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: 419071|回复: 239
go

Renal effects of Tamm-Horsfall protein (uromodulin) deficiency in mice [复制链接]

Rank: 1

积分
威望
0  
包包
0  
楼主
发表于 2009-4-21 13:05 |只看该作者 |倒序浏览 |打印
Medical Faculty of the Charité, Department of Anatomy, Nephrology/Hypertension, Franz Volhard Clinic, HELIOS Clinics and Max Delbrück Center for Molecular Medicine, and Department of Physiology, Berlin, Germany
  E, O" M$ R/ W7 _
+ O# {+ _2 i. {% m5 YUniversity of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma0 u* v$ e. A* Z$ y* p8 k2 h+ Z6 g

( L; \. d' v! }/ S( q1 t3 z- l3 JABSTRACT
) o' l% v' b& x. _4 s% H8 C
/ n. n8 C) ~' b$ s/ g( SThe Tamm-Horsfall protein (THP; uromodulin), the dominant protein in normal urine, is produced exclusively in the thick ascending limb of Henle's loop. THP mutations are associated with disease; however, the physiological role of THP remains obscure. We generated THP gene-deficient mice (THP –/–) and compared them with wild-type (WT) mice. THP –/– mice displayed anatomically normal kidneys. Steady-state electrolyte handling was not different between strains. Creatinine clearance was 63% lower in THP –/– than in WT mice (P ! p9 @9 u6 O# k: e+ t# }- ^
9 v1 ?9 n+ u/ g; v3 _' ?
gene-disrupted mice; thick ascending limb; renal concentrating mechanism
. O* h1 Y/ a4 R9 c5 T3 r* }% M. n/ r  S; ?3 y6 `
THE TAMM-HORSFALL PROTEIN (THP; uromodulin) is an 80-kDa glycoprotein synthesized exclusively in the thick ascending limb cells of Henle's loop (TAL) with exception of the macula densa (3, 4, 41). THP is the most abundant protein in the urine of normal mammals. THP production ranges from 30 to 60 mg/24 h in humans and from 0.3 to 0.5 mg﹞100 g body wt–1﹞24 h–1 in rats (2). Intracellularly, the protein resides along the basolateral membranes, in the subapical vesicle compartment, and to a lesser extent in or near the luminal plasma membrane (3, 38). Exocytosis and luminal secretion of THP are driven by a glycosylphosphatidylinositol (GPI) membrane anchor from which THP is cleaved and released into the tubular fluid (22). THP contains a COOH-terminal zona pellucida domain that allows the protein to polymerize and facilitates its gel-forming mucoid tendency (41). Interaction of THP with other proteins has thus far been reported only at the extracellular level and probably depends on the abundant oligosaccharide moieties of THP (48).. y7 @& \% [" u' ?
' u8 ~* c) }3 }- l; ~, c' F
THP may be involved in the pathogenesis of cast nephropathy, urolithiasis, and tubulointerstitial nephritis (34, 36, 41, 49). The THP gene is mutated in persons with familial juvenile hyperuricemic nephropathy and autosomal dominant medullary cystic kidney disease, suggesting a role for the protein in renal urate handling and perhaps in renal development (9, 15, 31, 45). In affected persons, THP accumulates in the TAL epithelia, apparently owing to the retention of mutated THP in the endoplasmic reticulum and in a delayed export of the protein to the plasma membrane (31). Increased proximal tubular reabsorption of uric acid has been postulated possibly due to a decrease in tubular NaCl reabsorption in Henle's loop (9). Changes in the quantity of THP synthesis or urinary excretion have been assessed in pathological conditions such as diabetic nephropathy, polycystic kidney disease, and lupus nephritis (21, 32, 44). TAL transporter mutations leading to the antenatal variant of Bartter's syndrome feature a near complete to absolute deficiency of cellular and urinary THP production (16, 30, 40). THP has therefore been proposed as a renal disease marker (49). The fact that THP competes efficiently with urothelial cell receptors, such as uroplakins, in adhering to bacteria suggests that THP may protect the organism from ascending urinary tract infection (29). The THP –/– mouse indeed confirmed this hypothesis by demonstrating that THP serves as a soluble receptor for type 1 fimbriated Escherichia coli and helps to eliminate bacteria from the urinary tract (5, 24).& {$ `1 e* N% }. `5 B! U) \" H

" O6 V: |+ ^8 J7 ZTHP may also play a role in regulating renal function. Hypothyroid rats revealed a decreased THP synthesis along with impaired TAL function and enhanced vasopressin availability (38). Vasopressin reduces THP production (2, 18), whereas dietary salt loading and furosemide administration increase renal THP mRNA expression (47) but not urinary THP excretion (2). To test the notion that THP plays a regulatory role in renal salt and water excretion, we performed functional and morphological studies in THP gene-deficient (–/–) mice and their wild-type (WT;  / ) siblings.( b" K) r9 e0 w" [7 V

5 h1 y! a* x0 X+ Y( dMATERIALS AND METHODS& Q! M% V/ n# }, R6 N' P# [! o: ]

* Y4 e+ w5 T. k# ]: ~/ ^" _8 n. p2 AAnimals. THP –/– and WT mice have been described (5). All mice were genotyped from tail tip-extracted DNA using polymerase chain reaction primers: 5'-TTGGGAAGACAATAGCAGGC-3' (neomycin resistence gene) and 5'-GCAAATGCAGTATGACCGCC-3' (THP intron) or the WT allele: 5'-ATGTGGATGAGTGCTCAGAG-3' (THP coding sequence) and 5'-GCAAATGCAGTATGACCGCC-3' (THP intron). The animals received standard mouse chow (SNIFF) and were allowed drinking water ad libitum. The local council on animal care approved the protocols. The standards correspond to the requirements of the American Physiological Society. For the physiological studies, routine parametric descriptive statistics were used. Groups were compared with two-way ANOVA and t-tests, Bonferroni corrected as appropriate. We also used t-tests to compare gene expression and Western blot quantification data. P
( `# O1 ?# B' z
6 X6 x4 B- H9 s$ f3 S- y: N4 pWater balance and creatinine clearance. THP –/– (total n = 14) and WT (n = 14) mice aged 8–13 wk were placed individually (10 AM) into metabolism cages for 24 h (Uno mouse metabolic cages, Uno, Zevenaar, The Netherlands). In the first protocol, we added sucrose (300 mmol/l) to the drinking water for 3 days. Mice received chow ad libitum. In the second protocol, mice received chow and water ad libitum for 2 days and on day 3, they were deprived of water but not chow. Urine volume, osmolality (freezing point depression), sodium, chloride, potassium, creatinine, uric acid, and total protein were determined by routine automated methods. The urine samples were checked for the presence of THP by Western blotting. Thereafter, the animals were killed under pentobarbital sodium anesthesia, and serum specimens and kidneys were obtained. Inter- and intrastrain kidney-to-body weight ratios were calculated. Serum sodium, chloride, and creatinine levels were determined; creatinine was measured by a modified Jaffé reaction. The creatinine clearance was determined in 12-wk-old mice and normalized for kidney weight. Plasma renin activity (PRA) was determined as the amount of formed ANG I (8).
2 b2 w8 R( m  o/ o( ?! o% f6 Z4 ^  g2 _
' E! m; z$ h+ E. }RT-PCR and Northern blot analysis. For RNA preparation, kidneys were rapidly dissected, homogenized, and total RNA was prepared using the RNeasy-total-RNA-kit (Qiagen). To perform semiquantitative RT-PCR, genomic DNA was digested by DNase. cDNA was synthesized by reverse transcription of 5 μg total RNA, using a cDNA synthesis kit (Invitrogen). Primers used for amplification of specific gene products are listed (Table 1). RT-PCR reactions were carried out in an automated thermal cycler (PerkinElmer, Boston, MA) using Taq polymerase (GIBCO). The number of cycles (between 23 and 35) was adapted for linear correlation between signal intensities of RT-PCR products and cDNA concentration. cDNA was adjusted to equal quantities by serial dilutions within linear range of these parameters. Reactions were controlled by PCR amplification of the housekeeping gene GAPDH. For Northern blot analysis, equal amounts of total RNA (30 μg per kidney as determined by UV at OD260 with Gene-Quant, Bio-Rad) were electrophoresed under denaturing conditions in a 1% agarose-formaldehyde gel at 40 V for 5 h. The separated RNA samples were then transferred in 10x SSC for 12 to 16 h onto a nylon membrane (Schleicher & Schull) by capillary blotting and cross-linked using a UV radiation chamber (Gene-Linker; Bio-Rad). A 383-bp mouse (furosimide/bumetanide-sensitive) Na -K -2Cl– transporter type 2 (NKCC2) cDNA fragment cloned into a in pBS vector and a 450 bp -actin fragment were prepared for hybridization using asymmetric PCR. Probes were labeled with alkaline phosphatase (Alk Phos Systems and CDP Star reagent, Amersham); hybridization and chemiluminescence detection were done according to the manufacturer's protocol. After hybridization with the NKCC2 probe, the membrane was stripped in 0.5% (wt/vol) SDS at 60°C for 1 h and reprobed with the -actin cDNA probe used as an internal standard.
5 D' b1 |% n$ p3 Y5 ?) k8 {
+ O! s2 P% i6 HView this table:. Q: P3 p& N; z& }* U" k

3 ]4 m; Z7 ?" z8 rWestern blot analysis. Methods were applied as described previously (37). Briefly, whole kidney homogenates (n = 6 per group) as obtained after cold homogenization in a sucrose-triethanolamine buffer using a tissue homogenizer were centrifuged at 1,000 g for 15 min at 4°C to remove whole cells, nuclei, mitochondria, and incompletely homogenized membrane fragments, and the supernatants were prepared for immunoblotting. Supernatants were solubilized, and 50 μg protein/lane as determined by a BCA protein assay reagent kit (Pierce) were run on 8 or 10% polacrylamide minigels. After electrophoretic transfer of the proteins to polyvinylidene fluoride membranes, equity in protein loading and blotting was verified by membrane staining using 0.1% Ponceau red. Coomassie staining was also performed to verify the equity in protein loading. Membranes were incubated with specific primary antibodies (Table 2) for 1 h at room temperature, following overnight incubation at 4°C, and then exposed to adequate horseradish peroxidase-conjugated secondary antibodies for 2 h at room temperature. Immunoreactive bands were detected by chemiluminescence, subsequently exposed to X-ray films, and the films were scanned and densitometrically evaluated.
/ I% {/ Q; p; Q3 }9 l( H1 p( z/ `# }8 l0 C! R/ J
View this table:
# K! N9 K3 \* u* b$ n; @. w9 N
. w+ J$ M0 ^4 v+ qTissue preparation for morphology. For morphological evaluation, male WT and THP –/– mice of different ages (total n = 12 per strain, age range 2 to 14 mo) were used. Mice were killed by in vivo perfusion fixation under pentobarbital sodium anesthesia. The kidneys were perfused retrogradely through the abdominal aorta using PBS adjusted to 330 mosmol/kgH2O with sucrose, pH 7.4, for 20 s. Next, 3% paraformaldehyde in PBS was infused for 5 min. The kidneys were removed and cut into slices. Slices were processed for embedding in Epon, LR white hydrophilic resin, or paraffin, or shock-frozen in liquid nitrogen-cooled isopentane for subsequent cryostat sectioning.
; V% U& t8 ~( N( I! f4 v  X7 b/ {5 l8 E
In situ hybridization. In situ hybridization was performed on perfusion-fixed, paraffin-embedded, or frozen tissue as described (38). In brief, digoxygenin (DIG)-11-UTP-labeled riboprobes were synthesized by in vitro transcription (DIG RNA labeling kit [Sp6/T7]; Roche) using a 492-bp fragment from mouse THP cDNA (between position 1170 and 1662) cloned into a pCRII-TOPO vector, a 300-bp rat renin cDNA fragment cloned into a pGEM3 vector, and the 383-bp mouse NKCC2 cDNA fragment applied for Northern blotting. After linearization with the appropriate restriction enzymes, sense or antisense riboprobes were generated with T7 or SP6 RNA polymerases. For in situ hybridization, 6-μm-thick paraffin sections were treated with proteinase K, hybridized for 18 h with a probe concentration of 2.5 ng/μl of hybridization mix at 40°C, rinsed, and incubated with sheep anti-DIG-alkaline phosphatase-conjugated antibody (DAKO) diluted 1:50 in blocking medium. Signal was generated using 4-nitroblue tetrazolium chloride. For control, sense probes were applied in parallel with antisense probes. Slides were rinsed with PBS, coverslipped with PBS-glycerol, and viewed in brightfield microscopy.' y3 |) j* [+ e; @' C

4 c  v: H" `& M' C) H  t( oImmunohistochemistry. For immunostaining, tissue sections were treated with 0.5% Triton X-100 in PBS for 30 min, washed in PBS, and blocked with 5% milk powder in PBS for 1 h at room temperature. Sections were then incubated with primary antibodies as listed (Table 2), diluted in blocking solution for 1 h at room temperature, followed by overnight incubation at 4°C. After being washed with PBS, appropriate secondary Cy3-conjugated (DIANOVA) or horseradish peroxidase-conjugated antibodies (DAKO) were incubated for 2 h. Slides were then rinsed in PBS, coverslipped with PBS-glycerol, and viewed with a Leica DMRB light microscope equipped with interference contrast optics and an HBO fluorescence lamp. Light microscopic images were obtained with a digital camera (Spot 32, Diagnostic Instruments) and processed with Meta View 3.6a software (Universal Imaging).
' d# L0 `1 U) U+ b* g9 y  M: T  d3 X) j" e" E; Z
Ultrastructural analysis. Ultrathin sections were viewed with an electron microscope. For ultrastructural immunogold localization of THP, LR white-embedded tissue was sectioned and incubated according to an established protocol (3).
# g! A% r+ Z7 ]0 V! l; v9 R
* s$ V- a) e! R2 S3 z: _6 {+ _NADPH-diaphorase reaction. For histochemical demonstration of nitric oxide synthase (NOS) tissue activity, the NADPH-diaphorase reaction was performed as described (6).
) ~7 P  }8 Z' d) z% q9 j% h, p2 i: l# ?( X1 S
Quantification of NOS, cyclooxygenase-2, and renin content. Cells of the macula densa and adjacent TAL portions that were histochemically positive for NADPH-diaphorase staining, NOS1-positive cells, or cyclooxygenase-2 (COX-2)-positive cells were counted, and renin-immunoreactive and renin mRNA-expressing arteriolar portions were quantified as detailed previously (6). Tissues from n = 4 mice per strain were used, and a total of eight coronary sections were evaluated for each parameter.
- V+ J! ^6 ?0 E0 w* `$ R! m% f5 @: @% {, G$ a+ `: L
RESULTS
4 ^( \: Y! h7 W& i0 @" C
+ ?7 ?  y9 U6 C9 jPhysiological parameters. No significant differences between THP –/– and WT mice were observed in serum electrolyte, liver function, and lipid values at 3 mo. The absence of THP did not significantly interfere with water consumption, urine flow, osmolar excretion, sodium, potassium, and uric acid excretion between strains (Fig. 1 and Table 3). Interstrain kidney-to-body weight ratios showed no significant differences. However, creatinine clearance was 63% lower in THP –/– than in WT mice (P
  n; O% C; ?, ?# i! y8 r6 y, z& H: x. f" H
View this table:3 }: q+ {5 D) D( U( {
, K, L6 z, J3 w4 a0 Y+ i6 a0 J
Renal morphology and THP staining. Kidneys and urinary tract epithelia were harvested from THP –/– and WT mice over an age range from 2 mo to over 1 yr. Tissues were screened for alterations induced by the gene disruption using light and electron microscopy and compared with age-matched WT controls. There were no obvious changes in the morphology of the renal tubules or the glomeruli. Sites of THP synthesis, i.e., the epithelium of the medullary and cortical TAL as well as tubular portions located downstream of TAL were apparently not altered by THP deficiency (Fig. 2). Ureter and bladder epithelia showed no major structural alterations either (not shown). Furthermore, there were no signs of epithelial or interstitial inflammation in THP –/– mice. Immunohistochemical staining revealed the total absence of THP from TAL epithelia in THP –/– compared with WT mice (Fig. 3, C and D). Fine structural immunogold THP staining of luminal and basolateral membranes, as well as of intracellular compartments of TAL cells, was absent (not shown). THP mRNA was also absent according to in situ hybridization (Fig. 3, A and B), although at high resolution an occasional remnant expression was evident. This finding may be due to nonfunctional, truncated mRNA still produced from the 3'-end of the targeted locus, probably owing to a partially ineffective stop site in the neo cassette of the transgenic construct. There was no generation of immunoreactive THP protein according to the immunohistochemical results using different polyclonal antibodies.8 |% p$ Y3 b4 N4 P8 n
# I: Q9 @: F  a+ Z8 ^
Major nephron transporters. We found significant increases in mRNA in THP –/– mice compared with WT mice for distal nephron transporters including -subunit of Na -K -ATPase (NKA;  36%), NKCC2 ( 33%), Na /H  exchange type 3 (NHE3;  83%), barttin ( 180%), K-type chloride channel (ClC-K2;  150%), ROMK ( 260%), (thiazide-sensitive) Na -Cl– cotransporter (NCC;  290%), and the -subunit of (amiloride-sensitive) epithelial Na  channel (ENaC;  80%) during steady state (Fig. 4). To confirm the increased NKCC2 mRNA expression as determined by semiquantitative RT-PCR, Northern blotting was used. An increase was found in THP –/– mice ( 25%; P
1 S* R: R, k& p9 r% t/ t2 V
" f# K8 m/ X$ T2 y& PJuxtaglomerular paracrine parameters. Semiquantitative evaluation of juxtaglomerular immunoreactive NOS1 and NADPH-diaphorase reactions revealed no strain differences during steady state (Figs. 6, E–H, and 7, A and B). COX-2 levels were significantly decreased in THP –/– mice compared with controls. Quantification of COX-2 mRNA by semiquantitative PCR from total kidney homogenate revealed a 51% decrease (P ( k# \$ d0 B+ F' E* ?
7 g! S& m& z$ Z
DISCUSSION) ]# R( c5 A0 T0 N
9 b0 l! U. Y* }: o& v; w
Our mouse model of THP deficiency revealed that kidneys were anatomically normal. This result implies that the only source of THP, namely, the TAL cells with their characteristic basolateral membrane interdigitation, densely packed flat mitochondria, and well-developed subapical vesicular compartment normally replete with immunoreactive THP, was left intact even in the absence of THP. Steady-state blood and urinary parameters were unchanged in THP –/– mice, even when fluid intake was increased by sucrose. However, three major findings were conspicuous. First, the glomerular filtration rate (GFR) in THP –/– mice as estimated by creatinine clearance was reduced. Second, the mice were less able to withstand water deprivation than WT mice. THP –/– mice could not concentrate their urine to the same degree under these circumstances and excreted higher urine volumes than the control mice. This increased urine output occurred without significantly elevated electrolyte losses. Third, the gene expression of paracrine/endocrine components of the juxtaglomerular apparatus and nephron-related ion transporters/channels was, in part, significantly altered in THP –/– mice, even at steady state.& h* X# ^% Y9 Y* G* o9 F/ ?4 d
  S6 t& c$ |3 Y2 j- i: R7 h' Z% T
The relationships of THP with ion transporters involved in the function of the TAL, chiefly with NKCC2, have been discussed in earlier studies (31, 38, 40). We speculated that more dramatic findings would be observed in THP –/– mice, such as NKCC2 dysfunction with a Bartter's-like syndrome, polyuria, dehydration, hypokalemia, nephrocalcinosis, and a stimulated prostaglandin synthesis (10, 16, 42). In Bartter's syndrome or after furosemide administration, the juxtaglomerular renin, COX-2, and NOS1 expression levels are strongly elevated along with high plasma renin concentrations (6, 14, 39). However, the phenotype we observed was much more innocuous than expected.8 K7 C5 d5 U, l. v6 r
1 _1 Q2 E- {, M  R; L7 ^, G" Y
Instead, we observed that in THP –/– mice COX-2 was lowered and renin-related parameters were reduced, whereas NOS1 expression/activity was unchanged. COX-2 normally shows a coordinate regulation with renin, which has given rise to earlier considerations regarding a functional relationship between renin expression and COX-2. In contrast to the effects of furosemide, an enhanced tubular NaCl load at the macula densa and subsequently stimulated NKCC2-dependent transport mechanisms are believed to be responsible for reduced juxtaglomerular COX-2 expression and decreased renin biosynthesis (14, 39, 43, 46). Because THP is physiologically absent from the macula densa and therefore cannot interfere with ion transporters at this site (3, 4), the macula densa in THP –/– mice would be expected to function normally. Thus a direct influence of THP deficiency on the local paracrine system of macula densa cells is not to be expected. However, in the TAL the absence of THP may well have an effect on transport. Intracellular THP functions have been postulated and relationships of THP with ion transporters involved in TAL transport, chiefly with NKCC2, have been discussed (31, 38, 40).# v* C( V/ j9 o% I3 Q
+ q) z$ H% B9 s! N2 b
We used biochemical and histochemical quantitative approaches to assess changes in the biosynthesis of products related to NaCl transport along the nephron in our mice (19, 25, 37). We found a significant upregulation of the major ion transporters/channels, namely the -subunit of NKA, NHE3, NKCC2, ClCK-2, the -subunit of ClC-K channels, barttin, ROMK, the -subunit of ENaC, and NCC in THP –/– mice compared with WT at steady-state conditions. Their segmental role and nephron distribution have been studied and reviewed extensively in previous studies (1, 7, 12, 20, 26–28, 33, 39).
. N* d: d5 O0 o1 E; L( G2 k. w+ x/ M' x+ w8 ?
Although somewhat difficult to reconcile with our functional data, an upregulation of TAL transporters in THP –/– mice may be related to the absence of THP. THP mutations in humans cause malfunction of the renal concentrating mechanism (31). Others and we found that NKCC2 and THP are both localized in detergent-resistant membrane domains (DRM, also termed lipid rafts) normally involved in apical trafficking events, and both products appear to interact with one another (13, 41). Possibly, the absence of THP reduces the efficiency of NKCC2 or related local products involved in transepithelial Na-K-Cl transport, thereby altering their biosynthesis rate. An altered TAL-reabsorptive capacity would be consistent with some of our functional findings. First, the concentrating defect with water deprivation suggests malfunctioning of the TAL in mice with THP deficiency. Second, a resulting increase in tubular ion load could, in turn, downregulate the juxtaglomerular mechanisms. The reduced COX-2 and renin mRNA levels thus may be indicative of an elevated NaCl load reaching the macula densa and probably also the distal convolutions. The significantly reduced GFR in THP –/– mice would further support this interpretation, namely that an enhanced load at the macula densa activates TGF, which in turn reduces GFR (39). The fact that electrolyte excretion was not different in THP –/– and WT mice is not a contraction as steady state was achieved and the medullary collecting duct presumably compensated for any differences in delivery.
3 a) q1 h$ g! ]% w' M. d( m$ q+ [. e! @: [5 p) k
TAL malfunction may also be related to upregulation of channels and transporters in the distal convolutions and collecting ducts in THP-deficient mice with altered solute reabsorption. We found upregulation of distal convoluted tubule-specific NCC in the distal convolutions. This cotransporter may have increased its biosynthesis rate in response to an enhanced solute load, as was observed previously at the mRNA level in chronically furosemide-treated rats (11, 27). In portions located further downstream, namely in the late distal convoluted tubule, the connecting tubule, and the cortical collecting duct, the observed upregulation of the -subunit of ENaC in THP knockouts may also be the result of altered tubular flow. Increased solute load has been shown to determine the expression of the channel (35). Of course, the higher expression levels may also depend on altered aldosterone release (23) that was not explored in our study. However, the suppression of the juxtaglomerular renin mRNA status we observed does not speak for increased mineralocorticoid availability. Taken together, there are signs of adaptive mechanisms in THP –/– mice during steady state that suggest an altered TAL-reabsorptive capacity. The failure to concentrate the urine to control levels in the absence of THP underscores this interpretation and may provide a hint at compensatory mechanisms in these mice., E* Z1 z( _2 |) ~2 H% W% e' e
0 `2 j4 J  d; T% ], u  F
Finally, our model provides some major inequities in terms of a human comparison. The human disease does feature a concentrating defect but also has clear morphological abnormalities that lead to interstitial nephritis with cyst formation (9, 15, 31, 45). These disease features did not occur in the mice. The human disease is further characterized by hyperuricemia, also not a feature of the mouse model. Mice are amply supplied with hepatic uricase (17), an enzyme that does not function in humans, so that disorders in urate handling are not to be expected. The human disease is characterized by a failure of transport of THP out of the cells in which it is synthesized. In the mouse, the gene was completely disrupted. The near complete absence of THP found in humans with the neonatal form of Bartter's syndrome is based on primary deficits of one of the essential NaCl transporters of TAL. Therefore, insufficient THP synthesis is probably not the initial event leading to the disease (16, 40, 42)., s  w: u. I) D0 |( q' g# h, Z* R
$ a+ L2 N9 c  N6 i, v6 I/ b" u+ R
In sum, we believe that our findings are functionally and clinically relevant. We provide the first hard evidence that THP is important to regulatory physiology and actually participates in transporter function. The THP –/– model promises to be important in the evaluation of human disease ranging from myeloma cast nephropathy to familial juvenile hyperuricemic nephropathy. The fact that THP is relevant to epithelial transport regulation is an observation certainly deserving of further study.0 x" [/ }. t7 i4 |7 b& ~5 g' K
* R7 g/ K) m1 L: Y* W
ACKNOWLEDGMENTS
4 u( [8 ?' w0 Q  d$ H+ @3 O; k* O
4 O4 |0 Z0 F+ i  J, c4 {We thank Dr. L. Bankir (Institut National de la Santé et de la Recherche Médicale Paris) for helpful advice in the evaluation of the urinary parameters, Dr. D. H. Ellison and Dr. T. Jentsch for providing the antibodies, and K. Riskowsky, P. Landmann, P. Schrade, and F. Serowka for expert technical assistence.
: E! e2 [+ R; {8 G& \  ?# x- W) j9 t2 A) M$ L% X. D+ t
FOOTNOTES
7 t7 X  B: f) k. S8 d( S: ]+ g; T
2 O$ E7 X/ X; ]4 X) eThe costs of publication of this article were defrayed in part by the payment of page charges. The article must therefore be hereby marked "advertisement" in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.2 d, R9 J1 g0 F% U, A2 \
( D" a7 L8 G- p8 Z# @9 S
S. Bachmann and K. Mutig contributed equally to this study.% i% R+ @/ }) E* e& p9 v, J& ~

6 x5 C8 k# p2 E1 cREFERENCES
2 u. `1 C3 `) ?6 I" X  b% C  U, N) Z/ d5 t
Bachmann S, Bostanjoglo M, Schmitt R, and Ellison DH. Sodium transport-related proteins in the mammalian distal nephron-distribution, ontogeny and functional aspects. Anat Embryol (Berl) 200: 447–468, 1999.# m0 n) h7 }: j7 \
6 b2 H6 n; _; i: ^+ @3 e
Bachmann S, Dawnay AB, Bouby N, and Bankir L. Tamm-Horsfall protein excretion during chronic alterations in urinary concentration and protein intake in the rat. Renal Physiol Biochem 14: 236–245, 1991.
' C/ P0 F/ X& v0 C3 S( T& g) P  j% p/ L8 d, D% s
Bachmann S, Koeppen-Hagemann I, and Kriz W. Ultrastructural localization of Tamm-Horsfall glycoprotein (THP) in rat kidney as revealed by protein A-gold immunocytochemistry. Histochemistry 83: 531–538, 1985.# k" P/ u, R( W% ^) G' B% A
8 W5 \& b" @6 w# R5 }3 x! Z/ h$ U" s5 j
Bachmann S, Metzger R, and Bunnemann B. Tamm-Horsfall protein-mRNA synthesis is localized to the thick ascending limb of Henle's loop in rat kidney. Histochemistry 94: 517–523, 1990.
% Q9 v* L; h. f5 l6 p- _4 q0 k6 V; b! o2 W3 \% q
Bates JM, Raffi HM, Prasadan K, Mascarenhas R, Laszik Z, Maeda N, Hultgren SJ, and Kumar S. Tamm-Horsfall protein knockout mice are more prone to urinary tract infection. Kidney Int 65: 791–797, 2004." L4 R/ V0 J) l2 v4 k
$ p, l5 p5 @8 I% Y
Bosse HM, Bhm R, Resch S, and Bachmann S. Parallel regulation of constitutive NO synthase and renin at the juxtaglomerular apparatus of rat kidney under various stimuli. Am J Physiol Renal Fluid Electrolyte Physiol 269: F793–F805, 1995.
: y) x! J7 @. f6 H' ^. O' C" U, e5 l4 [5 ~% \8 p3 g
Cmpean V, Kricke J, Ellison DH, Luft F, and Bachmann S. Localization of thiazide-sensitive Na -Cl– cotransport and associated gene products in mouse DCT. Am J Physiol Renal Physiol 281: F1028–F1035, 2001.
  [8 t1 y. d( E9 P. |: X: M# ?8 N, R/ n3 y4 J/ H+ Q2 ]2 M3 u
Castrop H, Klar J, Wagner C, Hcherl K, and Kurtz A. General inhibition of renocortical cyclooxygenase-2 expression by the renin-angiotensin system. Am J Physiol Renal Physiol 284: F518–F524, 2003.) N8 [. R6 x! h% n# P3 J  W
- W- }( I1 k8 [
Dahan K, Devuyst O, Smaers M, Vertommen D, Loute G, Poux JM, Viron B, Jacquot C, Gagnadoux MF, Chauveau D, Büchler M, Cochat P, Cosyns JP, Mougenot B, Rider MH, Antignac C, Verellen-Dumoulin C, and Pirson Y. A cluster of mutations in the UMOD gene causes familial juvenile hyperuricemic nephropathy with abnormal expression of uromodulin. J Am Soc Nephrol 14: 2883–2893, 2003.1 t! g9 Y, k. R/ V% ^( x$ x- x

" ?. G9 s& T3 wEllison DH. Divalent cation transport by the distal nephron: insights from Bartter's and Gitelman's syndromes. Am J Physiol Renal Physiol 279: F616–F625, 2000.
; q! m9 U8 C. A" T4 h' O% ?1 j) s6 n) H
Ellison DH, Vel角zquez H, and Wright FS. Adaptation of distal convoluted tubule of the rat. J Clin Invest 83: 113–126, 1989.; t. \1 x/ n4 c( G5 X, U7 }" Y9 q
. ?' ~( g4 \$ J" J/ @1 B$ N
Estevez R, Boettger T, Stein V, Birkenhager R, Otto E, Hildebrandt F, and Jentsch TJ. Barttin is a Cl– channel -subunit crucial for renal Cl– reabsorption and inner ear K  secretion. Nature 414: 558–561, 2001.
' f1 g5 q: J  _' T0 z
9 d, \  g/ N. @7 r0 f' `& g' JFrühauf JH, Welker P, Mutig K, Jentsch TJ, Kinne-Safran E, and Bachmann S. Lipid raft association of essential proteins of the thick ascending limb. J Am Soc Nephrol 14: 561A, 2003.
  w1 n5 ?; {) D' G% A8 r+ B
- C4 z5 |' h' NHarris RC Jr. Cyclooxygenase-2 inhibition and renal physiology. Am J Cardiol 89: 10D–17D, 2002.
0 L5 d- r3 v: }) a0 S$ o3 x1 g
0 t2 i3 Y7 C% D8 W+ Y' ~Hart TC, Gorry MC, Hart PS, Woodard AS, Shihabi Z, Sandhu J, Shirts B, Hy L, Zhu H, Barmada MM, and Bleyer AJ. Mutations in UMOD gene are responsible for medullary cystic kidney disease 2 and familial juvenile hyperuricaemic nephropathy. J Med Genet 39: 882–892, 2002.
" ^& d' g) I  _4 I& M1 c* p9 x! \8 O
# d! E) p" C% t8 E  K0 I4 LHebert SC. Bartter syndrome. Curr Opin Nephrol Hypertens 12: 527–532, 2003.9 J0 |1 T, Q* S7 h" J
: b8 v- Y* h5 U  Q5 A
Kelly SJ, Delnomdedieu M, Oliverio MI, Williams LD, Saifer MGP, Sherman MR, Coffman TM, Johnson GA, and Hershfield MS. Diabetes insipidus in uricase-deficient mice: a model for evaluating therapy with poly(ethylene glycol)-modified uricase. J Am Soc Nephrol 12: 1001–1009, 2001.7 o* \5 h$ C' r, B9 n
7 m1 L2 V2 |3 d) j5 T0 Q
Kim GH, Ecelbarger CA, Mitchell C, Packer RK, Wade JB, and Knepper MA. Vasopressin increases Na-K-2Cl cotransporter expression in thick ascending limb of Henle's loop. Am J Physiol Renal Physiol 276: F96–F103, 1999.
& e: Z" i# u  C
) ]' a) n% g. L1 }9 o" mKnepper MA and Masilamani S. Targeted proteomics in the kidney using ensembles of antibodies. Acta Physiol Scand 173: 11–21, 2001.
3 l1 |+ `( S# U; ^
; E9 n. J! s! N8 JKobayashi K, Uchida S, Okamura HO, Marumo F, and Sasaki S. Human CLC-KB gene promoter drives the EGFP expression in the specific distal nephron segments and inner ear. J Am Soc Nephrol 13: 1992–1998, 2002.
0 Q! U) y% e1 l  _0 m
# u" T# J" v8 nLynn KL and Marshall RD. Excretion of Tamm-Horsfall glycoprotein in renal disease. Clin Nephrol 22: 253–257, 1984.
" _/ Y+ t% d: Z8 C" q5 Y
; H4 W, _! T* S: s7 v* }Malagolini N, Cavallone D, and Serafini-Cessi F. Intracellular transport, cell-surface exposure and release of recombinant Tamm-Horsfall glycoprotein. Kidney Int 52: 1340–1350, 1997.) P& w7 J9 N/ g+ p8 [; l

- S. u8 _- m3 |* EMasilamani S, Kim GH, Mitchell C, Wade JB, and Knepper MA. Aldosterone-mediated regulation of ENaC , , and  subunit proteins in rat kidney. J Clin Invest 104: R19–R23, 1999.5 O; F8 r: \$ T; a& A1 B3 e

# l- J. r% I) Y+ X' ?Mo L, Zhu XH, Huang HY, Shapiro E, Hasty DL, and Wu XR. Ablation of the Tamm Horsfall protein gene increases susceptibility of mice to bladder colonization by type 1-fimbriated Escherichia coli. Am J Physiol Renal Physiol 286: F786–F795, 2004.
5 t; r. I! Z! L
- \3 x% j! o9 v3 u4 H6 m/ N) aNielsen J, Kwon TH, Masilamani S, Beutler K, Hager H, Nielsen S, and Knepper MA. Sodium transporter abundance profiling in kidney: effect of spironolactone. Am J Physiol Renal Physiol 283: F923–F933, 2002.2 p. @  Y1 A/ S9 p9 d$ l8 n

; f: E: }& F( k# r' e3 ZNielsen S, Maunsbach AB, Ecelbarger CA, and Knepper MA. Ultrastructural localization of Na-K-2Cl cotransporter in thick ascending limb and macula densa of rat kidney. Am J Physiol Renal Physiol 275: F885–F893, 1998.  O- f1 l5 V1 b" [( M& g" g
: D! Z- s7 q  o; j2 y
Obermüller N, Kunchaparty S, Ellison DH, and Bachmann S. Expression of the Na-K-2Cl cotransporter by macula densa and thick ascending limb cells of rat and rabbit nephron. Rapid publication. J Clin Invest 98: 635–640, 1996.5 y* _+ z9 D8 H
+ o8 h$ j- \6 A% b% e$ |2 ^7 {! s1 [
Obermüller N, Moser D, Kunchaparty S, Bernstein PL, Vel芍zquez H, Ellison DH, and Bachmann S. Expression of the thiazide sensitive Na/Cl cotransporter in rat and human kidney. Am J Physiol Renal Fluid Electrolyte Physiol 269: F900–F910, 1995.
- E1 O! i  _+ d+ b% ]
/ q; r5 s# D$ U0 ~4 E9 FPak J, Pu Y, Zhang ZT, Hasty DL, and Wu XR. Tamm-Horsfall protein binds to type 1 fimbriated Escherichia coli and prevents E. coli from binding to uroplakin Ia and Ib receptors. J Biol Chem 276: 9924–9930, 2001.5 h: B4 s* a+ {' |4 s6 i* A

, M  k/ y. v, j2 g* g* u$ `3 y. GPeters M, Ermert S, Jeck N, Derst C, Pechmann U, Weber S, Schlingmann KP, Seyberth HW, Waldegger S, and Konrad M. Classification and rescue of ROMK mutations underlying hyperprostaglandin E syndrome/antenatal Bartter syndrome. Kidney Int 64: 923–932, 2003.1 r  M) [% P8 u+ |( L8 s

6 d/ L' P* x1 e/ iRampoldi L, Caridi G, Santon D, Boaretto F, Bernascone I, Lamorte G, Tardanico R, Dagnino M, Colussi G, Scolari F, Ghiggeri GM, Amoroso A, and Casari G. Allelism of MCKD, FJHN and GCKD caused by impairment of uromodulin export dynamics. Hum Mol Genet 15: 3369–3384, 2003.
  \8 i4 o9 T1 R' o% y7 U: D. O2 M6 f, F
Rasch R, Torffvit O, Bachmann S, Jensen PK, and Jacobsen NO. Tamm-Horsfall glycoprotein in streptozotocin diabetic rats: a study of kidney in situ hybridization, immunohistochemistry, and urinary excretion. Diabetologia 38: 525–535, 1995.& a4 Z: R: z8 O* Q5 K, j+ t. z

+ C. |& F0 s. I! _Rotin D, Kanelis V, and Schild L. Trafficking and cell surface stability of ENaC. Am J Physiol Renal Physiol 281: F391–F399, 2001.  q; d1 L" b% S4 q. O
% o6 W9 \+ |  l% j
Sanders PW, Booker BB, Bishop JB, and Cheung HC. Mechanisms of intranephronal proteinaceous cast formation by low molecular weight proteins. J Clin Invest 85: 570–576, 1990.
+ w1 X. O+ g$ j  K* f6 P
% Y2 ~3 Y) x; E, g5 sSatlin LM, Sheng S, Woda CB, and Kleyman TR. Epithelial Na  channels are regulated by flow. Am J Physiol Renal Physiol 280: F1010–F1018, 2001.
, l& j: j) m* @& j* W
9 H- e0 o8 n  ]) R! VSato K, Oguchi H, Yoshie T, and Toshihiko K. Tubulointerstitial nephritis induced by Tamm-Horsfall protein sensitization in guinea pigs. Virchows Arch 58: 357–363, 1990.
! X- q9 A3 ?3 d$ t1 C+ Z1 D! c! L
Schmitt R, Ellison DH, Klussmann E, and Bachmann S. Renal expression of sodium transporters and aquaporin-2 in hypothyroid rats. Am J Physiol Renal Physiol 284: F1097–F1104, 2003.
) f- O6 ?$ r: P4 t# n# u, _+ ]# z0 e7 P# N
Schmitt R, Kahl T, Mutig K, and Bachmann S. Selectively reduced expression of thick ascending limb Tamm-Horsfall protein in hypothyroid kidneys. Histochem Cell Biol 121: 319–327, 2004." E# y* J4 |  a. G. v

# x1 W4 E& e( ]' ~+ A. X7 ~# PSchnermann J. The juxtaglomerular apparatus: from anatomical peculiarity to physiological relevance. J Am Soc Nephrol 14: 1681–1694, 2003.
; ^+ Y+ `8 T- k  C: a8 a7 `$ {! d: t8 ^# P; q3 I0 [3 E% g5 D: s2 p
Schrter J, Timmermans G, Seyberth HJ, Greven J, and Bachmann S. Marked reduction of Tamm-Horsfall protein expression and urinary excretion in hyperprostaglandin E-syndrome. Kidney Int 44: 401–410, 1993.
5 F& b! o# |9 I% l/ W
4 h) f* g& |! l3 d4 o. M  u! hSerafini-Cessi F, Malagolini N, and Cavallone D. Tamm-Horsfall glycoprotein: biology and clinical relevance. Am J Kidney Dis 42: 658–676, 2003.+ z3 v' V0 W( }; h- v0 K, n8 I$ e

- Z5 U0 D+ x8 c' t1 |2 iTakahashi N, Chernavvsky DR, Gomez RA, Igarashi P, Gitelman HJ, and Smithies O. Uncompensated polyuria in a mouse model of Bartter's syndrome. Proc Natl Acad Sci USA 97: 5434–5439, 2000.
! }1 h. [! X  n( A2 ~; P: I. m& D7 e7 @
Traynor TR, Smart A, Briggs JP, and Schnermann J. Inhibition of macula densa-stimulated renin secretion by pharmacological blockade of cyclooxygenase-2. Am J Physiol Renal Physiol 277: F706–F710, 1999.- p- P& C) B$ _; C( D

# i! P9 C3 U7 P' q) E: r+ s$ k1 oTsai CY, Wu TH, Yu CL, Lu JY, and Tsai YY. Increased excretions of 2-microglobulin, IL-6, and IL-8 and decreased excretion of Tamm-Horsfall glycoprotein in urine of patients with active lupus nephritis. Nephron 85: 207–214, 2000.
7 s6 Z  h0 ]& q' V# P
5 M4 H" W* }9 Z# P* |Wolf TF, Mucha BE, Attanasio M, Zalewski I, Karle SM, Neumann HPH, Rahman N, Bader B, Baldamus CA, Otto E, Witzgall R, Fuchshuber A, and Hildebrandt F. Mutations of the Uromodulin gene in MCKD type 2 patients cluster in exon 4, which encodes three EGF-like domains. Kidney Int 64: 1580–1587, 2003.
' C  Q$ W4 {0 S: C4 A! J1 U4 g/ B. Z9 R* {. S( w" C5 W& B
Yang T, Singh I, Pham H, Sun D, Smart A, Schnermann JB, and Briggs JP. Regulation of cyclooxygenase expression in the kidney by dietary salt intake. Am J Physiol Renal Physiol 274: F481–F489, 1998." D0 |/ k( W3 d' a
  x% \3 {" u' l  q7 L) {1 ?2 w8 [
Ying WZ and Sanders PW. Dietary salt regulates expression of Tamm-Horsfall glycoprotein in rats. Kidney Int 54: 1150–1156, 1998.
3 V5 S/ ]4 e4 y7 L4 l! \
8 c8 M) H$ E/ C* r7 N' xYing WZ and Sanders PW. Mapping the binding domain of immunoglobulin light chains for Tamm-Horsfall protein. Am J Pathol 158: 1859–1866, 2001.
, G1 F( f9 h' z- }# Y6 p  `: \6 z; ]
  M, t% b# |4 ~5 O' PZimmerhackl LB. Evaluation of nephrotoxicity with renal antigens in children: role of Tamm-Horsfall protein. Eur J Clin Pharmacol 44, Suppl 1: S39–S42, 1993.(Sebastian Bachmann, Kerim)

Rank: 2

积分
79 
威望
79  
包包
1769  
沙发
发表于 2015-5-30 10:09 |只看该作者
真是天底下好事多多  

Rank: 2

积分
72 
威望
72  
包包
1730  
藤椅
发表于 2015-7-15 09:36 |只看该作者
今天没事来逛逛  

Rank: 2

积分
166 
威望
166  
包包
1997  
板凳
发表于 2015-7-21 12:18 |只看该作者
干细胞之家微信公众号
干细胞之家

Rank: 2

积分
98 
威望
98  
包包
2211  
报纸
发表于 2015-7-27 19:36 |只看该作者
回复一下  

Rank: 2

积分
129 
威望
129  
包包
1788  
地板
发表于 2015-8-17 00:14 |只看该作者
老大,我好崇拜你哟  

Rank: 2

积分
69 
威望
69  
包包
1788  
7
发表于 2015-9-5 13:26 |只看该作者
看或者不看,贴子就在这里,不急不忙  

Rank: 2

积分
132 
威望
132  
包包
1727  
8
发表于 2015-9-14 07:27 |只看该作者
勤奋真能造就财富吗?  

Rank: 2

积分
66 
威望
66  
包包
1790  
9
发表于 2015-9-15 11:10 |只看该作者
今天临床的资料更新很多呀

Rank: 2

积分
72 
威望
72  
包包
1859  
10
发表于 2015-9-15 17:41 |只看该作者
今天无聊来逛逛  
‹ 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, 2024-4-25 19:54

Powered by Discuz! X1.5

© 2001-2010 Comsenz Inc.