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: 417533|回复: 253
go

Pravastatin treatment attenuates interstitial inflammation and fibrosis in a rat [复制链接]

Rank: 1

积分
威望
0  
包包
0  
楼主
发表于 2009-4-22 08:17 |只看该作者 |倒序浏览 |打印
作者:Can Li,, Chul Woo Yang, Joo Hyun Park, Sun Woo Lim, Bo Kyung Sun, Ju Young Jung, Soon Bae Kim, Yong Soo Kim, Jin Kim, and Byung Kee Bang作者单位:Departments of 1 Internal Medicine and 3 Anatomy, Cell Death Disease Research Center, The Catholic University of Korea, Seoul 137-040; 2 Nephrology and Dialysis Unit, Department of Internal Medicine, Affiliated Hospital, YanBian University Medical College, YanJi 13300 People‘s Republic of Chin 5 f0 R0 L6 l- c7 C5 @
                  
: N$ Q; w4 Y# {& X' b                  $ L( f6 H2 U# }3 o; f% ]
         
# d7 d$ [& s4 b( g$ v* |                        
' V% G- g! v1 V* }' n$ `            
/ {+ i# c1 `. _            
/ }9 ~! O5 w5 t& r5 s            
8 `; B5 s: p3 b: j9 ^            5 @5 e0 r$ T  S" f) y! C
                      1 ~; I# c$ d" l( I- _
        / C+ {. q+ P6 r
        9 x3 g' Y3 k$ ]! x! U( c5 J: s
        ' Y4 H" v- L+ x
          【摘要】
3 ^; ~3 b& F( x' b      We investigated the effects of pravastatin, a competitive inhibitor of 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase, on interstitial inflammation and fibrosis, using an animal model of chronic cyclosporine A (CsA)-induced nephropathy. Sprague-Dawley rats were maintained on a low-salt diet (0.05% sodium) and treated daily for 1 or 4 wk with vehicle (olive oil; 1 ml/kg sc), CsA (15 mg/kg sc), or both CsA and pravastatin (5 or 20 mg/kg in the drinking water). Anti-inflammatory and antifibrotic effects of pravastatin were studied by evaluating the concentrations of the inflammatory mediators osteopontin (OPN) and C-reactive protein (CRP), of fibrotic cytokine-transforming growth factor (TGF)- 1, and the presence of ED-1-positive cells (macrophages). In addition, renal function, serum lipid levels, histopathology (arteriolopathy and tubulointerstitial fibrosis), and the expression of the vasoactive factors endothelial nitric oxide synthase (eNOS) and renin protein were also compared for different treatment groups. Pravastatin induced dose-dependent decreases in the expression of OPN, intrarenal CRP, and TGF- 1, and in the numbers of ED-1-positive cells at 1 and 4 wk. These were accompanied by a significant attenuation of tubulointerstitial fibrosis at 4 wk. The downregulation of eNOS protein expression in CsA-treated rat kidney was markedly upregulated by pravastatin treatment, although intrarenal renin expression was unaffected. Renal dysfunction induced by CsA significantly improved with administration of pravastatin at a dose of 20 mg/kg. Neither CsA nor pravastatin influenced serum lipid or high-sensitivity CRP levels in the treatment groups. Thus in chronic CsA nephropathy, pravastatin effectively abrogates the progression of tubulointerstitial inflammation and fibrosis. This may support the clinical use of pravastatin. " U( L7 `2 e: {
          【关键词】 transforming growth factor osteopontin nitric oxide Creactive protein" s5 g# S- M% k& f- i
                  CHRONIC CYCLOSPORINE A (CsA)-induced nephropathy is characterized by progressive renal insufficiency, afferent arteriolopathy, tubulointerstitial inflammation, and striped fibrosis ( 36 ). The mechanisms leading to chronic CsA-induced nephropathy are not fully understood. However, utilizing a well-established animal model, we have previously demonstrated, as have others, that a complex causative network may involve inflammatory mediators ( 30, 42 ), transforming growth factor (TGF)- 1 ( 49, 54 ), the renin-angiotensin system (RAS) ( 3, 55 ), and nitric oxide (NO)/NO synthase (NOS) ( 1, 56 ).
) T$ r8 h/ Y6 V: I  t
+ {; F' B; J! r! zInterstitial inflammation is the predominant feature of chronic CsA nephropathy that precedes fibrosis ( 59 ). Osteopontin (OPN) is a potent chemoattractant and adhesion molecule for monocytes and macrophages, and there is overwhelming evidence that the upregulation of tubular OPN expression is closely associated with macrophage influx and renal scarring in a wide range of kidney diseases including chronic CsA-induced nephropathy ( 12, 22, 30, 31, 60 ). This concept is supported by studies using OPN knockout mice or anti-OPN antibody-treated animals ( 34, 40 ). TGF- 1, a well-known profibrogenic cytokine, has been implicated in the fibrosis of chronic CsA nephropathy, probably via its effects on extracellular matrix synthesis and degradation ( 48 ). Administration of neutralizing anti-TGF- antibody thus improves CsA-induced renal dysfunction and structural damage ( 32 ).
6 g9 ^9 r' p( f! B& t
! F% V* B$ n5 p% L' W! rStatins are competitive inhibitors of 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase, the key enzyme that regulates the synthesis of cholesterol from mevalonic acid by suppressing the conversion of HMG-CoA ( 13 ). Because of this activity, the clinical use of statins has produced a significant reduction in cardiovascular-related morbidity and mortality in patients with established cardiovascular disease and hypercholesterolemia ( 4, 47 ). However, mevalonate is the precursor not only of cholesterol but also of many nonsteroidal compounds; inhibition of HMG-CoA reductase by statins may therefore result in pleiotropic effects ( 8, 46 ). Statins may thus exert anti-inflammatory and antiarteriosclerotic actions beyond lipid reduction ( 37 ). The renoprotective effects of statins have also been reported in ischemia-reperfusion injury ( 21 ), subtotal renal ablation ( 29 ), streptozotocin-induced diabetic nephropathy ( 25 ), puromycin-induced nephrosis ( 15 ), and unilateral ureteral obstruction ( 35 ). Nevertheless, the benefits of statins in a rat model of chronic CsA-induced nephropathy have yet to be reported.
8 _3 `, C; P' {
8 x- a+ z6 W" h2 y& H# f8 YWe hypothesized that statins may have anti-inflammatory and antifibrotic effects on chronic CsA-induced nephropathy. To test this hypothesis, we investigated the effects of pravastatin on inflammatory mediators and profibrotic cytokine in a well-described rat model of chronic CsA-induced nephropathy.; s6 h3 H! R0 w9 s9 p
8 j" _  r4 R: L& d. C; K. L8 M
MATERIALS AND METHODS
4 {7 o/ `/ V8 {, j- G3 r
" O. C$ |/ I! W7 |Animals and drugs. Male Sprague-Dawley rats (Charles River Technology), initially weighing 200 to 220 g, were housed in cages in a temperature- and light-controlled environment and allowed free access to a low-salt diet (0.05% sodium, Teklad Premier, Madison, WI) and tap water. Cyclosporine (Novartis Pharma, Basle, Switzerland) was diluted in olive oil to a final concentration of 15 mg/ml. Pravastatin sodium (Bristol-Myers Squibb Pharmaceutical) was dissolved in drinking water and administered at two doses of 5 or 20 mg/kg. This dosage and method of pravastatin administration were chosen according to previous reports showing that pravastatin at a dose of 20 mg/kg did not alter blood lipid levels ( 7, 9 ).
0 ~# f' A0 B. w. i. \- H
" d) |& [: ]- S) ]6 J, ?+ |1 @, ^1 \Experimental design. The experimental protocol was approved by the Animal Care Committee of the Catholic University of Korea. Rats were randomized into six groups and treated daily for 1 or 4 wk as follows:
9 W8 V7 J8 u- O' n2 J5 Z2 b% l8 a  J8 c0 v
Vehicle (VH; n = 6): subcutaneous injection with olive oil (1 ml/kg).- M: Y* K) C$ {5 k/ f/ |- O8 D

  b# @2 A' J1 R9 e. J% xVehicle plus pravastatin (VH P5; n = 6): simultaneous treatment with olive oil and pravastatin (5 mg/kg in drinking water).
" x4 B* C* t1 [0 b
  Q$ L  M5 \! o) S- z  T8 bVehicle plus pravastatin (VH P20; n = 6): simultaneous treatment with olive oil and pravastatin (20 mg/kg in drinking water).
/ U. l- V7 J8 M  O# h; [6 c) h
0 z3 o7 }9 u+ S8 c7 P3 |: WCsA ( n = 6): subcutaneous injection with CsA (15 mg/kg).& d9 `+ }! u0 j; T$ I& M1 Y

, u" l: o5 i9 ^' a: O' c3 S; X. U" ?CsA plus pravastatin (CsA P5; n = 8): simultaneous treatment with CsA and pravastatin (5 mg/kg).: I/ f1 _* H: h
; C2 o( ]( z0 y$ Q; h" `
CsA plus pravastatin (CsA P20; n = 8): simultaneous treatment with CsA and pravastatin (20 mg/kg).9 _! X+ }* ~: `2 x# w  b  t
% y" o  r' X' q
Animals were euthanized under ketamine anesthesia at 1 or 4 wk, and the kidneys were rapidly removed for morphological and molecular studies.
! b3 u$ p0 [9 h( ?1 A" g2 q- M* N# _1 d7 H# V
Renal function. Before death, animals were individually housed in metabolic cages (Tecniplast Gazzada, Beguggiate, Italy) for 24-h urine collection, and blood samples were obtained to evaluate serum creatinine (S cr ). The creatinine clearance rate (C cr ) was calculated with the standard formula.
3 r3 M% o* R$ d2 M# P1 I: f% V/ H3 @: H- m
Miscellaneous measurements. After the treatment was started, rats were pair-fed and daily body weight was monitored. Systolic blood pressure was recorded in conscious rats by the tail-cuff method with plethysmography using a tail manometer-tachometer system (BP-2000, Visitech system, Apex); at least three readings for each rat were averaged. Whole blood CsA levels were measured by a monoclonal radioimmunoassay (Incstar, Stillwater, MN). Serum total cholesterol and triglycerides levels were determined with an auto-analyzer (Coulter-STKS, Coulter Electronics). High-sensitivity C-reactive protein (hs-CRP) was measured by a particle-enhanced immunoturbidometric method using a Cobas Integra 700 (Roche Diagnostic System, Basel, Switzerland) ( 5 ).
2 G' E% W) X- c
5 `% h/ e- V* X) OHistopathology. Kidney tissues were fixed in periodate-lysineparaformaldehyde (PLP) solution and embedded in wax. After dewaxing, 4-µm sections were processed and stained with periodic acid-Schiff or Masson's trichrome and hematoxylin. Arteriolopathy of the afferent arterioles was manifested by expansion of the cell cytoplasm of terminal arteriolar smooth muscle cells by eosinophilic, granular material and semiquantitatively evaluated by counting the percentage of juxtaglomerular afferent arterioles with arteriolopathy per total number of juxtaglomerular afferent arterioles available for examination, using a x 20 objective; at least 60 glomeruli were assessed per specimen. A finding of tubulointerstitial fibrosis (TIF) was defined as a matrix-rich expansion of the interstitium with tubular dilatation, tubular atrophy, tubular cast formation, sloughing of tubular epithelial cells, or thickening of the tubular basement membrane. A minimum of 20 fields/section was assessed using a color image analyzer (TDI Scope Eye Version 3.0 for Windows, Olympus). Briefly, the image was captured, and the extent of TIF was quantified using the Polygon program by counting the percentage of areas injured per field of cortex under x 100 magnification. Histopathological analyses were performed in randomly selected cortical fields of sections by a pathologist blinded to the identity of the treatment groups./ v' m0 l! r2 z- L3 i/ }" _
4 w' L5 f) n2 r- g& X# }0 ]+ ~
Immunohistochemistry. After dewaxing, sections were incubated with 0.5% Triton X-100/PBS solution for 30 min and washed three times with PBS. Nonspecific binding sites were blocked with normal horse serum diluted 1:10 in 0.3% BSA for 30-60 min and then incubated for 2 h at 4°C in mouse antiserum against OPN (MPIIIB10; obtained from the Developmental Studies Hybridoma Bank, Univ. of Iowa, Iowa City, IA) diluted in 1:1,000 in a humid environment. After being rinsed in Tris-buffered saline (TBS), sections were incubated in peroxidase-conjugated rabbit anti-mouse IgG (Amersham Pharmacia Biotech, Piscataway, NJ) for 30 min. For coloration, sections were incubated with a mixture of 0.05% 3,3'-diaminobenzidine containing 0.01% H 2 O 2 at room temperature until a brown color was visible, washed with TBS, counterstained with hematoxylin, and examined under light microscopy. The procedure of immunostaining for ED-1 (Serotec), CRP (Sigma), and renin was similar to that for OPN. The antibody against renin was kindly provided by Dr. Kirsten Madsen (Dept. of Medicine, Univ. of Florida, Gainesville, FL). This antibody is known to recognize rat renin and prorenin ( 39 ). In addition, immunohistochemistry for TGF- 1 (Santa Cruz Biotechnology, Santa Cruz, CA) was performed in cryostat sections. The number of ED-1- or CRP-positive cells was counted in at least 20 fields of cortex/section under x 200 magnification, and renin-positive glomeruli were evaluated by counting a minimum of 50 glomeruli/specimen.+ x2 I2 `! Z4 j
$ F! G) L- t/ S9 m0 @, F- D3 ^
Immunoblotting. For immunoblotting analysis, kidney cortex tissue was homogenized in lysis buffer [20 mM/l Tris·Cl (pH 7.6), 150 mM/l NaCl, 1% wt/vol sodium deoxycholate, 1% vol/vol Triton X-100, 0.1% SDS, 2 mM/l NaVO 3, and freshly added 1% vol/vol aprotinin, leupeptin (1 µg/ml), pepstatin (1 µg/ml), and 1 mM PMSF]. Homogenates were centrifuged at 3,000 rpm for 15 min at 4°C, and the protein concentration of the lysate was determined using a protein microassay of the Bradford method (Bio-RAD, Hercules, CA). Protein samples were resolved on 15% SDS-PAGE gels and then electroblotted onto Bio-Blot nitrocellulose membrane (Bio-Rad). An equal amount of protein loading (50 µg) was verified by Ponceau S staining. The membrane was blocked for 1 h in TBS-added Tween 20 [TBS-T; 10 mM Tris·Cl, 150 mM NaCl (pH 8.0), 0.05% Tween 20] containing 5% nonfat powdered milk. Endothelial NOS (eNOS) was detected by incubation for 1 h with a monoclonal anti-eNOS antibody (Transduction Laboratories) diluted 1:1,000. Primary antibody incubation was followed by six washes with TBS-T. The blot was then incubated with secondary antibody (goat anti-mouse IgG-horseradish peroxidase) conjugate at 1:1,000 (Bio-Rad) for 30 min. Antibody-reactive protein was detected using enhanced chemiluminescence (Amersham Life Science). Optical densities were obtained using the VH group as 100% reference and normalized with -actin.* F1 @+ Y9 v' C4 m0 V' K+ I
$ j& Q7 q% G. ~* d' L8 n4 U' S
Northern blot analysis. A 1-kb cRNA probe was generated from the 2B7 cDNA clone of rat smooth muscle OPN ( 11 ). Sense and antisense cRNA probe were labeled with digoxigenin (DIG)-UTP using a T7 RNA polymerase kit (Boehringer Mannheim, Mannheim, Germany). Probes were precipitated, and incorporation of DIG was determined by dot blotting. Northern blotting was performed as previously described by others ( 60 ). Briefly, kidney cortex was homogenized. Total RNA was extracted using the RNAzol reagent (TEL-TEST), and 20-µg samples were denatured with glyoxal and dimethylsulphoxide, size fractionated on 1.2% agarose gels, and capillary blotted onto positively charged nylon membranes (Boehringer Mannheim). Membranes were hybridized overnight at 68 or 42°C with DIG-labeled cRNA (or 32 P-labeled cDNA probes for TGF- 1 ) in a DIG wash and Block Buffer Set solution (Boehringer Mannheim). After hybridization, membranes were finally washed in 0.1 x SSC/0.1% SDS at 68°C or 0.2 x SSC/0.1% SDS at 42°C. Bound probes were detected using sheet anti-DIG antibody (Fab) conjugated with alkaline phosphatase (Boehringer Mannheim) and developed with CSPD-star enhanced chemiluminescence (Boehringer Mannheim). The densitometry analysis was performed using the NIH Image program. Three determinations for each band were averaged and referenced to 18S.7 c! {+ x: r9 a0 `

/ A, q6 P; S* R* H/ R! F) wStatistical analysis. Data are expressed as means ± SE. Multiple comparisons among groups were performed by one-way ANOVA with the post hoc Bonferroni test (SPSS software version 9.0, Microsoft). Statistical significance was assumed to be P
8 k( \( G0 P7 ^" p# b- ~: m: Y, B, E' [( X% a0 Y0 p, s
RESULTS  Q7 l' y9 _/ j+ R
7 }$ N) J& x( I9 V) ~/ G  P
Effect of pravastatin on basic parameters in chronic CsA nephropathy. Table 1 summarizes basic parameters for each group. The mean body weight of CsA-treated rats was significantly lower than that for the VH-treated rats (255 ± 5 vs. 295 ± 4 g, P
% G, [4 i6 Q- G1 T2 q
5 b" ]) k% Q( `, v; k4 y+ w6 wTable 1. Basic parameters in different groups5 f; d& h- w% f

6 u6 N: `4 X5 j, V9 [Effect of pravastatin on renal function in chronic CsA nephropathy. CsA treatment for 1 wk did not cause renal dysfunction (S cr : 0.63 ± 0.12 vs. 0.58 ± 0.11 mg/dl; C cr : 0.48 ± 0.06 vs. 0.52 ± 0.02 ml·min -1 ·100 g -1, P 0.05 vs. VH, respectively). Table 1 shows 4-wk data on renal function in the experimental groups. Daily treatment of rats with CsA for 4 wk caused higher levels of S cr (1.21 ± 0.07 vs. 0.55 ± 0.11 mg/dl, P ' C$ L; E1 q8 g! ~( L3 J
0 i& f1 R6 b* E6 y: J0 a
Effect of pravastatin on histopathology in chronic CsA nephropathy. CsA-treated rats showed the typical histopathological features of chronic CsA nephrotoxicity (afferent arteriolopathy, striped interstitial fibrosis, and tubular atrophy with focal inflammatory cell accumulation). Afferent arteriolopathy was manifested as the replacement of smooth muscle cells of the afferent glomerular arterioles by periodic acid-Schiff-positive material, resulting in a typical circumferential appearance of the lesion ( Fig. 1 ). In our semiquantitative scoring system, the arteriolopathy was significantly higher in the CsA group than the VH group (28 ± 3 vs. 11 ± 2, P
) N" A6 U. {0 W* a. j  m7 U: r" E: E$ [, w+ e& E
Fig. 1. Representative photomicrographs of arteriolopathy and semiquantitative analysis. A : normal glomerulus. Cyclosporine A (CsA) treatment for 4 wk induces a typical afferent arteriolopathy ( B and C ). The micrograph shows a glomerulus with expansion of the cell cytoplasm of terminal arteriolar smooth muscle cells by eosinophilic, granular material (arrow). Using semiquantitative analysis, the increased arteriolopathy seen in CsA-treated rat kidneys was significantly reduced with pravastatin administration. Periodic acid-Schiff staining; original magnification: x 400 ( A and B ); x 1,000 ( C ). P5 and P20, vehicle (VH) plus 5 and 20 mg/kg in drinking water, respectively. # P
. [0 N' ]! b$ ~4 \% }' j. N2 N" l, T2 S+ N  k
Table 2. Histological analyses2 i" b' d' q' }* t# L1 o
7 H. J# P8 E7 x* ]" T0 W6 ~
Fig. 2. Representative photomicrographs showing the degree of renal tubulointerstitial fibrosis. CsA treatment for 4 wk induced a typical interstitial fibrosis, inflammatory cell infiltration, and tubular atrophy. Note that pravastatin significantly decreased tubulointerstitial fibrosis in accordance with its dosage. Trichrome stain; original magnification: x 100.9 `; I: j# ?/ @+ @, u% T5 A

' `8 r% r( P$ K3 jEffect of pravastatin on serum hs-CRP and intrarenal CRP expression in chronic CsA-induced nephropathy. Table 1 outlines the hs-CRP concentrations in each group. The mean hs-CRP concentration in the VH group was 0.25 ± 0.04 mg/l. Similarly, the mean hs-CRP in the CsA group was 0.23 ± 0.03 mg/l, and there was no significance in serum hs-CRP between the VH and the CsA groups ( P = 0.7 vs. VH). Pravastatin at either dose did not alter the hs-CRP concentrations (CsA P5: 0.23 ± 0.04 mg/l; CsA P20: 0.23 ± 0.03 mg/l, P 0.7 vs. CsA, respectively).
3 H: G" c) D( n: E* _
2 f+ {0 U$ l% AImmunohistochemistry showed a significant increase in CRP expression and immunoreactivity in the CsA-treated rat kidneys, which were confined to cortical tubular epithelial cells ( Fig. 3 ). In our quantitative system ( Table 2 ), the number of CRP-positive cells was dramatically increased in the CsA group compared with the VH group (59 ± 9 vs. 25 ± 3, P 9 ~( y( d; f& s" T8 ]0 `

- T5 k6 `7 ^: k1 {7 WFig. 3. Representative photomicrographs of immunohistochemistry for C-reactive protein (CRP). A : VH-treated rat kidneys. CsA treatment led to a significant increase in CRP immunoreactivity (arrows, B and C ). Pravastatin decreased CRP immunoreactivity in accordance with dosage (CsA P20; D ). Immunohistochemical staining; original magnification: x 400 ( A, B, and D ); x 1,000 ( C ).
2 O! v0 a/ o% y1 B  z2 Y  Z! ^, I' e" p' ~" f& ]1 W% F. d
Fig. 4. Representative photomicrographs of immunohistochemistry for ED-1. ED-1-positive cells (macrophages) were rarely observed in VH-treated rat kidney ( A ), but their numbers increased with CsA treatment for 1 wk ( B ), and a further increase was observed at 4 wk ( C ). Pravastatin treatment [CsA P5 ( D ); CsA P20 ( E )] significantly decreased ED-1-positive cells in accordance with dosage at 4 wk. Immunohistochemical staining; original magnification: x 400.
9 o; D& p2 U# y! A: n- I3 d) b
8 ]: b/ ~/ ~) G8 k" b: k  vEffect of pravastatin on OPN mRNA and protein expression in chronic CsA-induced nephropathy. Northern blot analysis of the kidney cortex revealed a dramatic increase in OPN mRNA in the CsA group at 1 (956 ± 117 vs. 109 ± 17, P 2 j, T7 ?4 D- m* K( h
! `2 r9 @) n! h
Fig. 5. Northern blotting and quantitative analysis of osteopontin (OPN) mRNA expression. OPN mRNA expression was significantly increased in rat kidneys with CsA treatment at 1 or 4 wk, whereas pravastatin markedly decreased its expression in a dose-dependent manner. Relative OPN mRNAs values are represented with the VH group designated as 100% reference and are normalized to 18S. # P
3 `6 S) q; D4 O! J
2 k" P8 G" m1 N. l2 NFig. 6. Representative photomicrographs of immunohistochemistry for OPN. A : normal rat kidney. B : CsA treatment for 4 wk. C : CsA P5 for 4 wk. D : CsA P20 for 4 wk. Note that pravastatin significantly decreased OPN protein expression and its immunoreactivity. Immunohistochemical staining; original magnification: x 400.9 p3 s. @3 B+ H, J7 z3 X

" Y9 B; g9 c" w9 b+ FEffect of pravastatin on TGF- 1 mRNA and protein expression in chronic CsA-induced nephropathy. CsA treatment induced a gradual increase in TGF- 1 mRNA expression at 1 (447 ± 66 vs. 105 ± 10, P + g: x5 ~' C6 y  O( Z$ r4 s
% }# }2 g, U' d4 A8 v# K: M9 v' n
Fig. 7. Northern blotting and quantitative analysis of transforming growth factor (TGF)- 1 mRNA expression. TGF- 1 mRNA expression was significantly increased in rat kidneys with CsA treatment at 1 or 4 wk, whereas pravastatin markedly decreased its expression in a dosedependent manner. Relative TGF- 1 mRNA values are represented with the VH group designated as 100% reference and are normalized to 18S. # P
# M0 O5 {1 T9 P9 ^, X! E  E3 G0 ]5 K4 W- @/ \
Fig. 8. Representative photomicrographs of immunohistochemistry for TGF- 1. Immunostaining was performed on cryostat sections. A : normal rat kidney. B : CsA-treated rat kidney. C : CsA P20. Immunohistochemical staining; original magnification: x 200., {$ }; S; n7 t& ?* k2 W1 y* P& u
$ @% U! h- @$ u2 R3 b2 ?
Effect of pravastatin on eNOS protein expression and intrarenal renin expression in chronic CsA-induced nephropathy. Figure 9 shows immunoblotting of eNOS protein. Four weeks of CsA treatment induced a dramatic decrease in eNOS protein expression (19 ± 5 vs. 104 ± 4, P
9 m# b) w, _# k& F$ t! i# V. d/ ~7 U: s0 J' x) S
Fig. 9. Immunoblotting of endothelial nitric oxide synthase (eNOS) protein in each group. Note that CsA treatment dramatically decreased eNOS protein expression but that expression was increased in accordance with the dosage of pravastatin. The expression of eNOS protein was referenced with -actin, and the relative optical densities (%) are presented with the VH group designated as 100%. # P " Q! G! Z' [0 L" l- n4 w) ~

9 J) @2 S3 I( ]We used a well-documented immunohistochemical technique for renin detection and for semiquantitative analysis of renin-positive glomeruli to represent the status of the RAS ( 30 ). Intrarenal expression of renin was minimal in the VH group, but its immunoreactivity and the number of reninpositive glomeruli significantly increased in the CsA group compared with the VH group ( Fig. 10, 32 ± 3 vs. 9 ± 1, P ! N6 s" B" |3 R

7 I% w# U; y* kFig. 10. Representative photomicrographs of immunohistochemistry for intrarenal renin and semiquantitative analysis of renin-positive glomeruli (arrows). CsA treatment for 4 wk significantly increased intrarenal immunoreactivity of renin and renin-positive glomeruli, but pravastatin did not influence them. Immunohistochemical staining; original magnification: x 400. # P
- L* j* l8 g( a/ g4 y4 f4 ]9 }! ]* ?9 `
DISCUSSION, i! y; X) S4 g- M0 J7 h6 M
, R, v  p3 C; \$ w0 x
The present study clearly demonstrates that, in a rat model of chronic CsA-induced nephropathy, pravastatin attenuates tubulointerstitial inflammation and TIF probably by downregulating the expression of OPN, intrarenal CRP, and TGF- 1 and by upregulating eNOS protein expression. Interestingly, there were no significant differences in systemic lipid or CRP levels.( R! ^& ^% b  d1 o7 D

) v" q8 l* d$ [# cPravastatin inhibits inflammatory mediators (e.g., monocyte chemoattractant protein-1 and TNF- ) and macrophage infiltration ( 17, 33, 34, 50 ) and suppresses mesangial cell proliferation, TGF- 1 expression, and extracellular matrix (fibronectin and type IV collagen) production in cultured mesangial cells ( 6, 38, 57 ). Thus pravastatin may have both anti-inflammatory and antifibrotic potential in the kidney. In the present study, we observed that pravastatin significantly decreased OPN and TGF- 1 expression and the number of ED-1-positive cells at 1 and 4 wk in accordance with the dosage administered, which was paralleled by a significant attenuation of TIF at 4 wk. However, there were no significant differences in lipid parameters in any treatment groups. Our results suggest that pravastatin suppresses the progression of tubulointerstitial inflammation and TIF through a mechanism involving decreased OPN and TGF- 1 expression in chronic CsA-induced nephropathy.
3 ?% e4 `0 k7 t& G
" ~% \( F( @: D0 ~: }: `' _- BCRP is an inflammatory biomarker and a strong predictor of cardiovascular diseases and renal function abnormalities in humans ( 2, 51 ). In general, CRP is the main acute reactant, and its production is presumably restricted to the liver, but a recent study has suggested that the kidney may be a second site of CRP formation ( 18 ). In rats, CRP is not a typical acute phase protein ( 43 ), and a role for CRP is not well characterized. However, a recent publication demonstrated that rat CRP, similarly to human CRP, can activate autologous complement ( 41 ). This finding suggests that biological functions of CRP are conserved among species. In this study, we measured systemic hs-CRP level and intrarenal CRP expression to evaluate antiinflammatory effects of pravastatin on chronic CsA nephropathy. The results of this study revealed that pravastatin treatment decreased CsA-induced overexpression of intrarenal CRP expression. On the other hand, neither CsA nor pravastatin influenced serum hs-CRP concentrations in the treatment groups. On the basis of our study and previous reports, we propose that the attenuation of tubulointerstitial inflammation by pravastatin treatment in CsA-treated rat kidneys may be related, in part, to the decrease in intrarenal CRP expression.
9 W. H# k: z: {) {# a1 ^( n8 ]4 S9 W6 d+ k
The mechanism by which pravastatin attenuates interstitial inflammation and fibrosis in this model may be multifactorial, but two possibilities should be considered. First, chronic CsA-induced nephropathy is associated with afferent arteriolopathy ( 10 ), which ultimately leads to hypoxia-induced renal tubulointerstitial inflammation and TIF. Statins modulate vascular remodeling by inhibiting smooth muscle cell proliferation, migration, and extracellular matrix synthesis ( 14, 27, 44 ). Second, endothelial NO dysregulation has been linked with CsA-related vasoconstriction, inflammatory reaction, and fibrosis ( 16, 56 ). Recently, in vivo and in vitro studies have confirmed that statins improve endothelium function through enhancing eNOS expression and its activity ( 20, 28, 37 ). Here, we found that pravastatin significantly augmented eNOS protein expression and decreased afferent arteriolopathy. Therefore, the anti-inflammatory and antifibrotic effects of pravastatin in this study may be associated with its effects on arteriolopathy and endothelial NO.: f% g. O! N6 q) d, x

, L. j( |7 y$ U3 P. ^In addition, the RAS plays an important role in the pathogenesis of chronic CsA nephropathy ( 55 ). This is supported by the observation that blockade of the RAS with either angiotensinconverting enzyme inhibitors or angiotensin type I-receptor antagonists attenuates fibrosis in chronic CsA nephropathy ( 3 ). In the present study, we evaluated the status of the intrarenal RAS using immunohistochemistry, assuming that the effect of pravastatin on chronic CsA nephropathy is mediated by inhibition of the RAS. However, we observed no significant differences in intrarenal renin immunoreactivity between the CsA and CsA P groups. Thus it is likely that the renoprotective effect of pravastatin in this model is unrelated to the RAS, which is consistent with data from another study that indicate that pravastatin cannot influence cardiac ACE activity ( 37 ).. Z) [' e* q" h+ s0 g" ]
% Y0 o5 f! |9 D5 `- R" n
The types and dosage of statins are major limiting factors in the use of these drugs in clinical transplantation, because of adverse effects (myositis and rhabdomyolysis) or drug-drug interactions. Unlike lipophilic HMG-CoA reductase inhibitors, pravastatin is hydrophilic, and its metabolism is independent of that of cytochrome P -450 3A4 in the liver ( 53 ). As a result, pravastatin may have fewer toxic effects than other statins. It has been reported to be efficacious and tolerated well by transplant recipients undergoing CsA therapy ( 58 ). In the present study, we chose to compare two doses of pravastatin (5 or 20 mg/kg). Interestingly, the lower dose only improved renal histopathology, whereas the higher dose improved both histopathology and renal function. This observation suggests that, although a low dose of pravastatin (5 mg/kg) failed to overcome the hemodynamic changes caused by CsA, it was still effective in preventing the progression of chronic CsA-induced nephropathy.6 H  y" N, d' }: h  g' m. h
: f0 D& c# w  K2 Z0 `
The functions of pravastatin also extend to immunomodulation, as demonstrated by the inhibition of the expression of class II major histocompatibility antigens and of natural killer cell activity ( 52 ). Indeed, experimental and clinical studies in cardiac and renal transplant recipients show that the administration of pravastatin successfully decreases acute or chronic rejection episodes and improves graft survival ( 19, 24, 26 ). Furthermore, combined treatment with pravastatin and CsA produced additional immunosuppression in patients with kidney transplants ( 24 ), which was related to the synergistic inhibition of cytotoxic T lymphocyte activity ( 23 ). From this study and other reports, we suggest that adding pravastatin to CsA-based immunosuppressants may achieve enhanced immunosuppression in addition to lessening CsA-associated nephrotoxicity. Future clinical study is required to investigate this issue.
) _9 b2 V* [% E0 E: q3 g
! E, W- F% N1 \1 I* K1 J( hIn summary, this study demonstrates that pravastatin elicits anti-inflammatory and antifibrotic effects in a rat model of chronic CsA-induced nephropathy. Suppression of OPN, TGF- 1, and intrarenal CRP expression along with higher eNOS expression may be one of the mechanisms underlying the renoprotective properties of pravastatin. Our findings provide a potential rationale for the clinical use of pravastatin in reducing chronic CsA-induced nephropathy.
/ q% X2 Y' u, f6 s0 \1 m: I3 I3 N; \9 i8 V. C8 y4 o) ~" K
DISCLOSURES9 ~$ j6 [2 |1 |; d/ y* E

+ q) k% r7 e. i6 n; f  O" X: i# x( zA portion of this study was presented at the anuual meeting of the American Society of Nephrology (October 2002, Philadelphia, PA).
" u; J" I, A9 W: _! V# P1 a1 {  j9 Y
ACKNOWLEDGMENTS
/ I1 J5 J1 j$ u7 G, r2 Y3 V5 ~$ p  U+ I4 B; `. x$ u0 a
GRANTS
" i/ r: D2 u: w3 `2 e8 {( Y  p1 Q5 ^3 l
This work was supported by the Korea Science and Engineering Foundation (R13-2002-005-0000-1) through the Cell Death Disease Research Center at The Catholic University of Korea.0 l( i$ D( y5 b8 D  l9 g
          【参考文献】) Y, E8 X( q5 R6 Y
Andoh TF, Gardner MP, and Bennett WM. Protective effects of dietary L -arginine supplementation on chronic cyclosporine nephrotoxicity. Transplantation 64: 1236-1240, 1997.
+ ~  i- X4 ^/ c
* d, b- ?3 P( b# A# i5 c/ p. ]8 [/ @

7 c% O2 q/ R" G2 k( |Blake GJ and Ridker PM. Inflammatory bio-markers and cardiovascular risk prediction. J Intern Med 252: 283-294, 2002.7 ^3 g% c9 y/ O' Y9 s
1 q* X  z6 a- n: n1 Y- x2 o
( Q# d2 I. O2 M7 P
' S7 Q3 u7 q8 r' J8 j- B3 l
Burdmann EA, Andoh TF, Nast CC, Evan A, Connors BA, Coffman TM, Lindsley J, and Bennett WM. Prevention of experimental cyclosporin-induced interstitial fibrosis by losartan and enalapril. Am J Physiol Renal Fluid Electrolyte Physiol 269: F491-F499, 1995.
( s$ [% H2 z& ~5 L& L0 Y4 ]
/ m% H" I/ y( z1 e) X
' Y7 W5 U2 _8 M; F7 y* D) c3 @7 k9 q$ n' [4 a7 Z! C
Byington RP, Jukema JW, Salonen JT, Pitt B, Bruschke AV, Hoen H, Furberg CD, and Mancini GB. Reduction in cardiovascular events during pravastatin therapy. Pooled analysis of clinical events of the Pravastatin Atherosclerosis Intervention Program. Circulation 92: 2419-2425, 1995.
% c3 N7 ^. h" W0 j3 W- Q
' |% p# M8 }5 d3 {) u5 I! k0 j& f3 f! ^" p5 i4 ?
: d* U  h8 Q/ N. ?5 r+ k
Chang JW, Yang WS, Min WK, Lee SK, Park JS, and Kim SB. Effects of simvastatin on high-sensitivity C-reactive protein and serum albumin in hemodialysis patients. Am J Kidney Dis 39: 1213-1217, 2002.
. c3 F3 x3 G5 M2 e1 h$ S
0 C- M( f' o( E* t# v. F( Y' O& G% `  {
% i# x& k- `# R( }
Chen HC, Guh JY, Shin SJ, and Lai YH. Pravastatin suppress superoxide and fibronectin production of glomerular mesangial cells induced by oxidized-LDL and high glucose. Atherosclerosis 160: 141-146, 2002.' k7 g/ j( g/ t" j, H0 [" [& `

5 L$ a  U( y4 u
7 b5 G) w" B& [6 i  S
8 o" b! k; e1 O8 z- b! F0 Q, x# U5 jCoelho-Filho OR, De Luca IM, Tanus-Santos JE, Cittadino M, Sampaio RC, Coelho OR, Hyslop S, and Moreno H Jr. Pravastatin reduces myocardial lesions induced by acute inhibition of nitric oxide biosynthesis in normocholesterolemic rats. Int J Cardiol 79: 215-221, 2001.' W8 m2 G& \2 B% i

$ b! |+ G. [: p3 \$ c
: L4 e1 }! a3 q2 ?: b; T
" g! n5 K2 U. v4 [8 UFaggiotto A and Paoletti R. Statins and blockers of the renin-angiotensin system: vascular protection beyond their primary mode of action. Hypertension 34: 987-996, 1999.
1 e# l) P* M# s5 w1 y" |0 d7 x4 l# S6 m$ M2 C
! ~  ~. S& x; |
  B0 M# C( y% Y! `/ J
Fontaine D, Fontaine J, Dupont I, Dessy C, Piech A, Carpentier Y, and Berkenboom G. Chronic hydroxymethylglutaryl coenzyme A reductase inhibition and endothelial function of the normocholesterolemic rat: comparison with angiotensin-converting enzyme inhibition. J Cardiovasc Pharmacol 40: 172-180, 2002.
; u& h/ c% I% q" W8 S& h/ v8 b; `- j( Q) B
- T+ K+ ?# N& F  y2 W

+ t" e! b% c9 ]8 O3 V5 wFranceschini N, Alpers CE, Bennett WM, and Andoh TF. Cyclosporine arteriolopathy: effects of drug withdrawal. Am J Kidney Dis 32: 247-253, 1998.
) t! _" \" y3 `& M5 p6 U* B+ L% w2 W5 Q
+ ^' Q6 y: v& g$ e9 [% o! I" ?+ _
9 ^, c1 ?& ?$ x: [- V
Giachelli C, Bae N, Lombardi D, Majesky M, and Schwartz S. Molecular cloning and characterization of 2B7, a rat mRNA which distinguishes smooth muscle cell phenotypes in vitro and is identical to osteopontin (secreted phosphoprotein I, 2aR). Biochem Biophys Res Commun 177: 867-873, 1991.8 g' Q+ p( y% C

- b0 J7 F( H/ Q1 u: X8 q3 R1 Y; F% Q9 o' H& d
4 H  ?+ Z$ f1 r. k
Giachelli CM, Lombardi D, Johnson RJ, Murry CE, and Almeida M. Evidence for a role of osteopontin in macrophage infiltration in response to pathological stimuli in vivo. Am J Pathol 152: 353-358, 1998.
6 @' o" |* v( Z% S1 B. _& n
& q9 r7 q/ j% y3 t1 v: d% y, ~
+ Z7 J! F/ i! J6 i0 b2 B
- U" B8 ?) E* G7 ^Goldstein JL and Brown MS. Regulation of the mevalonate pathway. Nature 343: 425-430, 1990.
# B5 b8 ^; ^/ t9 F& C
4 p+ h0 J: A1 ~" V  [
  K8 W5 v, S; b* ?! o. ~
, L. J9 ~1 O7 I/ b+ F. YGuijarro C, Blanco-Colio LM, Ortego M, Alonso C, Ortiz A, Plaza JJ, Diaz C, Hernandez G, and Egido J. 3-Hydroxy-3-methylglutaryl coenzyme a reductase and isoprenylation inhibitors induce apoptosis of vascular smooth muscle cells in culture. Circ Res 83: 490-500, 1998.
( x$ }: m1 A) ^6 S
7 H2 p4 h! s8 j( Y8 D! d! ~) T( Q1 I& d5 v" {

; m  X; ]5 H- r6 g9 D& u, w$ YHarris KP, Purkerson ML, Yates J, and Klahr S. Lovastatin ameliorates the development of glomerulosclerosis and uremia in experimental nephrotic syndrome. Am J Kidney Dis 15: 16-23, 1990.
. u  V! `+ D: v8 x% m6 V1 M8 y3 F; L; [9 R. z0 x

0 h$ D3 K: Y6 i5 {& r, M; Q3 r
( h# J7 S, a& S1 |$ gHeeringa P, van Goor H, Itoh-Lindstrom Y, Maeda N, Falk RJ, Assmann KJ, Kallenberg CG, and Jennette JC. Lack of endothelial nitric oxide synthase aggravates murine accelerated anti-glomerular basement membrane glomerulonephritis. Am J Pathol 156: 879-888, 2000.
5 i2 d$ V5 k: Y" j4 q& T" G  _2 I0 F( |7 ?- P
  V1 c' G9 h) P, L2 T+ W: G
% _- c; `1 B, o. i( W
Holm T, Andreassen AK, Ueland T, Kjekshus J, Froland SS, Kjekshus E, Simonsen S, Aukrust P, and Gullestad L. Effect of pravastatin on plasma markers of inflammation and peripheral endothelial function in male heart transplant recipients. Am J Cardiol 87: 815-818, 2001.8 N/ I/ L+ X; Y, _! W

6 K. M3 E0 e/ N+ h) Y3 v# E" K: d5 h( q7 Q- z9 z7 h' v  d
. |! K1 M' M- y
Jabs WJ, Logering BA, Gerke P, Kreft B, Wolber EM, Klinger MH, Fricke L, and Steinhoff J. The kidney as a second site of human C-reactive protein formation in vivo. Eur J Immunol 33: 152-161, 2003.
- h+ A. l2 X1 u0 f. I. ~2 S2 ?6 O8 N/ `$ \( }, Y

. m* `, R7 J3 M) z; J. N% N" d* i) S# u. S' G
Ji P, Si MS, Podnos Y, Chow H, Steward E, and Imagawa DK. Prevention of chronic rejection by pravastatin in a rat kidney transplant model. Transplantation 74: 821-827, 2002./ [) ]) K. o% v/ @- @2 |2 m" \
8 p8 t7 b+ b+ n0 T

( ?6 a! x& D: m$ U. [# `) r
* o: V1 g7 k. k. lJorge PA, Osaki MR, and de Almeida E. Rapid reversal of endothelial dysfunction in hypercholesterolaemic rabbits treated with simvastatin and pravastatin. Clin Exp Pharmacol Physiol 24: 948-953, 1997.; r) x7 J$ W" w2 X7 z

$ I8 p% i% K0 i* c% T$ L- n9 f  Z5 X/ B0 n- x: J

$ O9 I2 q7 h4 o* [! w. m/ \Joyce M, Kelly C, Winter D, Chen G, Leahy A, and Bouchier-Hayes D. Pravastatin, a 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor, attenuates renal injury in an experimental model of ischemiareperfusion. J Surg Res 101: 79-84, 2001.% J; Q1 Z4 ]) T  \8 a" Q6 T
! U6 k7 N9 n7 H: s

# j4 i) r' X) {5 J7 k0 i1 U) c, z
; h  }4 i" w! ^" @, `9 Q: G3 O& h- EKaneto H, Morrissey J, McCracken R, Reyes A, and Klahr S. Osteopontin expression in the kidney during unilateral ureteral obstruction. Miner Electrolyte Metab 24: 227-237, 1998.
2 c/ ]0 y* q  K# V: P9 N7 n, `9 [5 R- H5 q5 Y( `

" f* o2 W2 E' g. Y2 |8 U3 m6 L4 E3 m1 u  v1 V! e* G3 H8 g
Katznelson S, Wang XM, Chia D, Ozawa M, Zhong HP, Hirata M, Terasaki PI, and Kobashigawa JA. The inhibitory effects of pravastatin on natural killer cell activity in vivo and on cytotoxic T lymphocyte activity in vitro. J Heart Lung Transplant 17: 335-340, 1998.
2 B$ e2 a& Z- ^: k+ d
6 s0 l$ z& @0 k4 _0 F- @3 F/ [# `. ~6 a

' F( P0 K, I( F2 O2 uKatznelson S, Wilkinson AH, Kobashigawa JA, Wang XM, Chia D, Ozawa M, Zhong HP, Hirata M, Cohen AH, Teraski PI, and Danovitch GM. The effect of pravastatin on acute rejection after kidney transplantation-a pilot study. Transplantation 61: 1469-1474, 1996.! b. D9 L: e9 T. m6 w

% P$ X* Q0 W6 G; r5 S5 B
1 S' ?, T- W8 f8 V. P# z
7 [$ j7 @& s9 q% c" R- FKim SI, Han DC, and Lee HB. Lovastatin inhibits transforming growth factor- 1 expression in diabetic rat glomeruli and cultured rat mesangial cells. J Am Soc Nephrol 11: 80-87, 2000.
* r- ?: Y, k9 z
; }4 e: n, D0 o& v
% @$ p  X2 ~  w: l8 m8 ~* _' Q0 v/ ~! X
Kobashigawa JA, Katznelson S, Laks H, Johnson JA, Yeatman L, Wang XM, Chia D, Terasaki PI, Sabad A, Cogert GA, Trosian K, Hamilton MA, Moriguchi JD, Kawata N, Hage A, Drinkwater DC, and Stevenson LW. Effect of pravastatin on outcomes after cardiac transplantation. N Engl J Med 333: 621-627, 1995.5 v, H" b+ B: ]9 U, Q

5 e# m1 k6 L) U/ `; n% w0 x. l5 v1 I! H6 d

; Z+ M6 v3 g9 RKomukai M, Wajima YS, Tashiro J, Shinomiya M, Saito Y, and Morisaki N. Carvastatin suppresses intimal thickening of rabbit carotid artery after balloon catheter injury probably through the inhibition of vascular smooth muscle cell proliferation and migration. Scand J Clin Lab Invest 59: 159-166, 1999.
& N8 Q6 P6 _$ n( b% E/ ]
& @' K+ K% d  x; [' e! W/ y1 Z9 ^. q, _; _, W6 k/ A2 K5 z$ ]/ E# F$ }
! o7 G) S- a" e0 B
Laufs U, La Fata V, Plutzky J, and Liao JK. Upregulation of endothelial nitric oxide synthase by HMG CoA reductase inhibitors. Circulation 97: 1129-1135, 1998.! t' R. A4 x' I3 H2 `4 ?

; r2 @% t9 @2 z: M) [+ I# I
/ a% x7 X: L) T0 ~5 U
6 `9 g. |" F! w# k; }, n5 U9 ZLee SK, Jin SY, Han DC, Hwang SD, and Lee HB. Effects of delayed treatment with enalapril and/or lovastatin on the progression of glomerulosclerosis in nephrectomized rats. Nephrol Dial Transplant 8: 1338-1343, 1993./ G8 R& W6 Y) p: e% ~" ]* l
+ i) s) D% z# f0 n3 T
: q  L4 Q( M+ ~+ `; p6 z

  h" n& q; R% ?; E3 g8 b( v; R4 |Li C, Yang CW, Kim WY, Jung JY, Cha JH, Kim YS, Kim J, Bennett WM, and Bang BK. Reversibility of chronic cyclosporine nephropathy in rats after withdrawal of cyclosporine. Am J Physiol Renal Physiol 284: F389-F398, 2003.5 T8 l- \( Q* q& z; }+ k

' l( s% s, W+ V3 _0 V" X5 D3 ^0 X1 g7 j; p9 ~9 G$ B
' C) }0 T9 \3 L
Li C, Yang CW, Park CW, Ahn HJ, Kim WY, Yoon KH, Suh SH, Lim SW, Cha JH, Kim YS, Kim J, Chang YS, and Bang BK. Long-term treatment with ramipril attenuates renal osteopontin expression in diabetic rats. Kidney Int 63: 454-463, 2003.- g2 e/ w3 M( s. r4 D- {  J
9 _  ~, |9 P9 R5 o5 p  }
( h- j, e0 M7 g& A, H  A
: ?; }/ W1 e1 Z) ?
Ling H, Li X, Jha S, Wang W, Karetskaya L, Pratt B, and Ledbetter S. Therapeutic role of TGF- -neutralizing antibody in mouse cyclosporin A nephropathy: morphologic improvement associated with functional preservation. J Am Soc Nephrol 14: 377-388, 2003.
; |3 @& P$ j. H. Q! d8 U0 B$ J( q4 u$ v* x$ ~+ @3 u+ W! [
8 T7 \5 x. `% R: R2 T3 q  o

" c+ \. k- \$ \3 h, Q  w' M6 }1 V0 sMartinez-Gonzalez J, Alfon J, Berrozpe M, and Badimon L. HMG-CoA reductase inhibitors reduce vascular monocyte chemotactic protein-1 expression in early lesions from hypercholesterolemic swine independently of their effect on plasma cholesterol levels. Atherosclerosis 159: 27-33, 2001.
3 I5 F" ]! Q( E6 `. S
$ Y5 Q7 l3 `" H3 R! e0 r9 F( n0 B
' w" G9 ~7 r' [( F& w8 j. q( v& M/ S+ G7 L
Mazzali M, Hughes J, Dantas M, Liaw L, Steitz S, Alpers CE, Pichler RH, Lan HY, Giachelli CM, Shankland SJ, Couser WG, and Johnson RJ. Effects of cyclosporine in osteopontin null mice. Kidney Int 62: 78-85, 2002.( f7 H$ R* K0 y/ ~- e

( K& l2 T3 ?' d% N- t, H* D" L
9 C+ _9 m  ~( u4 e' A, e6 A) z
9 e% c+ P5 }& s1 @" A0 E# hMoriyama T, Kawada N, Nagatoya K, Takeji M, Horio M, Ando A, Imai E, and Hori M. Fluvastatin suppresses oxidative stress and fibrosis in the interstitium of mouse kidneys with unilateral ureteral obstruction. Kidney Int 59: 2095-2103, 2001.
/ l: `; b4 b  l3 c  G
; H1 i% w; @, Y0 L2 r# i3 {9 W1 [5 H4 S
7 I- M: _) c" H2 s2 \5 e9 m
Myers BD, Sibley R, Newton L, Tomlanovich SJ, Boshkos C, Stinson E, Luetscher JA, Whitney DJ, Krasny D, Coplon NS, and Perlroth MG. The long-term course of cyclosporine-associated chronic nephropathy. Kidney Int 33: 590-600, 1988.
' j4 T) Z. ~4 n" x1 o6 h5 Q1 p7 c& u9 g# ^. O

& V* ?$ n* {: n1 n0 @. H% x$ r! j" P0 _- s" n/ ]
Ni W, Egashira K, Kataoka C, Kitamoto S, Koyanagi M, Inoue S, and Takeshita A. Antiinflammatory and antiarteriosclerotic actions of HMG-CoA reductase inhibitors in a rat model of chronic inhibition of nitric oxide synthesis. Circ Res 89: 415-421, 2001.
# I" ?/ |$ }, @: N7 j7 Y/ b
9 z# x% Y9 \; U# S9 ]5 f3 m. i; [6 V; b

7 S" J& Q  `9 z. e- e  |Nishimura M, Tanaka T, Yasuda T, Kurakata S, Kitagawa M, Yamada K, Saito Y, and Hirai A. Effect of pravastatin on type IV collagen secretion and mesangial cell proliferation. Kidney Int Suppl 71: S97-S100, 1999.+ Y* L: A5 V; ]4 C/ m$ }+ X0 [: ?
- X9 m" s- E+ [' X
6 p% N' I# v7 e/ G" I( @0 p
! H$ r( s! d% ]
Oliverio MI, Madsen K, Best CF, Ito M, Maeda N, Smithies O, and Coffman TM. Renal growth and development in mice lacking AT 1A receptors for angiotensin II. Am J Physiol Renal Physiol 274: F43-F50, 1998.
" C/ `% }0 D8 Y7 z' u2 M/ s  L) _3 p& p& [9 b( _# `
9 o9 ~8 Y, l' L

' ^- x1 M/ p$ w! kOphascharoensuk V, Giachelli CM, Gordon K, Hughes J, Pichler R, Brown P, Liaw L, Schmidt R, Shankland SJ, Alpers CE, Couser WG, and Johnson RJ. Obstructive uropathy in the mouse: role of osteopontin in interstitial fibrosis and apoptosis. Kidney Int 56: 571-580, 1999.
+ O2 _. S! N) Q6 a2 x# G8 J
1 K, R: H- a, K0 |5 r
& s0 j; q4 D6 e/ j3 ~0 \' }2 o% T
Padilla ND, Bleeker WK, Lubbers Y, Rigter GM, Van Mierlo GJ, Daha MR, and Hack CE. Rat C-reactive protein activates the autologous complement system. Immunology 109: 564-571, 2003.; F1 a+ @$ U" I1 L# H6 o" C% u
; E- y& w8 k( `/ |7 g; J/ Z
% x% n, k+ @! o  _! c9 P  D$ F: Q

9 `& Z# z1 N% \3 x6 gPichler RH, Franceschini N, Young BA, Hugo C, Andoh TF, Burdmann EA, Shankland SJ, Alpers CE, Bennett WM, and Couser WG. Pathogenesis of cyclosporine nephropathy: roles of angiotensin II and osteopontin. J Am Soc Nephrol 6: 1186-1196, 1995.
% t% ]+ j  z/ ^: v- l; C# h+ L2 d& b" f9 x+ H* b0 r# S4 G
+ T" o) o5 U) v8 `
2 H% a/ q9 U" C3 w
Rassouli M, Sambasivam H, Azadi P, Dell A, Morris HR, Nagpurkar A, Mookerjea S, and Murray RK. Derivation of the amino acid sequence of rat C-reactive protein from cDNA cloning with additional studies on the nature of its dimeric component. J Biol Chem 267: 2947-2954, 1992.
# W* E% h7 q; O
/ E* }" ]( ~& e2 W2 {, \; m
) _+ N% v% R, x- b- P/ b1 ?7 v/ L( a, m6 |7 i" w
Riessen R, Axel DI, Fenchel M, Herzog UU, Rossmann H, and Karsch KR. Effect of HMG-CoA reductase inhibitors on extracellular matrix expression in human vascular smooth muscle cells. Basic Res Cardiol 94: 322-332, 1999.) ~5 @! t. ?! y/ H& B8 I/ F2 j

0 v/ ?$ M, w+ \. S. x" j+ f
! ^. d7 h; B; |) R3 Y% v( P4 l
9 T! \" K' [1 _9 S/ O0 F- Z; o5 g# {Romano M, Diomede L, Sironi M, Massimiliano L, Sottocorno M, Polentarutti N, Guglielmotti A, Albani D, Bruno A, Fruscella P, Salmona M, Vecchi A, Pinza M, and Mantovani A. Inhibition of monocyte chemotactic protein-1 synthesis by statins. Lab Invest 80: 1095-1100, 2000.
/ \' j* u$ l& `2 O
* [  J' R+ ]+ h2 V( ?
( o5 I  V4 ?% Y; A. ]; P# o3 V
& _5 x9 z. |5 W: ?2 m6 E% BRosenson RS and Tangney CC. Antiatherothrombotic properties of statins: implications for cardiovascular event reduction. JAMA 279: 1643-1650, 1998.
8 M" ^# w: y; M' T1 _  o1 f, I1 r8 k1 v% ~$ `2 X6 I5 z
; c9 Y3 T  q1 o( O; N* H

/ S% s$ @1 s4 O! k& }* LShepherd J, Cobbe SM, Ford I, Isles CG, Lorimer AR, MacFarlane PW, McKillop JH, and Packard CJ. Prevention of coronary heart disease with pravastatin in men with hypercholesterolemia. West of Scotland Coronary Prevention Study Group. N Engl J Med 333: 1301-1307, 1995.
$ W) f0 @/ ], `0 @$ P! n9 M4 e
0 t& v5 F* z  R8 U  D# c; L" i! I+ w" Z# Q# p: k# }. [" {
+ y; k9 S. Z! L- l3 v' I- I
Shihab FS, Andoh TF, Tanner AM, and Bennett WM. Sodium depletion enhances fibrosis and the expression of TGF- 1 and matrix proteins in experimental chronic cyclosporine nephropathy. Am J Kidney Dis 30: 71-81, 1997.8 t. r7 Y+ _; {' J9 T/ j
* w6 Q. W. c3 ~2 v

, R9 B) @: Y9 K, u$ y* S1 G, b
4 M, h. r" Z3 p/ NShihab FS, Andoh TF, Tanner AM, Noble NA, Border WA, Franceschini N, and Bennett WM. Role of transforming growth factor- 1 in experimental chronic cyclosporine nephropathy. Kidney Int 49: 1141-1151, 1996.
9 q. q, n1 Y5 Q- ?8 _* Z7 E- H6 T# F# g0 Y
" k, i7 {8 a: \  y: N: x# R/ s' d
6 d5 }6 J2 b3 x
Solheim S, Seljeflot I, Arnesen H, Eritsland J, and Eikvar L. Reduced levels of TNF- in hypercholesterolemic individuals after treatment with pravastatin for 8 weeks. Atherosclerosis 157: 411-415, 2001.
* e% A" ~/ ~5 j& j3 s- S) ?; W2 X/ m2 N/ \7 k/ d

) ~  d5 w8 \3 R4 D6 F" d
+ }$ U2 c- d, K  {2 ~Stuveling EM, Hillege HL, Bakker SJ, Gans RO, De Jong PE, and De Zeeuw D. C-reactive protein is associated with renal function abnormalities in a non-diabetic population. Kidney Int 63: 654-661, 2003.
% Q4 ~: |, u; r. g
' z6 }" E! W0 @8 Z' d) t( y& c2 C  d$ u$ y. R8 W: g" t
3 }, a1 G4 u  u, X% C! G
Weitz-Schmidt G, Welzenbach K, Brinkmann V, Kamata T, Kallen J, Bruns C, Cottens S, Takada Y, and Hommel U. Statins selectively inhibit leukocyte function antigen-1 by binding to a novel regulatory integrin site. Nat Med 7: 687-692, 2001." v" ?1 N" C& ^8 W8 ~; t
! o2 k9 k. h) A; U, m- I# G5 u% o

" q; q: |! p2 o$ e/ d
: U9 ?9 k) E, X8 I: qWilliams D and Feely J. Pharmacokinetic-pharmacodynamic drug interactions with HMG-CoA reductase inhibitors. Clin Pharmacokinet 41: 343-370, 2002.
+ e' p! K# ^; ~
: }' ^+ O8 d9 |) l. f+ _( ?( @8 }. R, M, }5 s

; ~( |: u% `0 V8 e7 WYang CW, Ahn HJ, Kim WY, Li C, Jung JY, Yoon SA, Kim YS, Cha JH, Kim J, and Bang BK. Synergistic effects of mycophenolate mofetil and losartan in a model of chronic cyclosporine nephropathy. Transplantation 75: 309-315, 2003.0 X8 Z& F8 g' A& ]# l; K

. K7 F/ [9 U. w( w1 V
" U( n8 i0 r1 P% A. x' ~( v6 s, w5 D, Y9 S. P% m5 S& q/ O
Yang CW, Ahn HJ, Kim WY, Shin MJ, Kim SK, Park JH, Kim YO, Kim YS, Kim J, and Bang BK. Influence of the renin-angiotensin system on epidermal growth factor expression in normal and cyclosporine-treated rat kidney. Kidney Int 60: 847-857, 2001.7 \6 a: R; k8 j

0 R9 z: ~) V+ B% X; k: J8 B' R
$ f% r. ], W, H5 Q- ^) q
Yang CW, Kim YS, Kim J, Kim YO, Min SY, Choi EJ, and Bang BK. Oral supplementation of L -arginine prevents chronic cyclosporine nephrotoxicity in rats. Exp Nephrol 6: 50-56, 1998.
7 c5 [# P4 V* l, L+ d8 v
! M- r7 ]! x! ]* d7 l) ~5 v, m, Y9 A9 w

) Z2 z+ N& p( ?Yokota T, Utsunomiya K, Murakawa Y, Kurata H, and Tajima N. Mechanism of preventive effect of HMG-CoA reductase inhibitor on diabetic nephropathy. Kidney Int Suppl 71: S178-S181, 1999.
2 o' S! n1 ~6 D: o3 i/ ]: M8 V& \
2 u! s, L/ I  Q" @+ ]' y, K  B5 b  Y( Q& E$ w) S0 ?

, }8 [+ k7 p7 Q) _) eYoshimura N, Oka T, Okamoto M, and Ohmori Y. The effects of pravastatin on hyperlipidemia in renal transplant recipients. Transplantation 53: 94-99, 1992.
. p' S2 |5 W' M9 M' Z7 B
2 d& D. Y1 T6 }: E% Z* @% ~" I. g$ W+ v" `  a
* [3 S! U1 ~* _3 d; w& d
Young BA, Burdmann EA, Johnson RJ, Alpers CE, Giachelli CM, Eng E, Andoh T, Bennett WM, and Couser WG. Cellular proliferation and macrophage influx precede interstitial fibrosis in cyclosporine nephrotoxicity. Kidney Int 48: 439-448, 1995.7 L3 h7 Q7 ~/ t3 ~+ H# }4 [
4 C  |4 C9 u, y$ {5 I+ S5 y
$ K- X  M4 I9 I+ C2 e+ M3 h8 [

! Y- K3 u" ^7 z0 E( dYu XQ, Wu LL, Huang XR, Yang N, Gilbert RE, Cooper ME, Johnson RJ, Lai KN, and Lan HY. Osteopontin expression in progressive renal injury in remnant kidney: role of angiotensin II. Kidney Int 58: 1469-1480, 2000.

Rank: 2

积分
136 
威望
136  
包包
1877  
沙发
发表于 2015-6-17 14:43 |只看该作者
干细胞与动物克隆

Rank: 2

积分
101 
威望
101  
包包
1951  
藤椅
发表于 2015-7-8 13:48 |只看该作者
昨晚多几分钟的准备,今天少几小时的麻烦。  

Rank: 2

积分
64 
威望
64  
包包
1769  
板凳
发表于 2015-7-15 10:10 |只看该作者
干细胞之家微信公众号
偶啥时才能熬出头啊.  

Rank: 2

积分
70 
威望
70  
包包
1809  
报纸
发表于 2015-7-15 20:38 |只看该作者
谢谢哦  

Rank: 2

积分
64 
威望
64  
包包
1734  
地板
发表于 2015-7-17 19:54 |只看该作者
这个贴好像之前没见过  

Rank: 2

积分
166 
威望
166  
包包
1997  
7
发表于 2015-7-18 12:26 |只看该作者
神经干细胞

Rank: 2

积分
61 
威望
61  
包包
1757  
8
发表于 2015-7-23 17:12 |只看该作者
顶一个先  

Rank: 2

积分
79 
威望
79  
包包
1769  
9
发表于 2015-7-28 14:00 |只看该作者
宁愿选择放弃,不要放弃选择。  

Rank: 2

积分
98 
威望
98  
包包
2211  
10
发表于 2015-8-6 12:43 |只看该作者
帮你顶,人还是厚道点好  
‹ 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, 2024-4-30 06:39

Powered by Discuz! X1.5

© 2001-2010 Comsenz Inc.