干细胞之家 - 中国干细胞行业门户第一站

标题: Candesartan prevents long-term impairment of renal function in response to neona [打印本页]

作者: 轻羽    时间: 2009-4-22 09:44     标题: Candesartan prevents long-term impairment of renal function in response to neona

作者:Sukru Oguzkan Topcu,, Michael Pedersen, Rikke Nørregaard,, Guixian Wang,, Mark Knepper, Jens Christian Djurhuus, Søren Nielsen,, Troels Munch Jørgensen, and Jørgen Frøkiær,,作者单位:1 The Water and Salt Research Center, 2 Institute of Clinical Medicine, 5 Institute of Anatomy, University of Aarhus, 7 Department of Clinical Physiology, 3 The MR Research Centre, and 6 Department of Urology, Aarhus University Hospital-Skejby, Aarhus, Denmark; and 4 Laboratory of Kidney and Electro
3 T" S+ B$ N' v3 g4 K/ J# m& @. p                  3 s$ u+ D. I3 u' n! J3 o4 v
                  
( k  s& @2 u% a! M7 X/ Z         
; N; ~, c' T$ s+ |                         3 ^9 ]7 V) H4 P2 V2 q  H
            
2 C9 ~; ~$ D- V* `& M* M            
2 f0 C( s# l7 U  x7 x            
' G4 y5 G8 {' M+ b            ' y9 |- [+ {, B/ Y5 o# v7 E$ x
                     
* f, k! H6 Q' [        / R. ]4 N+ ]5 g, l! w8 c: y4 ^6 ?
        
* n) S4 C* g  U4 x+ u        
  j0 C( z9 b6 T6 T          【摘要】
& l  E* @  r+ b! I5 {      Angiotensin II (ANG II) plays an important role in the development of obstructive nephropathy. Here, we examined the effects of the ANG II receptor type 1 (AT1R) blockade using candesartan on long-term renal molecular and functional changes in response to partial unilateral ureteral obstruction (PUUO). Newborn rats were subjected to severe PUUO or sham operation (Sham) within the first 48 h of life. Candesartan was provided in the drinking water (10 mg·kg -1 ·day -1 ) from day 21 of life until 10 wk of age. Renal blood flow (RBF) was evaluated by MRI, glomerular filtration rate (GFR) was measured using the renal clearance of 51 Cr-EDTA, and the renal expression of Na-K-ATPase and the collecting duct water channel aquaporin-2 (AQP2) was examined by immunoblotting and immunocytochemistry. At 10 wk of age, PUUO significantly reduced RBF (0.8 ± 0.1 vs. 1.6 ± 0.1 ml·min -1 ·100 g body wt -1; P < 0.05) and GFR (37 ± 16 vs. 448 ± 111 µl·min -1 ·100 g body wt -1; P < 0.05) compared with Sham. Candesartan prevented the RBF reduction (PUUO CAN: 1.6 ± 0.2 vs. PUUO: 0.8 ± 0.1 ml·min -1 ·100 g body wt -1; P < 0.05) and attenuated the GFR reduction (PUUO CAN: 265 ± 68 vs. PUUO: 37 ± 16 µl·min -1 ·100 g body wt -1; P < 0.05). PUUO was also associated with a significant downregulation in the expression of Na-K-ATPase (75 ± 12 vs. 100 ± 5%, P < 0.05) and AQP2 (52 ± 15 vs. 100 ± 4%, P < 0.05), which were also prevented by candesartan (Na-K-ATPase: 103 ± 8 vs. 100 ± 5% and AQP2: 74 ± 13 vs. 100 ± 4%). These findings were confirmed by immunocytochemistry. Consistent with this, candesartan treatment partly prevented the reduction in solute free water reabsorption and attenuated fractional sodium excretion in rats with PUUO. In conclusion, candesartan prevents or attenuates the reduction in RBF, GFR and dysregulation of AQP2 and Na-K-ATPase in response to congenital PUUO in rats, suggesting that AT1R blockade may protect the neonatally obstructed kidney against development of obstructive nephropathy. ; ~$ R6 s1 [' J9 A, E
          【关键词】 congenital ureteral obstruction newborn rat aquaporin water channels sodium transporter AT receptor blockage
9 Q/ b; k, D; i- Z                  CONGENITAL OBSTRUCTIVE NEPHROPATHY is the primary cause for end-stage renal disease (ESRD) in children ( 2 ). As many as 1% of newborn infants have a prenatal diagnosis of hydronephrosis or significant renal pelvic dilation ( 11 ). The management of these patients remains controversial, some advocating for an early surgical repair whereas others favoring observation unless renal function decreases ( 21 )." m. w9 T9 W6 k, E

! ?& C, d* h2 d- DSeveral animal models have been developed to understand the development and pathophysiology of congenital obstructive uropathy. More than 90% of nephrogenesis in the rat takes place postnatally in the first 10 days of life, resembling the midtrimester in the human fetus ( 5 ), which makes neonatal ureteral obstruction (UO) attractive as an experimental model for congenital obstructive nephropathy. Recently, Wen et al. ( 48 ) developed a newborn rat model with severe partial obstruction, which was associated with the development of progressive hydronephrosis.
9 c, H9 P( f2 v7 \; y3 K6 V/ i6 z* J3 @
ANG II, the primary product of the renin-angiotensin system (RAS), is thought to play a crucial pathophysiological role in the functional changes in response to urinary tract obstruction ( 10 ). Furthermore, ANG II has also been demonstrated to play a critical role in kidney development ( 39 ). There are two major types of ANG II receptors, type 1 (AT 1 ) and type 2 (AT 2 ), but the AT 1 receptor is much better characterized then the AT 2 receptor. The AT 1 receptor is localized in proximal tubules, cortical and medullary collecting duct, and glomeruli, where it is primarily present on the mesangial cells. Furthermore, the AT 1 receptor is found on vascular smooth muscle cells in the afferent arteriole, interlobular, and arcuate arteries ( 16 ).
1 {0 d, g3 C  B" B" V
: f/ N9 m5 U9 n+ FInhibition of the RAS by both AT 1 blockade and angiotensin I-converting enzyme (ACE) inhibitors shows renoprotective effects in rats with chronic renal failure ( 38 ). In addition, Moriyama et al. ( 31 ) found that the ANG II receptor antagonist TCV-116 ameliorates the interstitial fibrosis and the progression of morphological changes in rat kidneys in response to complete UUO. Other studies have also shown that AT 1 -receptor blockade improved tubulointerstitial fibrosis in rats subjected to UUO ( 18, 47 ).
9 f. F+ R1 F( J/ A! N, X; k: r# c9 G& N# j5 c/ K- i
Inflammatory cell infiltration plays a key role in the onset and progression of renal injury in response to obstruction. Nuclear factor- B participates in the inflammatory response, regulating many proinflammatory genes. In a recent study, it was demonstrated that ANG II, both via AT 1 and AT 2 receptors, activates NF- B in response to obstruction in a mouse model ( 13 ). Pharmacological blockage of the RAS prevented NF- B activation and upregulation of NF- B-related proinflammatory genes ( 32 ), and inhibition of NF- B activation prevents inflammatory injury in UUO ( 30 ). Thus NF- B may play a pivotal role in the kidney injury observed in response to congenital induced ureteral obstruction.4 X: L4 }% {( r6 [% ?! z7 P0 H
. p+ @: d' x' `+ S
Aquaporins (AQPs) are a family of membrane water channels that mediate rapid water transport across the cell membrane. Recent studies have demonstrated that AQP1-4 were reduced in response to both bilateral ureteral obstruction (BUO) and UUO ( 14, 23, 35, 37 ). Additionally, Li et al. ( 24, 25 ) demonstrated that BUO and UUO in adult rats were associated with a reduced abundance of several major renal sodium transporters. Thus it is highly likely that dysregulation of renal AQPs and sodium transporters is responsible for the altered renal handling of water and sodium in response to ureteral obstruction. Importantly, this view was supported by studies in rats subjected to congenital UUO which demonstrated significant downregulation of AQP1, AQP3, and Na-K-ATPase and, consistent with these molecular changes, the obstructed kidneys had reduced solute-free water reabsorption and natriuresis ( 42 ). Importantly, this study demonstrated that release of obstruction after 1 wk, but not after 4 wk, prevented the majority of the changes in kidney function induced by the partial UUO, suggesting that early release of an obstruction protected the kidney from obstructive damage. Moreover, we have recently demonstrated in adult rats that treatment with candesartan partially prevented the decrease in AQP2 and Na-K-2Cl cotransporter expression in the kidney 2 days after release of BUO ( 19 ). This was paralleled by prevention of an impairment of phosphate, sodium, and water reabsorption in the tubules of the postobstructed kidney, demonstrating an association between the molecular and functional changes at these sites of the nephron. The findings indicated that intrarenal ANG II plays an important role in regulation of sodium transporters and AQPs in obstructive nephropathy ( 19 ).
: O  d0 E  v! h9 y' W8 w
7 W4 m9 ]7 D& f; v& Z8 xThe aims of the present study were therefore to examine whether blockade of the AT 1 receptor after completion of nephrogenesis in newborn rats with severe partial obstruction prevented the reduction in renal function as well as dysregulation of renal AQPs and sodium transporters and to explore potential pathways involved in this.. @" {# u; i# s

5 M8 h; G% [' ZMATERIALS AND METHODS
$ t- _6 a+ s! O- l& g. o8 x$ q1 @
* P! H# Z: s: p# H( U3 {Experimental Protocol1 ^2 n( e; T$ i. T4 N6 x6 m

/ P5 i* A( L1 N3 _- l/ Q( C$ BThis study included 44 (2-day-old) female Munich-Wistar rats (Møllegaard, Aarhus, Denmark), weighing 6-8 g, randomized into two groups: 1 ) severe PUUO ( n = 21), and 2 ) sham operation ( n = 23).
9 Z' H& M, M# Y, |7 Y
  m. w" c* m# BNewborn rats are poikilothermic, and adult thermoregulatory capabilities do not develop until week 3 of life. Because of their small body mass, rapid core cooling can be achieved by surface cooling ( 40 ). Accordingly, the newborn rat was placed on crushed ice for about 8-10 min. When the newborn rat was deeply asleep, the rat was removed to the operating table. If needed, additional crushed ice was placed around the body and neck region during the operation. Normally, placement of a newborn rat on crushed ice for 8-10 min is sufficient to maintain anesthesia for 30-40 min.
4 Z) R0 Q; l' \6 k
0 ]* u% i, M$ D3 \. J% Q6 KSevere partial obstruction of the left ureter was produced according to a modification of the technique of Ulm and Miller ( 46 ). The left ureter was exposed via an abdominal transperitoneal incision using a microscope with x 25 magnification. The underlying psoas muscle was split longitudinally to form a groove, into which the upper two-thirds of the left ureter was embedded. The muscle edges were closed by ethilon sutures (9-0). In the sham group, a laparotomy was performed and the left ureter was exposed. Afterward, the rats were placed in an incubator with a temperature of 28°C until they totally awakened.
( q1 v4 H8 p- u8 D  S  ?7 q) q2 T; G7 J. G2 e
The mother rats had free access to a standard rodent diet and were kept in an animal facility with a controlled room temperature of 21 ± 2°C and 12:12-h dark-light cycle.; j% q* o3 [% Z1 @. H+ n1 P
! U5 h3 b9 W( C- K& W( r
Two rats from the PUUO group and two rats from the sham group were eaten by mother rats during the preweanling term (before day 21 of life). Two rats from the PUUO group had a wound infection, so they were excluded from the study at day 10 of life. At day 21 of life, the rats were separated from their mother and divided randomly into the following four protocols.2 }5 x  K! z# f6 P+ `7 |+ {; @

# o, n2 {& C/ v) O" }0 iProtocol 1 ( n = 8): severe PUUO.
9 n2 `* d. _3 `& q+ X+ ]' o# H8 h
1 r0 g2 R  ]0 o& FProtocol 2 ( n = 9): severe PUUO treated with the AT 1 receptor antagonist candesartan (PUUO CAN).! Q0 z% D2 v. x9 \
8 d8 W- F" d! L3 c/ y& ^2 B( L7 T
Protocol 3 ( n = 10): sham rats treated with candesartan (Sham CAN).& H7 N) C  ~8 ?. u+ I5 g, L
* a1 T4 u1 m& C# ?9 Y4 ?/ A1 P
Protocol 4 ( n = 11): sham-operated rats (Sham).
+ m( l/ U: E7 J5 s3 N. b* D' ~# B' u, ?3 }" @, u0 E
All procedures conformed with the Danish National Guidelines for care and handling of animals and to the published guidelines from the National Institutes of Health. The experimental animal protocols were approved by the board of the Institute of Clinical Medicine, University of Aarhus, according to rules issued by the Danish Ministry of Justice.  n: z1 _4 J% ?8 T4 |& [

) x* o, s9 D& h4 q  jCandesartan cilexetil (10 mg·kg -1 ·day -1, AT 1 receptor antagonist, batch no: 300087-05, AstraZeneca, Molndal, Sweden) was suspended in 5% gum arabic (Sigma). From day 21 to week 10, candesartan cilexetil was provided in the drinking water daily at a dosage of 10 mg·kg -1 ·day -1, which complies with the pharmacological profile previously reported ( 15, 31, 33, 34 ).$ v6 n# [+ K- k' j% N
3 @1 `& }$ k# Z; l& J
At 10 wk of age, each rat was placed in an individual metabolic cage and allowed 2 days of acclimatization followed by three consecutive 24-h urine collections. Moreover, the intake of water and food was measured daily./ s4 U, N! f& U0 x9 S: M7 |

2 C2 N; ~9 F& U# VMRI was performed with a small-bore 7-T system (Varian, Palo Alto, CA). The rat was placed supine in a Helmholtz head-coil of 4 cm in diameter and subjected to an imaging protocol including measurements of single-kidney renal blood flow (RBF) and total kidney volume (TKV). Rats were anesthetized with 50 mg/kg body wt pentobarbital sodium (ip), and body temperature was maintained at 37°C during the studies.1 d' s" \  L# n4 w
0 G( B/ W( Y( y1 V9 y9 P7 g* F
RBF measurements were performed using a velocity-sensitive technique, involving a gradient echo-pulse sequence employed with bipolar gradients. The strengths of the velocity-encoding gradients were set according to the values from previous studies ( 42, 43, 49, 50 ). In brief, 10 slices of 1.2-mm thickness were prescribed perpendicularly to the renal veins. Each slice had a 7 x 7-cm 2 field of view and a resolution of 350 x 350 pixels to ensure appropriate pixels to find and derive the blood velocity in the renal veins. Other parameters included repetition time (TR) = 150 ms, echo time (TE) = 5.5 ms, flip angle = 55°, and no. of data averages = 4. Acquired phase images were subtracted, and the vein flow was determined by multiplication with the renal vein area in each available slice. The single-kidney RBF was then calculated as the average of the flow values found in all slices ( 43 ).
) M( f& P* D6 e+ |. i6 j) P$ q# L! R) ^& b+ u5 S
A gradient echo sequence was used to obtain a series of axial slices through both kidneys to determine TKV. Dependent on the kidney size, 20-30 equidistant slices of 1.0-mm thickness were employed to sufficiently cover both kidneys. The field of view and pixel size were identical as described for the RBF measurements, and other parameters were: TR = 125 ms and TE = 4 ms. Postprocessing included manual identification of each kidney for all slices, and, by careful encompassing of the regions of interest, TKV was measured by the sum-of-areas principle ( 43, 50 ).
* |/ S3 I) q+ w8 p$ n. U! q* u& d/ i
Measurement of GFR2 W4 }0 [9 c# k  \
' R9 h3 F, L) }6 {; A6 M& s4 c
GFR was evaluated from the renal clearance of 51 Cr-EDTA as previously described ( 43 ). In brief, the left femoral artery and vein were catheterized under inhalation anesthesia with isoflurane (Abbott Scandinavia). The anesthetized rats were placed on a heating table to maintain a rectal temperature of 37°C. The arterial and venous catheters were fixed as described by Shi et al. ( 43 ). To collect urine, both ureters were exposed and catheterized (0.8-mm flexible plastic tubing, Tygon, Weyerhauser, Cleveland, OH) through a midline incision. Afterward, a priming dose of 51 Cr-EDTA (0.2 MBq) was given (iv) for 15 min, followed by a sustained infusion (0.005 MBq/min) during a 75-min equilibration period, which was then followed by two 1-h urine collection periods. An intravenous infusion of 25 mM glucose (40 µl/min) was provided simultaneously to keep an adequate minimum urine flow rate necessary for biochemical analysis of the collected urine. Timed blood samples (150 µl) were withdrawn from the arterial catheter every hour during the urine collection periods and replaced immediately with a similar volume of heparinized donor blood without changing hematocrit. Timed urine samples were gravimetrically collected every hour from both ureters. During the experiment, the incision was closed to prevent loss of body fluid. The plasma and urine samples were diluted, and 51 Cr-EDTA was counted in a scintillation system (Cobra, Packard Instrument, Meriden, CT ) ( 42, 43 ).: z4 p% y$ r6 w2 K

5 Y2 ]4 {  f$ n- a5 o  A7 \4 nBefore the rats were killed, 3-4 ml of blood were collected in a heparinized tube for determination of plasma electrolytes and osmolality. Another 2-3 ml of blood were collected in an EDTA tube for aldosterone determination. The plasma and urinary concentrations of creatinine, urea, and phosphate and the plasma concentrations of sodium and potassium were determined (Vitros 950, Johnson & Johnson). The concentrations of urinary sodium and potassium were determined by standard flame photometry (Eppendorf FCM6341). The urine and plasma osmolalities were measured with a vapor pressure osmometer (Osmomat 030-D, Gonotec, Berlin, Germany). The harvested kidneys were rapidly frozen in liquid nitrogen and kept at -80°C until assayed.+ M/ X! D& c. h
9 `) g$ W# [. R7 d: o  e4 m
Analysis of Renal AQPs and Key Sodium Transport Proteins
5 C) o$ [& J* B! v" H( W' @/ B0 L* I9 p1 t: E& G
Membrane fractionation for immunoblotting. Kidneys were minced finely and homogenized in 9 ml of dissecting buffer (0.3 M sucrose, 25 mM imidazole, 1 mM EDTA, pH 7.2, containing protease inhibitors 8.5 µM leupeptin and 1 mM phenylmethylsulfonyl fluoride) with five strokes of a motor-driven Ultra-Turrax homogenizer (IKA Labortechnik, Staufen, Germany) at 1,250 rpm. This homogenate was centrifuged in a Beckman L8M centrifuge at 4,000 g for 15 min at 4°C, and gel samples (Laemmli sample buffer containing 2% SDS) were made of this pellet. The total protein concentration of the homogenate was measured using a Pierce BCA protein assay kit (Roche).) d8 G3 _' t" ~( t1 t7 v
$ x8 n# q' N5 R- v8 K$ S4 ?5 g( e9 d
Primary antibodies. For semiquantitative immunoblotting and immunocytochemistry, we used the characterized monoclonal and polyclonal antibodies as summarized below.4 Z* J5 a% T0 |" \- N, S5 p, m

: K6 ~( s' |  e0 tAQP1 (CHIP serum or LL266AP): immune serum or an affinity-purified antibody to AQP1 ( 44 ).
  n7 W- |% s, W$ E9 J+ i7 P
2 k# C# E3 N- e# p# yAQP2 (LL127 serum or LL127AP): immune serum or an affinity-purified antibody to AQP2 ( 7 ).
' Y, ^) y. m& z: U3 l
' x' h: ^9 B, B! ~" iAQP3 (LL178AP): an affinity-purified polyclonal antibody to AQP3 has previously been described ( 8 ).. C! G% V3 Z) |3 k: H% `1 G0 I

' A6 J0 s5 W7 y/ w5 ENa-K-ATPase: a monoclonal antibody against the 1 -subunit of Na-K-ATPase has previously been described ( 20 ).3 K. j- K) A9 J7 x+ g0 z

3 v5 f( z5 g3 l9 w7 wNKCC2 (LL320AP): an affinity-purified polyclonal antibody to the apical Na-K-2Cl cotransporter of the thick ascending limb has previously been described ( 9 ).
5 u! N/ z3 P# w0 x( S" m7 X5 A6 P* c" z/ v1 Q4 o
NF- B (DO604): immune serum or an affinity-purified antibody to NF- B ( 13 )
4 ?5 _& o2 b5 V1 z0 B4 c, f. m3 B  a0 ?9 I8 n3 f" ^" H
Electrophoresis and immunoblotting. Samples of membrane fractions from whole kidney were run on 9 or 12% polyacrylamide minigels (Bio-Rad Mini Protean II). For each gel an identical gel was run in parallel and subjected to Coomassie staining. The Coomassie-stained gel was used to ascertain identical loading or to allow for potential correction for minor differences in loading after scanning and densitometry of major bands. The other gel was subjected to blotting. After being transferred by electroelution to nitrocellulose membranes, blots were blocked with 5% milk in PBS-T (80 mM Na 2 HPO 4, 20 mM NaH 2 PO 4, 100 mM NaCl, 0.1% Tween 20, pH 7.5) for 1 h and incubated with primary antibodies overnight at 4°C. After being washed, the blots were incubated with horseradish peroxidase-conjugated secondary antibody (Dako, Glostrup, Denmark). After a final washing, antibody binding was visualized using the enhanced chemiluminescence (ECL) system (Amersham International). ECL films were scanned using a Hewlett-Packard Scanjet scanner and Adobe Photoshop software. The labeling density was determined from blots, where samples of kidneys from each group were run. The labeling density was corrected by densitometry of Coomassie brilliant blue-stained gels to detect minor differences in protein loading.
- L* D8 \* j$ a6 Q% F
$ M+ W$ m7 e2 Z; V( o- g9 OImmunocytochemistry- [. S8 ^7 b' c( f) h5 `# T5 k
6 p8 Y9 Z9 S" u3 N' @8 @
The kidneys from experimental rats and sham-operated rats were fixed by retrograde perfusion via the abdominal aorta with 3% paraformaldehyde in 0.1 M cacodylate buffer, pH 7.4. For immunoperoxidase microscopy, the blocks of the kidneys containing all zones were dehydrated and embedded in paraffin. The paraffin-embedded tissues were cut on a rotary microtome (Leica, Heidelberg, Germany). The sections were subsequently deparaffinated and rehydrated. For immunoperoxidase labeling, endogenous peroxidase was blocked by 0.5% H 2 O 2 in absolute methanol for 10 min at room temperature. To reveal antigens, the sections were put in a 1 mM Tris solution (pH 9.0) supplemented with 0.5 mM EDTA and heated in a microwave oven for 10 min. Nonspecific binding of immunoglobulin was prevented by incubating the sections in 50 mM NH 4 Cl for 30 min followed by blocking in PBS supplemented with 1% BSA, 0.05% saponin, and 0.2% gelatin. Finally, sections were incubated overnight at 4°C with primary antibodies diluted in PBS-T supplemented with 0.1% BSA and 0.3% Triton X-100. The following day, the sections were washed 3 x 10 min in PBS-T with 1% BSA and 0.3% Triton X-100 before being incubated in horseradish peroxidase-conjugated secondary antibodies (Dako) diluted in PBS-T supplemented with 0.1% BSA and 0.3% Triton X-100. Afterward, the sections were incubated in diaminobenzidine. Microscopy was carried out using a Leica DMRE light microscope (Leica).
+ i3 O; r$ I8 {1 G# k
7 g/ c5 `) ~" r% p7 _5 PStatistical Analysis  H! f% D: I0 \$ U: ?

. \; k+ G$ _' Z' ~  j. j9 b# w8 Q  J- hValues are expressed as means ± SE. Statistical analyses were carried out by one-way ANOVA followed by appropriate post hoc tests with the Bonferroni test, and P / Z" Z, \' }% C6 v* L
, b! K; b) U% Q: h0 l  q! Q
RESULTS+ g1 D: W& D; O# e

9 F3 K# C  A# aCandesartan Prevents a Significant Reduction in GFR
) D1 S$ B1 m9 z# D( f. Q
& a* h* Y. p7 FKidney GFR was measured using renal clearance of 51 Cr-EDTA at 10 wk after the onset of PUUO. In the PUUO group, total GFR was significantly reduced compared with sham-operated rats ( P
6 e$ v  z2 o9 ]# }; k
% Y, H4 I. V5 Y: V/ d% e. PFig. 1. Glomerular filtration rate (GFR) changes in rats at 10 wk of age. A : total GFR was calculated as a sum of obstructed (OBS) and nonobstructed single kidney GFR. At 10 wk after onset of partial unilateral ureteral obstruction (PUUO), total GFR was decreased to 51% of total GFR in sham-operated controls (SHAM). B : neonatally induced partial ureteral obstruction results with a marked GFR reduction in the obstructed kidney. Open bars, GFR changes in the obstructed kidney; filled bars, changes in the nonobstructed kidney. * P
! c. k  I0 C, [* y1 Q: Y! p
" s; e  X9 L' m% i9 ?3 O5 n6 q# yCandesartan Prevents PUUO-Induced RBF Reduction
) p( w9 r- E- C
( z' \9 m- Q5 `; WRBF measurements were obtained using a phase-contrast MRI technique involving a gradient echo sequence with bipolar flow-sensitive gradients. RBF was markedly decreased in the obstructed kidney of PUUO compared with sham-operated rats ( P " y" k2 b: l2 x/ H; _8 j; `
6 G( h" u8 ]( ]) j6 W
Fig. 2. Renal blood flow (RBF) and total kidney volume (TKV) in rats at 10 wk of age subjected to neonatal PUUO, PUUO CAN, SHAM, and SHAM CAN. * P
3 u/ y) j1 @4 f2 L! M
! n3 N  X( P) }. p( |# oTable 2. Changes in biochemical values at 10 wk of age in rats subjected to neonatal PUUO, PUUO   CAN, Sham   CAN, and sham operation
0 x% k, q7 I  Q, k
1 n9 B5 C; w" Y. X$ WLong-Term Candesartan Administration Prevents Progression of Hydronephrosis
& ~+ \* K, e% e* x3 a) C" W  q; i- x! I" }5 M/ l
The average TKV was also measured using MRI. This demonstrated a significant increase in TKV in the obstructed kidney of PUUO compared with sham-operated rats ( P
% Q" C% e* u! `9 K# w
! T. N8 U( {5 Z0 x2 b' Q9 HFig. 3. Representative MRI images. A : MRI of a rat with left severe PUUO. Enlarged obstructed pelvis is clearly visible. B : MRI of a SHAM CAN rat. The 2 kidneys are clearly identified. C : MRI of a 10-wk-old PUUO CAN rat with the obstruction of the left kidney.
" M% |  @# R. p4 E: `' V! N
: g0 `* k+ O& y$ ^" wTable 1. Body weight, kidney weight, protein concentration, and total protein in rats at 10 wk of age subjected to neonatal PUUO, PUUO   CAN, Sham   CAN, and sham operation
2 {, u$ h4 s1 Q( }  j( x( g& E+ s8 m; w
Candesartan Prevents PUUO-Induced Downregulation of AQP2 and Alters Renal Water Handling. J- {( l; |3 K0 t( w: ^6 r5 K
+ P! I% p8 H& d& }/ d, P
AQP2 was expressed in the apical plasma membrane and subapical vesicles of collecting duct principal cells. The affinity-purified anti-AQP2 antibody exclusively recognizes 29- and 35- to 50-kDa bands, corresponding to nongylcosylated and glycosylated forms of AQP2. Semiquantitative immunoblotting using membrane fractions of whole kidney revealed that the abundance of AQP2 in the obstructed kidney of rats with PUUO was decreased compared with sham levels (52 ± 15 vs. 100 ± 4%, P 6 @# X: k% Z% k. u9 t+ e

% a4 x6 L9 _9 v! a4 yFig. 4. Semiquantitative immunoblotting of membrane fractions of whole kidney from rats at 10 wk of age subjected to PUUO, PUUO CAN, SHAM, and SHAM CAN. A : immunoblot was reacted with anti-aquaporin-2 (AQP2) antibody. B : densitometric analysis revealed that AQP2 abundance in whole kidney was significantly decreased in rats with PUUO. Long-term candesartan intervention prevented the decrease in AQP2 expression of the obstructed kidney. * P 2 {1 n0 M5 j" o1 Y
/ N, X- y$ Q7 e& T/ k, b! F
Table 3. Functional data in rats at 10 wk of age subjected to neonatal PUUO, PUUO   CAN, Sham   CAN, and sham operation
* e0 x0 A2 J7 ?. M! p6 E
8 j; o' f" J4 V2 R7 ?6 ]Table 4. Renal tubular function in rats at 10 wk of age subjected to PUUO, PUUO   CAN, Sham   CAN, and sham operation9 M$ k2 G9 B2 I7 \  u5 A

( _8 [0 y7 n: Q" ^+ |0 MTable 5. Summary of densitometric changes in the expression of renal aquaporins and major sodium transporters in rats subjected to PUUO, PUUO   CAN, Sham   CAN, and sham operation+ X* i& f4 F2 D- y9 V

! d" c+ \! L) U% {* b& P. w* KWater transport across the basolateral membrane of the collecting duct principal cells is mediated by AQP3. Consistent with the findings on AQP1 abundance, total kidney AQP3 abundance did change significantly in either PUUO or PUUO CAN rats ( Table 5 ).
9 y: e5 _0 `" v1 V7 \& d2 m- e
/ N& p& [% Q& }' q- qCandesartan Prevents PUUO-Induced Downregulation of Na-K-ATPase and Decreases Fractional Sodium Excretion) P) M1 y, _* O
! ?* p4 N+ T6 N2 Q9 Y8 N& R8 t
The active transport of sodium occurs mainly via the key sodium transporters and cotransporters in the nephron, and we therefore examined the expression of key sodium transporters in whole kidney samples by semiquantitative immunoblotting. The abundance of Na-K-ATPase in the obstructed kidney of PUUO rats was reduced compared with sham-operated animals (75 ± 12 vs. 100 ± 5%, P % ?4 p3 p8 v3 K# D
7 [( n; Y! `+ O. S  S
Fig. 5. Immunoblot of membrane fractions of total kidneys from rats at 10 wk of age subjected to PUUO, PUUO CAN, SHAM, and SHAM CAN. A : immunoblot was reacted with anti-Na-K-ATPase ( 1 -subunit) and revealed an 96-kDa band. * P
: j: K) Z: K. \8 }3 [& X' T3 I- U( M8 G/ M
To investigate the molecular mechanisms involved in the impaired sodium reabsorption in the obstructed kidney after neonatal PUUO, expression of NKCC2 was examined in whole kidney samples. NKCC2 did not significantly change in the obstructed kidney, and candesartan treatment of PUUO rats revealed that NKCC2 did not differ from that in PUUO rats ( Table 5 ).
: B# Y0 P; q9 I9 u9 v! R$ ?8 F  j  F9 y/ Z. R/ y
Renal sodium handling was calculated from both obstructed and nonobstructed kidneys. Importantly, neonatally induced PUUO did not change plasma concentrations of sodium ( Table 2 ). In accordance with the reduced GFR, the filtered load of sodium was severely reduced in the obstructed kidney in PUUO rats compared with sham-operated rats (5.8 ± 2.6 vs. 29.3 ± 7.6 µmol·min -1 ·100 g -1, P 6 |( ]5 M; A) e: E# ~, Y, v$ m* ?

! o6 [" f2 h5 x: J3 I2 X  R9 [ANG II Activates NF- B in Response to PUUO# |5 U  C$ a, ^  M1 f. T! @% ]

% X6 D/ F+ F2 Q7 ]3 pTo evaluate the contribution of the NF- B pathway of renal changes in this model, we examined whole kidney NF- B expression in the different groups. NF- B expression in the obstructed kidney of PUUO rats was markedly elevated compared with sham-operated control rats (192 ± 38 vs. 100 ± 11%, P ( U. A5 i, g. l
% S7 d* ]- S0 C$ f5 J" q& W
Fig. 6. Immunoblot of membrane fractions of total kidneys from rats at 10 wk of age subjected to PUUO, PUUO CAN, SHAM, and SHAM CAN. A : immunoblot was reacted with anti-NF- B and revealed an 65-kDa band. * P
' Q! A- q" [# _) O6 m% j
5 n: E* b+ o/ Q3 T) @. a' \" G9 w' z( XPlasma Aldosterone Levels Are Reduced in Response to Candesartan Treatment
1 }0 ^0 G  p9 D  c5 h$ k/ }/ L
: N& U; B9 J  c5 xANG II stimulates aldosterone release from the adrenal gland via AT 1 receptors. The plasma concentration of aldosterone was used to verify inhibition of the AT 1 receptor by candesartan. In the present study plasma aldosterone levels were reduced in the candesartan-treated PUUO group compared with sham-operated rats ( Table 2 ). The candesartan-treated sham group showed a statistically significant decrease in aldosterone levels compared with those observed in the sham group ( Table 2 ), demonstrating that ANG II via the AT 1 receptors stimulates aldosterone release.8 F2 v, _; R/ M* H
" _2 j+ Z! C% K* Y; v
DISCUSSION1 a6 j' u5 D& j, F) \
8 Q; L3 [0 M- ]+ G
The main results of the present study demonstrated that neonatally induced PUUO for 10 wk resulted in a decrease in GFR and RBF in the obstructed kidney, paralleled by a marked increase in TKV in the obstructed kidney of PUUO rats. Consistent with these findings, AQP2 and Na-K-ATPase expression was downregulated in response to PUUO. Administration of the AT 1 receptor antagonist candesartan attenuated the decrease in ipsilateral RBF, GFR, and prevented the increase in TKV. Importantly, candesartan partly prevented the decrease in AQP2 and Na-K-ATPase expression in newborn rats subjected to PUUO. These findings indicate that ANG II is an important mediator of the hemodynamic changes during PUUO. The progression of hemodynamic and tubular changes was at least partly mediated via the angiotensin AT 1 receptor, and thus candesartan appears to be useful in the prevention of these changes.* b/ S) `& _% `* g
% R+ u5 d& G, }& M) y  Y1 `1 r
Candesartan Prevents PUUO-Induced GFR and RBF Reduction
: y) Z" I" I$ ~" Q/ R  x) }0 E9 W
, j" G' R0 T( a, }% v4 e" I; lThe present study demonstrated that PUUO induced within 2 days of life was associated with a marked GFR reduction. This is consistent with our previous studies demonstrating that GFR is markedly reduced at week 24 after onset of neonatal PUUO ( 42, 43 ). As suggested by a recent study using a model of variable PUUO in the neonatal rat in which GFR was also reduced by 80% and a reduced number of nephrons ( 45 ), this indicates that neonatal PUUO is associated with a dramatic impact on glomerulogenesis, which has also been demonstrated in pigs subjected to PUUO ( 12 ).
7 P- M# E( X. S; O( Z# Q& _- L5 _8 b
Candesartan attenuated the GFR reduction in the obstructed kidney of PUUO rats. This finding is also consistent with a previous study from our lab demonstrating prevention of the GFR reduction after treatment with candesartan in adult rats subjected to BUO for 24 h followed by a 2-day release ( 19 ). Moreover, Pimentel et al. ( 18, 41 ) also administered an ACE inhibitor or an AT 1 receptor inhibitor to rats before UUO and showed that both drugs improved the hemodynamics of the postobstructed kidney. The marked reduction in the mean arterial pressure by candesartan is also consistent with its ability to inhibit the vasoconstrictor effect of ANG II. Candesartan-induced augmentation of renal function is most likely related to antagonism of the renal AT 1 receptors. Consistent with previous studies indicating progressive RBF reduction in chronic PUUO, the present study confirmed that PUUO resulted in a dramatic decrease in RBF and candesartan treatment attenuated the reduction in RBF. Thus the results of the present paper support the view that ANG II is an important mediator of the vasoconstriction associated with ureteral obstruction of the kidney. Despite the profound reduction in mean arterial blood pressure in response to candesartan, the parallel increase in both RBF and GFR suggests a preponderant ANG II-dependent afferent arteriolar vasoconstriction of the partially obstructed kidney.9 U3 O! Z4 o- q3 Y

" o4 R  P& I2 X5 p0 ERecently, it was demonstrated that both mild and severe PUUO induced a significant decrease in RBF and the magnitude of the decrease was dependent on both the severity and duration of the PUUO ( 50 ). Additionally, MRI measurements in a 24-wk-old rat PUUO model revealed that both TKV and RBF increased significantly in the contralateral nonobstructed kidney. Consistent with this, Shi and co-workers ( 43 ) also demonstrated compensatory changes in RBF of the intact kidney in response to neonatal UUO, which persisted up to 24 wk of age. Interestingly, in the present study RBF of the contralateral kidney did not increase at 10 wk, suggesting that the compensatory increase in RBF is detectable at a later stage. However, our study demonstrated that RBF in the contralateral kidney of PUUO CAN rats increased, supporting the view that there is ANG II-dependent vasoconstriction of the contralateral kidney in response to PUUO, as suggested in a previous study where the effect of an ACE inhibitor was evaluated in a guinea pig model of partial ureteral obstruction ( 4 ). The effect of candesartan cilexetil treatment on RBF is likely due to antagonism of AT 1 receptors located on the afferent and efferent arterioles. Consistent with this, previous studies have demonstrated similar renoprotective effects of AT 1 receptor antagonist treatment in several animal models ( 28, 31, 33 ). Despite a significant reduction in mean arterial blood pressure, renal functions were unchanged in the sham-operated rats, suggesting that blood pressure remained within the limits of renal autoregulation. This is consistent with previous data demonstrating that inhibition of AT 1 receptors with candesartan cilexetil provides protection against ANG II mediated increases in arterial pressure and prevents the associated deterioration of renal autoregulatory responsiveness ( 17 ).
. a6 c! l: e6 \1 M8 S7 D: a8 t: S2 Q( t7 `+ g
Long-Term Candesartan Administration Prevents Progression of Hydronephrosis  W6 r. g7 {. D1 d# P: U% v
9 E* D6 {- B' w
Partial ureteral obstruction is associated with pelvic dilatation, and the degree of dilatation may reflect the severity of the obstruction. To examine the effect of obstruction on pelvic dilatation, total kidney volume was determined by MRI. Consistent with previous in vivo and in vitro studies, measurements of kidney volume correlated significantly, demonstrating that MRI is a reliable technique for evaluation of the degree of hydronephrosis ( 6, 50 ). Thus the present study revealed a significant hydronephrosis in obstructed PUUO kidneys which is consistent with previous findings demonstrating a similar increase in response to severe neonatal PUUO ( 43, 49, 50 ). In sham-operated rats, the TKV of the left and right kidneys did not differ. However, an important and novel finding of the present study was that long-term candesartan treatment markedly prevented the increase in TKV. In the candesartan-treated rats, TKV of the obstructed kidney was diminished by 50%. This result indicates that candesartan cilexetil may play an important role in reduction in TKV of the obstructed kidney in rats with PUUO, most likely by blocking detrimental ANG II-induced cellular effects in the obstructed kidney. Moreover, candesartan treatment prevented the reduction in PC and TP of the obstructed kidney, supporting the view from previous studies that sustained enhanced intrarenal ANG II generation is detrimental for renal functional development in response to unilateral ureteral obstruction and that blockade of the RAS may prevent the progression of proteinuria ( 38 ) and fibrosis ( 26, 29 ). Consistent with this, a recent study by Beharrie and co-workers ( 3 ) demonstrated that enalapril treatment affords protection by attenuating proteinuria, promoting uricosuria, and diverting solute diuresis. Thus there is now evidence that angiotensin blockade during chronic obstruction protects kidney function in the young rat. In a number of studies, it has been demonstrated that AT 1 receptor blockade ameliorates the obstruction-induced fibrosis, most likely via inhibition of the NF- B pathway, which participates in the regulation of renal monocyte recruitment ( 13, 27 ). At present, it is unclear whether the same pathways are of similar importance in response to congenital obstruction in the rat, and this needs to be further addressed in a future study. However, the obstruction-induced tubulointerstitial fibrosis is a complex renal disease, where multiple hormonal systems are activated ( 2 ).
( b. e. Y" _. _, q$ w: ?, ^; @4 b1 H  p) {) c" |% z
Candesartan Partly Prevents PUUO-Induced Downregulation of AQP2 and Alters Renal Water Handling
5 ~7 W! n4 r- l) i" [) D- _& l
$ @; j- R2 d* gAQP2 is expressed in the apical plasma membrane and subapical vesicles of the collecting duct principal cells, and this protein is responsible for water transport across the apical membrane in the collecting duct ( 35, 36 ). The present study demonstrated that AQP2 expression is downregulated in the obstructed kidneys of the rats with neonatally induced PUUO. In parallel, solute-free water reabsorption was severely reduced in obstructed kidneys, demonstrating a functional association between AQP2 downregulation and water reabsorption at the collecting duct level. This finding is consistent with previous studies demonstrating that unilateral ureteral obstruction downregulates several AQPs in the kidney ( 22, 23, 42, 43 ).
. p: [- Z. E, j3 w9 \) e; M7 T& o( l" p' y. l
Candesartan treatment partially prevented the reduction in AQP2 expression in PUUO rats. Consistent with this, candesartan treatment was associated with an increase in solute-free water reabsorption, demonstrating a functional association between the molecular changes in AQP2 and the physiological change in collecting duct water handling. Thus the present findings support the view that ANG II may play an important role in the regulation of collecting duct function, as recently demonstrated by Jensen et al. ( 19 ), who showed that candesartan administration to adult rats subjected to 24-h BUO followed by a 2-day release partially prevented the reduction in AQP2 expression levels in the obstructed kidney.; v& _' g/ X4 j" t/ C

- t/ G" ]7 Y" k1 Q9 S6 fCandesartan Prevents PUUO-Induced Downregulation of Na-K-ATPase and Decreased Fractional Sodium Excretion
, y/ o' N9 v- ~" T5 a- f
( M+ e; m. ?$ X3 S$ GThe active transport of sodium occurs mainly via the key sodium transporters: the basolateral Na-K-ATPase ( 20 ), the type 3 Na/H exchanger (NHE3) ( 1 ), and the apical BSC-1 (or NKCC2) ( 9 ). This study demonstrated that Na-K-ATPase abundance in the obstructed kidney was decreased after 10 wk of obstruction. There was a defective reabsorption of sodium in the obstructed kidney, which was evidenced by the increase in sodium excretion. Thus it is likely that the reduced abundance of Na-K-ATPase plays a significant role in the increased urinary excretion of sodium from the obstructed kidney in PUUO rats. The present results support the view that renal sodium transport is critically affected by ureteral obstruction, as previously demonstrated in a similar model ( 42, 43 ), and underscore the role of an intact expression of renal sodium transporters in maintaining an intact renal epithelial sodium transport in response to neonatal PUUO. Downregulation of Na-K-ATPase was prevented by candesartan treatment, demonstrating at the molecular level that blockade of ANG II-induced effects in the obstructed kidney is important in protecting the developing tubule system from damage. Candesartan treatment also attenuated the natriuresis from the obstructed kidney, demonstrating a functional association between the abundance of Na-K-ATPase and epithelial sodium transport. The Na-K-ATPase is distributed along all nephron segments and functions in basolateral transport of sodium in the kidney tubule ( 20 ). This protein is involved in establishing the driving force promoting sodium reabsorption in the kidney tubule ( 20 ). The mechanisms involved in downregulation of the Na-K-ATPase are not fully understood. The reduced GFR and associated reduction in the filtered load of sodium may directly regulate the expression of Na-K-ATPase. Alternatively, the progressive hydronephrosis and development of obstructive nephropathy due to the direct effects of the increased interstitial pressure may also be important factors in the dysregulation of Na-K-ATPase. Because candesartan treatment was associated with prevention of the GFR decrease as well as progression of hydronephrosis, this study cannot explain which factor(s) is the most significant.6 {. r3 s% s5 z2 m1 e* ]! Q8 I

& Y# C+ X& ]6 R# FCandesartan Attenuates the PUUO-Induced Increase in NF- B Expression
! m( {  g; E, V8 y5 u7 w2 l1 k5 s  q0 Z
Previous studies have demonstrated that ANG II activates NF- B in the kidney, via stimulation of both AT 1 and AT 2 receptors. Consistent with previous studies, we confirmed that NF- B expression in the obstructed kidney of PUUO rats was markedly elevated compared with sham-operated control rats. Importantly, candesartan treatment prevented the PUUO-induced increase in NF- B expression, suggesting that the beneficial effects of candesartan on kidney function in response to PUUO may be mediated via blockade of the NF- B pathway.0 {* ]+ @. G, r2 j" T

1 X+ D9 X& P! u( _) t9 XConclusion
$ u* a; X. b, i9 l  p
( G9 w' d3 I3 v9 GIn summary, this study confirmed that neonatal PUUO in rats is associated with marked long-term changes in renal functions. AT 1 receptor blockade prevented the decrease in GFR and RBF in the obstructed kidney in neonatally induced PUUO rats. Moreover, it also effectively prevented the kidney protein reduction, reduced hydronephrosis, and prevented downregulation of Na-K-ATPase and partially prevented AQP2 downregulation. Consistent with this, AT 1 receptor blockade prevented impairment of tubular transport of water and sodium from the obstructed kidney. Candesartan also attenuated the increase in NF- B expression, suggesting that NF- B may play a role in the renal injury in response to congenital ureteral obstruction.
7 `& P! {; {8 F  a5 W& n3 i8 M
8 @' j9 K2 U3 }# Q: |GRANTS  j* a* q& E% i, f
, D" j$ n5 e! P2 D; ^
The Water and Salt Research Centre at the University of Aarhus was established and supported by the Danish National Research Foundation (Danmarks Grundforskningsfond). Support for this study was also provided by The Danish Medical Research Council, The Karen Elise Jensen Foundation, The Novo Nordisk Foundation, WIRED program (Nordic Council and the Nordic Centre of Excellence Program in Molecular Medicine), and The Commission of the European Union (EU-Aquaplugs and EU-Action Programs), The Danish Research Academy, and the intramural budget of the National Heart, Lung, and Blood Institute, National Institutes of Health.7 ]5 U: @  W% f/ a
6 R2 D- {: b1 x# j+ q
ACKNOWLEDGMENTS6 t; k: ^. {. E! X, b3 o' s

2 z6 |5 ?  D& I/ E2 n. m7 s2 ]' \The expert technical assistance of Gitte Kall, Inger Merete Paulsen, and Dorte Wulff is highly appreciated.
+ f1 H5 R/ I/ X: ~: F7 p6 Z2 \3 Z          【参考文献】
" X. \7 x# |; _/ S, U Aronson PS. Role of ion exchangers in mediating NaCl transport in the proximal tubule. Kidney Int 49: 1665-1670, 1996.
) I, F4 u, i( n' g( M  t' V4 ^: w9 X6 V0 ^  X& q
. {7 c- u' q7 m: J+ o+ h
$ }# |% t1 P9 T3 ~9 s
Bascands JL, Schanstra JP. Obstructive nephropathy: insights from genetically engineered animals. Kidney Int 68: 925-937, 2005.0 ], }/ N5 q$ y1 M# |

* M; v* T7 g( z& V: b+ g
) G/ e6 p; ?# ~9 X% |) Y
  W) b6 {! I/ y+ Z! c, ~: ?Beharrie A, Franc-Guimond J, Rodriguez MM, Au J, Zilleruelo G, Abitbol CL. A functional immature model of chronic partial ureteral obstruction. Kidney Int 65: 1155-1161, 2004.! L  j" T5 t4 `* i8 D8 w

" N, X9 N1 ~0 Z! ?
  D8 D, {# {9 e/ \' S
* ~6 S& B9 @0 qChevalier RL, Peach MJ. Hemodynamic effects of enalapril on neonatal chronic partial ureteral obstruction. Kidney Int 28: 891-898, 1985.
# S' {8 ~/ V8 m8 K: f. {' O+ L* s
$ l4 J* G  @- N$ t. v% w$ [) s2 Z: W) z/ L( S0 j# v
0 ~% U- P" l/ w4 E8 J6 u
Chevalier RL, Thornhill BA, Wolstenholme JT, Kim A. Unilateral ureteral obstruction in early development alters renal growth: dependence on the duration of obstruction. J Urol 161: 309-313, 1999.
2 _9 I+ T9 n1 V% q) ?3 o) v6 {
1 h+ |& H8 `. _0 k* w
# {4 c6 a" A( Z- u9 P2 h& }2 h# f: M; _2 `( @
Christiansen T, Rasch R, Stodkilde-Jorgensen H, Flyvbjerg A. Relationship between MRI and morphometric kidney measurements in diabetic and non-diabetic rats. Kidney Int 51: 50-56, 1997.& M0 Z# Q& B4 z0 \6 A
( e- d4 q+ Z6 {! S8 O9 ~; Q

" T; `  W. M# [9 n9 b& ~! M' @( W0 M. R9 K+ T+ U( g5 o
DiGiovanni SR, Nielsen S, Christensen EI, Knepper MA. Regulation of collecting duct water channel expression by vasopressin in Brattleboro rat. Proc Natl Acad Sci USA 91: 8984-8988, 1994.
* P/ C1 Q( q1 }  x* u" N4 F: Q
" k- u+ B( h: n2 C' E* G
- F8 _, r9 y& M# u, S. a- Q& O. n: c5 d8 f) t* |6 o1 G
Ecelbarger CA, Terris J, Frindt G, Echevarria M, Marples D, Nielsen S, Knepper MA. Aquaporin-3 water channel localization and regulation in rat kidney. Am J Physiol Renal Fluid Electrolyte Physiol 269: F663-F672, 1995.
) W- w" z1 ^0 y  p( e2 G& b
; t) N8 @* }/ d0 j, \6 x
, L3 t. s3 |7 a
7 e; ]* Y7 i& g2 z, r# m7 YEcelbarger CA, Terris J, Hoyer JR, Nielsen S, Wade JB, Knepper MA. Localization and regulation of the rat renal Na   -K   -2Cl - cotransporter, BSC-1. Am J Physiol Renal Fluid Electrolyte Physiol 271: F619-F628, 1996.
/ u* k6 `: t; y4 @2 `+ m2 `: [" [$ r
5 K2 a3 F+ L  j7 D( R& u# |" ^- {! Z, R- S& Y: {
# {1 q5 l$ K* P8 Q
el-Dahr SS, Gee J, Dipp S, Hanss BG, Vari RC, Chao J. Upregulation of renin-angiotensin system and downregulation of kallikrein in obstructive nephropathy. Am J Physiol Renal Fluid Electrolyte Physiol 264: F874-F881, 1993.
$ e; s/ L' A- E; p, I. \6 V" Q) i1 ^8 _5 t- H- W, W- h+ R& \

+ \! e" @/ D. Z6 \" [! i6 V9 _% s2 o" q  ]3 b% A
Elder Antenatal hydronephrosis JS. Fetal and neonatal management. Pediatr Clin North Am 44: 1299-1321, 1997.
3 k; @8 w- a; @/ }0 l/ m
+ U) {; O) l2 m5 A6 _% a2 a& D9 w6 E2 T9 |& m
, C# ]+ ^5 s/ T. Z
Eskild-Jensen A, Frøkiær J, Djurhuus JC, Jorgensen TM, Nyengaard JR. Reduced number of glomeruli in kidneys with neonatally induced partial ureteropelvic obstruction in pigs. J Urol 167: 1435-1439, 2002.2 f+ _, Z0 E5 U
4 @0 w' Y( l" U7 h. @
4 Z: A7 i, k" _; L. {
" S% _) k% o- R% Z. x
Esteban V, Lorenzo O, Ruperez M, Suzuki Y, Mezzano S, Blanco J, Kretzler M, Sugaya T, Egido J, Ruiz-Ortega M. Angiotensin II, via AT1 and AT2 receptors and NF-kappaB pathway, regulates the inflammatory response in unilateral ureteral obstruction. J Am Soc Nephrol 15: 1514-1529, 2004.0 F3 [9 R1 Q7 t, ?
& w6 g' [. o  T% B* C7 K

: V$ v( I% `' n- z0 I, E" M2 x7 d  d6 Y3 I. q0 \4 o& l7 z
Frøkiær J, Christensen BM, Marples D, Djurhuus JC, Jensen UB, Knepper MA, Nielsen S. Downregulation of aquaporin-2 parallels changes in renal water excretion in unilateral ureteral obstruction. Am J Physiol Renal Physiol 273: F213-F223, 1997.1 B- ]4 z  [7 t$ M1 J

4 l  @' l, W; B6 m* d& l* }4 h, Q8 z. K8 ^0 ~6 U- O) M& e
' [  `% ~- e2 B2 Z5 f
Gohlke P, von KS, Jurgensen T, Kox T, Rascher W, Culman J, Unger T. Effects of orally applied candesartan cilexetil on central responses to angiotensin II in conscious rats. J Hypertens 20: 909-918, 2002.  ?  z# u9 W6 J( R
( F5 X" W; r3 X/ Y0 z9 n) \

* C9 o) T. p. r" d# g; _' h
( C; [' y) y7 s7 B/ n  |* aHarrison-Bernard LM, Navar LG, Ho MM, Vinson GP, el-Dahr SS. Immunohistochemical localization of ANG II AT1 receptor in adult rat kidney using a monoclonal antibody. Am J Physiol Renal Physiol 273: F170-F177, 1997.0 \) B5 l& Y6 Z

$ E/ }  z8 }2 E* T4 v0 s6 K8 J+ Z4 G- u7 u
+ |2 o  F; Z: G8 g' y2 c" p
Inscho EW, Imig JD, Deichmann PC, Cook AK. Candesartan cilexetil protects against loss of autoregulatory efficiency in angiotensin II-infused rats. J Am Soc Nephrol 10, Suppl 11: S178-S183, 1999.5 x2 a0 J7 c" s& o% H4 b2 S

- ]4 D9 v% n% ?( a% g
, Y: N; C! }' k, \2 _# E1 m+ H3 H3 ~: {2 `% h) D- S8 k
Ishidoya S, Morrissey J, McCracken R, Reyes A, Klahr S. Angiotensin II receptor antagonist ameliorates renal tubulointerstitial fibrosis caused by unilateral ureteral obstruction. Kidney Int 47: 1285-1294, 1995.
/ c2 L  Z( ^/ `/ r$ u# p5 u, y" E; d; R
4 ^4 c! `' @" x; T3 O; f# \
" \" z* s1 }$ r2 H% @% y4 f
Jensen AM, Li C, Praetorius HA, Norregaard R, Frische S, Knepper MA, Nielsen S, Frøkiær J. Angiotensin II mediates downregulation of aquaporin water channels and key renal sodium transporters in response to urinary tract obstruction. Am J Physiol Renal Physiol 291: F1021-F1032, 2006.
0 t6 Z, M, _3 f) \% z" F  z
+ m- k( F( J2 S0 Z; w. }# e7 `. B0 ~( O3 B0 n  N0 v
7 v) |/ _' [, L5 D6 _: v
Kashgarian M, Biemesderfer D, Caplan M, Forbush B III. Monoclonal antibody to Na,K-ATPase: immunocytochemical localization along nephron segments. Kidney Int 28: 899-913, 1985." O* f) ^8 M% Q

7 Y0 C$ E: D9 v- L2 w! t, p; S9 j* o6 j0 z4 O* x

2 s( K1 n' r; B; I9 IKoff SA. Pathophysiology of ureteropelvic junction obstruction. Clinical and experimental observations. Urol Clin North Am 17: 263-272, 1990.
, F' @9 E) i, N, |
3 p/ _" U5 O0 y# D8 r* i9 R7 _5 o- W' A; o; z3 p: H

7 d/ c2 k& q+ ^- F+ cLi C, Wang W, Knepper MA, Nielsen S, Frøkiær J. Downregulation of renal aquaporins in response to unilateral ureteral obstruction. Am J Physiol Renal Physiol 284: F1066-F1079, 2003.
* N6 T  T- Q! Z0 ^+ `
. c1 e, G$ {; k8 {! l& G' o' [, O' _7 \: v

( a# y7 d( l' k+ D3 R! u4 L' y( [Li C, Wang W, Kwon TH, Isikay L, Wen JG, Marples D, Djurhuus JC, Stockwell A, Knepper MA, Nielsen S, Frøkiær J. Downregulation of AQP1, -2, and -3 after ureteral obstruction is associated with a long-term urine-concentrating defect. Am J Physiol Renal Physiol 281: F163-F171, 2001.
7 E$ b' C- X+ S) }
" r2 G4 r( B  U. w6 Y! v& B- B# I
7 q( V* j) x8 Z) }2 R
' b9 @# [5 F% m8 J6 G: VLi C, Wang W, Kwon TH, Knepper MA, Nielsen S, Frøkiær J. Altered expression of major renal Na transporters in rats with unilateral ureteral obstruction. Am J Physiol Renal Physiol 284: F155-F166, 2003.
- i# r2 j! v  ?6 d! {
2 K5 F# k/ b* \( j; U2 p4 u( i0 u  U/ D3 [% ~& ]# l
+ A: o' N' S5 R% b% P  r  a
Li C, Wang W, Kwon TH, Knepper MA, Nielsen S, Frøkiær J. Altered expression of major renal Na transporters in rats with bilateral ureteral obstruction and release of obstruction. Am J Physiol Renal Physiol 285: F889-F901, 2003.
+ E. j7 m) W" u: A7 W; R( m* J3 h0 Q: V; A2 Z  ~9 n" u& a

, u; r5 J4 _0 h8 {8 w( s/ L/ d( A+ r( Z
Manucha W, Carrizo L, Ruete C, Molina H, Valles P. Angiotensin II type I antagonist on oxidative stress and heat shock protein 70 (HSP 70) expression in obstructive nephropathy. Cell Mol Biol (Noisy -le-grand) 51: 547-555, 2005.9 @7 M% V/ h6 }9 B

& I% Z$ w- }: D6 S# m  H) b3 _
+ v1 z; f; x9 f# {/ b* y% ~' G5 t- z; l) ]9 U
Manucha W, Oliveros L, Carrizo L, Seltzer A, Valles P. Losartan modulation on NOS isoforms and COX-2 expression in early renal fibrogenesis in unilateral obstruction. Kidney Int 65: 2091-2107, 2004.
9 _/ U! N8 C( [9 f  \" [- I0 r9 @6 M: `4 r9 f1 @, q' H* l

$ V: u3 r7 O) P( D- @$ l$ }0 ]% A- c8 [7 l3 T
Matsuo T, Ishikawa E, Ohta M, Shibouta Y, Ishimura Y, Imura Y, Sugiyama Y. Renal protective effect of candesartan cilexetil in spontaneously hypercholesterolemic rats. Jpn J Pharmacol 88: 300-306, 2002.
' ^+ D  h2 ~# c+ v  Z& E6 K" W4 O4 L& N$ u, Z. H
; ^8 K* f- a" _# n% s  r: M* [1 T
$ [# p2 f! U" w, P
Misseri R, Meldrum KK. Mediators of fibrosis and apoptosis in obstructive uropathies. Curr Urol Rep 6: 140-145, 2005.
7 `/ a. E6 N( \) y5 {! D
* V% `+ {+ ?) }6 q% g5 J( W5 c- _% u
2 F. a1 M8 I' L  J8 v7 \
Miyajima A, Kosaka T, Seta K, Asano T, Umezawa K, Hayakawa M. Novel nuclear factor kappa B activation inhibitor prevents inflammatory injury in unilateral ureteral obstruction. J Urol 169: 1559-1563, 2003.
2 V. l: |  ~: i8 H( a! b% W$ I7 l1 F& c: E5 x: Y
: L! {$ i1 C2 Q$ m$ R8 u% z
& i; m7 h* k8 k0 ?" l
Moriyama T, Kawada N, Akagi Y, Ando A, Horio M, Yamauchi A, Nagata K, Imai E, Hori M. TCV-116 inhibits interstitial fibrosis and HSP47 mRNA in rat obstructive nephropathy. Kidney Int Suppl 63: S232-S235, 1997.$ o2 E% i7 W8 \7 x

% @' S3 V* r" H9 L7 r- o( Y5 j
: s! X5 G! V1 t" M& v5 x( |6 c9 `2 L) W/ J) s0 Y$ h3 b9 b8 F
Morrissey JJ, Klahr S. Enalapril decreases nuclear factor kappa B activation in the kidney with ureteral obstruction. Kidney Int 52: 926-933, 1997.( F/ _8 y: r3 K6 M: j

  \7 q5 a/ {0 f% h/ F: p
7 X6 R* I& r  C9 Z0 [8 {9 ~& E; z# ]# U/ @  ?: p
Mozaffari MS, Patel KB, Schaffer SW. Renoprotective effects of chronic candesartan treatment in uninephrectomized rat. J Cardiovasc Pharmacol 41: 81-88, 2003." p# \4 @2 Z9 D5 p" {
4 G) p" D% {4 {% [7 U
$ E- c2 x# G, t0 u+ `, P

/ s- R+ l3 ?6 w- y; ANakamura F, Nagano M, Kobayashi R, Higaki J, Mikami H, Kawaguchi N, Onishi S, Ogihara T. Chronic administration of angiotensin II receptor antagonist, TCV-116, in cardiomyopathic hamsters. Am J Physiol Heart Circ Physiol 267: H2297-H2304, 1994.0 U0 H6 S, {, n- O8 J0 d

- c) n0 b) m: c7 P/ k& B) o
3 E8 w4 n$ |: H8 H. N/ Y* [) }5 {" |# f; [8 t1 m- S  R/ n
Nielsen S, Frøkiær J, Marples D, Kwon TH, Agre P, Knepper MA. Aquaporins in the kidney: from molecules to medicine. Physiol Rev 82: 205-244, 2002.+ k- u, ?0 I4 C5 U9 S5 B1 }

4 t7 l9 D5 \  ~  k) O' I7 Y* S, A- P/ _/ u7 G
9 w$ [: @0 K! o: V9 c0 E! j
Nielsen S, Kwon TH, Christensen BM, Promeneur D, Frøkiær J, Marples D. Physiology and pathophysiology of renal aquaporins. J Am Soc Nephrol 10: 647-663, 1999.+ I& `1 z$ n  q

) D0 A0 q' n+ Z& E
' K0 T# L3 S: l: @* q
' H9 r+ }8 ?0 S9 g4 r: g( B4 J" tNielsen S, Marples D, Frøkiær J, Knepper M, Agre P. The aquaporin family of water channels in kidney: an update on physiology and pathophysiology of aquaporin-2. Kidney Int 49: 1718-1723, 1996.) g, b& J1 r) y" x" l' H6 P! m

  q5 I  l1 d; B; h8 a- w8 d, k3 O$ Q' [; }8 Y
. ^" |$ ^1 A8 d( d# @7 S# @  q3 e
Noda M, Matsuo T, Fukuda R, Ohta M, Nagano H, Shibouta Y, Naka T, Nishikawa K, Imura Y. Effect of candesartan cilexetil (TCV-116) in rats with chronic renal failure. Kidney Int 56: 898-909, 1999.
; k) I2 ^# L$ o" b! [7 [: o7 e5 I0 A  }6 b* f
8 d; T+ L6 Q- c0 ?* n6 y6 D

9 a7 h  l( ^  D5 z4 [9 nOliverio MI, Madsen K, Best CF, Ito M, Maeda N, Smithies O, Coffman TM. Renal growth and development in mice lacking AT 1A receptors for angiotensin II. Am J Physiol Renal Physiol 274: F43-F50, 1998.$ f' X! [- t" b6 s, B& h

: T3 p% K$ H) E* f
6 k* n6 a1 h) m% y1 J4 F+ V$ i. J/ j: g7 l8 Y5 p
Phifer CB, Terry LM. Use of hypothermia for general anesthesia in preweanling rodents. Physiol Behav 38: 887-890, 1986.
5 h! P5 H) m7 @
  y5 p+ M- b! w6 h4 b$ o2 \$ n6 [4 a5 c+ v/ p" u
5 U5 i  J# o$ e; l
Pimentel JL Jr, Martinez-Maldonado M, Wilcox JN, Wang S, Luo C. Regulation of renin-angiotensin system in unilateral ureteral obstruction. Kidney Int 44: 390-400, 1993.
/ t" |8 d1 v' I1 D
: J1 v! |# {* f8 v
% K. w: J% I1 U6 t  P4 p3 @  \; V/ [* |* W: W* ^* O4 B7 H+ `% Q
Shi Y, Li C, Thomsen K, Jorgensen TM, Knepper MA, Nielsen S, Djurhuus JC, Frøkiær J. Neonatal ureteral obstruction alters expression of renal sodium transporters and aquaporin water channels. Kidney Int 66: 203-215, 2004., {. I6 Z& v5 Q& Y: o6 Z. U1 X9 z
4 f4 I) T  t( {* [3 z

$ T9 ?: j6 ]& N, M4 O  o' p$ x$ M  [+ M1 |) B7 y& V: y
Shi Y, Pedersen M, Li C, Wen JG, Thomsen K, Stodkilde-Jorgensen H, Jorgensen TM, Knepper MA, Nielsen S, Djurhuus JC, Frøkiær J. Early release of neonatal ureteral obstruction preserves renal function. Am J Physiol Renal Physiol 286: F1087-F1099, 2004.8 g1 l1 j% T1 f: v* B9 r" f
" h9 l  g/ f- N  l  ~# f

  w! f5 _1 ^/ p* K0 k8 t5 ~# k& d/ y; \
Terris J, Ecelbarger CA, Nielsen S, Knepper MA. Long-term regulation of four renal aquaporins in rats. Am J Physiol Renal Fluid Electrolyte Physiol 271: F414-F422, 1996.
' [2 H. j7 x5 M# t+ w9 f% b; j& {' f) o' \2 n5 @* F8 L+ i; {8 |6 D
' w9 D; c) A; N+ j5 Z0 V
7 m- @% U, W+ z/ \2 q$ d8 r/ `
Thornhill BA, Burt LE, Chen C, Forbes MS, Chevalier RL. Variable chronic partial ureteral obstruction in the neonatal rat: a new model of ureteropelvic junction obstruction. Kidney Int 67: 42-52, 2005.
, c, t+ g% `* b9 `  Q8 s* Z1 n1 i- Y0 g1 d

6 ^1 a3 O' G- l8 b4 r' b- V7 P! T) \/ Y) b9 j
Ulm AH, Miller F. An operation to produce experimental reversible hydronephrosis in dogs. J Urol 88: 337-341, 1962.
9 O% k& _8 v0 p# I8 Y8 I
" G# N: q/ [- _& }* c* J0 K: ?& X7 @; {6 B3 ]

+ A3 p/ Y% ~) ]9 f, y0 jWamsley-Davis A, Padda R, Truong LD, Tsao CC, Zhang P, Sheikh-Hamad D. AT 1A -mediated activation of kidney JNK1 and SMAD2 in obstructive uropathy: preservation of kidney tissue mass using candesartan. Am J Physiol Renal Physiol 287: F474-F480, 2004.
" }, r, X5 Q3 y  H; s0 M2 i
3 ^- n' o8 D9 a9 _
+ {) V% s$ M. h  s4 h) z: G- |0 E% b; u2 Q( W7 Q
Wen JG, Chen Y, Frokiaer J, Jorgensen TM, Djurhuus JC. Experimental partial unilateral ureter obstruction. I. Pressure flow relationship in a rat model with mild and severe acute ureter obstruction. J Urol 160: 1567-1571, 1998.
+ |; P- X- G) x0 x8 y1 i- ?
4 L+ |* d" K* j( {& A6 f( B0 Y5 D. a7 H( ~
4 C4 ~" ~4 ]6 \2 ?8 r1 f
Wen JG, Ringgaard S, Frøkiær J, Jorgensen TM, Stodkilde-Jorgensen H, Djurhuus JC. Contralateral compensatory kidney growth in rats with partial unilateral ureteral obstruction monitored by magnetic resonance imaging. J Urol 162: 1084-1089, 1999.  B3 s% H( N# g& C
5 T1 f; I5 o* Z+ z! h. e

/ |3 B6 U: H/ F1 x5 o: r. o8 o! A0 t# U* x: M
Wen JG, Ringgaard S, Jorgensen TM, Stodkilde-Jorgensen H, Djurhuus JC, Frøkiær J. Long-term effects of partial unilateral ureteral obstruction on renal hemodynamics and morphology in newborn rats: a magnetic resonance imaging study. Urol Res 30: 205-212, 2002.
作者: 杏花    时间: 2015-6-16 11:17

问渠哪得清如许,为有源头活水来。  
作者: 我心飞翔    时间: 2015-7-19 16:43

肿瘤干细胞
作者: nauticus    时间: 2015-7-26 16:18

楼主福如东海,万寿无疆!  
作者: 张佳    时间: 2015-7-27 16:43

肿瘤干细胞
作者: 龙水生    时间: 2015-8-6 11:54

我帮你 喝喝  
作者: laoli1999    时间: 2015-8-15 12:06

帮你项项吧  
作者: aakkaa    时间: 2015-8-28 15:35

很好!很强大!  
作者: tempo    时间: 2015-9-5 16:43

希望可以用些时间了~````  
作者: 石头111    时间: 2015-9-15 11:10

干细胞治疗  
作者: 剑啸寒    时间: 2015-11-19 18:35

哦...............  
作者: 科研人    时间: 2015-12-28 09:01

(*^__^*) 嘻嘻……  
作者: 昕昕    时间: 2016-1-6 08:18

进行溜达一下  
作者: 科研人    时间: 2016-1-10 18:35

支持~~  
作者: IPS干细胞    时间: 2016-3-1 16:27

人气还要再提高  
作者: tempo    时间: 2016-3-17 10:27

设置阅读啊  
作者: xm19    时间: 2016-4-2 17:53

几头雾水…  
作者: dongmei    时间: 2016-4-12 09:27

我回不回呢 考虑再三 还是不回了吧 ^_^  
作者: happyboy    时间: 2016-5-3 21:01

干细胞研究还要面向临床
作者: hmhy    时间: 2016-5-26 10:17

好啊,,不错、、、、  
作者: hmhy    时间: 2016-6-14 19:10

淋巴细胞
作者: 龙水生    时间: 2016-6-17 20:23

终于看完了~~~  
作者: 红旗    时间: 2016-6-28 21:35

拿把椅子看表演
作者: 草长莺飞    时间: 2016-7-9 17:00

终于看完了~~~  
作者: dongmei    时间: 2016-8-6 08:43

彪悍的人生不需要解释。  
作者: 983abc    时间: 2016-8-21 20:08

我起来了 哈哈 刚才迷了会  
作者: 兔兔    时间: 2016-9-4 19:01

我帮你 喝喝  
作者: lalala    时间: 2016-9-20 12:27

表观遗传学
作者: 123456zsz    时间: 2016-10-11 19:54

呵呵,支持一下哈  
作者: 丸子    时间: 2016-10-19 17:27

严重支持!
作者: 小丑的哭泣    时间: 2016-10-27 21:01

一个人最大的破产是绝望,最大的资产是希望。  
作者: 若天涯    时间: 2016-12-15 22:18

ding   支持  
作者: 生科院    时间: 2017-1-1 11:54

顶下再看  
作者: dr_ji    时间: 2017-1-6 11:25

长时间没来看了 ~~  
作者: dmof    时间: 2017-1-10 12:01

真的有么  
作者: Whole    时间: 2017-1-13 22:26

我帮你 喝喝  
作者: 修复者    时间: 2017-1-18 09:10

一定要回贴,因为我是文明人哦  
作者: DAIMAND    时间: 2017-1-19 20:54

活着,以死的姿态……  
作者: youngcell    时间: 2017-1-31 14:27

谢谢分享了!   
作者: 榴榴莲    时间: 2017-2-7 22:10

很有吸引力  
作者: 小敏    时间: 2017-2-10 03:17

顶也~  
作者: IPS干细胞    时间: 2017-3-12 18:52

回复一下  
作者: sshang    时间: 2017-3-15 07:05

这贴?不回都不行啊  
作者: biobio    时间: 2017-3-26 23:22

我等你哟!  
作者: foxok    时间: 2017-4-1 02:33

干细胞我这辈子就是看好你
作者: highlight    时间: 2017-4-17 00:53

我也来顶一下..  
作者: 追风    时间: 2017-4-30 23:43

好啊,谢楼主
作者: 张佳    时间: 2017-6-2 04:20

生殖干细胞
作者: qibaobao    时间: 2017-7-9 02:31

不对,就是碗是铁的,里边没饭你吃啥去?  
作者: beautylive    时间: 2017-8-21 03:22

干细胞之家是不错的网站
作者: chinagalaxy    时间: 2017-8-21 23:27

不错,看看。  
作者: tuanzi    时间: 2017-8-23 02:07

神经干细胞
作者: IPS干细胞    时间: 2017-9-3 08:40

这贴子你会收藏吗  
作者: 小小C    时间: 2017-9-22 17:43

我好想升级  
作者: 未必温暖    时间: 2017-9-22 23:25

鉴定完毕.!  
作者: dd赤焰    时间: 2017-9-29 01:05

顶也~  
作者: laoli1999    时间: 2017-10-24 05:00

哈哈 瞧你说的~~~  
作者: 老农爱科学    时间: 2017-11-9 22:07

神经干细胞
作者: 修复者    时间: 2017-11-17 01:01

呵呵 都没人想我~~  
作者: pspvp    时间: 2017-11-17 10:54

勤奋真能造就财富吗?  
作者: sshang    时间: 2017-11-18 07:24

在线等在线等  
作者: 365wy    时间: 2017-11-21 06:46

偶啥时才能熬出头啊.  
作者: 水木清华    时间: 2017-11-26 07:38

我的啦嘿嘿  
作者: keanuc    时间: 2017-12-15 23:34

今天没事来逛逛  
作者: nosoho    时间: 2017-12-20 18:18

ips是诱导多能干细胞induced pluripotent stem cells iPS
作者: whyboy    时间: 2018-1-13 04:26

不错啊! 一个字牛啊!  
作者: 若天涯    时间: 2018-1-17 06:57

看完了这么强的文章,我想说点什么,但是又不知道说什么好,想来想去只想  
作者: 安生    时间: 2018-2-6 20:39

我等你哟!  
作者: marysyq    时间: 2018-2-8 07:53

干细胞分化技术
作者: renee    时间: 2018-2-9 17:30

顶下再看  
作者: xm19    时间: 2018-2-14 23:27

哈哈 我支持你
作者: 狂奔的蜗牛    时间: 2018-2-22 09:27

这个贴好像之前没见过  
作者: 张佳    时间: 2018-2-25 10:10

拿把椅子看表演
作者: feixue66    时间: 2018-3-20 18:02

很好!很强大!  
作者: 王者之道    时间: 2018-3-28 20:18

帮你顶,人还是厚道点好  
作者: aliyun    时间: 2018-4-13 02:59

帮你顶,人还是厚道点好  
作者: www1202000    时间: 2018-4-30 20:01

真的有么  
作者: tempo    时间: 2018-5-10 23:59

好啊,,不错、、、、  
作者: 983abc    时间: 2018-5-15 03:32

任何的限制,都是从自己的内心开始的。  
作者: sshang    时间: 2018-6-1 16:54

也许似乎大概是,然而未必不见得。  
作者: biopxl    时间: 2018-6-14 07:10

似曾相识的感觉  
作者: abc987    时间: 2018-6-27 22:15

祝干细胞之家 越办越好~~~~~~~~~`  
作者: 草长莺飞    时间: 2018-7-4 08:00

说的真有道理啊!
作者: IPS干细胞    时间: 2018-7-12 20:55

既然来了,就留个脚印  
作者: 黄山    时间: 2018-8-12 17:13

挤在北京,给首都添麻烦了……  
作者: 罗马星空    时间: 2018-8-16 11:18

哈哈,看的人少,回一下  
作者: na602    时间: 2018-8-27 14:11

自己知道了  
作者: 安生    时间: 2018-9-8 02:40

怎么就没人拜我为偶像那?? ~  
作者: lab2010    时间: 2018-9-24 01:27

一楼的位置好啊..  
作者: 小倔驴    时间: 2018-10-14 04:34

说的不错  
作者: 心仪    时间: 2018-10-30 15:53

有空一起交流一下  
作者: 墨玉    时间: 2018-10-30 21:23

楼主good  
作者: kaikai    时间: 2018-11-1 00:34

造血干细胞
作者: doc2005    时间: 2018-11-2 16:33

我回不回呢 考虑再三 还是不回了吧 ^_^  
作者: foxok    时间: 2018-11-28 12:10

长时间没来看了 ~~  
作者: s06806    时间: 2018-12-6 19:53

顶也~  
作者: whyboy    时间: 2018-12-8 12:10

希望大家帮我把这个帖发给你身边的人,谢谢!  
作者: doors    时间: 2018-12-11 03:36

其实回帖算是一种没德德,所以我快成圣人了  
作者: www1202000    时间: 2018-12-17 12:26

初来乍到,请多多关照。。。  
作者: youngcell    时间: 2018-12-25 08:35

初来乍到,请多多关照。。。  




欢迎光临 干细胞之家 - 中国干细胞行业门户第一站 (http://stemcell8.cn/) Powered by Discuz! X1.5