干细胞之家 - 中国干细胞行业门户第一站

 

 

搜索
朗日生物

免疫细胞治疗专区

欢迎关注干细胞微信公众号

  
查看: 484743|回复: 232
go

PPAR agonists exert antifibrotic effects in renal tubular cells exposed to high [复制链接]

Rank: 1

积分
威望
0  
包包
0  
楼主
发表于 2009-4-21 13:02 |只看该作者 |倒序浏览 |打印
Department of Medicine, University of Sydney, and Renal Research Group, Kolling Institute of Medical Research, Royal North Shore Hospital, New South Wales, Australia
; D/ K' K  |) l/ g, ]* D  w( ~' s5 I
ABSTRACT
0 F" m8 y0 C5 d: h! z7 b, k4 ^
0 ?, I0 {7 x! b  ~- |' `5 BPeroxisome proliferator-activated receptor- (PPAR) are ligand-activated transcription factors that regulate cell growth, inflammation, lipid metabolism, and insulin sensitivity. We recently demonstrated that PPAR agonists limit high glucose-induced inflammation in a model of proximal tubular cells (PTC; Panchapakesan U, Pollock CA, and Chen XM. Am J Physiol Renal Physiol 287: F528–F534, 2004). However, the role of PPAR in the excess extracellular matrix production is largely unknown. We evaluated the effect of 24- to 48-h 8 μM L-805645 or 10 μM pioglitazone on 25 mM D-glucose-induced markers of fibrosis in HK-2 cells. High D-glucose induced nuclear binding of activator protein-1 (AP-1) to 140.8 ± 10.9% (P
9 a6 K+ o( O( Q- T
- o+ [7 Y- x: D( Pthiazolidinediones; diabetic nephropathy; proximal tubular cells
9 C5 @( |8 ~/ O! w
* @6 }% c- |' X9 S2 jDIABETES MELLITUS (DM) IS a major global health issue with up to one-third of patients suffering end-stage renal disease. The major burden of disease is derived from patients with type 2 DM (25), resulting in up to 50% of adult patients on dialysis in Westernized countries having diabetic nephropathy as their primary diagnosis.; B) }+ I8 e6 j) I: ^5 y

+ s4 j* }* ^7 c6 C) F6 D6 cThe interventions shown to prevent the onset or attenuate the progression of diabetic nephropathy include glycemic and blood pressure control (10, 32), specific interruption of the renin-angiotensin system with either angiotensin-converting enzyme inhibitors (30) or angiotensin II receptor blockers (6, 20) and potentially lipid-lowering therapy (3, 9). Understanding the mechanisms underlying the development of diabetic nephropathy is essential in establishing novel therapeutic strategies for the prevention or arrest of progressive renal failure. The search for specific molecular targets is ongoing, and PPAR agonists may show promise.' w  p/ k1 W' B$ O2 R" A

* q5 m& d3 r- @$ sPPAR are members of the nuclear hormone receptor superfamily of ligand-activated transcription factors that have a central role in regulating insulin sensitivity, adipocyte differentiation, cell growth, and inflammation (13). Synthetic agonists of PPAR like the thiazolidinediones (TZDs) are widely used as insulin-sensitizing agents in patients with type 2 DM. On ligand binding, PPAR form heterodimers with the retinoic acid receptor (23). This complex then binds to the PPAR response elements (PPREs) within the promoter region of target genes. Regulation of these target genes depends on the binding of ligands as well as complex interaction between resident coactivators and corepressors (36). Endogenous ligands include 15-deoxy-12,14-prostaglandin J2 (11, 17) and metabolites of oxidized low-density lipoproteins (LDL) (24, 31). Synthetic ligands include the TZDs and phenylacetic acid derivatives such as L-805645, which are specific, and selective PPAR ligands (5).0 ~  ~& e+ `3 a3 f
9 }% ^& f$ X" X6 ^
There are several clinical studies demonstrating a beneficial trend, with a reduction in albuminuria in patients with type 2 DM treated with TZDs (5, 15, 18). This finding is also reflected in animal models of experimental diabetic nephropathy where treatment with TZDs reduces albuminuria and decreases glomerular matrix deposition and glomerulosclerosis (7, 8, 22, 39). These benefits appear to be independent of glycemic control. Haplo-insuffficient PPAR /– db/db mice exhibit more severe hyperglycemia, albuminuria, and glomerular pathology (39). The renoprotective benefit of PPAR agonists is further suggested by studies in nondiabetic models of renal injury, such as the 5/6-nephrectomy model, where activation of PPAR reduced glomerulosclerosis (21). In vitro studies have focused on the use of mesangial cells where PPAR has been well characterized (2, 26), with specific PPAR activation exerting an antiproliferative (12, 26) and antifibrotic effect, reducing type 1 collagen synthesis and secretion (29) presumed due to a transforming growth factor (TGF)-1-dependent mechanism (34). In mesangial cells, pioglitazone has been shown to inhibit TGF--induced fibronectin production (14). In our own study using an immortalized proximal tubular cell model (opossum kidney cell line) under normal (5 mM)-glucose conditions, we have previously demonstrated that PPAR agonists stimulate tubular albumin uptake without provoking an inflammatory response (37). However, whether they protect the kidney from tubulointerstitial fibrosis, the hallmark of a progressive decline in renal function under hyperglycemic conditions, is unknown. Thickening of the basement membrane due to an increase in extracellular matrix production by renal proximal tubular cells is the initial pathological abnormality evident in diabetic nephropathy and central to the development of tubulointerstitial injury. Hence, our studies were designed to assess the effects of PPAR agonists on the transcription factors activator protein-1 (AP-1) and NF-B, the profibrotic cytokine TGF-1, and the extracellular matrix proteins fibronectin and collagen IV in proximal tubular cells after short-term exposure to high-glucose conditions.
7 e5 H+ L) y7 c6 ~, Y$ L+ d
6 B  U4 x# g9 ^METHODS- G! c: Z& z1 X4 V& V

* }. T8 e7 M- H) QCell culture. HK-2 cells, a primary human proximal tubular cell line (a gift from Prof. J Charlesworth, Sydney, Australia), were grown in keratinocyte serum-free media (KSFM) supplemented with bovine pituitary extract (20–30 μg/ml) and epidermal growth factor (0.1–0.2 ng/ml, GIBCO). Cell culture media was changed every 48–72 h. These cells were grown at 37°C in a humidified 5% CO2 incubator and were subcultured at 50–80% confluence using 0.05% trypsin-0.02% EDTA (GIBCO).  m' J' r9 B3 n* y4 k3 ?# ]* `
3 D! H6 A% h, @& @, `
The clinically available thiazolidinedione pioglitazone (10 μM, Cayman Chemical) and the more selective PPAR agonist L-805645 (8 μM, Merck) were used to determine the specific effects of PPAR activation in this proximal tubular model. These concentrations were chosen based on their ability to significantly upregulate PPAR expression in this model and have been used in previous studies (27).* D7 s  L0 U( {: G; X  A, m: l' C# y
' g' h8 l0 }  G" i+ T% `) g! |
When 80% confluent, HK-2 cells were exposed to the following experimental conditions for 24 h. For TGF- ELISA, cells were exposed for 48 h: 1) 5 mM D-glucose (control media); 2) 30 mM D-glucose (ICN Biomedical); 3) 5 mM D-glucose and 25 mM L-glucose (osmotic control, ICN Biomedical); 4) 8 μM L-805645 in 5 mM D-glucose; 5) 8 μM L-805645 in 30 mM D-glucose; 6) 10 μM pioglitazone in 5 mM D-glucose; and 7) 10 μM pioglitazone in 30 mM D-glucose.: o7 ]" W  e# O

, D) g& X) h- B" ^3 G* G, XAs L-805645 and pioglitazone were dissolved in 0.016 and 0.13% DMSO, respectively, additional controls were undertaken to evaluate independent effects of the DMSO at 0.13%.7 m5 x/ i2 ]/ y; L% u* |

4 D8 T% J0 I" P, TNuclear extraction and EMSA for AP-1 and NF-B. After exposure to the above-mentioned experimental conditions, nuclear extracts were prepared using a NucBuster Protein Extraction Kit (Novagen, Darmstadt, Germany) as per the manufacturer*s instructions. A digoxygenin (DIG) Gel Shift Kit (Roche Applied Science, Indianapolis, IN) was used in the EMSA. In brief, 25 μg of nuclear extract were incubated with 1 μg poly [d(I-C)] as the nonspecific competitor, 1 μg poly L-lysine in a binding buffer [(in mM) 100 HEPES, pH 7.6, 5 EDTA, 50 (NH4)2SO4, 5 DTT, and 150 KCl, as well as 1% Tween 20, wt/vol] and DIG-labeled AP-1 (5'-CGC TTG ATG AGT CAG CCG GAA-3') or NF-B (5'-AGT TGA GGG GAC TTT CCC AGG C-3') consensus oligonucleotide (Promega) for 30 min at room temperature. Unlabeled AP-1 and NF-B consensus oligonucleotides were used as specific competitors, respectively. The reaction mixture was electrophoresed through 6% polyacrylamide gels, transferred onto positively charged nylon membrane (Roche Applied Science), and then cross-linked using an UV transilluminator for 3 min. The membrane was subjected to immunological detection using anti-DIG-AP conjugate and chemiluminescence. Results were analyzed using Image J software, and shift bands were measured., t8 N: ~9 A, T# U+ d
' R" X, Z$ B7 X! F
TGF-1. Cells were seeded at 2 x 105 cells/well in a 24-well plate and grown in KSFM without growth factors for 24 h. At 70% confluence, cells were exposed to the experimental conditions as defined above for 48 h in quadruplicate. Supernatants were then collected, spun, and stored at –20~ C until TGF-1 levels were determined by immunoassay (Promega) as per the manufacturer*s instructions. Cell lysate protein concentration was determined (Bio-Rad), and TGF-1 levels were corrected for protein content per well., {  P1 X7 ]6 J: s8 a: V: v

9 i1 T# n$ W* h& J& c3 x& y, KWestern blot analysis. Cells were collected, and the cell pellet was resuspended in cell lysis buffer containing 50 mM Tris﹞HCl, 150 mM NaCl, 5 mM EDTA (pH 7.4), 0.5% Triton X-100, and protease inhibitors (Roche Diagnostics, Mannheim, Germany). Cell lysate was then sonicated to release total cell proteins, spun at 12,000 rpm at 4°C, and stored at –80°C.7 U: n  G5 W) A5 J' B% J( X6 c* F
* [# d4 U' X( e' ]* L& X' i4 e
Protein assay (Bio-Rad) was done to determine the protein concentration of the cell lysate. Eighty micrograms of total cell protein were mixed with 6x Laemmli sample buffer containing mercaptoethanol and heated at 95°C for 10 min. Samples were then analyzed by SDS-PAGE in 7.5% gels and electroblotted to Hybond Nitrocellulose membranes (Amersham Pharmacia Biotech, Bucks, UK). Membranes were blocked in Tris-buffered saline containing 0.2% Tween 20 (TTBS) in 5% skim milk for 2–3 h, incubated overnight at 4°C with fibronectin (1:100, NeoMarkers), collagen IV (1:5,000, Abcam, Cambridge, UK), then reprobed with actin (1:300, Santa Cruz) in TTBS containing 5% skim milk. Membranes were washed with TTBS and incubated with horseradish peroxidase-conjugated secondary antibody. Proteins were visualized using the enhanced chemiluminescence detection system (Amersham Pharmacia Biotech). Results were corrected for actin.8 [! h. k% ~2 s, W1 n# s, D8 U

( j/ r& z/ T* c3 _$ ]Statistical analysis. Results are expressed as a percentage of the mean ± SE of control values. Experiments were performed at least in triplicate or as detailed in the individual experimental protocols. Statistical comparisons between groups were made by ANOVA or unpaired t-tests where appropriate. Analyses were performed using the software package StatView version 5.0 (Abacus Concepts, Berkley, CA). P values
5 w- D+ h5 {: v5 L6 P- P7 y/ q! V9 p7 \& J. C9 d/ O2 A
RESULTS( ?- X& K" r: t
6 {) M' t2 c- n! v
AP-1. High D-glucose increased the DNA binding of DIG-labeled AP-1 to 140.8 ± 10.9% of control (P
! ]$ v! l  ~' \5 }; M0 o8 Q4 C5 ^* A+ T$ ?3 r" j3 z
TGF-1. High D-glucose increased total TGF-1 secretion to 4.82 pg﹞ml–1﹞unit protein cell lysate–1 or 139.6 ± 6.5% of control (P " D" s8 T0 L* S9 N  n+ J- z
3 G" t3 r. I8 Z$ J
Fibronectin. The protein expression of fibronectin, an extracellular matrix protein downstream to TGF-1, was increased in the presence of high glucose to 156.0 ± 24.9% of control (P " U1 U1 x+ o0 j. t

9 F8 o3 B- }& uCollagen IV. There was no significant upregulation of collagen IV expression following short-term exposure to high glucose (110 ± 23.2%; P = not significant), suggesting that the initial increase in extracellular matrix is largely as a result of deposition of noncollagen proteins. Nevertheless, both classes of PPAR agonists were able to suppress collagen IV expression to levels below that observed in high-glucose conditions (68.0 ± 14.5%, P 4 k/ I' X: d& C
* H# h( o8 \) K. ~
NF-B. High glucose increased the binding of DIG-labeled NF-B to 167.3 ± 22.4% of control (P ! V3 h8 T$ Z& {' ?
# b, Z6 r, p, W% P( e! N4 O9 Q
DISCUSSION1 F. m6 V7 {6 V8 s4 p9 n: G/ [! ^( ?

; J0 }9 {8 A( T: m* b0 s6 ?8 t& xTDZs are widely used as insulin-sensitizing agents in the treatment of type 2 diabetes. They appear to exert a renoprotective effect in animal models and in vitro studies, which until recently have focused on mesangial cells (2, 12, 14, 26, 34). Recent work from our laboratory has demonstrated that PPAR agonists limit LDL- and albumin-induced proinflammatory responses in the human proximal tubule (37) and reduce extracellular matrix production by human cortical fibroblasts (38). However, their role and function in human proximal tubular cells under high-glucose conditions inherent in diabetic nephropathy have not been well defined. In the present study, we demonstrate that PPAR ligands reverse the high glucose-induced profibrotic responses in the proximal tubule. As thickening of the basement membrane of the proximal tubule is the earliest pathological response observed in the development of diabetic nephropathy, these findings suggest that early use of PPAR agonists may limit the development of diabetic nephropathy.
; W! y; v2 g: ^4 w' z+ R3 `+ G6 X4 j; S9 x7 i+ o  M
The results of these studies suggest that the AP-1 pathway is upregulated following exposure to high-glucose conditions, an effect that is due, at least in part, to the hyperosmolar effect. The TGF- gene is a known target gene for the transcription factor AP-1 (35). Furthermore, in mesangial cells TZDs prevented high-glucose induction of TGF-1 promoter activity and elevation of nuclear c-fos (subunit of AP-1) protein levels. TGF-1 stimulates various extracellular matrix genes, including fibronectin. TZDs also inhibit TGF-1-induced fibronectin expression in mesangial cells (14). Clearly, our studies demonstrated that TGF-1 was increased specifically by exposure to high glucose, suggesting downstream specificity of AP-1 activation with respect to target cytokines. Both the TZD and non-TZD PPAR agonists reduced AP-1 expression and reversed the high glucose-induced increase in TGF-1, although the effect was more marked in the presence of the TZD pioglitazone. The increased expression of both AP-1 and TGF-1 was translated into an increase in fibronectin expression, which was also reversed in the presence of both the TZD and non-TZD agonists. This effect was seen despite the increase in TGF- seen with DMSO, the vehicle in which L-805645 and pioglitazone were dissolved (data not shown). Hence, these results taken together strongly support the notion that these agonists exert antifibrotic effects in proximal tubular cells by attenuating AP-1, its target, i.e., TGF-1, and the downstream fibronectin.% P# i# v5 Y: |) ?% K

) c5 b' _8 a8 f0 h, I6 fWe previously showed that short-term exposure to high D-glucose upregulates PPAR (27). This could be viewed as a protective compensatory response, which on further upregulation with the use of synthetic agonists, exerts renoprotective effects. This upregulation may be selective or sufficient in limiting the early expression of collagen IV on exposure to high glucose but not fibronectin. Alternatively, collagen IV may be deposited later in the course of fibrosis.4 Y3 P+ r* A  K! P0 ^

0 \; ~4 K& |3 c" oMonocyte chemotactic protein-1 (MCP-1) is known to be increased in diabetic nephropathy and considered to play an important role in the development of progressive tubulointerstitial fibrosis. Specifically, using immunohistochemical and in situ hybridization analyses, MCP-1-positive cells were found to be present in the advanced tubulointerstitial lesions of diabetic nephropathy and correlated with urinary MCP-1 levels (33). Part of the therapeutic benefit of angiotensin-converting enzyme inhibitors is considered to be mediated by a reduction in renal MCP-1 production (1, 16). We have previously demonstrated that a reduction in tubular production of MCP-1 is associated with an upregulation of PPAR (27). From our current data, this is independent of NF-B regulation. This is in keeping with previous data from our group showing that PPAR activation similarly reduces an LDL-induced increase in MCP-1, independently of modification of NF-B transcriptional pathway (37). The signaling pathways that govern MCP-1 expression in the human kidney are unknown. Our results suggest that the AP-1 pathway, modified by PPAR agonist activity, is likely to be at least in part responsible for reduction of transcription factors known to be involved in profibrotic and also proinflammatory pathways. This is consistent with the known murine MCP-1 promoter, which contains AP-1 and SP-1, in addition to NF-B promoter hypermethylation and orphan sites, all of which regulate MCP-1 activity (28). Hence, its modification is of key therapeutic significance. Of note, we found that DMSO vehicle increased NF-B binding (data not shown), consistent with recently reported data (19). This is important as it may be a confounding factor limiting the effects of the PPAR agonists.5 S* ~: i2 l3 n! q3 N
  z. J* V! J! X# f
Therapeutic strategies to delay or attenuate the progression of diabetic nephropathy are essential to the treatment of patients with DM. These results provide new knowledge as to whether targeting PPAR activation in patients with DM will ultimately reduce the burden of diabetic nephropathy.
: p' m. p+ J5 A8 C' u0 t
# S8 S7 B2 z: L. y3 V9 V1 ~: DGRANTS
( A6 O2 [* d; J2 U9 V) F( ]4 k' s5 d5 W0 V; v6 M* O2 x
U. Panchapakesan was funded by a Jacquot Research Entry Scholarship. We also acknowledge support by the National Health and Medical Research Council of Australia, Juvenile Diabetes Research Foundation, Diabetes Australia Research Trust, and Merck Laboratories for generously providing us with L-805645.
! g& ?: d- a2 V7 r+ o3 h
/ h- a: e0 Q* Z) U+ t6 dFOOTNOTES( |* _! J3 F* O" r& M0 r

5 K  H" f+ _# R; N" U  sThe costs of publication of this article were defrayed in part by the payment of page charges. The article must therefore be hereby marked "advertisement" in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.6 r8 u1 z4 T4 i& v

+ E; p1 a" K: j) o6 O, mREFERENCES% Q# e9 E. K  B9 x4 ~
8 K, }$ w( I& Q7 f8 \
Amann B, Tinzmann R, and Angelkort B. ACE inhibitors improve diabetic nephropathy through suppression of renal MCP-1. Diabetes Care 26: 2421–2425, 2003.
' @" _( V# V6 n, Q* U  E8 v; [" i6 q1 d) _0 f1 y. Q
Asano T, Wakisaka M, Yoshinari M, Iino K, Sonoki K, Iwase M, and Fujishima M. Peroxisome proliferator-activated receptor 1 (PPAR1) expresses in rat mesangial cells and PPAR agonists modulate its differentiation. Biochim Biophys Acta 1497: 148–154, 2000.' q! y/ r4 G4 G( H+ j

1 @+ q: e" X  x+ O: ?$ sAthyros VG, Mikhailidis DP, Papageorgiou AA, Symeonidis AN, Pehlivanidis AN, Bouloukos VI, and Elisaf M. The effect of statins versus untreated dyslipidaemia on renal function in patients with coronary heart disease. A subgroup analysis of the Greek atorvastatin and coronary heart disease evaluation (GREACE) study. J Clin Pathol 57: 728–734, 2004.
3 H( v1 T+ s- t
$ W9 S6 W) l6 x. \3 YBakris G, Viberti G, Weston WM, Heise M, Porter LE, and Freed MI. Rosiglitazone reduces urinary albumin excretion in type II diabetes. J Hum Hypertens 17: 7–12, 2003.
/ X- U* D0 D6 J; |5 k3 {  y& |8 Q% G$ e; i
Berger J and Wagner JA. Physiological and therapeutic roles of peroxisome proliferator-activated receptors. Diabetes Technol Ther 4: 163–174, 2002.
. B7 }2 ^; h7 ]2 j$ v  N, g! L5 f/ U& K/ @& l/ K
Brenner BM, Cooper ME, de Zeeuw D, Keane WF, Mitch WE, Parving HH, Remuzzi G, Snapinn SM, Zhang Z, and Shahinfar S. Effects of losartan on renal and cardiovascular outcomes in patients with type 2 diabetes and nephropathy. N Engl J Med 345: 861–869, 2001.
- q" C7 V0 N7 ^) U+ |- j+ t) a& H
Buckingham RE, Al-Barazanji KA, Toseland CD, Slaughter M, Connor SC, West A, Bond B, Turner NC, and Clapham JC. Peroxisome proliferator-activated receptor-gamma agonist, rosiglitazone, protects against nephropathy and pancreatic islet abnormalities in Zucker fatty rats. Diabetes 47: 1326–1334, 1998.: x3 K/ ]$ b  B$ Z6 e& C) F3 z
' c/ O$ Q9 e% Y2 U/ U+ X: }
Chen J, Sun Z, Ma J, Gu Y, Yang H, and Lin S. Abstract: non-metabolic protective effects of pioglitazone on diabetic nephropathy induced by streptozotocin in uninephrectomized rat (Abstract). J Am Soc Nephrol 14: 392A, 2003.  J: L2 S3 h/ {" c) ~6 `: P
% ^. I- X7 ~9 Q3 z5 i1 o* s
Collins R, Armitage J, Parish S, Sleigh P, and Peto R. MRC/BHF Heart Protection Study of cholesterol-lowering with simvastatin in 5,963 people with diabetes: a randomised placebo-controlled trial. Lancet 361: 2005–2016, 2003.
) v4 {. D/ _5 \4 H% a" X' d9 h& F2 u
The Diabetes Control and Complications Research Group. Effect of intensive therapy on the development, and progression of diabetic nephropathy in the Diabetes Control and Complications Trial. The Diabetes Control and Complications (DCCT) Research Group. Kidney Int 47: 1703–1720, 1995.
: T+ K' s3 P" j: u
6 N2 q% R$ D6 n: K6 o4 R2 ]7 g* pForman BM, Tontonoz P, Chen J, Brun RP, Spiegelman BM, and Evans RM. 15-Deoxy-delta 12, 14-prostaglandin J2 is a ligand for the adipocyte determination factor PPAR. Cell 83: 803–812, 1995.
0 ^5 W* C+ T1 W8 V7 C: k, p( L1 k3 N: |% ?# Y
Ghosh SS, Gehr TW, Ghosh S, Fakhry I, Sica DA, Lyall V, and Schoolwerth AC. PPAR ligand attenuates PDGF-induced mesangial cell proliferation: role of MAP kinase. Kidney Int 64: 52–62, 2003.! V% F' E" s7 m# q

5 J4 }8 Z3 D; |, q  wGuan Y, Zhang Y, and Breyer MD. The role of PPARs in the transcriptional control of cellular processes. Drug News Perspect 15: 147–154, 2002.
; r# O4 U. `! @6 f% R
$ }6 Y' _4 o1 r# R' P" zGuo B, Koya D, Isono M, Sugimoto T, Kashiwagi A, and Haneda M. Peroxisome proliferator-activated receptor-gamma ligands inhibit TGF-1-induced fibronectin expression in glomerular mesangial cells. Diabetes 53: 200–208, 2004.
9 G7 a7 k1 z" F) e+ T
, Q6 K6 p( b7 B5 K; t: A8 @Imano E, Kanda T, Nakatani Y, Nishida T, Arai K, Motomura M, Kajimoto Y, Yamasaki Y, and Hori M. Effect of troglitazone on microalbuminuria in patients with incipient diabetic nephropathy. Diabetes Care 21: 2135–2139, 1998.- y# E: e$ s- G% C

% \' \: ]# O% V8 h' c3 aKato S, Luyckx VA, Ots M, Lee KW, Ziai F, Troy JL, Brenner BM, and MacKenzie HS. Renin-angiotensin blockade lowers MCP-1 expression in diabetic rats. Kidney Int 56: 1037–1048, 1999.. G$ i9 l# P* E+ x- M. u# |
& c+ E9 B* j; j9 \: S  C
Kliewer SA, Lenhard JM, Willson TM, Patel I, Morris DC, and Lehmann JM. A prostaglandin J2 metabolite binds peroxisome proliferator-activated receptor  and promotes adipocyte differentiation. Cell 83: 813–819, 1995.! z: J* Q( O. Y4 K# D

8 N: @+ m$ r5 ^+ B+ Y9 O4 d- bLebovitz HE, Dole JF, Patwardhan R, Rappaport EB, and Freed MI. Rosiglitazone monotherapy is effective in patients with type 2 diabetes. J Clin Endocrinol Metab 86: 280–288, 2001./ i, h! b' x$ l- d4 O5 ?  ]' q
& G9 Q9 j% y! f+ E" X# a
Lee YR, Shim HJ, Yu HN, Song EK, Park J, Kwon KB, Park JW, Rho HW, Park BH, Han MK, and Kim JS. Dimethylsulfoxide induces upregulation of tumor suppressor protein PTEN through nuclear factor-B activation in HL-60 cells. Leuk Res 29: 401–405, 2005.
1 _( x+ V! F8 [) e" u& v7 X: k& |% p9 N3 c% u* I3 r& m9 d) y
Lewis EJ, Hunsicker LG, Clarke WR, Berl T, Pohl MA, Lewis JB, Ritz E, Atkins RC, Rohde R, and Raz I. Renoprotective effect of the angiotensin-receptor antagonist irbesartan in patients with nephropathy due to type 2 diabetes. N Engl J Med 345: 851–860, 2001.
9 |7 A/ s$ s) }8 p4 g% J" Z2 S
- U7 ?& e" J1 E+ e" xMa LJ, Marcantoni C, Linton MF, Fazio S, and Fogo AB. Peroxisome proliferator-activated receptor- agonist troglitazone protects against nondiabetic glomerulosclerosis in rats. Kidney Int 59: 1899–1910, 2001.
# R2 c3 y% r0 T$ J! Z- R& D2 D* F9 Z% |, ]
McCarthy KJ, Routh RE, Shaw W, Walsh K, Welbourne TC, and Johnson JH. Troglitazone halts diabetic glomerulosclerosis by blockade of mesangial expansion. Kidney Int 58: 2341–2350, 2000.% Q: {" b. T' V. N9 S# B! o

3 `' F/ S3 R! d0 }Miyata KS, McCaw SE, Marcus SL, Rachubinski RA, and Capone JP. The peroxisome proliferator-activated receptor interacts with the retinoid X receptor in vivo. Gene 148: 327–330, 1994.
7 G% X( s3 T  F9 B, G. X0 V3 R- s. }- F0 C0 j4 P7 ?
Nagy L, Tontonoz P, Alvarez JG, Chen H, and Evans RM. Oxidized LDL regulates macrophage gene expression through ligand activation of PPAR. Cell 93: 229–240, 1998.; z6 b; s9 b- u, I! I2 Z

( u; E' W$ d' b* I. ~0 FNational Institute of Diabetes and Digestive and Kidney Diseases. US Renal Data System. Annual Data Report: Atlas of End-Stage Renal Disease in the United States. Bethesda, MD: National Institute of Diabetes and Digestive and Kidney Diseases, 2003.; j6 r/ |6 ?6 H# Z( ]( ?8 F+ X

. t, l. t' K" m4 q. i9 INicholas SB, Kawano Y, Wakino S, Collins AR, and Hsueh WA. Expression and function of peroxisome proliferator-activated receptor- in mesangial cells. Hypertension 37: 722–727, 2001.
, j+ Q1 f1 o  R3 H
. r. ?+ i' y9 U7 E% B0 a# {Panchapakesan U, Pollock CA, and Chen XM. The effect of high glucose and PPAR- agonists on PPAR- expression and function in HK-2 cells. Am J Physiol Renal Physiol 287: F528–F534, 2004.! v- X+ H: k$ G7 H2 U% @
* M+ A3 h! f4 b3 ]3 k+ r! b
Ping D, Boekhoudt GH, Rogers EM, and Boss JM. Nuclear factor-B p65 mediates the assembly and activation of the TNF-responsive element of the murine monocyte chemoattractant-1 gene. J Immunol 162: 727–734, 1999.! J+ P& h3 b2 I1 ~

* u  a( }- S& i! P, ?Routh RE, Johnson JH, and McCarthy KJ. Troglitazone suppresses the secretion of type I collagen by mesangial cells in vitro. Kidney Int 61: 1365–1376, 2002.: l" B. \7 _8 X4 r4 E! B/ [
; U8 A8 V+ M: X& ?' @
Ruggenenti P, Fassi A, Ilieva AP, Bruno S, Iliev IP, Brusegan V, Rubis N, Gherardi G, Arnoldi F, Ganeva M, Ene-Iordache B, Gaspari F, Perna A, Bossi A, Trevisan R, Dodesini AR, and Remuzzi G. Preventing microalbuminuria in type 2 diabetes. N Engl J Med 351: 1941–1951, 2004.
' M, p3 ?6 |% |2 X- j1 L# y; ~: T4 q" @0 _1 T+ a
Tontonoz P, Nagy L, Alvarez JG, Thomazy VA, and Evans RM. PPAR promotes monocyte/macrophage differentiation and uptake of oxidized LDL. Cell 93: 241–252, 1998./ M  E- c# k6 O$ s( b; d( A

& K! j8 `: [( p. W& eUK Prospective Diabetes Study Group. Tight blood pressure control, and risk of macrovascular and microvascular complications in type 2 diabetes: UKPDS 38. UK Prospective Diabetes Study Group. BMJ 317: 703–713, 1998.
4 z6 X' k# k; F6 j: M$ n  |/ w. K4 [- @# c$ h* ~
Wada T, Furuichi K, Sakai N, Iwata Y, Yoshimoto K, Shimizu M, Takeda SI, Takasawa K, Yoshimura M, Kida H, Kobayashi KI, Mukaida N, Naito T, Matsushima K, and Yokoyama H. Up-regulation of monocyte chemoattractant protein-1 in tubulointerstitial lesions of human diabetic nephropathy. Kidney Int 58: 1492–1499, 2000.
; j5 r/ V& X* ~' E. [0 Z5 T
# R1 {$ J2 ^2 @* h0 V" T/ E3 e0 m7 g9 eWeigert C, Brodbeck K, Bierhaus A, Haring HU, and Schleicher ED. c-Fos-driven transcriptional activation of transforming growth factor -1: inhibition of high glucose-induced promoter activity by thiazolidinediones. Biochem Biophys Res Commun 304: 301–307, 2003.! F# `+ k5 ~' h  \
! B+ y. _9 r2 C1 m/ c, x0 l
Weigert C, Sauer U, Brodbeck K, Pfeiffer A, Haring HU, and Schleicher ED. AP-1 proteins mediate hyperglycemia-induced activation of the human TGF-1 promoter in mesangial cells. J Am Soc Nephrol 11: 2007–2016, 2000.
" Z( W' |, @* b7 [) K5 n) E. Z" B% y
0 k1 Y+ C) R- W% MXu L, Glass CK, and Rosenfeld MG. Coactivator and corepressor complexes in nuclear receptor function. Curr Opin Genet Dev 9: 140–147, 1999.
. s/ A5 w2 S* f* V- ?. Z1 A  c" j" S+ w" E2 |8 l
Zafiriou S, Stanners SR, Polhill TS, Poronnik P, and Pollock CA. Pioglitazone increases renal tubular cell albumin uptake but limits proinflammatory and fibrotic responses. Kidney Int 65: 1647–1653, 2004.( @; }* B$ Q6 g5 Q

  O1 N6 J7 l* Y% WZafiriou S, Stanners SR, Saad S, Polhill TS, Poronnik P, and Pollock CA. Pioglitazone inhibits cell growth and reduces matrix production in human kidney fibroblasts. J Am Soc Nephrol 16: 638–645, 2005.. C0 S& k, b" f" `5 N& H
" z9 d& `- x4 K% a$ J( F$ z3 Z
Zhang Y, Park CW, Zheng F, Fan X, Striker GE, Breyer MD, and Guan Y. Endogenous PPAR activity ameliorates diabetic nephropathy (Abstract). J Am Soc Nephrol 14: 392A, 2003.(U. Panchapakesan, S. Sumu)

Rank: 2

积分
116 
威望
116  
包包
1832  
沙发
发表于 2015-7-15 21:21 |只看该作者
今天无聊来逛逛  

Rank: 2

积分
107 
威望
107  
包包
1889  
藤椅
发表于 2015-7-21 19:15 |只看该作者
终于看完了~~~  

Rank: 2

积分
72 
威望
72  
包包
1730  
板凳
发表于 2015-7-30 20:51 |只看该作者
干细胞之家微信公众号
回贴赚学识,不错了  

Rank: 2

积分
75 
威望
75  
包包
2193  
报纸
发表于 2015-8-10 18:01 |只看该作者
好人一个  

Rank: 2

积分
97 
威望
97  
包包
1738  
地板
发表于 2015-8-14 12:26 |只看该作者
只有一条路不能选择——那就是放弃的路;只有一条路不能拒绝——那就是成长的路。  

Rank: 2

积分
70 
威望
70  
包包
1809  
7
发表于 2015-9-13 15:40 |只看该作者
我在顶贴~!~  

Rank: 2

积分
68 
威望
68  
包包
1752  
8
发表于 2015-9-16 08:43 |只看该作者
世界上那些最容易的事情中,拖延时间最不费力。  

Rank: 2

积分
61 
威望
61  
包包
1757  
9
发表于 2015-9-16 17:54 |只看该作者
继续查找干细胞研究资料

Rank: 2

积分
162 
威望
162  
包包
1746  
10
发表于 2015-9-25 08:35 |只看该作者
这个贴不错!!!!!看了之后就要回复贴子,呵呵  
‹ 上一主题|下一主题
你需要登录后才可以回帖 登录 | 注册
验证问答 换一个

Archiver|干细胞之家 ( 吉ICP备2021004615号-3 )

GMT+8, 2024-4-20 03:06

Powered by Discuz! X1.5

© 2001-2010 Comsenz Inc.