Skip to main content
Erschienen in: Pediatric Nephrology 4/2018

Open Access 13.03.2017 | Review

The molecular biology of pelvi-ureteric junction obstruction

verfasst von: Laura Jackson, Mark Woodward, Richard J. Coward

Erschienen in: Pediatric Nephrology | Ausgabe 4/2018

Abstract

Over recent years routine ultrasound scanning has identified increasing numbers of neonates as having hydronephrosis and pelvi-ureteric junction obstruction (PUJO). This patient group presents a diagnostic and management challenge for paediatric nephrologists and urologists. In this review we consider the known molecular mechanisms underpinning PUJO and review the potential of utilising this information to develop novel therapeutics and diagnostic biomarkers to improve the care of children with this disorder.

Introduction

Antenatally detected hydronephrosis is a major clinical dilemma for paediatric nephrologists and urologists (incidence of 1 in 200) [1]. This condition has become more prevalent in recent years as antenatal scanning has become more sensitive and widely used. Approximately one in seven neonates with antenatally detected hydronephrosis has pelvi-ureteric junction obstruction (PUJO) [24], making PUJO one of the most common causes of congenital urinary tract obstruction, with an incidence of one in 1000 to one in 2000 live births [35]. Interestingly, males are affected approximately threefold more frequently than females by this condition [4]. The reason for this difference is unknown.
Intrinsic obstruction due to an adynamic stenotic segment at the PUJ is the most common aetiology (75% of cases) [4], with failure of peristalsis producing an incomplete, functional obstruction. Other causes include: crossing vessels (20%), peripelvic fibrosis, abnormal ureteric insertion, fibroepithelial polyps and anatomical variants, such as retrocaval ureter, horseshoe and duplex kidneys [4, 6, 7].
The major challenge for clinicians is deciding which of these children, who are largely asymptomatic, require a pyeloplasty to relieve the obstruction. This is because two-thirds of children with PUJO do not sustain renal damage or need surgery, and their hydronephrosis spontaneously improves [810].
Currently, serial ultrasound and invasive isotope studies are performed to guide surgical management of PUJO [4]. However, the ability of these diagnostic modalities to accurately detect obstruction, identify children at risk of functional deterioration and predict the need for surgery is questionable. Additionally, there remains debate regarding the parameters which indicate clinically significant obstruction [9, 1113].
In general a pyeloplasty is performed for [6]:
  • differential renal function deterioration (differential function of <40% or a fall of >10% on serial MAG3 renograms)
  • significant hydronephrosis with a renal pelvis anteroposterior diameter of >3 cm on ultrasound scan
  • increasing hydronephrosis with an increasing anteroposterior diameter on serial ultrasound scan
  • symptomatic children.
Our current understanding of the natural history of PUJO as well as our ability to distinguish which children require surgery is inadequate. Available diagnostic tests cannot accurately discern between children with PUJO that will resolve spontaneously and those with PUJO that will persist, causing functional impairment. Consequently, despite radiological monitoring, there is a risk of loss of function in the affected kidney while the patient is under observation [14].
In this review we discuss the currently known molecular mechanisms underlying intrinsic PUJO and whether this information could contribute to the future development of novel therapies and diagnostic biomarkers.

Anatomy of the upper urinary tract

The PUJ is a region of gradual transition from the funnel-shaped renal pelvis to the proximal ureter [15] (Fig. 1). It is a physiologic sphincter [16] that is characterised by prominent luminal folds with increased muscle thickness capable of creating a high-pressure zone to regulate urine flow. Similar to the adjacent renal pelvis and ureter, the PUJ comprises three main layers: the inner urothelium, middle smooth muscle and outer adventitia [15]. Smooth muscle contraction propels urine from the renal pelvis to the bladder [17], coordinated by submucosal and intra-muscular nerve plexi [18] and modulated by autonomic innervation involving a range of neurotransmitters that include acetylcholine, noradrenaline, substance P, neurokinin A, calcitonin gene-related peptide, neuropeptide Y, vasoactive intestinal peptide and nitric oxide (NO) [17].

Embryology of the ureter and PUJ

Understanding the normal embryology of PUJ formation is vital when considering where development may proceed incorrectly in congenital abnormalities such as PUJO. The kidney develops from metanephric mesoderm as far along the nephron as the distal tubules. The collecting duct onwards, including the major and minor calyces, renal pelvis and ureter has a different embryological origin, arising from the ureteric bud [19, 20]. Thus, the PUJ does not represent an embryological fusion site, rather it is derived exclusively from the ureteric bud. The important molecular pathways that form the ureter and PUJ are shown in Fig. 2 and Table 1 [15, 2628]. Briefly, the ureteric bud, consisting of a simple epithelial layer extending into loose mesenchyme, arises from the mesonephric duct during the fifth week of gestation in humans [26]. Epithelial cell proliferation and differentiation then results in the formation of the transitional epithelium. Epithelial paracrine and mesenchymal autocrine signalling stimulates the formation of smooth muscle cells from mesenchyme, which begins at 12 weeks of gestation in humans [26, 29]. Mouse models have implicated a number of signalling molecules in this process of proliferation, aggregation, differentiation and orientation of smooth muscle cells as they encircle the urothelial tube (Fig. 2, Table 1). A second phase of smooth muscle differentiation that particularly affects the renal pelvis and proximal ureter occurs in postnatal mice (equivalent to the second trimester of gestation in humans) and is regulated by calcineurin and angiotensin II signalling [30, 31].
Table 1
Proteins/molecular pathways involved in ureteric development
Protein
Full protein name
Function
Reference
SHH
Sonic hedgehog
Morphogen which stimulates peri-urothelial mesenchymal cell proliferation and regulates timing of smooth muscle cell differentiation
[21]
PTCH1 receptor
Protein patched homolog 1
Receptor for SHH, functions as tumour suppressor when unbound
[21]
BMP4
Bone morphogenetic protein 4
Growth factor, necessary for smooth muscle cell differentiation and ureter morphogenesis
[22]
TSHZ3
Teashirt zinc finger homeobox 3
Transcription factor-like protein necessary for myocardin expression and ureteric smooth muscle cell differentiation
[23]
MYOCD
Myocardin
Transcriptional co-activator, necessary for expression of contractile proteins
[23]
TBX18
T Box protein 18
Transcription factor necessary for correct localisation and aggregation of smooth muscle cells around ureteric urothelium
[24]
DLGH1
Disks large homolog 1
Scaffolding protein, regulates smooth muscle cell orientation
[25]

Pathologic features of intrinsic PUJO

Inflammatory cell infiltration [32], varying degrees of fibrosis, excess collagen deposition [3235] and abnormal muscle fibre arrangement [36] are present in human intrinsic PUJ obstruction. Both muscular hypertrophy/hyperplasia [32, 34, 37] and atrophy/hypoplasia [32, 36] are reported alongside depletion of nerves to the muscular layer [33]. These findings are noted when the PUJ is excised at pyeloplasty and therefore represent late features of PUJ obstruction (Fig. 3). Although the time course of PUJ disease progression is unknown in humans, genetic mouse models of hydronephrosis show abnormalities of peri-urothelial mesenchymal organisation as early as embryonic day (E) 12.5 (approximately equivalent to 35 days of gestation in humans) [24] and smooth muscle cell differentiation at E15.5 (approximately equivalent to 12 weeks of gestation in humans) [23]. One week postnatally (approximately equivalent to humans at birth) mice with Id2 haploinsufficiency show smooth muscle irregularity and hypertrophy at the PUJ [38], features which are common to human PUJO. The possible mechanisms underlying this pathology are described later in this review.

Modelling PUJO to understand its molecular biology

Adult and neonatal rodent models of complete and partial unilateral ureteric obstruction (UUO) have been extensively used to investigate the molecular biology of congenital obstructive nephropathy. Neonatal models are particularly helpful because rodent nephrogenesis continues for 1 week postnatally and nephron maturation over the subsequent week. Thus, at birth and 1 week of age, rodent kidney development is equivalent to humans at the second trimester of gestation and birth, respectively [11]. This gives a window in which surgery can be performed on the animals to mimic in utero obstruction in humans. Adult obstructive models show a broadly similar pathologic progression to neonatal models with the exception that neonatal obstruction impedes normal maturation and growth of the kidney and leads to early nephron loss. The renal pathologic findings in neonatal and adult UUO models and the timescale of their development are presented in Fig. 4 [3947].
A comprehensive review comparing neonatal models with human disease confirms their validity for investigating obstructive nephropathy and will not be further discussed in this review [48].

Proposed molecular mechanisms underpinning PUJO

In the following subsections we highlight some of the molecular steps that may lead to the development of intrinsic PUJO and subsequent obstructive nephropathy. Data have been obtained from both adult and neonatal models of complete and partial ureteric obstruction alongside evaluation of tissue obtained at pyeloplasty for human PUJ obstruction.

Neurogenic factors

Light microscopy studies have revealed reduced innervation within the muscular layer of the PUJ in human specimens excised at pyeloplasty for PUJO [33]. This is associated with reduced expression of molecular markers, including glial cell line-derived neurotrophic factor (survival factor for neurons), protein gene product 9.5 (general neuronal marker), and nerve growth factor receptor protein, in the muscle layers of the stenotic PUJ compared to controls. Although it is speculated that these neuronal changes may contribute to the pathogenesis of PUJO, there is as yet no evidence to confirm or refute this notion. Conflicting changes in synaptophysin (e.g. major synaptic vesicle protein p38) expression in terms of both amount (increased and decreased) and distribution (localisation to the nucleus) are reported in PUJO compared to controls and are of uncertain significance. S-100 (schwann cell marker) and neurofilament (neuronal protein) expression are unchanged, demonstrating there is not a global reduction in neuronal components [34, 49].

Myogenic factors

Together with increased smooth muscle cell apoptosis, phenotypic and cytoskeletal smooth muscle cell changes are seen in the human PUJ excised at pyeloplasty for PUJO. The stenotic PUJ shows significantly increased expression of smooth muscle myosin heavy chain isoforms 1 and 2 [37], as well as an altered ratio of integrin (transmembrane signalling receptor) isoform expression compared to control samples [50]. The preferential expression of immature integrins in the stenotic PUJ [50] may indicate developmental delay of the smooth muscle cells, potentially contributing to their altered function and increased apoptosis in PUJO.
Supporting a myogenic cause of PUJO, transgenic mouse models targeting smooth muscle differentiation generate a PUJ phenotype with hydronephrosis secondary to functional obstruction (Table 2).
Table 2
Evidence from animal and human studies of genes potentially involved in the pathogenesis of pelvi-ureteric junction obstruction
Gene
Full gene name
Animal
Features and mechanism
Human
Reference
Ace
Angiotensin converting enzyme
Ace −/− mice
Hydronephrosis, renal parenchymal atrophy
 
[51]
Adamts-1
A disintegrin-like and metallopeptidase with thrombospondin type 1 motif, 1
Adamts -1−/− mice
PUJ obstruction, increased collagen at PUJ. Other urogenital anomalies.
 
[52]
Agt
Angiotensin
Agt −/− mice
Hydronephrosis, renal parenchymal atrophy,
 
[53]
Agtr 1a/b
Angiotensin II receptor type 1 (1a and 1b)
Agtr1 −/− (1a and 1b) mice
Hydronephrosis in older mice, renal parenchymal atrophy, failure of renal pelvis development, ureteric smooth muscle hypoplasia and abnormal peristalsis
 
[31]
Aqp2
Aquaporin 2
Aqp 2S256L/S256L CPH mice
Mutation in CPH mice prevents Aqp2 phosphorylation and normal trafficking. Hydronephrosis secondary to polyuria
 
[54]
Calcineurin
Calcineurin. Also known as Protein phosphatase 3 (ppp3)
Pax3-Cre T/+; Cnb1 flox/ flox mice
Calcineurin inactivation in metanephric and ureteral mesenchyme giving hydronephrosis, abnormal pyeloureteral peristalsis with defective renal pelvis and smooth muscle development
 
[30]
Id2
Inhibitor of DNA binding 2
Id2 −/− and Id2 +/− mice
Hydronephrosis and PUJ development
 
[38]
Nfia
Nuclear factor I/A
Nfia +/− and Nfia −/− mice
Hydroureteronephrosis, VUR, abnormal PUJ and VUJ development. CNS malformations.
Nfia +/−due to chromosomal translocation and deletion. VUR and CNS malformations.
[55]
TBX18
T-box transcription factor
Tbx18 −/− mice
Hydroureteronephrosis, short ureters, ureteric smooth muscle defects due to abnormal smooth muscle cell differentiation and localisation
Hispanic family with autosomal dominant CAKUT predominantly PUJO. Heterozygous truncating mutation (c.1010delG) of Tbx18
[24, 56]
Tshz2 and 3
Teashirt zinc finger family member 2 and 3
Tshz3 −/− mice
Hydronephrosis with PUJ configuration, abnormal smooth muscle differentiation proximal ureter
Tshz2/Tshz3 mutations not cause of PUJO in Albanian/Macedonian population
[57, 58]
CAKUT, Congenital anomalies of the kidney and urinary tract; CNS, central nervous system; CPH, congenital progressive hydronephrosis; PUJO, pelvi-ureteric junction obstruction; VUJ, vesico-ureteric junction; VUR, vesico-ureteric reflux

Increased pressure, impeded blood supply and hypoxia

Obstructive hydronephrosis is associated with a doubling to trebling of renal pelvis pressure [16, 5961]. The resultant increased intratubular hydrostatic pressure [62] stimulates the renopathogenic effects of obstruction via three proposed mechanisms, namely, (1) tubular ischaemia due to hypoperfusion, (2) pressure-induced mechanical stretch/compression of tubular cells and (3) altered urinary shear stress. The latter two mechanisms are likely to be the primary inducers of obstructive renal injury [48], causing dysregulation of many cytokines, growth factors, enzymes and cytoskeletal proteins (Table 3), resulting in early renal haemodynamic changes followed by structural and functional alterations to the entire nephron. Figure 5 highlights the major mechanisms of renal injury in PUJO.
Table 3
Table showing the major cytokines, growth factors, chemokines, enzymes and cytoskeletal proteins which demonstrate altered intra-renal regulation in obstructive nephropathy, the timing of these changes and their mode of action
Proteina
Action
Change/timing
Species
Reference
Angiotensin II
Vasoregulatory, proinflammatory, proapoptotic, profibrotic
Increased 28 days
Increased 1 week and 5 weeks
Increased after mechanical stretch
Neonatal rat CUUO
Adult rat CUUO
In vitro podocytes
[63]
[64]
[65]
α-SMA
Increases myofibroblast contractility/EMT marker
Increased 5 days
Increased 4 days
Neonatal rat CUUO
Adult mouse CUUO
[39]
[66]
Caspases
Proapoptotic
Increased 14 days
Increased 1 day
Neonatal rat CUUO
Adult rat CUUO
[67]
[68]
Clusterin
Cytoprotective via pro-survival autophagy
Increased 5 days
Neonatal rat CUUO
[39]
COX-2
Polyuria and natriuresis, anti-apoptotic, antifibrotic
Increased 24 h
Increased 3 days (mRNA)
Adult rat CBUO
Adult mouse CUUO
[69]
[70]
CTGF
Profibrotic
Increased 2 days (mRNA)
Adult rat CUUO
[45]
EGF
Epithelial survival factor
Decreased 7 days (mRNA) (Undetectable expression in neonatal rat kidney before 4 days)
Decreased 33 days
Decreased at pyeloplasty (mean age 2 years) (mRNA)
Decreased at pyeloplasty (mean age 5 years)
Neonatal rat CUUO
Neonatal rat both CUUO and 5 day CUUO then release
Human renal biopsy
Human renal biopsy
[71]
[39]
[72]
[73]
ET-1
Vasoconstrictor
Increased 2 days (mRNA)
Adult rat CUUO
[45]
Fas-L
Proapoptotic
Increased 1 day (mRNA)
Adult rat CUUO
[68]
HSP-70
Antiapoptotic
Decreased 14 days
Neonatal CUUO
[67]
ICAM-1
Proinflammatory
Increased 3 days
Adult mouse CUUO
[74]
Il-6
Proinflammatory
Increased 2 days (mRNA)
Adult rat CUUO
[45]
Integrin (β1)
Profibrotic
Increased 3 days
Increased after mechanical stretch
Adult mouse CUUO
In vitro proximal tubular cells
[75]
[76]
MCP-1
Proinflammatory
Increased 12 days, no change 4 days
Increased 2 days (mRNA)
Increased at pyeloplasty (mean age 2 years) (mRNA)
Neonatal rat CUUO
Adult rat CUUO
Human renal biopsy
[77]
[45]
[72]
MMP 2 and 9
ECM degradation
Decreased 3 days
Adult mouse CUUO
[74]
PAI-1
Profibrotic, inhibits ECM degradation
Increased 7 days
Adult mouse CUUO
[78]
PDGF
Profibrotic
Increased 4 days
Adult mouse CUUO
[66]
NF-κB
Regulatory transcription factor
Increased 2 days
Adult mouse CUUO
[45]
Nitric oxide
Vasodilator, anti-apoptotic, antifibrotic
Decreased 14 days
Neonatal rat CUUO
[67, 79]
Renin
Cleaves angiotensinogen, upregulates renin–angiotensin system
Increased 3 days (mRNA)
Increased 5 days
Increased 14 days (mRNA)
Increased 4–5 weeks
Increased 24 h
Increased after mechanical stretch
Neonatal rat CUUO
Neonatal rat CUUO
Neonatal rat CUUO
Neonatal rat CUUO
Adult rat CUUO
In vitro proximal tubular cells
[71]
[39]
[63]
[80]
[81]
[82]
TGF-β
Proinflammatory, proapoptotic, profibrotic, stimulates EMT
Increased 1 day (mRNA)
Increased 33 days
Increased 3 days (mRNA)
Increased at pyeloplasty (mean age 5 years)
Neonatal rat CUUO
Neonatal rat both CUUO and 5 day CUUO then release
Adult rat CUUO
Human renal biopsy
[71]
[39]
[83]
[73]
TIMP-1
Profibrotic, inhibits ECM degradation
Increased 5 days
Increased 3 days
Adult rat CUUO
Adult mouse CUUO
[84]
[74]
TNF-α
Proapoptotic, proinflammatory
Increased 14 days (mRNA)
Increased 1 day
Increased 2 days (mRNA)
Increased 1 day
Neonatal rat CUUO
Adult rat CUUO
Adult rat CUUO
Adult rat CUUO
[85]
[68]
[45]
[68]
VCAM-1
Proinflammatory
Increased 3 days (mRNA)
Adult mouse CUUO
[86]
VEGF (podocytes)
Endothelial survival factor
Increased 28 days
Decreased 14 days
Neonatal PUUO
Neonatal CUUO
[87]
[87]
VEGF (tubules)
Endothelial survival factor
Variable expression
Decreased 14 days
Neonatal PUUO
Neonatal CUUO
[87]
[87]
Vimentin
Intermediate filament protein/ EMT marker
Increased 5 days
Neonatal rat CUUO
[39]
WT-1
Transcriptional regulator, key role in renal development
Decreased 14 days
Neonatal rat CUUO
[85]
Change is compared to sham animal or control human kidney and refers to protein expression unless otherwise stated. Timing is days after creation of unilateral ureteric obstruction (UUO)
CUUO, Complete UUO; CBUO complete bilateral ureteric obstruction; PUUO, partial UUO
aα-SMA, Alpha-smooth muscle actin; COX-2, cyclooxygenase 2; CTGF, connective tissue growth factor; ECM, extracellular matrix; EGF, epidermal growth factor; EMT, epithelial–mesenchymal transition; FasL, Fas ligand; HSP-70, heat shock protein 70; ICAM-1, intercellular adhesion molecule 1; IL-6, interleukin-6; MCP-1, monocyte chemoattractant protein 1; MMP, matrix metalloproteinase; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; TGF-β, transforming growth factor-beta; TIMP-1, tissue inhibitor of metalloproteinases 1; TNF-α , tumour necrosis factor-alpha; VCAM-1, vascular cell adhesion molecule 1; VEGF, vascular endothelial growth factor; WT-1, Wilms tumor protein
Following a short initial increase in renal blood flow related to local vasodilator production [48], the intra-renal renin–angiotensin–aldosterone system (RAAS) is activated causing pre- and post-glomerular vasoconstriction and a resultant fall in renal blood flow (RBF), medullary oxygen tension and glomerular filtration rate (GFR) [11, 48, 64, 80, 8890]. Proximal tubular hypoxia and necrosis in neonatal rats with UUO suggest that vasoconstriction causes segment-specific ischaemic injury [91]. Accordingly, angiotensin II receptor, type 1 (AT1 receptor) inhibition improves tubular function by increasing RBF and GFR [92].
Reduced urine production and continuing urine drainage by venous and lymphatic systems together with tubular and renal pelvis dilatation result in a subsequent decline in renal pelvic pressure [48, 89, 93], which may be a compensatory mechanism to limit damaging increased intra-renal pressure [93].

Initiation of proinflammatory cytokines

Cytokines in the stenotic PUJ

Transforming growth factor-beta (TGF-β) expression is noted in human stenotic PUJ compared to normal controls [94]. Furthermore, the smooth muscle regulators endothelin-1 (smooth muscle constrictor) and adrenomedullin (smooth muscle relaxant) have been shown to be increased and decreased, respectively, in stenotic PUJ disease [95].
Analysis of paediatric renal pelvis tissue proximal to the PUJO for cytokines that show altered renal expression in nephropathy demonstrates increased TGF-β and reduced macrophage inflammatory protein-1alpha (MIP-1α). In contrast, epidermal growth factor (EGF), monocyte chemotactic peptide 1, interferon-γ-inducible protein 10 and RANTES (regulated on activation normal T-cell expressed and secreted) mRNA expression are unchanged, suggesting that TGF-β and MIP-1α play important roles in the development of PUJO [88, 96].

Intra-renal cytokines

Increased intra-renal angiotensin II activates nuclear factor kappa B and ROCK (rho-associated coiled-coil-forming protein kinase), leading to cytokine release and interstitial macrophage infiltration and activation. Intra-renal selectins, integrins, intercellular-adhesion molecule 1, vascular cell adhesion molecule 1, interleukin 1, monocyte chemoattractant peptide 1, colony stimulating factor 1 and osteopontin expression are all involved in macrophage stimulation [11, 48, 88, 97]. Therefore, it appears that renal signals initiate and maintain the injurious inflammatory response to PUJO. Accordingly, both selectin and β2-integrin knockout mouse models show reduced macrophage infiltration into the obstructed kidney after UUO [43, 44].

Inflammatory infiltrates

Activated macrophages infiltrate the renal interstitium, sustaining the inflammatory response by releasing cytokines, such as TGF-β1, tumour necrosis factor-alpha (TNF-α), and platelet-derived growth factor [11, 88].

Profibrotic processes

Tubulointerstitial fibrosis is the final common pathway for many chronic kidney disorders, including obstructive uropathy, and is instigated by altered cytokine expression (Table 4). Activated resident interstitial myofibroblasts [98], expressing α-smooth muscle actin (boosts cell contractility) [99], aggregate, proliferate and produce extracellular matrix. Extracellular matrix consisting of collagens I, III and IV, fibronectin, laminin and proteoglycans accumulates due to increased synthesis and reduced degradation [74, 100, 101]. Myofibroblasts amplify fibrosis by producing cytokines, including TGF-β1 and TNF-α [11]. Parenchymal damage and renal dysfunction results, such that in children with PUJO the extent of fibrosis significantly correlates with differential renal function [102].
Table 4
Cytokines, growth factors, enzymes and adhesion molecules promoting or preventing tubulointerstitial fibrosis in ureteric obstruction
Molecules PROMOTING tubulointerstitial fibrosis in ureteric obstruction
Molecules PREVENTING tubulointerstitial fibrosis in ureteric obstruction
Angiotensin II
EGF
CTGF
MMP
ICAM-1
Nitric oxide
Integrins
VEGF
PAI-1
 
PDGF
 
TGF-β
 
TIMP-1
 
PAI-1, Plasminogen activator inhibitor 1; PDGF, platelet-derived growth factor
Angiotensin II upregulation is central to the pathogenesis of obstructive nephropathy (Fig. 6) [11, 41, 45, 64, 68, 83, 84, 91, 103112]. Angiotensinogen murine knockout studies have demonstrated that angiotensin II expression is responsible for at least 50% of renal fibrosis in chronic neonatal UUO [104]. Acting predominantly via the AT1 receptor [45, 105, 113] it regulates cytokine production and stimulates reactive oxygen species (ROS) generation, which in turn propagates the proinflammatory, fibrogenic state [48, 104]. The generation of ROS also causes proximal tubular degeneration by apoptosis, autophagy and necrosis, with consequent destruction of the glomerulotubular junction, resulting in the formation of atubular glomeruli [41, 109].
TGF-β1 is a profibrotic cytokine and fibroblast chemo-attractant which plays a major role in fibrosis development via SMAD-dependent and -independent pathways (Fig. 7) [7476, 114118]. Renal TGF-β expression is increased in experimental UUO [83, 103, 105, 107, 119121] and children with PUJO, being positively correlated with the histopathologic grade, radioisotope drainage half time (t1/2) and post-void washout and negatively correlated with pre-operative differential renal function [73, 122].
Nitric oxide is an endogenous vasodilator that protects against tubulointerstitial fibrosis and proximal tubular oxidant injury in obstructive nephropathy [79, 84, 123]. Animal models [111, 124, 125] and human studies of PUJO show altered endothelial NO synthase (eNOS) and inducible NO synthase (iNOS) expression/activity together with reduced NO production. Lower eNOS expression/activity is associated with worse creatinine clearance, reduced differential renal function [90, 126] and increased fibrosis [90, 126], oxidant injury and apoptosis [67, 79].

Antifibrotic processes

Renal cyclooxygenase 2 (COX-2) expression and prostaglandin production in experimental UUO is increased [69] and may be a protective response. COX-2 inhibition worsens obstructive nephropathy, while prostacyclin analogue (ONO-1301) supplementation alleviates UUO-induced fibrosis [127].

Cellular apoptosis

Apoptosis affects podocytes and endothelial and epithelial cells within the kidney, leading to loss of glomeruli, peritubular capillaries and tubules [11]. Tubular cell mechanical stretch is a potent stimulator of apoptosis [91, 128] that is mediated via TGF-β1 and TNF-α [68, 110] released from tubular cells and infiltrating macrophages [88]. Other proapoptotic factors increased after UUO include Fas-L [45], p53, caspases and ceramide [11].
Downregulation of anti-apoptotic factors, including EGF, eNOS, NO, vascular endothelial growth factor, heat shock protein 70 and Wilms tumour-1, compounds the renal injury [11, 67, 88, 128, 129].

Tubular function impairment

Ureteric obstruction leads to reduced renal expression of the V2 (vasopressin) receptor [130], renal sodium and urea transporters [131133] and aquaporins [134136]. Aquaporins are a family of transmembrane proteins normally expressed by mammalian kidney [137] and urothelium [138, 139] that mediate water movement across the cell membrane along an osmotic gradient [140]. Reduced renal aquaporin expression in experimental UUO is noted within 24 h of complete obstruction [134]. Similarly, renal aquaporins are downregulated in children undergoing pyeloplasty, and in both human and animal models this reduction is associated with polyuria and reduced concentrating ability following relief of obstruction [141143].

Genetic mechanistic clues in PUJO

Phenotypes similar to PUJO have been noted in numerous transgenic mouse models. Many genes involved in ureteric smooth muscle proliferation and differentiation are implicated, supporting a primary myogenic aetiology. Importantly, one of these genes has been implicated in human disease (Table 2).
Mutations in TBX18, the gene coding for T Box protein 18, have been reported in association with congenital anomalies of the kidney and urinary tract (CAKUT). In particular, a heterozygous TBX18 truncating mutation (c.1010delG) showing autosomal dominant inheritance has been described across four generations of a family with CAKUT, and predominantly PUJO [56]. The transcription factor TBX18 is necessary for normal smooth muscle cell proliferation, differentiation and localisation around the developing urothelial stalk [24]. TBX18 also directs epithelial proliferation and when absent leads to an abnormally short ureteric bud [28].
In the majority of patients, however, PUJO is a polygenic disorder without an obviously inherited genetic component [11].

Potential therapeutic molecular targets in PUJO

Human and animal studies have highlighted a number of potential therapeutic targets that could be manipulated to alleviate the nephropathy sustained secondary to PUJO. Several drugs targeting these pathways have been assessed in rodent UUO models as described below, however, to our knowledge none of these therapies have been trialed in childhood human PUJO.

Angiotensin-converting enzyme and AT1 receptor inhibitors

In adult rodent UUO models angiotensin-converting enzyme (ACE) inhibitors and AT1 receptor inhibitors given prophylactically (for the duration of obstruction) are beneficial in alleviating nephropathy. Specifically, they reduce TGF-β [121, 144] and TNF-α [106] expression, as well as macrophage infiltration and tubulointerstitial fibrosis [84, 105, 145]. Additionally, AT1 receptor inhibitors improve tubular function by improving RBF and GFR and attenuating the reduction in sodium transporter and aquaporin 2 (AQP2) expression, thus reducing polyuria and natriuresis [92, 112].
ACE inhibitors reduce both AT1 and AT2 receptor stimulation [146] and indirectly increase NO levels via bradykinin generation [84]. This may explain why they confer additional benefits, particularly anti-inflammatory, compared to AT1 receptor inhibitors [97]. Unfortunately, inhibition of angiotensin during either the period of nephrogenesis (first 10 days after UUO) or renal maturation (second 10 days after UUO) in neonatal partial UUO exacerbates renal injury in both the obstructed and contralateral kidney [147, 148]. Such studies highlight the importance of these pathways in normal kidney development and maturation.
However, it is important to remember that ACE inhibitors and AT1 receptor inhibitors are frequently used in children with chronic kidney disease, in whom they significantly reduce proteinuria [149] despite not significantly alleviating the natural decline in excretory function [150, 151]. They are largely well tolerated, with no apparent effect on growth and development and a low incidence of side effects such as hyperkalaemia, hypotension and renal injury [149].

Hydroxymethylglutaryl-CoA reductase inhibitors (statins)

Statins ameliorate nephropathy when administered prophylactically in adult and neonatal rodent UUO models by reducing renal cytokine production (TGF-β, TNF-α), macrophage infiltration, oxidative stress, apoptosis and tubulointerstitial fibrosis [85, 152, 153]. These pleiotropic effects are achieved through decreased Ras/ERK/Akt signalling [154] and increased NO bioavailability [155]. Importantly, statins remain beneficial in neonatal rodent UUO where an improvement in tubular dilatation and glomerular number and size are also seen [67, 79, 85]. Functionally, in UUO models, statins improve GFR and microalbuminuria [156] and increase urinary concentrating ability via boosting AQP2 expression [157].
Statins are commonly used and usually well tolerated in adults. Side effects of treatment include hepatic dysfunction, diabetes mellitus, benign proteinuria, peripheral neuropathy, myalgia and rhabdomyolysis [158]. A 10-year follow-up study of children (≥8 years) treated with statins for familial hypercholesterolaemia demonstrated that few discontinue therapy due to side effects and that there were no serious adverse reactions [159]. In that same study, growth, puberty and educational parameters were also unaffected compared to controls [159].

TGF-β modulation

Prophylactic TGF-β receptor inhibition is renoprotective in adult rodent UUO models, reducing apoptosis, macrophage infiltration, fibrosis, proximal tubular atrophy and atubular glomeruli formation [117, 160]. Similarly, anti-TGF-β antibody treatment increases NOS expression while reducing apoptosis and fibrosis [110]. Conversely, prophylactic TGF-β receptor inhibition in neonatal mouse UUO causes widespread renal necrosis, exacerbating the injury in the obstructed kidney and highlighting the differing responses to signalling cascades during renal development [117].
Anti-TGF-β antibody treatment (GC1008) has been trialled in human oncological disease and was generally well tolerated. However, side effects included gingivitis, fatigue and skin rashes, including keratoacanthoma and squamous cell carcinoma development (melanoma patients only). GC1008 treatment has not progressed beyond phase II clinical trials as drug development was discontinued by the manufacturer [161].

COX-2 inhibition

In adult rodent bilateral ureteric obstruction COX-2 inhibition alleviates AQP2 and sodium transporter downregulation and improves post-obstructive polyuria, which would appear to be beneficial [69]. Conversely, other studies have demonstrated that both genetic COX-2 knockout and prophylactic COX-2 inhibition in adult rodent UUO models increase tubular injury, apoptosis and fibrosis, thereby negating potential use in the clinical setting [70, 162].
Chronic celecoxib (COX-2 inhibitor) use in children demonstrates a similar frequency of adverse events to non-selective non-steroidal anti-inflammatory drugs, which are most frequently gastrointestinal side effects [163].

Other potential therapeutic options

Other potential therapeutic pathways include those that are able to increase the vasoactive molecule NO, as this has been shown to reduce tubulointerstitial fibrosis in adult rodent UUO models [84]. Although both ACE inhibitors and statins increase NO bioavailability, this is an indirect effect at the expense of drug-related side effects.
Dietary nitrate supplementation is a novel therapeutic option which directly targets the NO pathway, increasing NO generation via nitrite production. Nitrite also has cytoprotective effects independent of NO by influencing mitochondrial function [164], and when administered during rodent ischaemia reperfusion studies reduces renal injury [165].
Despite former concerns associating nitrates with methaemoglobinaemia and carcinogenesis, the nitrate–nitrite–NO pathway is increasingly implicated in a protective role in humans [166]. Further investigation of dietary nitrate supplementation as a potential therapy in obstructive nephropathy is warranted.

Urinary biomarkers

Identifying early urinary biomarkers in PUJO may be beneficial for the diagnosis, management and prognosis of this condition. Such biomarkers would enable timely detection of children with ‘damaging’ hydronephrosis who require surgery to protect renal function, while avoiding surgery in those with ‘safe’ hydronephrosis.

Urinary biomarkers in animal studies

There is little data on urinary biomarkers from animal studies. Proteomics using a rat UUO model demonstrated increased urinary and renal levels of alpha-actinin-1 and moesin at 1 week which corresponded with histological evidence of tubular injury. Following 3 weeks of UUO urine and renal levels of vimentin, annexin A1 and clusterin were significantly elevated, corresponding with substantial renal interstitial fibrosis [167].

Urinary biomarkers in human studies

Many urinary cytokines, growth factors, chemokines, tubular enzymes and tubular transport proteins have been investigated in children undergoing pyeloplasty for PUJO. Studies with conservatively managed PUJO as a comparator are most useful to identify biomarkers able to aid selection of patients for surgery. Potential urinary biomarker proteins measured in bladder urine samples are presented in Table 5.
Table 5
Urinary proteins from studies in children with pelvi-ureteric junction obstruction
Urinary protein (corrected for creatinine)a
Primary measured group
Comparators
Bladder urine protein level
Sensitivity/specificity/accuracyb
Post-operative bladder urine (compared to pre-operative)
Ref
ALP
Pyeloplasty
CMP
Increased pre-operative
Se 91.4%/ Sp 100%/ Ac 94%
Decreased 12 months post-operative
[168]
Angiotensinogen
Pyeloplasty
Healthy control
CMP
Increased pre-operative
Se 93.3%c/ Sp 60%c
 
[169]
B2-microglobulin
PUJO*
Healthy control
Increased
 
Decreased 42 months post-operative
[170]
B2-microglobulin
Pyeloplasty
Healthy control
No change
  
[171]
Ca19-9
Pyeloplasty
Healthy control
CMP
Increased pre-operative
Se 76%d/Sp 85%d
Decreased 3 months post-operative
[172]
Ca19-9
Pyeloplasty
Healthy control
Hydrocoele/renal cyst
Increased pre-operative
Se 100%e/ Sp 82.6%e
Decreased 3 months post-operative
[173]
CyC
Pyeloplasty
Healthy control
No change
  
[171]
EGF
PUJO*
Healthy control
Decreased (obstructed group only)
 
No change
[170]
EGF
Pyeloplasty
Healthy control
Decreased pre-operative
 
Increased
[174]
EGF
Pyeloplasty
Healthy control
Increased pre-operative
Se 70.4%/Sp 69.2%
Decreased 3 months and 1 year post-operative
[175]
EGF
Pyeloplasty
Healthy control
No change
  
[176]
ET-1
Pyeloplasty
Healthy control
VUR
Renal stones
Increased pre-operative
Se 74.3%/Sp 90%/ Ac 81.5%
Decreased 12 months post-operative
[177]
γGT
Pyeloplasty
CMP
Increased pre-operative
Se 62.9%/Sp 100%/Ac 74%
Decreased 12 months post-operative
[168]
HO-1
Pyeloplasty
Healthy control
CMP
Increased pre-operative
Se 72.2%c/Sp 78.1%c
Decreased 1 month post-operative
[178]
IP-10
Pyeloplasty
Healthy control
No change
  
[175]
KIM-1
Pyeloplasty
Healthy control
CMP
Increased pre-operative
Se 100%c/Sp 71.4%c
 
[179]
MCP-1
Pyeloplasty
Healthy control
Increased pre-operative
Se 77.8%/Sp 69.2%
Decreased 3 months and 1 year post-operative
[175]
MCP-1
PUJO*
Healthy control
Increased
 
Decreased 42 months post-operative
[170]
MCP-1
Pyeloplasty
Healthy control
Increased pre-operative
  
[174]
MCP-1
Pyeloplasty
Healthy control
CMP
Increased pre-operative
Se 100%c/Sp 0%c
Remains high 3 months post-operative
[180]
MIP-1α
Pyeloplasty
Healthy control
Decreased pre-operative
 
Increased 1 year post-operative
[175]
NAG
Pyeloplasty
CMP
Increased pre-operative
Se 97.1%/Sp 80%/Ac 92%
Decreased 12 months post-operative
[168]
NGAL
Pyeloplasty
Healthy control
No change
  
[171]
NGAL
Pyeloplasty
Healthy control
Increased pre-operative
  
[181]
NGAL
Pyeloplasty
Healthy control
CMP
Increased pre-operative
Se 100%c/Sp 28.6%c
Decreased 3 months post-operative
[179]
OPN
Pyeloplasty
Healthy control
No change
  
[171]
OPN
Pyeloplasty
Healthy control
CMP
Increased pre-operative
Se 98.5%c/Sp 10.5%c
Remains high 3 months post-operative
[180]
RANTES
Pyeloplasty
Healthy control
No change
  
[175]
TGF-β
Pyeloplasty
Healthy control
Increased pre-operative
Se 100%/Sp 80%/Ac 90.8%
Decreased 1 year post-operative
[176]
TGF-β
Pyeloplasty
CMP
Increased pre-operative
Se 82%/Sp 86%
 
[182]
Generally, the primary group measured is children undergoing pyeloplasty; these children are then compared to healthy controls and/or conservatively managed children with PUJO (CMP). The exception in the studies listed in the table is labelled PUJO*, which includes children with conservatively managed PUJO split into ‘functional’ (t1/2 of renogram < 0 min) and ‘obstructed’ (t1/2 of renogram > 20 min). In these studies voided urine from children undergoing pyeloplasty was only obtained 42 months post-operative
aALP, Alkaline phosphatase; Ca19-9, carbohydrate antigen 19–9; CyC, cystatin-C; HO-1, heme oxygenase-1; γGT, gamma-glutamyl transferase; IP-10, interferon-γ-inducible protein 10; KIM-1, kidney injury molecule-1; MIP-1α, macrophage inflammatory protein-1α; NAG, N-acetyl-beta-D-glucosaminidase; NGAL, neutrophil gelatinase-associated lipocali; OPN, osteopontinn, RANTES, regulated on activation normal T-cell expressed and secreted
bWhere applicable sensitivity (Se), specificity (Sp) and accuracy (Ac) of the test at best threshold value from receiver operating characteristic curve analysis is presented
cTo detect differential renal function (DRF) of <40% out of all hydronephrosis cases
dTo detect pyeloplasty cases out of all hydronephrosis cases
eTo detect pyeloplasty cases out of all cases
Finding a suitable biomarker test with high sensitivity, specificity and predictive value is challenging [88], not least because these markers are excreted in health as well as disease, show significant intra- and inter-patient variation and may be affected by patient age, the presence of urinary tract infection and other renal disorders [174, 183].
A recent systematic review of urinary and serum biomarkers included 14 studies which reported data on 380 surgically managed PUJO patients, 174 conservatively managed patients and 213 controls [184]. This review reported a wide-range of sometimes conflicting results, and the authors were unable to draw any firm conclusions, attributing this to differences in study design with heterogeneous age groups, various or absent control groups and often short durations of follow-up [184].
More successfully, proteomics of neonatal urine has identified a panel of 51 peptides which distinguish obstruction severity. When implemented in a prospective blinded study it had an accuracy of 94% to predict future need for surgery in newborns with PUJO [185]. However, beyond 1 year of age the sensitivity and specificity of this proteome profile diminished significantly [186].
A single biomarker able to guide selection of patients for pyeloplasty has not yet been identified, indicating a panel of biomarkers may be necessary to achieve this.

Conclusions

Managing children with asymptomatic intrinsic PUJO is a significant challenge for clinicians. Animal and human studies have expanded our understanding of the molecular mechanisms involved in the aetiology of obstruction and in particular the progression of the renal insult. Upregulation of the RAAS and TGF-β expression are fundamental to renal injury, which is attenuated in animal models by therapeutic inhibition of these pathways. Much, however, remains to be learned in order to identify molecular markers and targets useful in the day-to-day diagnosis and management of this condition.

Future perspectives and unanswered questions in PUJO

  • What is the underlying aetiology of intrinsic congenital PUJO? Does this explain the variable outcome of PUJO and can this be targeted therapeutically?
  • Does individual ability to relieve intra-renal pressure determine disease progression?
  • Are therapies tested in animals applicable in children to limit renal injury?
  • Can urinary biomarkers improve early identification and thus outcome of children requiring pyeloplasty?

Compliance with ethical standards

Funding

Laura Jackson is funded by a joint Royal College of Surgeons of England/British Association of Paediatric Surgeons Fellowship (Awarded 2014). Richard Coward is funded by the Medical Research Council as a Senior Clinical Fellow MR/K010492/1.

Conflict of interest

The authors declare that they have no conflict of interest.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

© Springer Medizin

Bis 11. April 2024 bestellen und im ersten Jahr 50 % sparen!

e.Med Pädiatrie

Kombi-Abonnement

Mit e.Med Pädiatrie erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Pädiatrie, den Premium-Inhalten der pädiatrischen Fachzeitschriften, inklusive einer gedruckten Pädiatrie-Zeitschrift Ihrer Wahl.

© Springer Medizin

Bis 11. April 2024 bestellen und im ersten Jahr 50 % sparen!

e.Med Urologie

Kombi-Abonnement

Mit e.Med Urologie erhalten Sie Zugang zu den urologischen CME-Fortbildungen und Premium-Inhalten der urologischen Fachzeitschriften.

© Springer Medizin

Bis 11. April 2024 bestellen und im ersten Jahr 50 % sparen!

Literatur
1.
Zurück zum Zitat Dudley JA, Haworth JM, McGraw ME, Frank JD, Tizard EJ (1997) Clinical relevance and implications of antenatal hydronephrosis. Arch Dis Child Fetal Neonatal Ed 76:F31–34PubMedPubMedCentralCrossRef Dudley JA, Haworth JM, McGraw ME, Frank JD, Tizard EJ (1997) Clinical relevance and implications of antenatal hydronephrosis. Arch Dis Child Fetal Neonatal Ed 76:F31–34PubMedPubMedCentralCrossRef
2.
Zurück zum Zitat Brussels Free University Perinatal Nephrology Study Group, Ismaili K, Avni FE, Wissing KM, Hall M (2004) Long-term clinical outcome of infants with mild and moderate fetal pyelectasis: validation of neonatal ultrasound as a screening tool to detect significant nephrouropathies. J Pediatr 144:759–765 Brussels Free University Perinatal Nephrology Study Group, Ismaili K, Avni FE, Wissing KM, Hall M (2004) Long-term clinical outcome of infants with mild and moderate fetal pyelectasis: validation of neonatal ultrasound as a screening tool to detect significant nephrouropathies. J Pediatr 144:759–765
3.
Zurück zum Zitat Jaswon MS, Dibble L, Puri S, Davis J, Young J, Dave R, Morgan H (1999) Prospective study of outcome in antenatally diagnosed renal pelvis dilatation. Arch Dis Child Fetal Neonatal Ed 80:F135–138PubMedPubMedCentralCrossRef Jaswon MS, Dibble L, Puri S, Davis J, Young J, Dave R, Morgan H (1999) Prospective study of outcome in antenatally diagnosed renal pelvis dilatation. Arch Dis Child Fetal Neonatal Ed 80:F135–138PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Woodward M, Frank D (2002) Postnatal management of antenatal hydronephrosis. BJU Int 89:149–156PubMedCrossRef Woodward M, Frank D (2002) Postnatal management of antenatal hydronephrosis. BJU Int 89:149–156PubMedCrossRef
5.
Zurück zum Zitat Morris RK, Kilby MD (2008) Congenital urinary tract obstruction. Best Pract Res Clin Obstet Gynaecol 22:97–122PubMedCrossRef Morris RK, Kilby MD (2008) Congenital urinary tract obstruction. Best Pract Res Clin Obstet Gynaecol 22:97–122PubMedCrossRef
6.
Zurück zum Zitat Corbett HJ, McCarthy L (2013) Hydronephrosis in children: pelviureteric junction dysfunction. Surgery (Oxford) 31:135–139CrossRef Corbett HJ, McCarthy L (2013) Hydronephrosis in children: pelviureteric junction dysfunction. Surgery (Oxford) 31:135–139CrossRef
7.
Zurück zum Zitat Hashim H, Woodhouse CRJ (2012) Ureteropelvic junction obstruction. Eur Urol Suppl 11:25–32CrossRef Hashim H, Woodhouse CRJ (2012) Ureteropelvic junction obstruction. Eur Urol Suppl 11:25–32CrossRef
8.
Zurück zum Zitat Ulman I, Jayanthi VR, Koff SA (2000) The long-term followup of newborns with severe unilateral hydronephrosis initially treated nonoperatively. J Urol 164:1101–1105PubMedCrossRef Ulman I, Jayanthi VR, Koff SA (2000) The long-term followup of newborns with severe unilateral hydronephrosis initially treated nonoperatively. J Urol 164:1101–1105PubMedCrossRef
9.
Zurück zum Zitat Hafez AT, McLorie G, Bagli D, Khoury A (2002) Analysis of trends on serial ultrasound for high grade neonatal hydronephrosis. J Urol 168(4 Pt 1):1518–1521PubMedCrossRef Hafez AT, McLorie G, Bagli D, Khoury A (2002) Analysis of trends on serial ultrasound for high grade neonatal hydronephrosis. J Urol 168(4 Pt 1):1518–1521PubMedCrossRef
10.
Zurück zum Zitat Chertin B, Pollack A, Koulikov D, Rabinowitz R, Hain D, Hadas-Halpren I, Farkas A (2006) Conservative treatment of ureteropelvic junction obstruction in children with antenatal diagnosis of hydronephrosis: lessons learned after 16 years of follow-up. Eur Urol 49:734–738PubMedCrossRef Chertin B, Pollack A, Koulikov D, Rabinowitz R, Hain D, Hadas-Halpren I, Farkas A (2006) Conservative treatment of ureteropelvic junction obstruction in children with antenatal diagnosis of hydronephrosis: lessons learned after 16 years of follow-up. Eur Urol 49:734–738PubMedCrossRef
11.
Zurück zum Zitat Chevalier RL, Thornhill BA, Forbes MS, Kiley SC (2010) Mechanisms of renal injury and progression of renal disease in congenital obstructive nephropathy. Pediatr Nephrol 25:687–697PubMedCrossRef Chevalier RL, Thornhill BA, Forbes MS, Kiley SC (2010) Mechanisms of renal injury and progression of renal disease in congenital obstructive nephropathy. Pediatr Nephrol 25:687–697PubMedCrossRef
12.
Zurück zum Zitat Gordon I, Dhillon HK, Gatanash H, Peters AM (1991) Antenatal diagnosis of pelvic hydronephrosis: assessment of renal function and drainage as a guide to management. J Nucl Med 32:1649–1654PubMed Gordon I, Dhillon HK, Gatanash H, Peters AM (1991) Antenatal diagnosis of pelvic hydronephrosis: assessment of renal function and drainage as a guide to management. J Nucl Med 32:1649–1654PubMed
13.
Zurück zum Zitat Ransley PG, Dhillon HK, Gordon I, Duffy PG, Dillon MJ, Barratt TM (1990) The postnatal management of hydronephrosis diagnosed by prenatal ultrasound. The J Urol 144(2 Pt 2):584–587, discussion 593–584PubMedCrossRef Ransley PG, Dhillon HK, Gordon I, Duffy PG, Dillon MJ, Barratt TM (1990) The postnatal management of hydronephrosis diagnosed by prenatal ultrasound. The J Urol 144(2 Pt 2):584–587, discussion 593–584PubMedCrossRef
14.
Zurück zum Zitat Csaicsich D, Greenbaum LA, Aufricht C (2004) Upper urinary tract: when is obstruction obstruction? Curr Opin Urol 14:213–217PubMedCrossRef Csaicsich D, Greenbaum LA, Aufricht C (2004) Upper urinary tract: when is obstruction obstruction? Curr Opin Urol 14:213–217PubMedCrossRef
15.
Zurück zum Zitat Stringer MD, Yassaie S (2013) Is the pelviureteric junction an anatomical entity? J Pediatr Urol 9:123–128PubMedCrossRef Stringer MD, Yassaie S (2013) Is the pelviureteric junction an anatomical entity? J Pediatr Urol 9:123–128PubMedCrossRef
16.
Zurück zum Zitat Shafik A, Al-Sherif A (1999) Ureteropelvic junction: a study of its anatomical structure and function. Ureteropelvic junction sphincter? Eur Urol 36:150–156, discussion 156–157PubMedCrossRef Shafik A, Al-Sherif A (1999) Ureteropelvic junction: a study of its anatomical structure and function. Ureteropelvic junction sphincter? Eur Urol 36:150–156, discussion 156–157PubMedCrossRef
17.
Zurück zum Zitat Santicioli P, Maggi CA (1998) Myogenic and neurogenic factors in the control of pyeloureteral motility and ureteral peristalsis. Pharmacol Rev 50:683–722PubMed Santicioli P, Maggi CA (1998) Myogenic and neurogenic factors in the control of pyeloureteral motility and ureteral peristalsis. Pharmacol Rev 50:683–722PubMed
18.
Zurück zum Zitat Nemeth L, O’Briain DS, Puri P (2001) Demonstration of neuronal networks in the human upper urinary tract using confocal laser scanning microscopy. J Urol 166:255–258PubMedCrossRef Nemeth L, O’Briain DS, Puri P (2001) Demonstration of neuronal networks in the human upper urinary tract using confocal laser scanning microscopy. J Urol 166:255–258PubMedCrossRef
19.
Zurück zum Zitat Takasato M, Little MH (2015) The origin of the mammalian kidney: implications for recreating the kidney in vitro. Development 142:1937–1947PubMedCrossRef Takasato M, Little MH (2015) The origin of the mammalian kidney: implications for recreating the kidney in vitro. Development 142:1937–1947PubMedCrossRef
20.
21.
Zurück zum Zitat Yu J, Carroll TJ, McMahon AP (2002) Sonic hedgehog regulates proliferation and differentiation of mesenchymal cells in the mouse metanephric kidney. Development 129:5301–5312PubMed Yu J, Carroll TJ, McMahon AP (2002) Sonic hedgehog regulates proliferation and differentiation of mesenchymal cells in the mouse metanephric kidney. Development 129:5301–5312PubMed
22.
Zurück zum Zitat Brenner-Anantharam A, Cebrian C, Guillaume R, Hurtado R, Sun TT, Herzlinger D (2007) Tailbud-derived mesenchyme promotes urinary tract segmentation via BMP4 signaling. Development 134:1967–1975PubMedCrossRef Brenner-Anantharam A, Cebrian C, Guillaume R, Hurtado R, Sun TT, Herzlinger D (2007) Tailbud-derived mesenchyme promotes urinary tract segmentation via BMP4 signaling. Development 134:1967–1975PubMedCrossRef
23.
Zurück zum Zitat Caubit X, Lye CM, Martin E, Core N, Long DA, Vola C, Jenkins D, Garratt AN, Skaer H, Woolf AS, Fasano L (2008) Teashirt 3 is necessary for ureteral smooth muscle differentiation downstream of SHH and BMP4. Development 135:3301–3310PubMedCrossRef Caubit X, Lye CM, Martin E, Core N, Long DA, Vola C, Jenkins D, Garratt AN, Skaer H, Woolf AS, Fasano L (2008) Teashirt 3 is necessary for ureteral smooth muscle differentiation downstream of SHH and BMP4. Development 135:3301–3310PubMedCrossRef
24.
25.
Zurück zum Zitat Mahoney ZX, Sammut B, Xavier RJ, Cunningham J, Go G, Brim KL, Stappenbeck TS, Miner JH, Swat W (2006) Discs-large homolog 1 regulates smooth muscle orientation in the mouse ureter. Proc Natl Acad Sci USA 103:19872–19877 Mahoney ZX, Sammut B, Xavier RJ, Cunningham J, Go G, Brim KL, Stappenbeck TS, Miner JH, Swat W (2006) Discs-large homolog 1 regulates smooth muscle orientation in the mouse ureter. Proc Natl Acad Sci USA 103:19872–19877
27.
Zurück zum Zitat Alcaraz A, Vinaixa F, Tejedo-Mateu A, Fores MM, Gotzens V, Mestres CA, Oliveira J, Carretero P (1991) Obstruction and recanalization of the ureter during embryonic development. J Urol 145:410–416PubMedCrossRef Alcaraz A, Vinaixa F, Tejedo-Mateu A, Fores MM, Gotzens V, Mestres CA, Oliveira J, Carretero P (1991) Obstruction and recanalization of the ureter during embryonic development. J Urol 145:410–416PubMedCrossRef
28.
29.
Zurück zum Zitat Matsuno T, Tokunaka S, Koyanagi T (1984) Muscular development in the urinary tract. J Urol 132:148–152PubMedCrossRef Matsuno T, Tokunaka S, Koyanagi T (1984) Muscular development in the urinary tract. J Urol 132:148–152PubMedCrossRef
30.
Zurück zum Zitat Chang CP, McDill BW, Neilson JR, Joist HE, Epstein JA, Crabtree GR, Chen F (2004) Calcineurin is required in urinary tract mesenchyme for the development of the pyeloureteral peristaltic machinery. J Clin Invest 113:1051–1058PubMedPubMedCentralCrossRef Chang CP, McDill BW, Neilson JR, Joist HE, Epstein JA, Crabtree GR, Chen F (2004) Calcineurin is required in urinary tract mesenchyme for the development of the pyeloureteral peristaltic machinery. J Clin Invest 113:1051–1058PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Miyazaki Y, Tsuchida S, Nishimura H, Pope JC 4th, Harris RC, McKanna JM, Inagami T, Hogan BL, Fogo A, Ichikawa I (1998) Angiotensin induces the urinary peristaltic machinery during the perinatal period. J Clin Invest 102:1489–1497PubMedPubMedCentralCrossRef Miyazaki Y, Tsuchida S, Nishimura H, Pope JC 4th, Harris RC, McKanna JM, Inagami T, Hogan BL, Fogo A, Ichikawa I (1998) Angiotensin induces the urinary peristaltic machinery during the perinatal period. J Clin Invest 102:1489–1497PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Zhang PL, Peters CA, Rosen S (2000) Ureteropelvic junction obstruction: morphological and clinical studies. Pediatr Nephrol 14:820–826PubMedCrossRef Zhang PL, Peters CA, Rosen S (2000) Ureteropelvic junction obstruction: morphological and clinical studies. Pediatr Nephrol 14:820–826PubMedCrossRef
33.
Zurück zum Zitat Murakumo M, Nonomura K, Yamashita T, Ushiki T, Abe K, Koyanagi T (1997) Structural changes of collagen components and diminution of nerves in congenital ureteropelvic junction obstruction. J Urol 157:1963–1968PubMedCrossRef Murakumo M, Nonomura K, Yamashita T, Ushiki T, Abe K, Koyanagi T (1997) Structural changes of collagen components and diminution of nerves in congenital ureteropelvic junction obstruction. J Urol 157:1963–1968PubMedCrossRef
34.
Zurück zum Zitat Demirbilek S, Edali MN, Gurunluoglu K, Turkmen E, Tas E, Karaman A, Akin M, Aksoy RT, Celbis O, Uzun I (2006) Glial cell line-derived neurotrophic factor and synaptophysin expression in pelviureteral junction obstruction. Urology 67:400–405PubMedCrossRef Demirbilek S, Edali MN, Gurunluoglu K, Turkmen E, Tas E, Karaman A, Akin M, Aksoy RT, Celbis O, Uzun I (2006) Glial cell line-derived neurotrophic factor and synaptophysin expression in pelviureteral junction obstruction. Urology 67:400–405PubMedCrossRef
35.
Zurück zum Zitat Ozel SK, Emir H, Dervisoglu S, Akpolat N, Senel B, Kazez A, Soylet Y, Cetin G, Danismend N, Buyukunal SN (2010) The roles of extracellular matrix proteins, apoptosis and c-kit positive cells in the pathogenesis of ureteropelvic junction obstruction. J Pediatr Urol 6:125–129PubMedCrossRef Ozel SK, Emir H, Dervisoglu S, Akpolat N, Senel B, Kazez A, Soylet Y, Cetin G, Danismend N, Buyukunal SN (2010) The roles of extracellular matrix proteins, apoptosis and c-kit positive cells in the pathogenesis of ureteropelvic junction obstruction. J Pediatr Urol 6:125–129PubMedCrossRef
36.
Zurück zum Zitat Kaneto H, Orikasa S, Chiba T, Takahashi T (1991) Three-D muscular arrangement at the ureteropelvic junction and its changes in congenital hydronephrosis: a stereo-morphometric study. J Urol 146:909–914PubMedCrossRef Kaneto H, Orikasa S, Chiba T, Takahashi T (1991) Three-D muscular arrangement at the ureteropelvic junction and its changes in congenital hydronephrosis: a stereo-morphometric study. J Urol 146:909–914PubMedCrossRef
37.
Zurück zum Zitat Hosgor M, Karaca I, Ulukus C, Ozer E, Ozkara E, Sam B, Ucan B, Kurtulus S, Karkiner A, Temir G (2005) Structural changes of smooth muscle in congenital ureteropelvic junction obstruction. J Pediatr Surg 40:1632–1636PubMedCrossRef Hosgor M, Karaca I, Ulukus C, Ozer E, Ozkara E, Sam B, Ucan B, Kurtulus S, Karkiner A, Temir G (2005) Structural changes of smooth muscle in congenital ureteropelvic junction obstruction. J Pediatr Surg 40:1632–1636PubMedCrossRef
38.
Zurück zum Zitat Aoki Y, Mori S, Kitajima K, Yokoyama O, Kanamaru H, Okada K, Yokota Y (2004) Id2 haploinsufficiency in mice leads to congenital hydronephrosis resembling that in humans. Genes Cells 9:1287–1296PubMedCrossRef Aoki Y, Mori S, Kitajima K, Yokoyama O, Kanamaru H, Okada K, Yokota Y (2004) Id2 haploinsufficiency in mice leads to congenital hydronephrosis resembling that in humans. Genes Cells 9:1287–1296PubMedCrossRef
39.
Zurück zum Zitat Chevalier RL, Kim A, Thornhill BA, Wolstenholme JT (1999) Recovery following relief of unilateral ureteral obstruction in the neonatal rat. Kidney Int 55:793–807PubMedCrossRef Chevalier RL, Kim A, Thornhill BA, Wolstenholme JT (1999) Recovery following relief of unilateral ureteral obstruction in the neonatal rat. Kidney Int 55:793–807PubMedCrossRef
40.
Zurück zum Zitat Chevalier RL, Thornhill BA, Wolstenholme JT (1999) Renal cellular response to ureteral obstruction: role of maturation and angiotensin II. Am J Physiol 277(1 Pt 2):F41–47PubMed Chevalier RL, Thornhill BA, Wolstenholme JT (1999) Renal cellular response to ureteral obstruction: role of maturation and angiotensin II. Am J Physiol 277(1 Pt 2):F41–47PubMed
41.
Zurück zum Zitat Forbes MS, Thornhill BA, Chevalier RL (2011) Proximal tubular injury and rapid formation of atubular glomeruli in mice with unilateral ureteral obstruction: a new look at an old model. Am J Physiol Ren Physiol 301:F110–117CrossRef Forbes MS, Thornhill BA, Chevalier RL (2011) Proximal tubular injury and rapid formation of atubular glomeruli in mice with unilateral ureteral obstruction: a new look at an old model. Am J Physiol Ren Physiol 301:F110–117CrossRef
42.
Zurück zum Zitat Forbes MS, Thornhill BA, Galarreta CI, Minor JJ, Gordon KA, Chevalier RL (2013) Chronic unilateral ureteral obstruction in the neonatal mouse delays maturation of both kidneys and leads to late formation of atubular glomeruli. Am J Physiol Ren Physiol 305:F1736–1746CrossRef Forbes MS, Thornhill BA, Galarreta CI, Minor JJ, Gordon KA, Chevalier RL (2013) Chronic unilateral ureteral obstruction in the neonatal mouse delays maturation of both kidneys and leads to late formation of atubular glomeruli. Am J Physiol Ren Physiol 305:F1736–1746CrossRef
43.
Zurück zum Zitat Lange-Sperandio B, Cachat F, Thornhill BA, Chevalier RL (2002) Selectins mediate macrophage infiltration in obstructive nephropathy in newborn mice. Kidney Int 61:516–524PubMedCrossRef Lange-Sperandio B, Cachat F, Thornhill BA, Chevalier RL (2002) Selectins mediate macrophage infiltration in obstructive nephropathy in newborn mice. Kidney Int 61:516–524PubMedCrossRef
44.
Zurück zum Zitat Lange-Sperandio B, Schimpgen K, Rodenbeck B, Chavakis T, Bierhaus A, Nawroth P, Thornhill B, Schaefer F, Chevalier RL (2006) Distinct roles of Mac-1 and its counter-receptors in neonatal obstructive nephropathy. Kidney Int 69:81–88PubMedCrossRef Lange-Sperandio B, Schimpgen K, Rodenbeck B, Chavakis T, Bierhaus A, Nawroth P, Thornhill B, Schaefer F, Chevalier RL (2006) Distinct roles of Mac-1 and its counter-receptors in neonatal obstructive nephropathy. Kidney Int 69:81–88PubMedCrossRef
45.
Zurück zum Zitat Esteban V, Lorenzo O, Ruperez M, Suzuki Y, Mezzano S, Blanco J, Kretzler M, Sugaya T, Egido J, Ruiz-Ortega M (2004) Angiotensin II, via AT1 and AT2 receptors and NF-kappaB pathway, regulates the inflammatory response in unilateral ureteral obstruction. J Am Soc Nephrol 15:1514–1529PubMedCrossRef Esteban V, Lorenzo O, Ruperez M, Suzuki Y, Mezzano S, Blanco J, Kretzler M, Sugaya T, Egido J, Ruiz-Ortega M (2004) Angiotensin II, via AT1 and AT2 receptors and NF-kappaB pathway, regulates the inflammatory response in unilateral ureteral obstruction. J Am Soc Nephrol 15:1514–1529PubMedCrossRef
46.
Zurück zum Zitat Thornhill BA, Burt LE, Chen C, Forbes MS, Chevalier RL (2005) Variable chronic partial ureteral obstruction in the neonatal rat: a new model of ureteropelvic junction obstruction. Kidney Int 67:42–52PubMedCrossRef Thornhill BA, Burt LE, Chen C, Forbes MS, Chevalier RL (2005) Variable chronic partial ureteral obstruction in the neonatal rat: a new model of ureteropelvic junction obstruction. Kidney Int 67:42–52PubMedCrossRef
47.
Zurück zum Zitat Thornhill BA, Forbes MS, Marcinko ES, Chevalier RL (2007) Glomerulotubular disconnection in neonatal mice after relief of partial ureteral obstruction. Kidney Int 72:1103–1112PubMedCrossRef Thornhill BA, Forbes MS, Marcinko ES, Chevalier RL (2007) Glomerulotubular disconnection in neonatal mice after relief of partial ureteral obstruction. Kidney Int 72:1103–1112PubMedCrossRef
48.
Zurück zum Zitat Klein J, Gonzalez J, Miravete M, Caubet C, Chaaya R, Decramer S, Bandin F, Bascands JL, Buffin-Meyer B, Schanstra JP (2011) Congenital ureteropelvic junction obstruction: human disease and animal models. Int J Exp Pathol 92:168–192PubMedPubMedCentralCrossRef Klein J, Gonzalez J, Miravete M, Caubet C, Chaaya R, Decramer S, Bandin F, Bascands JL, Buffin-Meyer B, Schanstra JP (2011) Congenital ureteropelvic junction obstruction: human disease and animal models. Int J Exp Pathol 92:168–192PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Wang Y, Puri P, Hassan J, Miyakita H, Reen DJ (1995) Abnormal innervation and altered nerve growth factor messenger ribonucleic acid expression in ureteropelvic junction obstruction. J Urol 154:679–683PubMedCrossRef Wang Y, Puri P, Hassan J, Miyakita H, Reen DJ (1995) Abnormal innervation and altered nerve growth factor messenger ribonucleic acid expression in ureteropelvic junction obstruction. J Urol 154:679–683PubMedCrossRef
50.
Zurück zum Zitat Cutroneo G, Arena S, Anastasi G, Cervellione RM, Grimaldi S, Di Mauro D, Speciale F, Arena F, Di Benedetto V, Favaloro A, Magno C (2011) Altered cytoskeletal structure of smooth muscle cells in ureteropelvic junction obstruction. J Urol 185:2314–2319PubMedCrossRef Cutroneo G, Arena S, Anastasi G, Cervellione RM, Grimaldi S, Di Mauro D, Speciale F, Arena F, Di Benedetto V, Favaloro A, Magno C (2011) Altered cytoskeletal structure of smooth muscle cells in ureteropelvic junction obstruction. J Urol 185:2314–2319PubMedCrossRef
51.
Zurück zum Zitat Esther CR Jr, Howard TE, Marino EM, Goddard JM, Capecchi MR, Bernstein KE (1996) Mice lacking angiotensin-converting enzyme have low blood pressure, renal pathology, and reduced male fertility. Lab Invest 74:953–965PubMed Esther CR Jr, Howard TE, Marino EM, Goddard JM, Capecchi MR, Bernstein KE (1996) Mice lacking angiotensin-converting enzyme have low blood pressure, renal pathology, and reduced male fertility. Lab Invest 74:953–965PubMed
52.
Zurück zum Zitat Shindo T, Kurihara H, Kuno K, Yokoyama H, Wada T, Kurihara Y, Imai T, Wang Y, Ogata M, Nishimatsu H, Moriyama N, Oh-hashi Y, Morita H, Ishikawa T, Nagai R, Yazaki Y, Matsushima K (2000) ADAMTS-1: a metalloproteinase-disintegrin essential for normal growth, fertility, and organ morphology and function. J Clin Invest 105:1345–1352PubMedPubMedCentralCrossRef Shindo T, Kurihara H, Kuno K, Yokoyama H, Wada T, Kurihara Y, Imai T, Wang Y, Ogata M, Nishimatsu H, Moriyama N, Oh-hashi Y, Morita H, Ishikawa T, Nagai R, Yazaki Y, Matsushima K (2000) ADAMTS-1: a metalloproteinase-disintegrin essential for normal growth, fertility, and organ morphology and function. J Clin Invest 105:1345–1352PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Nagata M, Tanimoto K, Fukamizu A, Kon Y, Sugiyama F, Yagami K, Murakami K, Watanabe T (1996) Nephrogenesis and renovascular development in angiotensinogen-deficient mice. Lab Invest 75:745–753PubMed Nagata M, Tanimoto K, Fukamizu A, Kon Y, Sugiyama F, Yagami K, Murakami K, Watanabe T (1996) Nephrogenesis and renovascular development in angiotensinogen-deficient mice. Lab Invest 75:745–753PubMed
54.
Zurück zum Zitat McDill BW, Li SZ, Kovach PA, Ding L, Chen F (2006) Congenital progressive hydronephrosis (cph) is caused by an S256L mutation in aquaporin-2 that affects its phosphorylation and apical membrane accumulation. Proc Natl Acad Sci USA 103:6952–6957PubMedPubMedCentralCrossRef McDill BW, Li SZ, Kovach PA, Ding L, Chen F (2006) Congenital progressive hydronephrosis (cph) is caused by an S256L mutation in aquaporin-2 that affects its phosphorylation and apical membrane accumulation. Proc Natl Acad Sci USA 103:6952–6957PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Lu W, Quintero-Rivera F, Fan Y, Alkuraya FS, Donovan DJ, Xi Q, Turbe-Doan A, Li QG, Campbell CG, Shanske AL, Sherr EH, Ahmad A, Peters R, Rilliet B, Parvex P, Bassuk AG, Harris DJ, Ferguson H, Kelly C, Walsh CA, Gronostajski RM, Devriendt K, Higgins A, Ligon AH, Quade BJ, Morton CC, Gusella JF, Maas RL (2007) NFIA haploinsufficiency is associated with a CNS malformation syndrome and urinary tract defects. PLoS Genet 3:e80PubMedPubMedCentralCrossRef Lu W, Quintero-Rivera F, Fan Y, Alkuraya FS, Donovan DJ, Xi Q, Turbe-Doan A, Li QG, Campbell CG, Shanske AL, Sherr EH, Ahmad A, Peters R, Rilliet B, Parvex P, Bassuk AG, Harris DJ, Ferguson H, Kelly C, Walsh CA, Gronostajski RM, Devriendt K, Higgins A, Ligon AH, Quade BJ, Morton CC, Gusella JF, Maas RL (2007) NFIA haploinsufficiency is associated with a CNS malformation syndrome and urinary tract defects. PLoS Genet 3:e80PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Vivante A, Kleppa MJ, Schulz J, Kohl S, Sharma A, Chen J, Shril S, Hwang DY, Weiss AC, Kaminski MM, Shukrun R, Kemper MJ, Lehnhardt A, Beetz R, Sanna-Cherchi S, Verbitsky M, Gharavi AG, Stuart HM, Feather SA, Goodship JA, Goodship TH, Woolf AS, Westra SJ, Doody DP, Bauer SB, Lee RS, Adam RM, Lu W, Reutter HM, Kehinde EO, Mancini EJ, Lifton RP, Tasic V, Lienkamp SS, Juppner H, Kispert A, Hildebrandt F (2015) Mutations in TBX18 cause dominant urinary tract malformations via transcriptional dysregulation of ureter development. Am J Hum Genet 97:291–301PubMedPubMedCentralCrossRef Vivante A, Kleppa MJ, Schulz J, Kohl S, Sharma A, Chen J, Shril S, Hwang DY, Weiss AC, Kaminski MM, Shukrun R, Kemper MJ, Lehnhardt A, Beetz R, Sanna-Cherchi S, Verbitsky M, Gharavi AG, Stuart HM, Feather SA, Goodship JA, Goodship TH, Woolf AS, Westra SJ, Doody DP, Bauer SB, Lee RS, Adam RM, Lu W, Reutter HM, Kehinde EO, Mancini EJ, Lifton RP, Tasic V, Lienkamp SS, Juppner H, Kispert A, Hildebrandt F (2015) Mutations in TBX18 cause dominant urinary tract malformations via transcriptional dysregulation of ureter development. Am J Hum Genet 97:291–301PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Jenkins D, Caubit X, Dimovski A, Matevska N, Lye CM, Cabuk F, Gucev Z, Tasic V, Fasano L, Woolf AS (2010) Analysis of TSHZ2 and TSHZ3 genes in congenital pelvi-ureteric junction obstruction. Nephrol Dial Transplant 25:54–60PubMedCrossRef Jenkins D, Caubit X, Dimovski A, Matevska N, Lye CM, Cabuk F, Gucev Z, Tasic V, Fasano L, Woolf AS (2010) Analysis of TSHZ2 and TSHZ3 genes in congenital pelvi-ureteric junction obstruction. Nephrol Dial Transplant 25:54–60PubMedCrossRef
58.
Zurück zum Zitat Lye CM, Fasano L, Woolf AS (2010) Ureter myogenesis: putting Teashirt into context. J Am Soc Nephrol 21:24–30PubMedCrossRef Lye CM, Fasano L, Woolf AS (2010) Ureter myogenesis: putting Teashirt into context. J Am Soc Nephrol 21:24–30PubMedCrossRef
59.
Zurück zum Zitat Pope JC 4th, Showalter PR, Milam DF, Brock JW (1994) Intrapelvic pressure monitoring in the partially obstructed porcine kidney. Urology 44:565–571PubMedCrossRef Pope JC 4th, Showalter PR, Milam DF, Brock JW (1994) Intrapelvic pressure monitoring in the partially obstructed porcine kidney. Urology 44:565–571PubMedCrossRef
60.
Zurück zum Zitat Holden D, George NJ, Rickards D, Barnard RJ, O’Reilly PH (1984) Renal pelvic pressures in human chronic obstructive uropathy. Br J Urol 56:565–570PubMedCrossRef Holden D, George NJ, Rickards D, Barnard RJ, O’Reilly PH (1984) Renal pelvic pressures in human chronic obstructive uropathy. Br J Urol 56:565–570PubMedCrossRef
61.
62.
Zurück zum Zitat Chevalier RL (1984) Chronic partial ureteral obstruction in the neonatal guinea-pig. 2. Pressure-gradients affecting glomerular-filtration rate. Pediatr Res 18:1271–1277 Chevalier RL (1984) Chronic partial ureteral obstruction in the neonatal guinea-pig. 2. Pressure-gradients affecting glomerular-filtration rate. Pediatr Res 18:1271–1277
63.
Zurück zum Zitat Yoo KH, Norwood VF, el-Dahr SS, Yosipiv I, Chevalier RL (1997) Regulation of angiotensin II AT1 and AT2 receptors in neonatal ureteral obstruction. Am J Physiol 273:R503–509PubMed Yoo KH, Norwood VF, el-Dahr SS, Yosipiv I, Chevalier RL (1997) Regulation of angiotensin II AT1 and AT2 receptors in neonatal ureteral obstruction. Am J Physiol 273:R503–509PubMed
64.
Zurück zum Zitat el-Dahr SS, Gee J, Dipp S, Hanss BG, Vari RC, Chao J (1993) Upregulation of renin–angiotensin system and downregulation of kallikrein in obstructive nephropathy. Am J Physiol 264:F874–881PubMed el-Dahr SS, Gee J, Dipp S, Hanss BG, Vari RC, Chao J (1993) Upregulation of renin–angiotensin system and downregulation of kallikrein in obstructive nephropathy. Am J Physiol 264:F874–881PubMed
65.
Zurück zum Zitat Durvasula RV, Petermann AT, Hiromura K, Blonski M, Pippin J, Mundel P, Pichler R, Griffin S, Couser WG, Shankland SJ (2004) Activation of a local tissue angiotensin system in podocytes by mechanical strain. Kidney Int 65:30–39PubMedCrossRef Durvasula RV, Petermann AT, Hiromura K, Blonski M, Pippin J, Mundel P, Pichler R, Griffin S, Couser WG, Shankland SJ (2004) Activation of a local tissue angiotensin system in podocytes by mechanical strain. Kidney Int 65:30–39PubMedCrossRef
66.
Zurück zum Zitat Taneda S, Hudkins KL, Topouzis S, Gilbertson DG, Ophascharoensuk V, Truong L, Johnson RJ, Alpers CE (2003) Obstructive uropathy in mice and humans: potential role for PDGF-D in the progression of tubulointerstitial injury. J Am Soc Nephrol 14:2544–2555PubMedCrossRef Taneda S, Hudkins KL, Topouzis S, Gilbertson DG, Ophascharoensuk V, Truong L, Johnson RJ, Alpers CE (2003) Obstructive uropathy in mice and humans: potential role for PDGF-D in the progression of tubulointerstitial injury. J Am Soc Nephrol 14:2544–2555PubMedCrossRef
67.
Zurück zum Zitat Manucha W, Kurban F, Mazzei L, Benardon ME, Bocanegra V, Tosi MR, Valles P (2011) eNOS/Hsp70 interaction on rosuvastatin cytoprotective effect in neonatal obstructive nephropathy. Eur J Pharmacol 650:487–495PubMedCrossRef Manucha W, Kurban F, Mazzei L, Benardon ME, Bocanegra V, Tosi MR, Valles P (2011) eNOS/Hsp70 interaction on rosuvastatin cytoprotective effect in neonatal obstructive nephropathy. Eur J Pharmacol 650:487–495PubMedCrossRef
68.
Zurück zum Zitat Misseri R, Meldrum DR, Dinarello CA, Dagher P, Hile KL, Rink RC, Meldrum KK (2005) TNF-alpha mediates obstruction-induced renal tubular cell apoptosis and proapoptotic signaling. Am J Physiol Ren Physiol 288:F406–411CrossRef Misseri R, Meldrum DR, Dinarello CA, Dagher P, Hile KL, Rink RC, Meldrum KK (2005) TNF-alpha mediates obstruction-induced renal tubular cell apoptosis and proapoptotic signaling. Am J Physiol Ren Physiol 288:F406–411CrossRef
69.
Zurück zum Zitat Norregaard R, Jensen BL, Li C, Wang W, Knepper MA, Nielsen S, Frokiaer J (2005) COX-2 inhibition prevents downregulation of key renal water and sodium transport proteins in response to bilateral ureteral obstruction. Am J Physiol Ren Physiol 289:F322–333CrossRef Norregaard R, Jensen BL, Li C, Wang W, Knepper MA, Nielsen S, Frokiaer J (2005) COX-2 inhibition prevents downregulation of key renal water and sodium transport proteins in response to bilateral ureteral obstruction. Am J Physiol Ren Physiol 289:F322–333CrossRef
70.
Zurück zum Zitat Nilsson L, Madsen K, Krag S, Frokiaer J, Jensen BL, Norregaard R (2015) Disruption of cyclooxygenase type 2 exacerbates apoptosis and renal damage during obstructive nephropathy. Am J Physiol Ren Physiol 309:F1035–1048CrossRef Nilsson L, Madsen K, Krag S, Frokiaer J, Jensen BL, Norregaard R (2015) Disruption of cyclooxygenase type 2 exacerbates apoptosis and renal damage during obstructive nephropathy. Am J Physiol Ren Physiol 309:F1035–1048CrossRef
71.
Zurück zum Zitat Chung KH, Chevalier RL (1996) Arrested development of the neonatal kidney following chronic ureteral obstruction. J Urol 155:1139–1144PubMedCrossRef Chung KH, Chevalier RL (1996) Arrested development of the neonatal kidney following chronic ureteral obstruction. J Urol 155:1139–1144PubMedCrossRef
72.
Zurück zum Zitat Bartoli F, Gesualdo L, Paradies G, Caldarulo E, Infante B, Grandaliano G, Monno R, Leggio S, Salzillo F, Schena FP, Leggio A (2000) Renal expression of monocyte chemotactic protein-1 and epidermal growth factor in children with obstructive hydronephrosis. J Pediatr Surg 35:569–572PubMedCrossRef Bartoli F, Gesualdo L, Paradies G, Caldarulo E, Infante B, Grandaliano G, Monno R, Leggio S, Salzillo F, Schena FP, Leggio A (2000) Renal expression of monocyte chemotactic protein-1 and epidermal growth factor in children with obstructive hydronephrosis. J Pediatr Surg 35:569–572PubMedCrossRef
73.
Zurück zum Zitat Yang Y, Hou Y, Wang CL, Ji SJ (2006) Renal expression of epidermal growth factor and transforming growth factor-beta1 in children with congenital hydronephrosis. Urology 67:817–821, discussion 821–812PubMedCrossRef Yang Y, Hou Y, Wang CL, Ji SJ (2006) Renal expression of epidermal growth factor and transforming growth factor-beta1 in children with congenital hydronephrosis. Urology 67:817–821, discussion 821–812PubMedCrossRef
74.
Zurück zum Zitat Cai G, Zhang X, Hong Q, Shao F, Shang X, Fu B, Feng Z, Lin H, Wang J, Shi S, Yin Z, Chen X (2008) Tissue inhibitor of metalloproteinase-1 exacerbated renal interstitial fibrosis through enhancing inflammation. Nephrol Dial Transplant 23:1861–1875PubMedCrossRef Cai G, Zhang X, Hong Q, Shao F, Shang X, Fu B, Feng Z, Lin H, Wang J, Shi S, Yin Z, Chen X (2008) Tissue inhibitor of metalloproteinase-1 exacerbated renal interstitial fibrosis through enhancing inflammation. Nephrol Dial Transplant 23:1861–1875PubMedCrossRef
75.
Zurück zum Zitat Yeh YC, Wei WC, Wang YK, Lin SC, Sung JM, Tang MJ (2010) Transforming growth factor-{beta}1 induces Smad3-dependent {beta}1 integrin gene expression in epithelial-to-mesenchymal transition during chronic tubulointerstitial fibrosis. Am J Pathol 177:1743–1754PubMedPubMedCentralCrossRef Yeh YC, Wei WC, Wang YK, Lin SC, Sung JM, Tang MJ (2010) Transforming growth factor-{beta}1 induces Smad3-dependent {beta}1 integrin gene expression in epithelial-to-mesenchymal transition during chronic tubulointerstitial fibrosis. Am J Pathol 177:1743–1754PubMedPubMedCentralCrossRef
76.
Zurück zum Zitat Hamzeh MT, Sridhara R, Alexander LD (2015) Cyclic stretch-induced TGF-beta1 and fibronectin expression is mediated by beta1-integrin through c-Src- and STAT3-dependent pathways in renal epithelial cells. Am J Physiol Ren Physiol 308:F425–436CrossRef Hamzeh MT, Sridhara R, Alexander LD (2015) Cyclic stretch-induced TGF-beta1 and fibronectin expression is mediated by beta1-integrin through c-Src- and STAT3-dependent pathways in renal epithelial cells. Am J Physiol Ren Physiol 308:F425–436CrossRef
77.
Zurück zum Zitat Silverstein DM, Travis BR, Thornhill BA, Schurr JS, Kolls JK, Leung JC, Chevalier RL (2003) Altered expression of immune modulator and structural genes in neonatal unilateral ureteral obstruction. Kidney Int 64:25–35PubMedCrossRef Silverstein DM, Travis BR, Thornhill BA, Schurr JS, Kolls JK, Leung JC, Chevalier RL (2003) Altered expression of immune modulator and structural genes in neonatal unilateral ureteral obstruction. Kidney Int 64:25–35PubMedCrossRef
78.
Zurück zum Zitat Samarakoon R, Overstreet JM, Higgins SP, Higgins PJ (2012) TGF-beta1-SMAD/p53/USF2-PAI-1 transcriptional axis in ureteral obstruction-induced renal fibrosis. Cell Tissue Res 347:117–128PubMedCrossRef Samarakoon R, Overstreet JM, Higgins SP, Higgins PJ (2012) TGF-beta1-SMAD/p53/USF2-PAI-1 transcriptional axis in ureteral obstruction-induced renal fibrosis. Cell Tissue Res 347:117–128PubMedCrossRef
79.
Zurück zum Zitat Garcia IM, Mazzei L, Benardon ME, Oliveros L, Cuello-Carrion FD, Gil Lorenzo A, Manucha W, Valles PG (2012) Caveolin-1-eNOS/Hsp70 interactions mediate rosuvastatin antifibrotic effects in neonatal obstructive nephropathy. Nitric Oxide 27:95–105PubMedCrossRef Garcia IM, Mazzei L, Benardon ME, Oliveros L, Cuello-Carrion FD, Gil Lorenzo A, Manucha W, Valles PG (2012) Caveolin-1-eNOS/Hsp70 interactions mediate rosuvastatin antifibrotic effects in neonatal obstructive nephropathy. Nitric Oxide 27:95–105PubMedCrossRef
80.
Zurück zum Zitat el-Dahr SS, Gomez RA, Gray MS, Peach MJ, Carey RM, Chevalier RL (1990) In situ localization of renin and its mRNA in neonatal ureteral obstruction. Am J Physiol 258:F854–862PubMed el-Dahr SS, Gomez RA, Gray MS, Peach MJ, Carey RM, Chevalier RL (1990) In situ localization of renin and its mRNA in neonatal ureteral obstruction. Am J Physiol 258:F854–862PubMed
81.
Zurück zum Zitat Pimentel JL Jr, Martinez-Maldonado M, Wilcox JN, Wang S, Luo C (1993) Regulation of renin–angiotensin system in unilateral ureteral obstruction. Kidney Int 44:390–400PubMedCrossRef Pimentel JL Jr, Martinez-Maldonado M, Wilcox JN, Wang S, Luo C (1993) Regulation of renin–angiotensin system in unilateral ureteral obstruction. Kidney Int 44:390–400PubMedCrossRef
82.
Zurück zum Zitat Ricardo SD, Franzoni DF, Roesener CD, Crisman JM, Diamond JR (2000) Angiotensinogen and AT(1) antisense inhibition of osteopontin translation in rat proximal tubular cells. Am J Physiol Ren Physiol 278:F708–716CrossRef Ricardo SD, Franzoni DF, Roesener CD, Crisman JM, Diamond JR (2000) Angiotensinogen and AT(1) antisense inhibition of osteopontin translation in rat proximal tubular cells. Am J Physiol Ren Physiol 278:F708–716CrossRef
83.
Zurück zum Zitat Kaneto H, Morrissey J, Klahr S (1993) Increased expression of TGF-beta 1 mRNA in the obstructed kidney of rats with unilateral ureteral ligation. Kidney Int 44:313–321PubMedCrossRef Kaneto H, Morrissey J, Klahr S (1993) Increased expression of TGF-beta 1 mRNA in the obstructed kidney of rats with unilateral ureteral ligation. Kidney Int 44:313–321PubMedCrossRef
84.
Zurück zum Zitat Morrissey JJ, Ishidoya S, McCracken R, Klahr S (1996) Nitric oxide generation ameliorates the tubulointerstitial fibrosis of obstructive nephropathy. J Am Soc Nephrol 7:2202–2212PubMed Morrissey JJ, Ishidoya S, McCracken R, Klahr S (1996) Nitric oxide generation ameliorates the tubulointerstitial fibrosis of obstructive nephropathy. J Am Soc Nephrol 7:2202–2212PubMed
85.
Zurück zum Zitat Mazzei LJ, Garcia IM, Altamirano L, Docherty NG, Manucha W (2012) Rosuvastatin preserves renal structure following unilateral ureteric obstruction in the neonatal rat. Am J Nephrol 35:103–113PubMedCrossRef Mazzei LJ, Garcia IM, Altamirano L, Docherty NG, Manucha W (2012) Rosuvastatin preserves renal structure following unilateral ureteric obstruction in the neonatal rat. Am J Nephrol 35:103–113PubMedCrossRef
86.
Zurück zum Zitat Han H, Zhu J, Wang Y, Zhu Z, Chen Y, Lu L, Jin W, Yan X, Zhang R (2017) Renal recruitment of B lymphocytes exacerbates tubulointerstitial fibrosis by promoting monocyte mobilization and infiltration after unilateral ureteral obstruction. J Pathol 241:80–90PubMedCrossRef Han H, Zhu J, Wang Y, Zhu Z, Chen Y, Lu L, Jin W, Yan X, Zhang R (2017) Renal recruitment of B lymphocytes exacerbates tubulointerstitial fibrosis by promoting monocyte mobilization and infiltration after unilateral ureteral obstruction. J Pathol 241:80–90PubMedCrossRef
87.
Zurück zum Zitat Burt LE, Forbes MS, Thornhill BA, Kiley SC, Chevalier RL (2007) Renal vascular endothelial growth factor in neonatal obstructive nephropathy. I. Endogenous VEGF. Am J Physiol Ren Physiol 292:F158–167CrossRef Burt LE, Forbes MS, Thornhill BA, Kiley SC, Chevalier RL (2007) Renal vascular endothelial growth factor in neonatal obstructive nephropathy. I. Endogenous VEGF. Am J Physiol Ren Physiol 292:F158–167CrossRef
88.
Zurück zum Zitat Madsen MG (2013) Urinary biomarkers in hydronephrosis. Dan Med J 60:B4582PubMed Madsen MG (2013) Urinary biomarkers in hydronephrosis. Dan Med J 60:B4582PubMed
89.
Zurück zum Zitat Moody TE, Vaughn ED Jr, Gillenwater JY (1975) Relationship between renal blood flow and ureteral pressure during 18 hours of total unilateral uretheral occlusion. Implications for changing sites of increased renal resistance. Invest Urol 13:246–251 Moody TE, Vaughn ED Jr, Gillenwater JY (1975) Relationship between renal blood flow and ureteral pressure during 18 hours of total unilateral uretheral occlusion. Implications for changing sites of increased renal resistance. Invest Urol 13:246–251
90.
Zurück zum Zitat Valles PG, Pascual L, Manucha W, Carrizo L, Ruttler M (2003) Role of endogenous nitric oxide in unilateral ureteropelvic junction obstruction in children. Kidney Int 63:1104–1115PubMedCrossRef Valles PG, Pascual L, Manucha W, Carrizo L, Ruttler M (2003) Role of endogenous nitric oxide in unilateral ureteropelvic junction obstruction in children. Kidney Int 63:1104–1115PubMedCrossRef
91.
Zurück zum Zitat Cachat F, Lange-Sperandio B, Chang AY, Kiley SC, Thornhill BA, Forbes MS, Chevalier RL (2003) Ureteral obstruction in neonatal mice elicits segment-specific tubular cell responses leading to nephron loss. Kidney Int 63:564–575PubMedCrossRef Cachat F, Lange-Sperandio B, Chang AY, Kiley SC, Thornhill BA, Forbes MS, Chevalier RL (2003) Ureteral obstruction in neonatal mice elicits segment-specific tubular cell responses leading to nephron loss. Kidney Int 63:564–575PubMedCrossRef
92.
Zurück zum Zitat Topcu SO, Pedersen M, Norregaard R, Wang G, Knepper M, Djurhuus JC, Nielsen S, Jorgensen TM, Frokiaer J (2007) Candesartan prevents long-term impairment of renal function in response to neonatal partial unilateral ureteral obstruction. Am J Physiol Ren Physiol 292:F736–748CrossRef Topcu SO, Pedersen M, Norregaard R, Wang G, Knepper M, Djurhuus JC, Nielsen S, Jorgensen TM, Frokiaer J (2007) Candesartan prevents long-term impairment of renal function in response to neonatal partial unilateral ureteral obstruction. Am J Physiol Ren Physiol 292:F736–748CrossRef
93.
94.
Zurück zum Zitat Yang Y, Zhou X, Gao H, Ji SJ, Wang C (2003) The expression of epidermal growth factor and transforming growth factor-beta1 in the stenotic tissue of congenital pelvi-ureteric junction obstruction in children. J Pediatr Surg 38:1656–1660PubMedCrossRef Yang Y, Zhou X, Gao H, Ji SJ, Wang C (2003) The expression of epidermal growth factor and transforming growth factor-beta1 in the stenotic tissue of congenital pelvi-ureteric junction obstruction in children. J Pediatr Surg 38:1656–1660PubMedCrossRef
95.
Zurück zum Zitat Knerr I, Nyul Z, Miller J, Rosch W, Dotsch J, Repp R, Weidner W, Rascher W (2001) Increased endothelin-1 and decreased adrenomedullin gene expression in the stenotic tissue of congenital pelvi-ureteric junction obstruction in children. BJU Int 87:667–671PubMedCrossRef Knerr I, Nyul Z, Miller J, Rosch W, Dotsch J, Repp R, Weidner W, Rascher W (2001) Increased endothelin-1 and decreased adrenomedullin gene expression in the stenotic tissue of congenital pelvi-ureteric junction obstruction in children. BJU Int 87:667–671PubMedCrossRef
96.
Zurück zum Zitat Seremetis GM, Maizels M (1996) TGF-beta mRNA expression in the renal pelvis after experimental and clinical ureteropelvic junction obstruction. J Urol 156:261–266PubMedCrossRef Seremetis GM, Maizels M (1996) TGF-beta mRNA expression in the renal pelvis after experimental and clinical ureteropelvic junction obstruction. J Urol 156:261–266PubMedCrossRef
97.
Zurück zum Zitat Klahr S, Ishidoya S, Morrissey J (1995) Role of angiotensin II in the tubulointerstitial fibrosis of obstructive nephropathy. Am J Kidney Dis 26:141–146PubMedCrossRef Klahr S, Ishidoya S, Morrissey J (1995) Role of angiotensin II in the tubulointerstitial fibrosis of obstructive nephropathy. Am J Kidney Dis 26:141–146PubMedCrossRef
98.
Zurück zum Zitat Picard N, Baum O, Vogetseder A, Kaissling B, Le Hir M (2008) Origin of renal myofibroblasts in the model of unilateral ureter obstruction in the rat. Histochem Cell Biol 130:141–155PubMedPubMedCentralCrossRef Picard N, Baum O, Vogetseder A, Kaissling B, Le Hir M (2008) Origin of renal myofibroblasts in the model of unilateral ureter obstruction in the rat. Histochem Cell Biol 130:141–155PubMedPubMedCentralCrossRef
99.
Zurück zum Zitat Hinz B, Celetta G, Tomasek JJ, Gabbiani G, Chaponnier C (2001) Alpha-smooth muscle actin expression upregulates fibroblast contractile activity. Mol Biol Cell 12:2730–2741PubMedPubMedCentralCrossRef Hinz B, Celetta G, Tomasek JJ, Gabbiani G, Chaponnier C (2001) Alpha-smooth muscle actin expression upregulates fibroblast contractile activity. Mol Biol Cell 12:2730–2741PubMedPubMedCentralCrossRef
100.
Zurück zum Zitat Strutz F, Zeisberg M (2006) Renal fibroblasts and myofibroblasts in chronic kidney disease. J Am Soc Nephrol 17:2992–2998PubMedCrossRef Strutz F, Zeisberg M (2006) Renal fibroblasts and myofibroblasts in chronic kidney disease. J Am Soc Nephrol 17:2992–2998PubMedCrossRef
101.
Zurück zum Zitat Zeisberg M, Strutz F, Muller GA (2001) Renal fibrosis: an update. Curr Opin Nephrol Hypertens 10:315–320PubMedCrossRef Zeisberg M, Strutz F, Muller GA (2001) Renal fibrosis: an update. Curr Opin Nephrol Hypertens 10:315–320PubMedCrossRef
102.
Zurück zum Zitat Han SW, Lee SE, Kim JH, Jeong HJ, Rha KH, Choi SK (1998) Does delayed operation for pediatric ureteropelvic junction obstruction cause histopathological changes? J Urol 160:984–988PubMedCrossRef Han SW, Lee SE, Kim JH, Jeong HJ, Rha KH, Choi SK (1998) Does delayed operation for pediatric ureteropelvic junction obstruction cause histopathological changes? J Urol 160:984–988PubMedCrossRef
103.
Zurück zum Zitat Border WA, Noble NA (1998) Interactions of transforming growth factor-beta and angiotensin II in renal fibrosis. Hypertension 31:181–188PubMedCrossRef Border WA, Noble NA (1998) Interactions of transforming growth factor-beta and angiotensin II in renal fibrosis. Hypertension 31:181–188PubMedCrossRef
104.
Zurück zum Zitat Fern RJ, Yesko CM, Thornhill BA, Kim HS, Smithies O, Chevalier RL (1999) Reduced angiotensinogen expression attenuates renal interstitial fibrosis in obstructive nephropathy in mice. J Clin Invest 103:39–46PubMedPubMedCentralCrossRef Fern RJ, Yesko CM, Thornhill BA, Kim HS, Smithies O, Chevalier RL (1999) Reduced angiotensinogen expression attenuates renal interstitial fibrosis in obstructive nephropathy in mice. J Clin Invest 103:39–46PubMedPubMedCentralCrossRef
105.
Zurück zum Zitat Ishidoya S, Morrissey J, McCracken R, Reyes A, Klahr S (1995) Angiotensin II receptor antagonist ameliorates renal tubulointerstitial fibrosis caused by unilateral ureteral obstruction. Kidney Int 47:1285–1294PubMedCrossRef Ishidoya S, Morrissey J, McCracken R, Reyes A, Klahr S (1995) Angiotensin II receptor antagonist ameliorates renal tubulointerstitial fibrosis caused by unilateral ureteral obstruction. Kidney Int 47:1285–1294PubMedCrossRef
106.
Zurück zum Zitat Guo G, Morrissey J, McCracken R, Tolley T, Liapis H, Klahr S (2001) Contributions of angiotensin II and tumor necrosis factor-alpha to the development of renal fibrosis. Am J Physiol Ren Physiol 280:F777–785CrossRef Guo G, Morrissey J, McCracken R, Tolley T, Liapis H, Klahr S (2001) Contributions of angiotensin II and tumor necrosis factor-alpha to the development of renal fibrosis. Am J Physiol Ren Physiol 280:F777–785CrossRef
107.
Zurück zum Zitat Fukuda K, Yoshitomi K, Yanagida T, Tokumoto M, Hirakata H (2001) Quantification of TGF-beta1 mRNA along rat nephron in obstructive nephropathy. Am J Physiol Ren Physiol 281:F513–521CrossRef Fukuda K, Yoshitomi K, Yanagida T, Tokumoto M, Hirakata H (2001) Quantification of TGF-beta1 mRNA along rat nephron in obstructive nephropathy. Am J Physiol Ren Physiol 281:F513–521CrossRef
108.
Zurück zum Zitat Klahr S, Morrissey J (1998) Angiotensin II and gene expression in the kidney. Am J Kidney Dis 31:171–176PubMedCrossRef Klahr S, Morrissey J (1998) Angiotensin II and gene expression in the kidney. Am J Kidney Dis 31:171–176PubMedCrossRef
109.
Zurück zum Zitat Chevalier RL, Forbes MS, Galarreta CI, Thornhill BA (2014) Responses of proximal tubular cells to injury in congenital renal disease: fight or flight. Pediatr Nephrol 29:537–541PubMedCrossRef Chevalier RL, Forbes MS, Galarreta CI, Thornhill BA (2014) Responses of proximal tubular cells to injury in congenital renal disease: fight or flight. Pediatr Nephrol 29:537–541PubMedCrossRef
110.
Zurück zum Zitat Miyajima A, Chen J, Lawrence C, Ledbetter S, Soslow RA, Stern J, Jha S, Pigato J, Lemer ML, Poppas DP, Vaughan ED, Felsen D (2000) Antibody to transforming growth factor-beta ameliorates tubular apoptosis in unilateral ureteral obstruction. Kidney Int 58:2301–2313PubMedCrossRef Miyajima A, Chen J, Lawrence C, Ledbetter S, Soslow RA, Stern J, Jha S, Pigato J, Lemer ML, Poppas DP, Vaughan ED, Felsen D (2000) Antibody to transforming growth factor-beta ameliorates tubular apoptosis in unilateral ureteral obstruction. Kidney Int 58:2301–2313PubMedCrossRef
111.
Zurück zum Zitat Manucha W, Oliveros L, Carrizo L, Seltzer A, Valles P (2004) Losartan modulation on NOS isoforms and COX-2 expression in early renal fibrogenesis in unilateral obstruction. Kidney Int 65:2091–2107PubMedCrossRef Manucha W, Oliveros L, Carrizo L, Seltzer A, Valles P (2004) Losartan modulation on NOS isoforms and COX-2 expression in early renal fibrogenesis in unilateral obstruction. Kidney Int 65:2091–2107PubMedCrossRef
112.
Zurück zum Zitat Jensen AM, Li C, Praetorius HA, Norregaard R, Frische S, Knepper MA, Nielsen S, Frokiaer J (2006) Angiotensin II mediates downregulation of aquaporin water channels and key renal sodium transporters in response to urinary tract obstruction. Am J Physiol Ren Physiol 291:F1021–1032CrossRef Jensen AM, Li C, Praetorius HA, Norregaard R, Frische S, Knepper MA, Nielsen S, Frokiaer J (2006) Angiotensin II mediates downregulation of aquaporin water channels and key renal sodium transporters in response to urinary tract obstruction. Am J Physiol Ren Physiol 291:F1021–1032CrossRef
113.
Zurück zum Zitat Kellner D, Chen J, Richardson I, Seshan SV, El Chaar M, Vaughan ED Jr, Poppas D, Felsen D (2006) Angiotensin receptor blockade decreases fibrosis and fibroblast expression in a rat model of unilateral ureteral obstruction. J Urol 176:806–812PubMedCrossRef Kellner D, Chen J, Richardson I, Seshan SV, El Chaar M, Vaughan ED Jr, Poppas D, Felsen D (2006) Angiotensin receptor blockade decreases fibrosis and fibroblast expression in a rat model of unilateral ureteral obstruction. J Urol 176:806–812PubMedCrossRef
114.
Zurück zum Zitat Derynck R, Zhang YE (2003) Smad-dependent and Smad-independent pathways in TGF-beta family signalling. Nature 425:577–584PubMedCrossRef Derynck R, Zhang YE (2003) Smad-dependent and Smad-independent pathways in TGF-beta family signalling. Nature 425:577–584PubMedCrossRef
115.
Zurück zum Zitat Lan HY, Mu W, Tomita N, Huang XR, Li JH, Zhu HJ, Morishita R, Johnson RJ (2003) Inhibition of renal fibrosis by gene transfer of inducible Smad7 using ultrasound-microbubble system in rat UUO model. J Am Soc Nephrol 14:1535–1548PubMedCrossRef Lan HY, Mu W, Tomita N, Huang XR, Li JH, Zhu HJ, Morishita R, Johnson RJ (2003) Inhibition of renal fibrosis by gene transfer of inducible Smad7 using ultrasound-microbubble system in rat UUO model. J Am Soc Nephrol 14:1535–1548PubMedCrossRef
116.
Zurück zum Zitat Meng XM, Huang XR, Xiao J, Chen HY, Zhong X, Chung AC, Lan HY (2012) Diverse roles of TGF-beta receptor II in renal fibrosis and inflammation in vivo and in vitro. J Pathol 227:175–188PubMedCrossRef Meng XM, Huang XR, Xiao J, Chen HY, Zhong X, Chung AC, Lan HY (2012) Diverse roles of TGF-beta receptor II in renal fibrosis and inflammation in vivo and in vitro. J Pathol 227:175–188PubMedCrossRef
117.
Zurück zum Zitat Galarreta CI, Thornhill BA, Forbes MS, Simpkins LN, Kim DK, Chevalier RL (2013) Transforming growth factor-beta1 receptor inhibition preserves glomerulotubular integrity during ureteral obstruction in adults but worsens injury in neonatal mice. Am J Physiol Ren Physiol 304:F481–490CrossRef Galarreta CI, Thornhill BA, Forbes MS, Simpkins LN, Kim DK, Chevalier RL (2013) Transforming growth factor-beta1 receptor inhibition preserves glomerulotubular integrity during ureteral obstruction in adults but worsens injury in neonatal mice. Am J Physiol Ren Physiol 304:F481–490CrossRef
118.
Zurück zum Zitat Sato M, Muragaki Y, Saika S, Roberts AB, Ooshima A (2003) Targeted disruption of TGF-beta1/Smad3 signaling protects against renal tubulointerstitial fibrosis induced by unilateral ureteral obstruction. J Clin Invest 112:1486–1494PubMedPubMedCentralCrossRef Sato M, Muragaki Y, Saika S, Roberts AB, Ooshima A (2003) Targeted disruption of TGF-beta1/Smad3 signaling protects against renal tubulointerstitial fibrosis induced by unilateral ureteral obstruction. J Clin Invest 112:1486–1494PubMedPubMedCentralCrossRef
119.
Zurück zum Zitat Yang SP, Woolf AS, Quinn F, Winyard PJ (2001) Deregulation of renal transforming growth factor-beta1 after experimental short-term ureteric obstruction in fetal sheep. Am J Pathol 159:109–117PubMedPubMedCentralCrossRef Yang SP, Woolf AS, Quinn F, Winyard PJ (2001) Deregulation of renal transforming growth factor-beta1 after experimental short-term ureteric obstruction in fetal sheep. Am J Pathol 159:109–117PubMedPubMedCentralCrossRef
120.
Zurück zum Zitat Chung KH, Gomez RA, Chevalier RL (1995) Regulation of renal growth factors and clusterin by AT1 receptors during neonatal ureteral obstruction. Am J Physiol 268:F1117–1123PubMed Chung KH, Gomez RA, Chevalier RL (1995) Regulation of renal growth factors and clusterin by AT1 receptors during neonatal ureteral obstruction. Am J Physiol 268:F1117–1123PubMed
121.
Zurück zum Zitat Pimentel JL, Sundell CL, Wang SS, Kopp JB, Montero A, Martinezmaldonado M (1995) Role of angiotensin-Ii in the expression and regulation of transforming growth-factor-beta in obstructive nephropathy. Kidney Int 48:1233–1246PubMedCrossRef Pimentel JL, Sundell CL, Wang SS, Kopp JB, Montero A, Martinezmaldonado M (1995) Role of angiotensin-Ii in the expression and regulation of transforming growth-factor-beta in obstructive nephropathy. Kidney Int 48:1233–1246PubMedCrossRef
122.
Zurück zum Zitat Murer L, Benetti E, Centi S, Della Vella M, Artifoni L, Capizzi A, Zucchetta P, Del Prete D, Carasi C, Montini G, Rigamonti W, Zaccello G (2006) Clinical and molecular markers of chronic interstitial nephropathy in congenital unilateral ureteropelvic junction obstruction. J Urol 176:2668–2673PubMedCrossRef Murer L, Benetti E, Centi S, Della Vella M, Artifoni L, Capizzi A, Zucchetta P, Del Prete D, Carasi C, Montini G, Rigamonti W, Zaccello G (2006) Clinical and molecular markers of chronic interstitial nephropathy in congenital unilateral ureteropelvic junction obstruction. J Urol 176:2668–2673PubMedCrossRef
123.
Zurück zum Zitat Forbes MS, Thornhill BA, Park MH, Chevalier RL (2007) Lack of endothelial nitric-oxide synthase leads to progressive focal renal injury. Am J Pathol 170:87–99PubMedPubMedCentralCrossRef Forbes MS, Thornhill BA, Park MH, Chevalier RL (2007) Lack of endothelial nitric-oxide synthase leads to progressive focal renal injury. Am J Pathol 170:87–99PubMedPubMedCentralCrossRef
124.
Zurück zum Zitat Sun D, Wang Y, Liu C, Zhou X, Li X, Xiao A (2012) Effects of nitric oxide on renal interstitial fibrosis in rats with unilateral ureteral obstruction. Life Sci 90:900–909PubMedCrossRef Sun D, Wang Y, Liu C, Zhou X, Li X, Xiao A (2012) Effects of nitric oxide on renal interstitial fibrosis in rats with unilateral ureteral obstruction. Life Sci 90:900–909PubMedCrossRef
125.
Zurück zum Zitat Chang B, Mathew R, Palmer LS, Valderrama E, Trachtman H (2002) Nitric oxide in obstructive uropathy: role of endothelial nitric oxide synthase. J Urol 168:1801–1804PubMedCrossRef Chang B, Mathew R, Palmer LS, Valderrama E, Trachtman H (2002) Nitric oxide in obstructive uropathy: role of endothelial nitric oxide synthase. J Urol 168:1801–1804PubMedCrossRef
126.
Zurück zum Zitat Valles PG, Manucha W, Carrizo L, Vega Perugorria J, Seltzer A, Ruete C (2007) Renal caveolin-1 expression in children with unilateral ureteropelvic junction obstruction. Pediatr Nephrol 22:237–248PubMedCrossRef Valles PG, Manucha W, Carrizo L, Vega Perugorria J, Seltzer A, Ruete C (2007) Renal caveolin-1 expression in children with unilateral ureteropelvic junction obstruction. Pediatr Nephrol 22:237–248PubMedCrossRef
127.
Zurück zum Zitat Nasu T, Kinomura M, Tanabe K, Yamasaki H, Htay SL, Saito D, Hinamoto N, Watatani H, Ujike H, Suzuki Y, Sugaya T, Sugiyama H, Sakai Y, Matsumoto K, Maeshima Y, Makino H (2012) Sustained-release prostacyclin analog ONO-1301 ameliorates tubulointerstitial alterations in a mouse obstructive nephropathy model. Am J Physiol Ren Physiol 302:F1616–1629CrossRef Nasu T, Kinomura M, Tanabe K, Yamasaki H, Htay SL, Saito D, Hinamoto N, Watatani H, Ujike H, Suzuki Y, Sugaya T, Sugiyama H, Sakai Y, Matsumoto K, Maeshima Y, Makino H (2012) Sustained-release prostacyclin analog ONO-1301 ameliorates tubulointerstitial alterations in a mouse obstructive nephropathy model. Am J Physiol Ren Physiol 302:F1616–1629CrossRef
128.
Zurück zum Zitat Miyajima A, Chen J, Poppas DP, Vaughan ED Jr, Felsen D (2001) Role of nitric oxide in renal tubular apoptosis of unilateral ureteral obstruction. Kidney Int 59:1290–1303PubMedCrossRef Miyajima A, Chen J, Poppas DP, Vaughan ED Jr, Felsen D (2001) Role of nitric oxide in renal tubular apoptosis of unilateral ureteral obstruction. Kidney Int 59:1290–1303PubMedCrossRef
129.
Zurück zum Zitat Mazzei L, Garcia IM, Cacciamani V, Benardon ME, Manucha W (2010) WT-1 mRNA expression is modulated by nitric oxide availability and Hsp70 interaction after neonatal unilateral ureteral obstruction. Biocell 34:121–132PubMed Mazzei L, Garcia IM, Cacciamani V, Benardon ME, Manucha W (2010) WT-1 mRNA expression is modulated by nitric oxide availability and Hsp70 interaction after neonatal unilateral ureteral obstruction. Biocell 34:121–132PubMed
130.
Zurück zum Zitat Jensen AM, Bae EH, Fenton RA, Norregaard R, Nielsen S, Kim SW, Frokiaer J (2009) Angiotensin II regulates V2 receptor and pAQP2 during ureteral obstruction. Am J Physiol Ren Physiol 296:F127–134CrossRef Jensen AM, Bae EH, Fenton RA, Norregaard R, Nielsen S, Kim SW, Frokiaer J (2009) Angiotensin II regulates V2 receptor and pAQP2 during ureteral obstruction. Am J Physiol Ren Physiol 296:F127–134CrossRef
131.
Zurück zum Zitat Li C, Wang W, Kwon TH, Knepper MA, Nielsen S, Frokiaer J (2003) Altered expression of major renal Na transporters in rats with unilateral ureteral obstruction. Am J Physiol Ren Physiol 284:F155–166CrossRef Li C, Wang W, Kwon TH, Knepper MA, Nielsen S, Frokiaer J (2003) Altered expression of major renal Na transporters in rats with unilateral ureteral obstruction. Am J Physiol Ren Physiol 284:F155–166CrossRef
132.
Zurück zum Zitat Li C, Wang W, Kwon TH, Knepper MA, Nielsen S, Frokiaer J (2003) Altered expression of major renal Na transporters in rats with bilateral ureteral obstruction and release of obstruction. Am J Physiol Ren Physiol 285:F889–901CrossRef Li C, Wang W, Kwon TH, Knepper MA, Nielsen S, Frokiaer J (2003) Altered expression of major renal Na transporters in rats with bilateral ureteral obstruction and release of obstruction. Am J Physiol Ren Physiol 285:F889–901CrossRef
133.
Zurück zum Zitat Li C, Klein JD, Wang W, Knepper MA, Nielsen S, Sands JM, Frokiaer J (2004) Altered expression of urea transporters in response to ureteral obstruction. Am J Physiol Ren Physiol 286:F1154–1162CrossRef Li C, Klein JD, Wang W, Knepper MA, Nielsen S, Sands JM, Frokiaer J (2004) Altered expression of urea transporters in response to ureteral obstruction. Am J Physiol Ren Physiol 286:F1154–1162CrossRef
134.
Zurück zum Zitat Li C, Wang W, Knepper MA, Nielsen S, Frokiaer J (2003) Downregulation of renal aquaporins in response to unilateral ureteral obstruction. Am J Physiol Ren Physiol 284:F1066–1079CrossRef Li C, Wang W, Knepper MA, Nielsen S, Frokiaer J (2003) Downregulation of renal aquaporins in response to unilateral ureteral obstruction. Am J Physiol Ren Physiol 284:F1066–1079CrossRef
135.
Zurück zum Zitat Shi Y, Li C, Thomsen K, Jorgensen TM, Knepper MA, Nielsen S, Djurhuus JC, Frokiaer J (2004) Neonatal ureteral obstruction alters expression of renal sodium transporters and aquaporin water channels. Kidney Int 66:203–215PubMedCrossRef Shi Y, Li C, Thomsen K, Jorgensen TM, Knepper MA, Nielsen S, Djurhuus JC, Frokiaer J (2004) Neonatal ureteral obstruction alters expression of renal sodium transporters and aquaporin water channels. Kidney Int 66:203–215PubMedCrossRef
136.
Zurück zum Zitat Frokiaer J, Christensen BM, Marples D, Djurhuus JC, Jensen UB, Knepper MA, Nielsen S (1997) Downregulation of aquaporin-2 parallels changes in renal water excretion in unilateral ureteral obstruction. Am J Physiol 273:F213–223PubMed Frokiaer J, Christensen BM, Marples D, Djurhuus JC, Jensen UB, Knepper MA, Nielsen S (1997) Downregulation of aquaporin-2 parallels changes in renal water excretion in unilateral ureteral obstruction. Am J Physiol 273:F213–223PubMed
137.
Zurück zum Zitat Chen YC, Cadnapaphornchai MA, Schrier RW (2005) Clinical update on renal aquaporins. Biol Cell 97:357–371PubMedCrossRef Chen YC, Cadnapaphornchai MA, Schrier RW (2005) Clinical update on renal aquaporins. Biol Cell 97:357–371PubMedCrossRef
138.
Zurück zum Zitat Rubenwolf PC, Georgopoulos NT, Clements LA, Feather S, Holland P, Thomas DF, Southgate J (2009) Expression and localisation of aquaporin water channels in human urothelium in situ and in vitro. Eur Urol 56:1013–1023PubMedCrossRef Rubenwolf PC, Georgopoulos NT, Clements LA, Feather S, Holland P, Thomas DF, Southgate J (2009) Expression and localisation of aquaporin water channels in human urothelium in situ and in vitro. Eur Urol 56:1013–1023PubMedCrossRef
139.
Zurück zum Zitat Spector DA, Wade JB, Dillow R, Steplock DA, Weinman EJ (2002) Expression, localization, and regulation of aquaporin-1 to −3 in rat urothelia. Am J Physiol Ren Physiol 282:F1034–1042CrossRef Spector DA, Wade JB, Dillow R, Steplock DA, Weinman EJ (2002) Expression, localization, and regulation of aquaporin-1 to −3 in rat urothelia. Am J Physiol Ren Physiol 282:F1034–1042CrossRef
140.
Zurück zum Zitat Agre P, King LS, Yasui M, Guggino WB, Ottersen OP, Fujiyoshi Y, Engel A, Nielsen S (2002) Aquaporin water channels—from atomic structure to clinical medicine. J Physiol 542:3–16PubMedPubMedCentralCrossRef Agre P, King LS, Yasui M, Guggino WB, Ottersen OP, Fujiyoshi Y, Engel A, Nielsen S (2002) Aquaporin water channels—from atomic structure to clinical medicine. J Physiol 542:3–16PubMedPubMedCentralCrossRef
141.
Zurück zum Zitat Wen JG, Li ZZ, Zhang H, Wang Y, Wang G, Wang Q, Nielsen S, Djurhuus JC, Frokiaer J (2009) Expression of renal aquaporins is down-regulated in children with congenital hydronephrosis. Scand J Urol Nephrol 43:486–493PubMedCrossRef Wen JG, Li ZZ, Zhang H, Wang Y, Wang G, Wang Q, Nielsen S, Djurhuus JC, Frokiaer J (2009) Expression of renal aquaporins is down-regulated in children with congenital hydronephrosis. Scand J Urol Nephrol 43:486–493PubMedCrossRef
142.
Zurück zum Zitat Frokiaer J, Marples D, Knepper MA, Nielsen S (1996) Bilateral ureteral obstruction downregulates expression of vasopressin-sensitive AQP-2 water channel in rat kidney. Am J Physiol 270:F657–668PubMed Frokiaer J, Marples D, Knepper MA, Nielsen S (1996) Bilateral ureteral obstruction downregulates expression of vasopressin-sensitive AQP-2 water channel in rat kidney. Am J Physiol 270:F657–668PubMed
143.
Zurück zum Zitat Li C, Wang W, Kwon TH, Isikay L, Wen JG, Marples D, Djurhuus JC, Stockwell A, Knepper MA, Nielsen S, Frokiaer J (2001) Downregulation of AQP1, −2, and −3 after ureteral obstruction is associated with a long-term urine-concentrating defect. Am J Physiol Ren Physiol 281:F163–171CrossRef Li C, Wang W, Kwon TH, Isikay L, Wen JG, Marples D, Djurhuus JC, Stockwell A, Knepper MA, Nielsen S, Frokiaer J (2001) Downregulation of AQP1, −2, and −3 after ureteral obstruction is associated with a long-term urine-concentrating defect. Am J Physiol Ren Physiol 281:F163–171CrossRef
144.
Zurück zum Zitat Ishidoya S, Morrissey J, McCracken R, Klahr S (1996) Delayed treatment with enalapril halts tubulointerstitial fibrosis in rats with obstructive nephropathy. Kidney Int 49:1110–1119PubMedCrossRef Ishidoya S, Morrissey J, McCracken R, Klahr S (1996) Delayed treatment with enalapril halts tubulointerstitial fibrosis in rats with obstructive nephropathy. Kidney Int 49:1110–1119PubMedCrossRef
145.
Zurück zum Zitat Kaneto H, Morrissey J, McCracken R, Reyes A, Klahr S (1994) Enalapril reduces collagen type IV synthesis and expansion of the interstitium in the obstructed rat kidney. Kidney Int 45:1637–1647PubMedCrossRef Kaneto H, Morrissey J, McCracken R, Reyes A, Klahr S (1994) Enalapril reduces collagen type IV synthesis and expansion of the interstitium in the obstructed rat kidney. Kidney Int 45:1637–1647PubMedCrossRef
146.
Zurück zum Zitat Klahr S, Morrissey J (2002) Comparative effects of ACE inhibition and angiotensin II receptor blockade in the prevention of renal damage. Kidney Int Suppl 82:S23–26 Klahr S, Morrissey J (2002) Comparative effects of ACE inhibition and angiotensin II receptor blockade in the prevention of renal damage. Kidney Int Suppl 82:S23–26
147.
Zurück zum Zitat Chen CO, Park MH, Forbes MS, Thornhill BA, Kiley SC, Yoo KH, Chevalier RL (2007) Angiotensin-converting enzyme inhibition aggravates renal interstitial injury resulting from partial unilateral ureteral obstruction in the neonatal rat. Am J Physiol Ren Physiol 292:F946–955CrossRef Chen CO, Park MH, Forbes MS, Thornhill BA, Kiley SC, Yoo KH, Chevalier RL (2007) Angiotensin-converting enzyme inhibition aggravates renal interstitial injury resulting from partial unilateral ureteral obstruction in the neonatal rat. Am J Physiol Ren Physiol 292:F946–955CrossRef
148.
Zurück zum Zitat Coleman CM, Minor JJ, Burt LE, Thornhill BA, Forbes MS, Chevalier RL (2007) Angiotensin AT1-receptor inhibition exacerbates renal injury resulting from partial unilateral ureteral obstruction in the neonatal rat. Am J Physiol Ren Physiol 293:F262–268CrossRef Coleman CM, Minor JJ, Burt LE, Thornhill BA, Forbes MS, Chevalier RL (2007) Angiotensin AT1-receptor inhibition exacerbates renal injury resulting from partial unilateral ureteral obstruction in the neonatal rat. Am J Physiol Ren Physiol 293:F262–268CrossRef
149.
Zurück zum Zitat Webb NJ, Shahinfar S, Wells TG, Massaad R, Gleim GW, Santoro EP, Sisk CM, Lam C (2012) Losartan and enalapril are comparable in reducing proteinuria in children. Kidney Int 82:819–826PubMedCrossRef Webb NJ, Shahinfar S, Wells TG, Massaad R, Gleim GW, Santoro EP, Sisk CM, Lam C (2012) Losartan and enalapril are comparable in reducing proteinuria in children. Kidney Int 82:819–826PubMedCrossRef
150.
Zurück zum Zitat ItalKid Project, Ardissino G, Vigano S, Testa S, Dacco V, Paglialonga F, Leoni A, Belingheri M, Avolio L, Ciofani A, Claris-Appiani A, Cusi D, Edefonti A, Ammenti A, Cecconi M, Fede C, Ghio L, La Manna A, Maringhini S, Papalia T, Pela I, Pisanello L, Ratsch IM (2007) No clear evidence of ACEi efficacy on the progression of chronic kidney disease in children with hypodysplastic nephropathy--report from the ItalKid Project database. Nephrol Dial Transplant 22:2525–2530CrossRef ItalKid Project, Ardissino G, Vigano S, Testa S, Dacco V, Paglialonga F, Leoni A, Belingheri M, Avolio L, Ciofani A, Claris-Appiani A, Cusi D, Edefonti A, Ammenti A, Cecconi M, Fede C, Ghio L, La Manna A, Maringhini S, Papalia T, Pela I, Pisanello L, Ratsch IM (2007) No clear evidence of ACEi efficacy on the progression of chronic kidney disease in children with hypodysplastic nephropathy--report from the ItalKid Project database. Nephrol Dial Transplant 22:2525–2530CrossRef
151.
Zurück zum Zitat Hari P, Sahu J, Sinha A, Pandey RM, Bal CS, Bagga A (2013) Effect of enalapril on glomerular filtration rate and proteinuria in children with chronic kidney disease: a randomized controlled trial. Indian Pediatr 50:923–928PubMedCrossRef Hari P, Sahu J, Sinha A, Pandey RM, Bal CS, Bagga A (2013) Effect of enalapril on glomerular filtration rate and proteinuria in children with chronic kidney disease: a randomized controlled trial. Indian Pediatr 50:923–928PubMedCrossRef
152.
Zurück zum Zitat Moriyama T, Kawada N, Nagatoya K, Takeji M, Horio M, Ando A, Imai E, Hori M (2001) Fluvastatin suppresses oxidative stress and fibrosis in the interstitium of mouse kidneys with unilateral ureteral obstruction. Kidney Int 59:2095–2103PubMedCrossRef Moriyama T, Kawada N, Nagatoya K, Takeji M, Horio M, Ando A, Imai E, Hori M (2001) Fluvastatin suppresses oxidative stress and fibrosis in the interstitium of mouse kidneys with unilateral ureteral obstruction. Kidney Int 59:2095–2103PubMedCrossRef
153.
Zurück zum Zitat Mizuguchi Y, Miyajima A, Kosaka T, Asano T, Asano T, Hayakawa M (2004) Atorvastatin ameliorates renal tissue damage in unilateral ureteral obstruction. J Urol 172:2456–2459PubMedCrossRef Mizuguchi Y, Miyajima A, Kosaka T, Asano T, Asano T, Hayakawa M (2004) Atorvastatin ameliorates renal tissue damage in unilateral ureteral obstruction. J Urol 172:2456–2459PubMedCrossRef
154.
Zurück zum Zitat Rodriguez-Pena AB, Fuentes-Calvo I, Docherty NG, Arevalo M, Grande MT, Eleno N, Perez-Barriocanal F, Lopez-Novoa JM (2014) Effect of angiotensin II and small GTPase Ras signaling pathway inhibition on early renal changes in a murine model of obstructive nephropathy. Biomed Res Int 2014:124902PubMedPubMedCentralCrossRef Rodriguez-Pena AB, Fuentes-Calvo I, Docherty NG, Arevalo M, Grande MT, Eleno N, Perez-Barriocanal F, Lopez-Novoa JM (2014) Effect of angiotensin II and small GTPase Ras signaling pathway inhibition on early renal changes in a murine model of obstructive nephropathy. Biomed Res Int 2014:124902PubMedPubMedCentralCrossRef
155.
Zurück zum Zitat Jasinska M, Owczarek J, Orszulak-Michalak D (2007) Statins: a new insight into their mechanisms of action and consequent pleiotropic effects. Pharmacol Rep 59:483–499PubMed Jasinska M, Owczarek J, Orszulak-Michalak D (2007) Statins: a new insight into their mechanisms of action and consequent pleiotropic effects. Pharmacol Rep 59:483–499PubMed
156.
Zurück zum Zitat Kamdar C, Chou SY, Mooppan UM, Kim H, Gulmi FA (2010) Atorvastatin protects renal function in the rat with acute unilateral ureteral obstruction. Urology 75:853–857PubMedCrossRef Kamdar C, Chou SY, Mooppan UM, Kim H, Gulmi FA (2010) Atorvastatin protects renal function in the rat with acute unilateral ureteral obstruction. Urology 75:853–857PubMedCrossRef
157.
Zurück zum Zitat Danilovic A, Lopes RI, Sanches TR, Shimizu MH, Oshiro FM, Andrade L, Denes FT, Seguro AC (2012) Atorvastatin prevents the downregulation of aquaporin-2 receptor after bilateral ureteral obstruction and protects renal function in a rat model. Urology 80(485):e415–420 Danilovic A, Lopes RI, Sanches TR, Shimizu MH, Oshiro FM, Andrade L, Denes FT, Seguro AC (2012) Atorvastatin prevents the downregulation of aquaporin-2 receptor after bilateral ureteral obstruction and protects renal function in a rat model. Urology 80(485):e415–420
158.
Zurück zum Zitat Ramkumar S, Raghunath A, Raghunath S (2016) Statin therapy: review of safety and potential side effects. Acta Cardiol Sin 32:631–639PubMedPubMedCentral Ramkumar S, Raghunath A, Raghunath S (2016) Statin therapy: review of safety and potential side effects. Acta Cardiol Sin 32:631–639PubMedPubMedCentral
159.
Zurück zum Zitat Kusters DM, Avis HJ, de Groot E, Wijburg FA, Kastelein JJ, Wiegman A, Hutten BA (2014) Ten-year follow-up after initiation of statin therapy in children with familial hypercholesterolemia. JAMA 312:1055–1057PubMedCrossRef Kusters DM, Avis HJ, de Groot E, Wijburg FA, Kastelein JJ, Wiegman A, Hutten BA (2014) Ten-year follow-up after initiation of statin therapy in children with familial hypercholesterolemia. JAMA 312:1055–1057PubMedCrossRef
160.
Zurück zum Zitat Moon JA, Kim HT, Cho IS, Sheen YY, Kim DK (2006) IN-1130, a novel transforming growth factor-beta type I receptor kinase (ALK5) inhibitor, suppresses renal fibrosis in obstructive nephropathy. Kidney Int 70:1234–1243PubMedCrossRef Moon JA, Kim HT, Cho IS, Sheen YY, Kim DK (2006) IN-1130, a novel transforming growth factor-beta type I receptor kinase (ALK5) inhibitor, suppresses renal fibrosis in obstructive nephropathy. Kidney Int 70:1234–1243PubMedCrossRef
161.
Zurück zum Zitat Morris JC, Shapiro GI, Tan AR, Lawrence DP, Olencki TE, Dezube BJ, Hsu FJ, Reiss M, Berzofsky JA (2008) Phase I/II study of GC1008: A human anti-transforming growth factor-beta (TGFβ) monoclonal antibody in patients with advanced malignant melanoma or renal cell carcinoma. J Clin Oncol 26[Suppl]:abstr 9028 Morris JC, Shapiro GI, Tan AR, Lawrence DP, Olencki TE, Dezube BJ, Hsu FJ, Reiss M, Berzofsky JA (2008) Phase I/II study of GC1008: A human anti-transforming growth factor-beta (TGFβ) monoclonal antibody in patients with advanced malignant melanoma or renal cell carcinoma. J Clin Oncol 26[Suppl]:abstr 9028
162.
Zurück zum Zitat Kamata M, Hosono K, Fujita T, Kamata K, Majima M (2015) Role of cyclooxygenase-2 in the development of interstitial fibrosis in kidneys following unilateral ureteral obstruction in mice. Biomed Pharmacother 70:174–180PubMedCrossRef Kamata M, Hosono K, Fujita T, Kamata K, Majima M (2015) Role of cyclooxygenase-2 in the development of interstitial fibrosis in kidneys following unilateral ureteral obstruction in mice. Biomed Pharmacother 70:174–180PubMedCrossRef
163.
Zurück zum Zitat Pediatric Rheumatology Collaborative Study Group, Sobel RE, Lovell DJ, Brunner HI, Weiss JE, Morris PW, Gottlieb BS, Chalom EC, Jung LK, Onel KB, Petiniot L, Goldsmith DP, Nanda K, Shishov M, Abramsky S, Young JP, Giannini EH (2014) Safety of celecoxib and nonselective nonsteroidal anti-inflammatory drugs in juvenile idiopathic arthritis: results of the Phase 4 registry. Pediatr Rheumatol Online J 12:29CrossRef Pediatric Rheumatology Collaborative Study Group, Sobel RE, Lovell DJ, Brunner HI, Weiss JE, Morris PW, Gottlieb BS, Chalom EC, Jung LK, Onel KB, Petiniot L, Goldsmith DP, Nanda K, Shishov M, Abramsky S, Young JP, Giannini EH (2014) Safety of celecoxib and nonselective nonsteroidal anti-inflammatory drugs in juvenile idiopathic arthritis: results of the Phase 4 registry. Pediatr Rheumatol Online J 12:29CrossRef
165.
Zurück zum Zitat Tripatara P, Patel NS, Webb A, Rathod K, Lecomte FM, Mazzon E, Cuzzocrea S, Yaqoob MM, Ahluwalia A, Thiemermann C (2007) Nitrite-derived nitric oxide protects the rat kidney against ischemia/reperfusion injury in vivo: role for xanthine oxidoreductase. J Am Soc Nephrol 18:570–580PubMedCrossRef Tripatara P, Patel NS, Webb A, Rathod K, Lecomte FM, Mazzon E, Cuzzocrea S, Yaqoob MM, Ahluwalia A, Thiemermann C (2007) Nitrite-derived nitric oxide protects the rat kidney against ischemia/reperfusion injury in vivo: role for xanthine oxidoreductase. J Am Soc Nephrol 18:570–580PubMedCrossRef
166.
Zurück zum Zitat Gilchrist M, Winyard PG, Benjamin N (2010) Dietary nitrate—good or bad? Nitric Oxide 22:104–109PubMedCrossRef Gilchrist M, Winyard PG, Benjamin N (2010) Dietary nitrate—good or bad? Nitric Oxide 22:104–109PubMedCrossRef
167.
168.
Zurück zum Zitat Taha MA, Shokeir AA, Osman HG, Abd El-Aziz Ael A, Farahat SE (2007) Obstructed versus dilated nonobstructed kidneys in children with congenital ureteropelvic junction narrowing: role of urinary tubular enzymes. J Urol 178:640–646PubMedCrossRef Taha MA, Shokeir AA, Osman HG, Abd El-Aziz Ael A, Farahat SE (2007) Obstructed versus dilated nonobstructed kidneys in children with congenital ureteropelvic junction narrowing: role of urinary tubular enzymes. J Urol 178:640–646PubMedCrossRef
169.
Zurück zum Zitat Taranta-Janusz K, Wasilewska A, Debek W, Filonowicz R, Michaluk-Skutnik J (2013) Urinary angiotensinogen as a novel marker of obstructive nephropathy in children. Acta Paediatr 102:e429–433PubMedCrossRef Taranta-Janusz K, Wasilewska A, Debek W, Filonowicz R, Michaluk-Skutnik J (2013) Urinary angiotensinogen as a novel marker of obstructive nephropathy in children. Acta Paediatr 102:e429–433PubMedCrossRef
170.
Zurück zum Zitat Bartoli F, Penza R, Aceto G, Niglio F, D’Addato O, Pastore V, Campanella V, Magaldi S, Lasalandra C, Di Bitonto G, Gesualdo L (2011) Urinary epidermal growth factor, monocyte chemotactic protein-1, and beta2-microglobulin in children with ureteropelvic junction obstruction. J Pediatr Surg 46:530–536PubMedCrossRef Bartoli F, Penza R, Aceto G, Niglio F, D’Addato O, Pastore V, Campanella V, Magaldi S, Lasalandra C, Di Bitonto G, Gesualdo L (2011) Urinary epidermal growth factor, monocyte chemotactic protein-1, and beta2-microglobulin in children with ureteropelvic junction obstruction. J Pediatr Surg 46:530–536PubMedCrossRef
171.
Zurück zum Zitat Madsen MG, Norregaard R, Palmfeldt J, Olsen LH, Frokiaer J, Jorgensen TM (2012) Urinary NGAL, cystatin C, beta2-microglobulin, and osteopontin significance in hydronephrotic children. Pediatr Nephrol 27:2099–2106PubMedCrossRef Madsen MG, Norregaard R, Palmfeldt J, Olsen LH, Frokiaer J, Jorgensen TM (2012) Urinary NGAL, cystatin C, beta2-microglobulin, and osteopontin significance in hydronephrotic children. Pediatr Nephrol 27:2099–2106PubMedCrossRef
172.
Zurück zum Zitat Atar A, Oktar T, Kucukgergin C, Kalelioglu I, Seckin S, Ander H, Ziylan O, Kadioglu TC (2015) The roles of serum and urinary carbohydrate antigen 19–9 in the management of patients with antenatal hydronephrosis. J Pediatr Urol 11(133):e1–5 Atar A, Oktar T, Kucukgergin C, Kalelioglu I, Seckin S, Ander H, Ziylan O, Kadioglu TC (2015) The roles of serum and urinary carbohydrate antigen 19–9 in the management of patients with antenatal hydronephrosis. J Pediatr Urol 11(133):e1–5
173.
Zurück zum Zitat Kajbafzadeh AM, Elmi A, Talab SS, Emami H, Esfahani SA, Saeedi P (2010) Urinary and serum carbohydrate antigen 19–9 as a biomarker in ureteropelvic junction obstruction in children. J Urol 183:2353–2360PubMedCrossRef Kajbafzadeh AM, Elmi A, Talab SS, Emami H, Esfahani SA, Saeedi P (2010) Urinary and serum carbohydrate antigen 19–9 as a biomarker in ureteropelvic junction obstruction in children. J Urol 183:2353–2360PubMedCrossRef
174.
Zurück zum Zitat Grandaliano G, Gesualdo L, Bartoli F, Ranieri E, Monno R, Leggio A, Paradies G, Caldarulo E, Infante B, Schena FP (2000) MCP-1 and EGF renal expression and urine excretion in human congenital obstructive nephropathy. Kidney Int 58:182–192PubMedCrossRef Grandaliano G, Gesualdo L, Bartoli F, Ranieri E, Monno R, Leggio A, Paradies G, Caldarulo E, Infante B, Schena FP (2000) MCP-1 and EGF renal expression and urine excretion in human congenital obstructive nephropathy. Kidney Int 58:182–192PubMedCrossRef
175.
Zurück zum Zitat Madsen MG, Norregaard R, Palmfeldt J, Olsen LH, Frokiaer J, Jorgensen TM (2013) Epidermal growth factor and monocyte chemotactic peptide-1: potential biomarkers of urinary tract obstruction in children with hydronephrosis. J Pediatr Urol 9:838–845PubMedCrossRef Madsen MG, Norregaard R, Palmfeldt J, Olsen LH, Frokiaer J, Jorgensen TM (2013) Epidermal growth factor and monocyte chemotactic peptide-1: potential biomarkers of urinary tract obstruction in children with hydronephrosis. J Pediatr Urol 9:838–845PubMedCrossRef
176.
Zurück zum Zitat Taha MA, Shokeir AA, Osman HG, Abd El-Aziz Ael A, Farahat SE (2007) Pelvi-ureteric junction obstruction in children: the role of urinary transforming growth factor-beta and epidermal growth factor. BJU Int 99:899–903PubMedCrossRef Taha MA, Shokeir AA, Osman HG, Abd El-Aziz Ael A, Farahat SE (2007) Pelvi-ureteric junction obstruction in children: the role of urinary transforming growth factor-beta and epidermal growth factor. BJU Int 99:899–903PubMedCrossRef
177.
Zurück zum Zitat Taha MA, Shokeir AA, Osman HG, Abd el-Aziz Ael A, Farahat SE (2007) Diagnosis of ureteropelvic junction obstruction in children: role of endothelin-1 in voided urine. Urology 69:560–564, discussion 564–565PubMedCrossRef Taha MA, Shokeir AA, Osman HG, Abd el-Aziz Ael A, Farahat SE (2007) Diagnosis of ureteropelvic junction obstruction in children: role of endothelin-1 in voided urine. Urology 69:560–564, discussion 564–565PubMedCrossRef
178.
Zurück zum Zitat Li Z, Liu X, Liu S, Gu C, Tian F, Wen J (2012) Urinary heme oxygenase-1 in children with congenital hydronephrosis due to ureteropelvic junction obstruction. Biomarkers 17:471–476PubMedCrossRef Li Z, Liu X, Liu S, Gu C, Tian F, Wen J (2012) Urinary heme oxygenase-1 in children with congenital hydronephrosis due to ureteropelvic junction obstruction. Biomarkers 17:471–476PubMedCrossRef
179.
Zurück zum Zitat Wasilewska A, Taranta-Janusz K, Debek W, Zoch-Zwierz W, Kuroczycka-Saniutycz E (2011) KIM-1 and NGAL: new markers of obstructive nephropathy. Pediatr Nephrol 26:579–586PubMedPubMedCentralCrossRef Wasilewska A, Taranta-Janusz K, Debek W, Zoch-Zwierz W, Kuroczycka-Saniutycz E (2011) KIM-1 and NGAL: new markers of obstructive nephropathy. Pediatr Nephrol 26:579–586PubMedPubMedCentralCrossRef
180.
Zurück zum Zitat Taranta-Janusz K, Wasilewska A, Debek W, Waszkiewicz-Stojda M (2012) Urinary cytokine profiles in unilateral congenital hydronephrosis. Pediatr Nephrol 27:2107–2113PubMedPubMedCentralCrossRef Taranta-Janusz K, Wasilewska A, Debek W, Waszkiewicz-Stojda M (2012) Urinary cytokine profiles in unilateral congenital hydronephrosis. Pediatr Nephrol 27:2107–2113PubMedPubMedCentralCrossRef
181.
Zurück zum Zitat Cost NG, Noh PH, Devarajan P, Ivancic V, Reddy PP, Minevich E, Bennett M, Haffner C, Schulte M, DeFoor WR Jr (2013) Urinary NGAL levels correlate with differential renal function in patients with ureteropelvic junction obstruction undergoing pyeloplasty. J Urol 190[4 Supp]:1462–1467 Cost NG, Noh PH, Devarajan P, Ivancic V, Reddy PP, Minevich E, Bennett M, Haffner C, Schulte M, DeFoor WR Jr (2013) Urinary NGAL levels correlate with differential renal function in patients with ureteropelvic junction obstruction undergoing pyeloplasty. J Urol 190[4 Supp]:1462–1467
182.
Zurück zum Zitat Almodhen F, Loutochin O, Capolicchio JP, Jednak R, El-Sherbiny M (2009) The role of bladder urine transforming growth factor-beta1 concentrations in diagnosis and management of unilateral prenatal hydronephrosis. J Urol 182:292–298, discussion 298PubMedCrossRef Almodhen F, Loutochin O, Capolicchio JP, Jednak R, El-Sherbiny M (2009) The role of bladder urine transforming growth factor-beta1 concentrations in diagnosis and management of unilateral prenatal hydronephrosis. J Urol 182:292–298, discussion 298PubMedCrossRef
183.
Zurück zum Zitat Madsen MG, Norregaard R, Frokiaer J, Jorgensen TM (2011) Urinary biomarkers in prenatally diagnosed unilateral hydronephrosis. J Pediatr Urol 7:105–112PubMedCrossRef Madsen MG, Norregaard R, Frokiaer J, Jorgensen TM (2011) Urinary biomarkers in prenatally diagnosed unilateral hydronephrosis. J Pediatr Urol 7:105–112PubMedCrossRef
184.
Zurück zum Zitat Papachristou F, Pavlaki A, Printza N (2014) Urinary and serum biomarkers in ureteropelvic junction obstruction: a systematic review. Biomarkers 19:531–540PubMedCrossRef Papachristou F, Pavlaki A, Printza N (2014) Urinary and serum biomarkers in ureteropelvic junction obstruction: a systematic review. Biomarkers 19:531–540PubMedCrossRef
185.
Zurück zum Zitat Decramer S, Wittke S, Mischak H, Zurbig P, Walden M, Bouissou F, Bascands JL, Schanstra JP (2006) Predicting the clinical outcome of congenital unilateral ureteropelvic junction obstruction in newborn by urinary proteome analysis. Nat Med 12:398–400PubMedCrossRef Decramer S, Wittke S, Mischak H, Zurbig P, Walden M, Bouissou F, Bascands JL, Schanstra JP (2006) Predicting the clinical outcome of congenital unilateral ureteropelvic junction obstruction in newborn by urinary proteome analysis. Nat Med 12:398–400PubMedCrossRef
186.
Zurück zum Zitat Drube J, Zurbig P, Schiffer E, Lau E, Ure B, Gluer S, Kirschstein M, Pape L, Decramer S, Bascands JL, Schanstra JP, Mischak H, Ehrich JH (2010) Urinary proteome analysis identifies infants but not older children requiring pyeloplasty. Pediatr Nephrol 25:1673–1678PubMedCrossRef Drube J, Zurbig P, Schiffer E, Lau E, Ure B, Gluer S, Kirschstein M, Pape L, Decramer S, Bascands JL, Schanstra JP, Mischak H, Ehrich JH (2010) Urinary proteome analysis identifies infants but not older children requiring pyeloplasty. Pediatr Nephrol 25:1673–1678PubMedCrossRef
Metadaten
Titel
The molecular biology of pelvi-ureteric junction obstruction
verfasst von
Laura Jackson
Mark Woodward
Richard J. Coward
Publikationsdatum
13.03.2017
Verlag
Springer Berlin Heidelberg
Erschienen in
Pediatric Nephrology / Ausgabe 4/2018
Print ISSN: 0931-041X
Elektronische ISSN: 1432-198X
DOI
https://doi.org/10.1007/s00467-017-3629-0

Weitere Artikel der Ausgabe 4/2018

Pediatric Nephrology 4/2018 Zur Ausgabe

Update Pädiatrie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.