segerer S, Dantas M, Brown PA, Hudkins KL, Goodpaster T, Kirk E, LeBoeuf RC, Alpers CE: Renal injury in apolipoprotein E-deficient mice. Lab Invest 82:999eC1006, 2002

    Wolf G, Ziyadeh, Stahl RA: Atrial natriuretic peptide stimulates the expression of transforming growth factor-beta in cultured murine mesangial cells: relationship to suppression of proliferation. J Am Soc Nephrol 6:224eC233, 1995

    Nagel T, Resnick N, Atkinson WJ, Dewey CF Jr, Gimbrone MA Jr: Sheer stress selectively upregulates intercellular adhesion molecule-1 expression in cultured human vascular endothelial cells. J Clin Invest 94:885eC891, 1994

    Djouadi F, Weinheimer CJ, Saffitz JE, Pitchford C, Bastin J, Gonzalez FJ, Kelly DP: A gender-related defect in lipid metabolism and glucose homeostasis in peroxisome proliferator-activated receptor alpha-deficient mice. J Clin Invest 102:1083eC1091, 1998

    Campbell FM, Kozak R, Wagner A, Altarejos JY, Dyck JR, Belke DD, Severson DL, Kelly DP, Lopaschuk GD: A role for peroxisome proliferator-activated receptor alpha (PPARalpha) in the control of cardiac malonyl-CoA levels: reduced fatty acid oxidation rates and increased glucose oxidation rates in the hearts of mice lacking PPARalpha are associated with higher concentrations of malonyl-CoA and reduced expression of malonyl-CoA decarboxylase. J Biol Chem 277:4098eC4103, 2002

    Schoonjans K, Painado-Onsurbe J, Lefebvre AM, Heyman RA, Briggs M, Deeb S, Staels B, Auwerx J: PPAR and PPAR activators direct a distinct tissue-specific transcriptional response via a PPRE in the lipoprotein lipase gene. EMBO J 15:5336eC5348, 1996

    Staels B, Vu-Dac N, Kosykh VA, Saladin R, Fruchart JC, Dallongevillae J, Auwerx J: Fibrates downregulate apolipoprotein C-III expression independent of induction of peroxisomal acyl coenzyme A oxidase. J Clin Invest 95:705eC712, 1995

    Mishra R, Emancipator SN, Miller C, Kern T, Simonson MS: Adipose differentiation-related protein and regulators of lipid homeostasis identified by gene expression profiling in the murine dbb/dbb diabetic kidney. Am J Physiol Renal Physiol 286:F913eCF921, 2004

    Finck BN, Han X, Courtois M, Aimond F, Nerbonne JM, Kovacs A, Gross RW, Kelly DP: A critical role for PPAR-mediated lipitoxicity in the pathogenesis of diabetic cardiamyopathy: modulation by dietary fat content. Proc Natl Acad Sci U S A 100:1226eC1231, 2003

    Fried LF, Orchard TJ, Kasiske BL: Effect of lipid reduction on the progression of renal disease: a meta analysis. Kidney Int 59:260eC269, 2001

    Smulders YM, van Eeden AE, Stehouwer CD, Weigers RN, Slaats EH, Silberbusch J: Can reduction in hypertriglyceridaemia slow progression of microalbuminuria in patients with non-insulin-dependent diabetes mellitus Eur J Clin Invest 27997eC1002, 2002

    Devchand PR, Keller H, Peters JM, Gonzalez FJ, Wahli W: The PPAR-leukotrien B4 pathway to inflammation control. Nature 384:39eC43, 1996

    Murao K, Imachi H, Momoi A, Sayo Y, Hosokawa H, Sato M, Ishida T, Takahara T: Thiazolidinedione inhibit the production of monocyte chemoattractant protein-1 in cytokine-treated human vascular endothelial cells. FEBS Lett 454:27eC30, 1999

    Jackson SM, Parhami F, Xi XP, Berliner JA, Law RE, Demer LL: Peroxisome proliferator-activated receptor activators target human endothelial cells to inhibit leukocyte-endothelial cell interaction. Arterioscler Throm Vasc Biol 19:2094eC2104, 1999

    Satoh H, Tsukamoto K, Hashimoto Y, Hashimoto N, Togo M, Hara M, Maekawa H, Isoo N, Kimura S, Watanabe T: Thiazolidinediones suppress endothelin-1 secrestion from bovine vascular endothelial cells: a new possible role of PPAR on vascular endothelial function. Biochem Biophys Res Commun 254:757eC763, 1999

    Chow F, Ozols E, Nikolic-Paterson DJ, Atkins RC, Tesch GH: Macrophages in mouse type 2 diabetic nephropathy: correlation with diabetic state and progressive renal injury. Kidney Int 65:116eC128, 2004

    Young BA, Johnson RJ, Apers CE, Eng E, Gordon K, Flooege J, Couser WG, Seidel K: Cellular event in the evolution of experimental diabetic nephropathy. Kidney Int 47: 935:944, 1995

    Wilmer WA, Dixon CL, Herbert C, Lu L, Rovin BH: PPAR- ligands inhibit H2O2-mediated activation of transforming growth factor-1 in human mesangial cells. Antioxid Redox Signal 4:877eC884, 2002

    Mishra R, Emancipator SN, Kern T, Simonson MS: High glucose evokes an intrinsic proapoptotic signaling pathway in mesangial cells. Kidney Int 67:82eC93, 2005

    Mishra R, Simonson MS: Saturated free fatty acids and apoptosis in microvascular mesangial cells: palmitate activates pro-apoptotic signaling involving caspase 9 and mitochondrial release of endonuclease G. Cardiovasc Diabetol 4:2, 2005

    Komers R, Anderson S: Paradoxes or nitric oxide in the diabetic kidney. Am J Physiol Renal Physiol 284:F1121eCF1137, 2003

    Kelly DJ, Chanty A, Gow RM, Zhang Y, Gilbert RE: Protein kinase C beta inhibition attenuates osteopontin expression, macrophage recruitment, and tubulointerstitial injury in advanced experimental nephropathy. J Am Soc Nephrol 16:1654eC1660, 2005

    Chow FY, Nikolic-Paterson DJ, Ozols E, Atkins RC, Tesch GH: Intracellular adhesion molecule-1 deficiency is protective against nephropathy in type 2 diabetic db/db mice. J Am Soc Nephrol 16:1711eC1722, 2005

    Park CW, Zhang Y, Zhang X, Wu J, Chen L, Cha DR, Su D, Hwang MT, Fan X, David L, Striker GE, Zheng F, Breyer MD, Guan Y: PPAR agonist fenofibrate improves diabetic nephropathy in db/db mice. Kidney Int. In press(Cheol Whee Park, Hyeong W)