51
|
Eskild-Jensen A, Paulsen LF, Wogensen L, Olesen P, Pedersen L, Frøkiaer J, Nyengaard JR. AT1 receptor blockade prevents interstitial and glomerular apoptosis but not fibrosis in pigs with neonatal induced partial unilateral ureteral obstruction. Am J Physiol Renal Physiol 2007; 292:F1771-81. [PMID: 17356126 DOI: 10.1152/ajprenal.00479.2006] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Obstruction-induced fibrosis is a leading cause of end-stage renal failure in children. The pathophysiological mechanisms may involve apoptosis and the renin-angiotensin system. We studied apoptosis and fibrosis in a well-established neonatal pig model with unilateral partial ureteral obstruction (PUUO) induced during ongoing nephrogenesis in 2-day-old piglets. The role of angiotensin II (ANG II) was studied using the AT(1) receptor blocker CV-11974 (0.12 mg/h candesartan from age 23 to 30 days). At day 30 the kidneys were perfusion fixed and fibrosis, apoptosis, and tubular lengths were quantitated using stereological methods, picro Sirius red staining, and immunohistochemical techniques identifying activated caspase 3, aquaporin-2 (AQP2), and von Willebrand factor. The collagen content was assessed by hydroxyproline density. Neonatal induced PUUO increased interstitial and glomerular cell apoptosis and fibrosis. At this stage, PUUO did not increase tubular cell apoptosis or decrease tubular length and cell number. AT(1) receptor blockade prevented the PUUO-induced interstitial and glomerular cell apoptosis but did not attenuate fibrosis. In conclusion, AT(1) receptor blockade after the end of nephrogenesis may prevent interstitial and glomerular cell apoptosis but not fibrosis, suggesting that pathways not involving AT(1) receptor stimulation contribute to neonatal obstruction-induced fibrosis or that prevention of interstitial cell apoptosis counteracts a potential antifibrotic effect of AT(1) receptor blockade in this pig model of congenital obstructive nephropathy. Our results demonstrate that ANG II plays a role in PUUO-induced glomerular cell apoptosis.
Collapse
Affiliation(s)
- Anni Eskild-Jensen
- Department of Clinical Physiology and Nuclear Medicine, Aarhus University Hospital, Aarhus C, Denmark.
| | | | | | | | | | | | | |
Collapse
|
52
|
Handtrack C, Cordasic N, Klanke B, Veelken R, Hilgers KF. Effect of the angiotensinogen genotype on experimental hypertension in mice. J Mol Med (Berl) 2007; 85:343-50. [PMID: 17333097 DOI: 10.1007/s00109-007-0166-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2006] [Revised: 12/10/2006] [Accepted: 01/02/2007] [Indexed: 10/23/2022]
Abstract
Polymorphisms of the angiotensinogen (Agt) gene may affect blood pressure. We used a mouse model to test for the role of the Agt genotype in low-renin or high-renin forms of hypertension. Mice bearing one, two, three, or four copies of the Agt gene underwent renal artery clipping to induce high-renin two-kidney, one-clip renovascular hypertension (2K1C), or uninephrectomy, salt loading, and application of deoxycorticosterone-acetate (DOCA) pellets to induce low-renin mineralocorticoid hypertension. Appropriate control animals were also studied. Blood pressure was measured by tail cuff as well as by direct intra-arterial recordings. There was a small effect of the Agt genotype on baseline blood pressure before induction of hypertension. The extent of 2K1C hypertension was not affected by the genotype. In contrast, there was a marked gene-dose effect on DOCA-hypertension (21.2 mmHg over all genotypes). Treatment of DOCA mice with the angiotensin II type 1 receptor antagonist abolished the genotype effect on blood pressure and left ventricular hypertrophy. There was a trend towards less suppression of endogenous aldosterone by DOCA treatment with increasing number of Agt gene copies. We conclude that the Agt genotype exerts a marked effect on blood pressure in a low-renin form of hypertension but no effect in the face of stimulated renin, at least in mice.
Collapse
Affiliation(s)
- Claudia Handtrack
- Department of Nephrology and Hypertension, University of Erlangen-Nuremberg, Erlangen, Germany
| | | | | | | | | |
Collapse
|
53
|
Carlström M, Sällström J, Skøtt O, Larsson E, Wåhlin N, Persson AEG. Hydronephrosis causes salt-sensitive hypertension and impaired renal concentrating ability in mice. Acta Physiol (Oxf) 2007; 189:293-301. [PMID: 17305709 DOI: 10.1111/j.1748-1716.2006.01637.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
AIM Hypertension is a common disease in the industrialized world and approximately 5% of all cases are secondary to kidney malfunction. We have recently shown that hydronephrosis due to partial unilateral ureteral obstruction (PUUO) causes salt-sensitive hypertension in rats. The mechanisms are still unclear, but appear to be intrarenal and primarily located to the diseased kidney. In the present study, we have developed a model for PUUO to study if hydronephrotic mice develop salt-sensitive hypertension. METHODS PUUO was created in 3-week-old mice (C57bl/6J). Blood pressure and heart rate were measured telemetrically in adult animals on normal and high salt diets. Metabolism cages were used to study the renal excretion of electrolytes and water. Plasma samples for renin analysis were collected and renal histological changes were evaluated. RESULTS All hydronephrotic animals developed salt-sensitive hypertension that correlated to the degree of hydronephrosis. In hydronephrotic animals, blood pressure increased from 114 +/- 1 mmHg on normal salt diet to 120 +/- 2 mmHg on high salt diet, compared with 103 +/- 1 to 104 +/- 1 in controls. Hydronephrotic animals showed increased diuresis and reduced ability to regulate electrolyte concentration. No differences in plasma renin concentration were found between the groups. The parenchymal weight and glomerular area of contralateral kidneys were significantly increased in the hydronephrotic animals. Histopathology of the hydronephrotic kidneys displayed areas with fibrosis, inflammation and glomerular changes. CONCLUSION This study provides a model for PUUO in mice and demonstrates the presence of salt-sensitive hypertension and an impaired renal concentrating ability in mice which has not been described before.
Collapse
Affiliation(s)
- M Carlström
- Department of Medical Cell Biology, Division of Integrative Physiology, Uppsala University, Uppsala, Sweden
| | | | | | | | | | | |
Collapse
|
54
|
Yoo KH, Thornhill BA, Forbes MS, Coleman CM, Marcinko ES, Liaw L, Chevalier RL. Osteopontin regulates renal apoptosis and interstitial fibrosis in neonatal chronic unilateral ureteral obstruction. Kidney Int 2006; 70:1735-41. [PMID: 17003824 DOI: 10.1038/sj.ki.5000357] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Congenital obstructive nephropathy is a major cause of renal insufficiency in children. Osteopontin (OPN) is a phosphoprotein produced by the kidney that mediates cell adhesion and migration. We investigated the role of OPN in the renal response to unilateral ureteral obstruction (UUO) in neonatal mice. OPN null mutant (-/-) and wild-type (+/+) mice were subjected to sham operation or UUO within the first 2 days of life. At 7 and 21 days of age, fibroblasts (fibroblast-specific protein (FSP)-1), myofibroblasts (alpha-smooth muscle actin (SMA)), and macrophages (F4/80) were identified by immunohistochemical staining. Apoptotic cells were detected by terminal deoxy transferase uridine triphosphate nick end-labeling technique and interstitial collagen by Masson trichrome or picrosirius red stain. Compared to sham-operated or contralateral kidneys, obstructed kidneys showed increases in all parameters by 7 days, with further increases by 21 days. After 21 days UUO, there was an increase in tubular and interstitial apoptosis in OPN -/- mice as compared to +/+ animals (P<0.05). However, FSP-1- and alpha-SMA-positive cells and collagen in the obstructed kidney were decreased in OPN -/- compared to +/+ mice (P<0.05), whereas the interstitial macrophage population did not differ between groups. We conclude that OPN plays a significant role in the recruitment and activation of interstitial fibroblasts to myofibroblasts in the progression of interstitial fibrosis in the developing hydronephrotic kidney. However, OPN also suppresses apoptosis. Future approaches to limit the progression of obstructive nephropathy in the developing kidney will require targeting of specific renal compartments.
Collapse
Affiliation(s)
- K H Yoo
- Department of Pediatrics, Korea University Guro Hospital, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
55
|
Chevalier RL. Obstructive nephropathy: towards biomarker discovery and gene therapy. ACTA ACUST UNITED AC 2006; 2:157-68. [PMID: 16932414 DOI: 10.1038/ncpneph0098] [Citation(s) in RCA: 143] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2005] [Accepted: 12/05/2005] [Indexed: 12/16/2022]
Abstract
Obstructive nephropathy is a major cause of renal failure, particularly in infants and children. Cellular and molecular mechanisms responsible for the progression of the tubular atrophy and interstitial fibrosis-processes that lead to nephron loss-have been elucidated in the past 5 years. Following urinary tract obstruction and tubular dilatation, a cascade of events results in upregulation of the intrarenal renin-angiotensin system, tubular apoptosis and macrophage infiltration of the interstitium. This is followed by accumulation of interstitial fibroblasts through proliferation of resident fibroblasts and epithelial-mesenchymal transformation of renal tubular cells. Under the influence of cytokines, chemokines and other signaling molecules produced by tubular and interstitial cells, fibroblasts undergo transformation to myofibroblasts that induce expansion of the extracellular matrix. The cellular interactions that regulate development of interstitial inflammation, tubular apoptosis and interstitial fibrosis are complex. Changes in renal gene expression and protein production afford many potential biomarkers of disease progression and targets for therapeutic manipulation. These include signaling molecules and receptors involved in macrophage recruitment and proliferation, tubular death signals and survival factors, and modulators of epithelial-mesenchymal transformation. Targeted gene deletion and various forms of gene therapy have been used in experimental obstructive nephropathy, mostly rodent models of unilateral ureteral obstruction or cell culture techniques. Further refinement of these models is needed to develop a matrix of biomarkers with clinical predictive value, as well as molecular therapies that will prevent or reverse the renal structural and functional consequences of obstructive nephropathy.
Collapse
Affiliation(s)
- Robert L Chevalier
- Department of Pediatrics at the University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
56
|
Kellner D, Chen J, Richardson I, Seshan SV, El Chaar M, Vaughan ED, Poppas D, Felsen D. Angiotensin receptor blockade decreases fibrosis and fibroblast expression in a rat model of unilateral ureteral obstruction. J Urol 2006; 176:806-12. [PMID: 16813952 DOI: 10.1016/j.juro.2006.03.076] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2005] [Indexed: 10/24/2022]
Abstract
PURPOSE Unilateral ureteral obstruction is characterized by histopathological changes including interstitial fibrosis, fibroblast specific protein expression, tubular atrophy and apoptosis, and macrophage infiltration. Angiotensin II has been implicated in some of these changes. We examined the effect of angiotensin blockade on markers of renal injury, including fibroblast specific protein expression, fibrosis, apoptosis and macrophage infiltration. We used losartan, an angiotensin II antagonist, in a unilateral ureteral obstruction model and studied animals 3 weeks after unilateral ureteral obstruction, a time at which renal damage is well established. MATERIALS AND METHODS Rats underwent unilateral ureteral obstruction and were given either drinking water or losartan for 21 days. Kidneys were harvested and examined for fibrosis (trichrome and the Sircol assay for collagen), apoptosis (TUNEL), and fibroblast specific protein expression and macrophage infiltration (immunohistochemistry). RESULTS Unilateral ureteral obstruction was found to induce fibrosis, apoptosis, fibroblast expression and macrophage in the obstructed kidney. Losartan significantly decreased apoptosis and macrophage infiltration in the obstructed kidney. It also decreased fibrosis, as measured by either trichrome staining assessed by a pathologist, the Sircol assay for collagen or fibroblast specific protein expression. However, approximately 50% of the changes were not affected by the current treatment, suggesting that other factors contribute to renal damage in unilateral ureteral obstruction. CONCLUSIONS We observed the direct contribution of angiotensin II to both apoptotic and cellular transition processes (epithelial mesenchymal transition) and fibrosis in unilateral ureteral obstruction. Because these processes are active not only in unilateral ureteral obstruction, but also in other renal diseases, the value of angiotensin II blockade as an important part of the antifibrotic armamentarium has been confirmed.
Collapse
Affiliation(s)
- Daniel Kellner
- Institute for Pediatric Urology, Weill Cornell Medical College, New York, New York 10021, USA
| | | | | | | | | | | | | | | |
Collapse
|
57
|
Ozawa Y, Kobori H, Suzaki Y, Navar LG. Sustained renal interstitial macrophage infiltration following chronic angiotensin II infusions. Am J Physiol Renal Physiol 2006; 292:F330-9. [PMID: 16804106 PMCID: PMC2001287 DOI: 10.1152/ajprenal.00059.2006] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Chronic angiotensin (ANG) II infusions into rats lead to augmented intrarenal levels of ANG II and inflammatory factors, impaired renal function, and progressive hypertension. Residual effects persist after cessation of ANG II infusions, as manifested by a hypertensive response to high-salt intake. This study was performed to determine the residual cytokines and chemokines following the cessation of ANG II infusion. Male Sprague-Dawley rats, maintained on a normal diet, received either a sham operation or continuous ANG II infusion (120 ng/min) subcutaneously via minipumps. The ANG II-infused rats were further subdivided into three subgroups. Minipumps were removed on day 12 with subsequent harvesting of kidneys at 0, 3, and 6 days after cessation of ANG II infusion. After 12 days of ANG II infusion, systolic blood pressure, interstitial fibrosis, preglomerular hypertrophy, and interstitial macrophage infiltration were significantly enhanced compared with the shams. By 3 days following the cessation of ANG II infusion, systolic blood pressure was normalized; however, interstitial fibrosis and preglomerular hypertrophy were still present. Furthermore, increased interstitial macrophage infiltration was still present 6 days after cessation of ANG II infusion. Importantly, augmented mRNA levels of monocyte chemotactic protein (MCP)-1 (1.55 +/- 0.15 vs. 1.00 +/- 0.13, relative ratio) and transforming growth factor (TGF)-beta(1) (1.52 +/- 0.16 vs. 1.00 +/- 0.08) persisted 6 days after the withdrawal of ANG II infusion (1.60 +/- 0.20 for MCP-1 and 1.43 +/- 0.17 for TGF-beta(1)). Thus, the ANG II-induced activation of MCP-1 and TGF-beta(1) is sustained and may account for the persistent effect of chronic ANG II infusions on interstitial macrophage infiltration, suggesting a possible mechanism for the development of salt sensitivity in ANG II-dependent hypertension.
Collapse
Affiliation(s)
- Yuri Ozawa
- Department of Physiology and Hypertension and Renal Center of Excellence, Tulane University Health Sciences Center, New Orleans, Louisiana 70112-2699, USA
| | | | | | | |
Collapse
|
58
|
Abstract
PURPOSE OF REVIEW This review focuses on recent advances in understanding the factors contributing to obstructive nephropathy, the most important cause of renal failure in children. The major focus is on renal cellular and molecular events, with emphasis on those affecting the developing kidney. RECENT FINDINGS Experiments in the fetal sheep or neonatal rat, mouse, or pig reveal dramatic effects of urinary tract obstruction on renal growth and development. Surgical relief of obstruction can reverse some of the structural and functional deficits, but cannot restore normalcy. Renal tubular apoptosis is a major factor leading to tubular atrophy following unilateral ureteral obstruction. Increased reactive oxygen species, and a renal environment favoring pro-apoptotic, over survival, signals, contribute to cell death. A variety of intrarenal factors lead to progressive interstitial fibrosis, including the newly described process of epithelial-mesenchymal transition, whereby tubular epithelial cells are transformed into activated fibroblasts. A number of endogenous antifibrotic counter-regulatory molecules have been identified, opening the possibility of enhancing the kidney's own defenses against progressive fibrosis. SUMMARY The renal response to urinary tract obstruction is complex and involves a wide array of interacting molecules. Elucidation of these interactions will lead to the identification of biomarkers that will allow a more precise prediction to the response to surgical intervention and, hopefully, to novel therapies to prevent renal deterioration.
Collapse
Affiliation(s)
- Robert L Chevalier
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia 22908, USA.
| |
Collapse
|
59
|
Lange-Sperandio B, Schimpgen K, Rodenbeck B, Chavakis T, Bierhaus A, Nawroth P, Thornhill B, Schaefer F, Chevalier RL. Distinct roles of Mac-1 and its counter-receptors in neonatal obstructive nephropathy. Kidney Int 2006; 69:81-8. [PMID: 16374427 DOI: 10.1038/sj.ki.5000017] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Urinary tract obstruction during renal development leads to tubular atrophy and interstitial fibrosis. Inflammatory macrophages are crucial in this process, and beta2-integrins play a major role in leukocyte recruitment. We investigated the role of beta2-integrins and their major counter-receptors (intercellular adhesion molecule-1 (ICAM-1), receptor for advanced glycation endproducts (RAGE), junctional adhesion molecule (JAM)-C) in obstructive nephropathy in neonatal mice. Two-day-old beta2-integrin-deficient mice (Mac-1-/- and LFA-1-/-(deficient for leukocyte function-associated antigen-1)) and wild-type mice (C57BL/6) underwent unilateral ureteral obstruction (UUO) or sham operation. After 1, 5 or 12 days of obstruction, renal macrophage infiltration and tubulointerstitial damage were quantitated. Tissue abundance of Mac-1 and its ligands ICAM-1, RAGE and JAM-C was examined by Western blot and immunoprecipitation. Deficiency of either integrin was associated with reduced early macrophage invasion into the obstructed kidney. After 12 days of UUO, macrophage infiltration and tubulointerstitial injury were reduced only in Mac-1-/- but not in LFA-1-/- mice. Besides ICAM-1, an upregulation of two novel Mac-1 ligands, RAGE and JAM-C were observed, however, with distinct time courses. We conclude that beta2-integrins mediate macrophage infiltration in UUO. Mac-1 is the predominant leukocyte integrin involved in leukocyte recruitment after obstruction. ICAM-1 and its new ligands RAGE and JAM-C are sequentially activated in UUO. Blocking of Mac-1 and its ligands may confer synergistic renoprotective effects in neonatal obstructive nephropathy.
Collapse
Affiliation(s)
- B Lange-Sperandio
- Department of Pediatrics, University of Heidelberg, Heidelberg, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
60
|
Abstract
Congenital obstructive nephropathy is the primary cause for end-stage renal disease (ESRD) in children. An increasingly used animal model of obstructive nephropathy is unilateral ureteral obstruction (UUO). This model mimics, in an accelerated manner, the different stages of obstructive nephropathy leading to tubulointerstitial fibrosis: cellular infiltration, tubular proliferation and apoptosis, epithelial-mesenchymal transition (EMT), (myo)fibroblast accumulation, increased extracellular matrix (ECM) deposition, and tubular atrophy. During the last decade genetically modified animals are increasingly used to study the development of obstructive nephropathy. Although the use of these animals (mainly knockouts) has highlighted some pitfalls of this approach (compensation by closely related gene products, absence of temporal knockouts) it has brought important information about the role of specific gene-products in the pathogenesis of obstructive nephropathy. Besides confirming the important pathologic role for angiotensin II (Ang II) and transforming growth factor-beta (TGF-beta) in obstructive nephropathy, these animals have shown the complexity of the development of tubulointerstitial fibrosis involving a large number of closely functionally related molecules. More interestingly, the use of these animals has led to the discovery of unexpected and contradictory roles (both potentially pro- and antifibrotic) for Ang II, for ECM degrading enzymes matrix metalloproteinase 9 (MMP-9) and tissue plasminogen activators (PAs), for plasminogen activator inhibitor 1 (PAI-1), and for the adhesion molecule osteopontin (OPN) in obstructive nephropathy. Further use of these animals, especially in combination with pharmacologic tools, should help to better identify potential antifibrotic strategies in obstructive nephropathy.
Collapse
|
61
|
Kiley SC, Thornhill BA, Belyea BC, Neale K, Forbes MS, Luetteke NC, Lee DC, Chevalier RL. Epidermal growth factor potentiates renal cell death in hydronephrotic neonatal mice, but cell survival in rats. Kidney Int 2005; 68:504-14. [PMID: 16014027 DOI: 10.1111/j.1523-1755.2005.00428.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Epidermal growth factor (EGF) markedly attenuates tubular apoptosis induced by unilateral ureteral obstruction (UUO) in the neonatal rat, and reduces apoptosis induced by mechanical stretch of cultured rat tubular cells. METHODS To investigate the role of EGF in modulating apoptosis resulting from UUO, neonatal wild type and mutant mice lacking EGF (knockout), or with diminished EGF receptor activity (waved-2 mutant) were compared to control mice for tubular apoptosis and atrophy. Rat and mouse kidneys were compared for localization of the EGF receptor. Apoptosis was also measured in cultured mouse tubular cells subjected to stretch and exposed to EGF. RESULTS UUO reduced endogenous renal EGF expression in wild-type mice. Unlike the rat, exogenous EGF did not decrease tubular apoptosis or atrophy in the obstructed kidney, and significantly increased stretch-induced apoptosis of cultured mouse tubular cells. Tubular apoptosis was 50% lower in the obstructed kidney of EGF knockout and waved-2 mice relative to wild type and heterozygous animals. Exogenous EGF increased tubular apoptosis and doubled atrophy in the obstructed kidney of waved-2 mice. Species differences in EGF receptor localization were detected in 3-day-old kidneys. CONCLUSION EGF acts as a survival factor in the neonatal rat, but potentiates tubular cell death in the neonatal mouse. Species differences are maintained in cultured cells, suggesting that differences in EGF receptor signaling underlie these opposing effects.
Collapse
Affiliation(s)
- Susan C Kiley
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia 22908, USA
| | | | | | | | | | | | | | | |
Collapse
|
62
|
Shin GT, Kim WH, Yim H, Kim MS, Kim H. Effects of suppressing intrarenal angiotensinogen on renal transforming growth factor-beta1 expression in acute ureteral obstruction. Kidney Int 2005; 67:897-908. [PMID: 15698429 DOI: 10.1111/j.1523-1755.2005.00154.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Angiotensin II (Ang II) mediates the up-regulation of fibrogenic factors such as transforming growth factor-beta1 (TGF-beta1) in chronic renal diseases. In addition, it has been proposed that the intrarenal renin-angiotensin system (RAS) is as important as the systemic RAS in kidney disease progression. METHODS We suppressed angiotensinogen (AGT) gene expression in the kidney by transferring recombinant adenoviral vectors carrying a transgene expressing AGT antisense mRNA, and determined the effect of the local inhibition of the RAS on TGF-beta1 synthesis in the kidneys of rats with unilateral ureteral obstruction (UUO). Immediately after UUO, recombinant adenovirus vectors were injected intraparenchymally into the cortex of obstructed kidneys. RESULTS beta-galactosidase (beta-gal)-stained kidney sections revealed the efficient transduction of the recombinant adenoviral vectors into tubular epithelial cells. Kidney cortex injected with AGT antisense showed significantly lower native AGT mRNA and protein expressions than control UUO kidneys at 24 hours and 5 days post-UUO. TGF-beta1 was significantly up-regulated in the renal cortex 24 hours and 5 days post-UUO, whereas AGT antisense-injected UUO rats showed significantly reduced TGF-beta1 expression compared to control UUO rats. Both fibronectin and collagen type I expressions were increased 24 hours and 5 days post-UUO, and these augmentations were considerably reduced by AGT antisense RNA treatment. CONCLUSION This study demonstrates that the suppression of intrarenal RAS prevents the formation of renal cortical TGF-beta1, and of related fibrogenic factors, in early UUO.
Collapse
Affiliation(s)
- Gyu-Tae Shin
- Department of Nephrology, Ajou University School of Medicine, Suwon, South Korea.
| | | | | | | | | |
Collapse
|
63
|
Simic P, Vukicevic S. Bone morphogenetic proteins in development and homeostasis of kidney. Cytokine Growth Factor Rev 2005; 16:299-308. [PMID: 15923134 DOI: 10.1016/j.cytogfr.2005.02.010] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2004] [Accepted: 02/17/2005] [Indexed: 01/01/2023]
Abstract
Bone morphogenetic proteins play a key role in kidney development and postnatal function. The kidney has been identified as a major site of bone morphogenetic protein (BMP)-7 synthesis during embryonic and postnatal development, which mediates differentiation and maintenance of metanephric mesenchyme. Targeted disruption of BMP-7 gene expression in mice resulted in dysgenic kidneys with hydroureters, causing uremia within 24h after birth. Several experimental animal models of acute and chronic renal injury have all unequivocally shown beneficial effect of BMP-7 in ameliorating the severity of damage by preventing inflammation and fibrosis. Apart from the beneficial effect on kidney disease itself, BMP-7 improves important complications of chronic renal impairment such as renal osteodystrophy and vascular calcification.
Collapse
Affiliation(s)
- Petra Simic
- Laboratory for Mineralized Tissues, Department of Anatomy, School of Medicine, University of Zagreb, Salata 11, 10 000 Zagreb, Croatia
| | | |
Collapse
|
64
|
Yu L, Border WA, Anderson I, McCourt M, Huang Y, Noble NA. Combining TGF-β inhibition and angiotensin II blockade results in enhanced antifibrotic effect. Kidney Int 2004; 66:1774-84. [PMID: 15496148 DOI: 10.1111/j.1523-1755.2004.00901.x] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Although angiotensin II (Ang II) blockade is rapidly becoming standard antifibrotic therapy in renal diseases, current data suggest that Ang II blockade alone cannot stop fibrotic disease. New therapies, such as antibodies to transforming growth factor-beta (TGF-beta), or drug combinations will be required to further slow or halt disease progression. Here, using the anti-Thy1 model of glomerulonephritis, the maximally therapeutic dose of the TGF-beta neutralizing mouse monoclonal antibody (1D11) was determined and compared with the maximally effective dose of enalapril. Then, the effect of combining both treatments at maximal doses was determined. METHODS After disease induction with the anti-Thy1 antibody, OX-7, increasing doses of 1D11 were given intraperitoneally (IP) on days 1, 3, and 5. Enalapril was administered in drinking water from day 1. The fibrotic response was assessed at day 6. RESULTS 1D11 dose-dependently reduced fibrosis, with the 0.5 and 5 mg/kg doses showing maximal therapeutic effects, reducing period-acid Schiff (PAS) staining by 56% and 45%, respectively. Fibronectin and collagen I staining was reduced by 32% to 36%, respectively. Glomerular mRNA and production of fibronectin, plasminogen activator inhibitor-1 (PAI-1), TGF-beta1, and p-Smad2 protein were also reduced. The maximal therapeutic effects of 1D11 and enalapril alone were very similar. However, combination therapy led to further reduction in disease. Notably, matrix deposition was reduced by 80%. CONCLUSION While 1D11 or enalapril at maximal doses reduce fibrosis equally, simultaneous blockade of Ang II and TGF-beta reduces fibrotic disease considerably more, offering hope that such drug combinations may confer a therapeutic advantage over angiotensin blockade alone.
Collapse
Affiliation(s)
- Ling Yu
- Fibrosis Research Laboratory, Division of Nephrology, University of Utah School of Medicine, Salt Lake City, Utah 84108, USA
| | | | | | | | | | | |
Collapse
|
65
|
Abstract
PURPOSE Congenital obstructive nephropathy constitutes one of the major causes of renal insufficiency in infants and children. This review addresses the need to define biomarkers that serve as surrogate end points for measuring the severity of obstruction, the evolution of renal maldevelopment and injury, and the response to medical or surgical intervention. MATERIALS AND METHODS The literature from the last 10 years was reviewed for biomarkers of congenital obstructive nephropathy. Sources of biomarkers included urine, blood, amniotic fluid, tissue and imaging techniques. RESULTS Previous markers of congenital obstructive nephropathy include sonographic renal pelvic diameter, quantitative diuretic renography, and markers of glomerular and tubular function. Attempts to correlate renal histological changes with differential renal function have been disappointing. Immunohistochemical analysis and laser capture microscopy should improve specificity. Most promising is the application of new insights into the cellular response of the developing kidney to urinary tract obstruction. These findings include components of the renin-angiotensin system, transforming growth factor-beta 1, monocyte chemoattractant protein-1 and epidermal growth factor. Microarray studies show unique patterns of gene expression by the neonatal rat kidney subjected to ureteral obstruction, and proteomics should provide even more sensitive biomarkers of obstructive nephropathy. CONCLUSIONS We must define the cellular and molecular bases of renal maldevelopment, focusing on the link between functional and developmental pathophysiology. These findings will lead to biomarkers that will optimize our management of congenital obstructive nephropathy.
Collapse
Affiliation(s)
- Robert L Chevalier
- Department of Pediatrics, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
66
|
Bohlega S, Abu-Amero SN, Wakil SM, Carroll P, Al-Amr R, Lach B, Al-Sayed Y, Cupler EJ, Meyer BF. Mutation of the slow myosin heavy chain rod domain underlies hyaline body myopathy. Neurology 2004; 62:1518-21. [PMID: 15136674 DOI: 10.1212/01.wnl.0000123255.92062.37] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To identify the gene and specific mutation underlying hyaline body myopathy in the family studied. METHODS A microsatellite-based whole genome scan was performed. Linkage analysis assumed autosomal dominant inheritance and equal allele frequencies. A candidate gene approach within the linked interval and direct sequencing were used for mutation detection. RESULTS Initial analysis indicated a maximum lod score of 3.01 at D14S1280. High-density mapping surrounding the linked locus was performed. Multipoint analysis showed that the linked region with a maximum lod score of 3.01 extended from D14S742 to D14S608 with a peak non-parametric linkage (NPL) score of 3.75 at D14S608. The myosin heavy chain genes MYH6 and MYH7 map to the region between D14S742 and D14S1280. Sequence analysis of the coding regions of MYH7 revealed an A-->T transversion at nucleotide position 25596 (M57965) resulting in a histidine-to-leucine amino acid change at residue 1904 (H1904L). CONCLUSION Pathogenicity of the MYH7 H1904L mutation most likely results from disruption of myosin heavy chain assembly or stability of the sarcomeric protein. The MYH7 tail domain mutation results in an inclusion body myopathy with an apparent absence of hypertrophic cardiomyopathy usually associated with mutations of this gene.
Collapse
Affiliation(s)
- S Bohlega
- Department of Neurosciences, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Abstract
Renal tubular epithelial cells (TEC) are thought to play an active role in tubulointerstitial inflammation. Various immune and non-immune factors activate TEC to produce a variety of cytokines and chemokines, contributing to attraction of inflammatory cells to the kidney. The proinflammatory transcription factor nuclear factor-kappaB (NF-kappaB) appears to be a key player in these responses and tubular expression of NF-kappaB has been demonstrated in vitro and in vivo. Although glucocorticoids are known to inhibit NF-kappaB activation at different levels, the proinflammatory capacity of TEC was not inhibited. In contrast, glucocorticoids seemed to enhance the profibrotic response of TEC, emphasizing the cell-type specific characteristics of glucocorticoid action. We propose that specific inhibition of NF-kappaB activation in TEC might be an attractive strategy for therapeutic intervention in renal inflammation.
Collapse
Affiliation(s)
- Simone de Haij
- Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | |
Collapse
|
68
|
Abstract
Significant advances have been made recently in elucidating the cellular consequences of urinary tract obstruction during renal development. Urinary tract obstruction impairs growth and maturation of the kidney, and can also cause renal maldevelopment. This includes a reduction in the number of nephrons, tubular atrophy, and progressive interstitial fibrosis. Apoptosis (programmed cell death) accounts for much of the loss of tubular epithelial cells. Factors contributing to apoptosis include stretching of cells in dilated tubules, altered renal production of growth factors, and infiltration of the renal interstitium by macrophages. Two major controversies remain regarding the surgical management of congenital obstructive nephropathy: first, which fetuses with bladder outlet obstruction should undergo prenatal intervention, and second, which infants should undergo early pyeloplasty for ureteropelvic junction obstruction? Even after successful surgery for congential obstructive nephropathy, all patients should be followed for hypertension, proteinuria, or renal deterioration.
Collapse
Affiliation(s)
- Robert L Chevalier
- Department of Pediatrics, Children's Medical Center, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
69
|
|
70
|
Stern JM, Chen J, Silver RB, Poppas DP, Vaughan ED, Felsen D. Effect of UUO on D1aR expression reveals a link among dopamine, transforming growth factor-β, and nitric oxide. Am J Physiol Renal Physiol 2004; 286:F509-15. [PMID: 14612384 DOI: 10.1152/ajprenal.00253.2002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Interactions between transforming growth factor-β (TGF-β) and nitric oxide (NO) are important in the pathophysiology of unilateral ureteral obstruction (UUO). Dopamine (DA) is a vasoactive renal mediator active at the D1Areceptor (D1AR), which has not been studied in UUO; therefore, we examined the interactions among DA, TGF-β, and NO in UUO. In vivo, UUO was carried out in rats with or without concurrent treatment with 1D11, a monoclonal antibody to TGF-β, for 14 days. In vitro, NRK-52E cells (normal rat kidney tubules) were treated with DA, and NO and TGF-β release were examined. UUO resulted in a 70% decrease in the expression of renal D1AR, confirmed by both Western blot analysis and immunohistochemistry. 1D11 treatment restored expression to 60% of control values. DA treatment decreased NRK-52E release of TGF-β by 80%; conversely, DA significantly increased NO release from NRK-52E cells. These results suggest that DA modulates the release of cytokines, which are involved in the fibrotic and apoptotic sequelae of UUO, and that these effects are independent of DA's known vasoactive properties.
Collapse
Affiliation(s)
- Joshua M Stern
- Department of Urology, New York Presbyterian Children's Hospital-Weill Cornell Medical School, New York, NY 10021, USA
| | | | | | | | | | | |
Collapse
|
71
|
Desmoulière A, Darby IA, Gabbiani G. Normal and pathologic soft tissue remodeling: role of the myofibroblast, with special emphasis on liver and kidney fibrosis. J Transl Med 2003; 83:1689-707. [PMID: 14691287 DOI: 10.1097/01.lab.0000101911.53973.90] [Citation(s) in RCA: 278] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Affiliation(s)
- Alexis Desmoulière
- Groupe de Recherches pour l'Etude du Foie (AD), INSERM E0362, Université Victor Segalen Bordeaux 2, Bordeaux, France.
| | | | | |
Collapse
|
72
|
Ma LJ, Yang H, Gaspert A, Carlesso G, Barty MM, Davidson JM, Sheppard D, Fogo AB. Transforming growth factor-beta-dependent and -independent pathways of induction of tubulointerstitial fibrosis in beta6(-/-) mice. THE AMERICAN JOURNAL OF PATHOLOGY 2003; 163:1261-73. [PMID: 14507636 PMCID: PMC1868298 DOI: 10.1016/s0002-9440(10)63486-4] [Citation(s) in RCA: 177] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Transforming growth factor-beta1 (TGF-beta1) and the renin-angiotensin-aldosterone system are key mediators in kidney fibrosis. Integrin alphavbeta6, a heterodimeric matrix receptor expressed in epithelia, binds and activates latent TGF-beta1. We used beta6 integrin-null mice (beta6(-/-)) to determine the role of local TGF-beta1 activation in renal fibrosis in the unilateral ureteral obstruction (UUO) model. Obstructed kidneys from beta6(-/-) mice showed less injury than obstructed kidneys from wild-type (WT) mice, associated with lower collagen I, collagen III, plasminogen activator inhibitor (PAI-1), and TGF-beta1 mRNA levels and lower collagen content. Infusion with either angiotensin II (Ang II) or aldosterone (Aldo) or combination in beta6(-/-) UUO mice significantly increased collagen contents to levels comparable to those in identically treated WT. Active TGF-beta protein expression in beta6(-/-) mice was less in UUO kidneys with or without Ang II infusion compared to matched WT mice. Activated Smad 2 levels in beta6(-/-) obstructed kidneys were lower than in WT UUO mice, and did not increase when fibrosis was induced in beta6(-/-) UUO mice by Ang II infusion. Anti-TGF-beta antibody only partially decreased this Ang II-stimulated fibrosis in beta6(-/-) UUO kidneys. In situ hybridization and immunostaining showed low expression of PAI-1 mRNA and protein in tubular epithelium in beta6(-/-) UUO kidneys, with increased PAI-1 expression in response to Ang II, Aldo, or both. Our results indicate that interruption of alphavbeta6-mediated activation of TGF-beta1 can protect against tubulointerstitial fibrosis. Further, the robust induction of tubulointerstitial fibrosis without increase in activated Smad 2 levels in obstructed beta6(-/-) mice by Ang II suggests the existence of a TGF-beta1-independent pathway of induction of fibrosis through angiotensin.
Collapse
Affiliation(s)
- Li-Jun Ma
- Department of Pathology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | | | | | | | | | | | | | | |
Collapse
|
73
|
[Experimental ureteral obstruction and knockout animals]. Arch Pediatr 2003; 10:903-10. [PMID: 14550981 DOI: 10.1016/s0929-693x(03)00398-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Obstructive uropathies caused by congenital malformations of the urinary tract are relatively frequent in newborn. These obstructive lesions are the main cause for renal disease in infancy. Most of these uropathies are treated by surgical interventions restoring the drainage function of the urinary tract. Clinically these patients are cured but the question remains wether these patients will develop renal disease in adult life, since it has been recently shown in animal models that transient, neonatal and prenatal, ureteral obstruction induces significant renal deterioration later in life. Except for angiotensin converting enzyme inhibitors that slow down the progression of renal disease, no specific drugs reducing renal fibrosis exist. Animal models of ureteral obstruction have allowed to clearly identify the events leading to tubulointerstitial fibrosis. Furthermore, more recently, the use of ureteral obstruction in genetically engineered animals has shown pro- and anti-fibrotic properties of a large number of molecules. These studies using genetically engineered animals have suggested several new future promising therapeutic directions to treat renal fibrosis.
Collapse
|
74
|
Eikmans M, Baelde JJ, de Heer E, Bruijn JA. ECM homeostasis in renal diseases: a genomic approach. J Pathol 2003; 200:526-36. [PMID: 12845620 DOI: 10.1002/path.1417] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Chronic renal disease is in general histologically accompanied by a vast amount of scar tissue, ie glomerulosclerosis and interstitial fibrosis. Scarring results from excessive accumulation of extracellular matrix (ECM) components, a process driven by a plethora of cytokines and growth factors. Studies in experimental renal disease which target these regulators using gene therapy limit or prevent the development of scarring. This review focuses specifically on the role of transforming growth factor-beta, platelet-derived growth factor, connective tissue growth factor, hepatocyte growth factor, and epidermal growth factor. The results obtained in animal models hold promise for molecular intervention strategies in human renal disease. Microarray technology allows large-scale gene expression profiling in kidney tissue to identify common molecular pathways in a step towards discovery of new drug targets. Molecular techniques are expected to be used for diagnostic and prognostic purposes in nephrological practice to supplement renal biopsy. Several studies already show that molecular techniques might be of use in routine diagnostic practice. Improvement of diagnosis and prediction of outcome in renal patients might lead to more efficient and earlier therapeutic intervention.
Collapse
Affiliation(s)
- M Eikmans
- Department of Pathology, Leiden University Medical Center, Building 1, LI-Q, PO Box 9600, 2300 RC Leiden, The Netherlands.
| | | | | | | |
Collapse
|
75
|
Yang J, Liu Y. Delayed administration of hepatocyte growth factor reduces renal fibrosis in obstructive nephropathy. Am J Physiol Renal Physiol 2003; 284:F349-F357. [PMID: 12529273 DOI: 10.1152/ajprenal.00154.2002] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Hepatocyte growth factor (HGF) is a renotropic protein that elicits antifibrogenic activity by preventing the activation of matrix-producing myofibroblast cells in animal models of chronic renal diseases. However, whether a delayed administration of HGF can still attenuate renal fibrosis remains uncertain. In this study, we examined the therapeutic potential of exogenous HGF on an established renal interstitial fibrosis induced by unilateral ureteral obstruction (UUO). Three days after UUO, the obstructed kidneys displayed interstitial fibrotic lesions with characteristic features of an established renal fibrosis, as manifested by myofibroblast activation, fibronectin overexpression, interstitial matrix deposition, and transforming growth factor-beta1 upregulation. Beginning at this time point, administration of recombinant HGF into mice by intravenous injections for 11 days markedly suppressed the progression of renal interstitial fibrosis. HGF significantly suppressed renal alpha-smooth muscle actin expression, total kidney collagen contents, interstitial matrix components, such as fibronectin, and renal expression of transforming growth factor-beta1 and its type I receptor. Compared with the starting point (3 days after UUO), HGF treatment largely blunted the progression of myofibroblast accumulation and collagen deposition but did not reverse it. Delayed administration of HGF also suppressed the myofibroblastic transdifferentiation from tubular epithelial cells in vitro, as demonstrated by a decline in alpha-smooth muscle actin and fibronectin expression. These results suggest that exogenous HGF exhibits potent therapeutic effects on retarding the progression of an established renal fibrosis.
Collapse
Affiliation(s)
- Junwei Yang
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA
| | | |
Collapse
|
76
|
Cachat F, Lange-Sperandio B, Chang AY, Kiley SC, Thornhill BA, Forbes MS, Chevalier RL. Ureteral obstruction in neonatal mice elicits segment-specific tubular cell responses leading to nephron loss. Kidney Int 2003; 63:564-75. [PMID: 12631121 DOI: 10.1046/j.1523-1755.2003.00775.x] [Citation(s) in RCA: 99] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND To elucidate the sequence of renal responses leading to nephron loss in obstructive nephropathy, we examined the evolution of segmental nephron cellular changes consequent to chronic unilateral ureteral obstruction (UUO) in the neonatal mouse. METHODS Neonatal mice were subjected to UUO or sham-operation, and kidneys were harvested 5, 12 or 19 days after surgery. Proximal tubules (PT), distal tubules (DT) and collecting ducts (CD) were identified with lectins. Histomorphometric quantitation was made for cellular necrosis, apoptosis, proliferation, tubular dilatation, tubular basement membrane (TBM) thickening, interstitial collagen, and glomerular maturation. The distribution of hypoxic tissue was determined using pimonidazole as a marker. Additional studies were performed by mechanically stretching monolayer cultures of mouse proximal tubular and collecting duct cells, and measuring apoptosis. RESULTS Neonatal UUO induced an arrest of glomerular maturation throughout the period of study. Chronic UUO induced hypoxia, tubular necrosis, proliferation, and TBM thickening in the PT, but stimulated apoptosis in the DT and CD. Tubular dilation in the obstructed kidney was most severe in CD and least severe in PT. Tubular cell apoptosis closely paralleled tubular dilation (P < 0.05), and fibrosis surrounding individual tubules also correlated with tubular dilation (P < 0.001). Mechanical stretching of cultured mouse tubular cells induced apoptosis directly proportional to the magnitude of axial strain: apoptosis was consistently greater in CD than in PT cells (P < 0.05). CONCLUSIONS Following UUO, the co-localization of hypoxia with cellular proliferation, necrosis, and TBM thickening of the PT is consistent with ischemic injury resulting from vasoconstriction. In contrast, a selective dilation of the distal portion of the nephron (DT and CD), which results from the greater tubular compliance there, leads to stretch-induced epithelial cell apoptosis, along with a progressive peritubular fibrosis. Nephron loss in the obstructed developing kidney likely results from complex, segment-specific cellular responses.
Collapse
Affiliation(s)
- François Cachat
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia 22908, USA
| | | | | | | | | | | | | |
Collapse
|
77
|
|
78
|
Affiliation(s)
- Juan A Oliver
- Department of Medicine, Columbia University, New York, New York, USA.
| |
Collapse
|
79
|
Affiliation(s)
- Juan A Oliver
- Department of Medicine, Columbia University, New York, New York, USA.
| |
Collapse
|
80
|
Sawada T, Ishii Y, Tojimbara T, Nakajima I, Fuchinoue S, Teraoka S. The ACE inhibitor, quinapril, ameliorates peritoneal fibrosis in an encapsulating peritoneal sclerosis model in mice. Pharmacol Res 2002; 46:505-10. [PMID: 12457623 DOI: 10.1016/s1043661802002281] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Encapsulating peritoneal sclerosis (EPS) is a serious complication of patients on continuous ambulatory peritoneal dialysis. In the present study, the inhibitory effect of angiotensin-converting enzyme inhibitor, quinapril, on the peritoneal fibrosis was examined in an experimental EPS model in mice. C57BL/6 mice were divided into three groups. Group 1 (n=20) mice received daily intraperitoneal injection of 0.3 ml of SH solution which consists of 0.1% chlorhexidine gluconate and 15% ethanol dissolved in saline. Group 2 (n=20) and group 3 (n=20) mice received SH solution by the same manner of group 1 mice, and were given orally 1 or 3 mg kg(-1) of quinapril, respectively, on daily basis. Five mice from each group were sacrificed on day 3, 7, 21, and 56, and evaluated macroscopically and histologically. Macroscopic examination revealed that fibrotic change in parietal peritoneum in group 1 was more severe than in group 2 and 3, accompanied with statistical significance. Histological examination demonstrated that peritoneal thickening in group 2 and 3 were markedly ameliorated than in group 1. Semi-quantitative analysis showed that histological fibrotic score was significantly higher in group 1 than in group 2 and 3. These results suggest that quinapril ameliorate the fibrotic change in parietal peritoneum in experimental EPS model in mice, and may have a clinical utility for the prevention of EPS.
Collapse
Affiliation(s)
- Tokihiko Sawada
- Department of Surgery, Kidney Center, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan.
| | | | | | | | | | | |
Collapse
|
81
|
Abstract
Interstitial fibrosis has a major role in the progression of renal diseases. Several animal models are available for the study of renal fibrosis. The models of aminonucleoside-induced nephrotic syndrome, cyclosporin nephrotoxicity, and passive Heyman nephritis are characterized by molecular and cellular events similar to those that occur in obstructive nephropathy. Additionally, inhibition of angiotensin-converting enzyme exerts salutary effects on the progression of renal fibrosis in obstructive nephropathy. Unilateral ureteral obstruction (UUO) has emerged as an important model for the study of the mechanisms of renal fibrosis and also for the evaluation of the impact of potential therapeutic approaches to ameliorate renal disease. Many quantifiable pathophysiological events occur over the span of 1 wk of UUO, making this an attractive model for study. This paper reviews some of the ongoing studies that utilized a rodent model of UUO. Some of the findings of the animal model have been compared with observations made in patients with obstructive nephropathy. Most of the evidence suggests that the rodent model of UUO is reflective of human renal disease processes.
Collapse
Affiliation(s)
- Saulo Klahr
- Department of Internal Medicine, Washington University School of Medicine at Barnes-Jewish Hospital, St. Louis, Missouri 63110-1092, USA.
| | | |
Collapse
|
82
|
Yang J, Dai C, Liu Y. Hepatocyte growth factor gene therapy and angiotensin II blockade synergistically attenuate renal interstitial fibrosis in mice. J Am Soc Nephrol 2002; 13:2464-2477. [PMID: 12239235 DOI: 10.1097/01.asn.0000031827.16102.c1] [Citation(s) in RCA: 115] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Tubulointerstitial fibrosis is considered to be common endpoint result of many forms of chronic renal diseases. Except for renal replacement, chronic renal fibrosis is presently incurable. This study demonstrates that the combination of hepatocyte growth factor (HGF) gene therapy with inhibition of the renin-angiotensin system produced synergistic beneficial effects leading to dramatic attenuation of renal tubulointerstitial fibrosis in obstructive nephropathy in mice. The combined treatment with human HGF gene and losartan, an angiotensin II (AngII) type I receptor blocker, preserved renal mass and gross morphology of the obstructed kidneys. Although HGF gene therapy alone inhibited the expression of alpha-smooth muscle actin (alpha SMA) by approximately 54% and 60% at day 7 and day 14 after surgery, respectively, its combination with losartan almost completely abolished alpha SMA induction in the obstructed kidneys. The combined therapy also synergistically inhibited the accumulation of interstitial matrix components, such as fibronectin and collagen I, and suppressed renal expression of transforming growth factor-beta1 (TGF-beta1) and its type I receptor. In vitro studies revealed that AngII by itself did not induce alpha SMA, but it drastically potentiated TGF-beta1-initiated alpha SMA expression in tubular epithelial cells. Furthermore, HGF abrogated de novo alpha SMA expression induced by TGF-beta1 plus AngII. These results suggest that many factors are implicated in the pathogenesis of renal interstitial fibrosis; therefore, a combined therapy aimed at simultaneously targeting multiple pathologic pathways may be necessary for halting the progression of chronic renal diseases. These findings may provide the basis for designing future therapeutic regimens for blocking progressive renal fibrosis in patients.
Collapse
Affiliation(s)
- Junwei Yang
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA
| | | | | |
Collapse
|
83
|
Abstract
The inhibition of angiotensin II through angiotensin converting enzyme inhibitors or angiotensin receptor blockers has become the foundation of medical treatment of progressive chronic renal disease. Although these drugs provide a significant improvement over earlier treatments, they only slow the progression of renal disease, implying the need for additional drugs that could be combined with antiangiotensin treatment. Potentially valuable novel drug targets include downstream mediators of angiotensin II such as transforming growth factor-b, plasminogen activator inhibitor-1, and endothelin-1. In addition, recent evidence points to aldosterone as a major player in progressive renal disease, indicating that multiple points of the renin-angiotensin-aldosterone system might have to be targeted. This paper reviews the experimental and clinical evidence indicating that targeting these cytokines and hormones could provide additional benefits to antiangiotensin treatment in chronic renal disease.
Collapse
Affiliation(s)
- Jens Gaedeke
- Fibrosis Research Laboratory, Salt Lake City, UT 84108, USA.
| | | | | |
Collapse
|
84
|
Crawford K, Flick R, Close L, Shelly D, Paul R, Bove K, Kumar A, Lessard J. Mice lacking skeletal muscle actin show reduced muscle strength and growth deficits and die during the neonatal period. Mol Cell Biol 2002; 22:5887-96. [PMID: 12138199 PMCID: PMC133984 DOI: 10.1128/mcb.22.16.5887-5896.2002] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
All four of the muscle actins (skeletal, cardiac, vascular, and enteric) in higher vertebrates show distinct expression patterns and display highly conserved amino acid sequences. While it is hypothesized that each of the muscle isoactins is specifically adapted to its respective tissue and that the minor variations among them have developmental and/or physiological relevance, the exact functional and developmental significance of these proteins remains largely unknown. In order to begin to assess these issues, we disrupted the skeletal actin gene by homologous recombination. All mice lacking skeletal actin die in the early neonatal period (day 1 to 9). These null animals appear normal at birth and can breathe, walk, and suckle, but within 4 days, they show a markedly lower body weight than normal littermates and many develop scoliosis. Null mice show a loss of glycogen and reduced brown fat that is consistent with malnutrition leading to death. Newborn skeletal muscles from null mice are similar to those of wild-type mice in size, fiber type, and ultrastructural organization. At birth, both hemizygous and homozygous null animals show an increase in cardiac and vascular actin mRNA in skeletal muscle, with no skeletal actin mRNA present in null mice. Adult hemizygous animals show an increased level of skeletal actin mRNA in hind limb muscle but no overt phenotype. Extensor digitorum longus (EDL) muscle isolated from skeletal-actin-deficient mice at day 2 to 3 showed a marked reduction in force production compared to that of control littermates, and EDL muscle from hemizygous animals displayed an intermediate force generation. Thus, while increases in cardiac and vascular smooth-muscle actin can partially compensate for the lack of skeletal actin in null mice, this is not sufficient to support adequate skeletal muscle growth and/or function.
Collapse
Affiliation(s)
- K Crawford
- Division of Developmental Biology, Children's Hospital Research Foundation, Cincinnati, Ohio 45229, USA
| | | | | | | | | | | | | | | |
Collapse
|
85
|
Chevalier RL, Thornhill BA, Chang AY, Cachat F, Lackey A. Recovery from release of ureteral obstruction in the rat: relationship to nephrogenesis. Kidney Int 2002; 61:2033-43. [PMID: 12028444 DOI: 10.1046/j.1523-1755.2002.00359.x] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Obstructive nephropathy is a major cause of renal insufficiency in infants and children. Despite release of unilateral ureteral obstruction (UUO) in the first five days of life in the rat, renal growth is impaired, while glomerular filtration rate (GFR) is preserved at one month, but decreases markedly by one year. To test the hypothesis that renal recovery from UUO depends on the stage of nephrogenesis at the time of relief of obstruction, renal recovery from relief of five days UUO following completion of nephrogenesis (days 14 to 19) was compared with UUO during nephrogenesis (days 1 to 5). METHODS Rats underwent UUO or sham operation at one day of age, with relief five days later. In additional groups of neonatal rats, the operation was at 14 days, with relief at 19 days. Three months later, blood pressure, GFR, urine flow, sodium and potassium excretion, and kidney weight were measured. In addition, the number of glomeruli, glomerular maturation, glomerular diameter, tubular atrophy, and interstitial fibrosis were determined in each kidney. The effects of five-day UUO on number of glomeruli was determined also in adult rats one month following relief of obstruction. RESULTS Three months following relief of UUO during days 14 to 19, renal growth was decreased by 50%, compared to a 30% reduction following relief of UUO during days 1 to 5 (P < 0.05). The number of glomeruli was reduced by approximately 50% regardless of the timing of UUO, but glomerular size was reduced only in rats with UUO from days 14 to 19. Blood pressure and tubular atrophy were increased, and GFR, urine flow, sodium and potassium excretion were decreased in the postobstructed kidney of both neonatal groups. In the adult rat, the five-day UUO did not result in a decrease in the number of glomeruli. CONCLUSIONS In the period immediately following nephrogenesis, the kidney is particularly susceptible to long-term injury from temporary UUO. This suggests that a delay in relief of significant ureteral obstruction should be avoided if diagnosed in the perinatal or neonatal period.
Collapse
Affiliation(s)
- Robert L Chevalier
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia 22908, USA.
| | | | | | | | | |
Collapse
|
86
|
Affiliation(s)
- Keith A Hruska
- Barnes-Jewish Hospital at Washington University, St. Louis, Missouri, USA.
| |
Collapse
|
87
|
Okada H, Inoue T, Kanno Y, Kobayashi T, Watanabe Y, Kopp JB, Carey RM, Suzuki H. Interstitial fibroblast-like cells express renin-angiotensin system components in a fibrosing murine kidney. THE AMERICAN JOURNAL OF PATHOLOGY 2002; 160:765-72. [PMID: 11891174 PMCID: PMC1867169 DOI: 10.1016/s0002-9440(10)64898-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Recently, the renin-angiotensin system (RAS) was implicated in organ fibrosis. However, few studies have examined the localization of RAS components, such as angiotensin II receptors, renin (REN), angiotensinogen (AGTN), and angiotensin-converting enzyme (ACE), in the fibrosing kidney. To localize these components in the fibrosing kidney, we used a murine model of renal fibrosis that shows an enhanced expression of angiotensin II type 1A receptor (AT(1A)R) and AGTN. Our results indicate that the overall expression of angiotensin II type 2 receptor (AT(2)R) and ACE was attenuated in this model, whereas REN expression was unchanged. In addition to tubular epithelial cells that were positive for AT(1A)R, AT(2)R, REN, and AGTN, interstitial fibroblast-like cells expressed AT(1A)R, REN, AGTN, and ACE in the fibrosing kidney. The interstitial fibroblast-like cells that were positive for AT(1A)R mRNA were further characterized as positive for the expression of vimentin and transforming growth factor-beta1. These data provide strong evidence for a tubulointerstitial RAS within the fibrosing kidney, and a linkage between the RAS and renal fibrogenesis.
Collapse
Affiliation(s)
- Hirokazu Okada
- Department of Nephrology, Saitama Medical College, Saitama, Japan
| | | | | | | | | | | | | | | |
Collapse
|
88
|
Lange-Sperandio B, Cachat F, Thornhill BA, Chevalier RL. Selectins mediate macrophage infiltration in obstructive nephropathy in newborn mice. Kidney Int 2002; 61:516-24. [PMID: 11849391 DOI: 10.1046/j.1523-1755.2002.00162.x] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Urinary tract obstruction during development leads to tubular atrophy and causes interstitial fibrosis. Macrophage infiltration into the interstitium plays a central role in this process. Selectins, a family of three adhesion molecules, are involved in leukocyte recruitment to sites of inflammation and immune activity. We investigated the role of selectins in obstructive nephropathy in newborn mice. METHODS Triple selectin-deficient mice (EPL-/-), L-selectin deficient mice (L-/-) and wild type mice (WT) were subjected to complete unilateral ureteral obstruction (UUO) or sham operation within the first 48 hours of life, and were sacrificed 5 and 12 days later. Kidneys were removed, and sections were stained for macrophage infiltration (mAb F4/80), apoptosis (TUNEL), tubular atrophy (periodic acid-Schiff) and interstitial fibrosis (Masson trichrome). RESULTS Selectin deficient mice showed a marked reduction in macrophage infiltration into the obstructed kidney compared to WT at day 5 and day 12 after UUO. Tubular apoptosis was strongly reduced in EPL-/- at day 5 after UUO, and in EPL-/- and L-/- at day 12 after UUO when compared to WT. The number of apoptotic tubular cells was correlated with macrophage infiltration, suggesting that macrophages stimulate tubular apoptosis in obstructive nephropathy. In addition, tubular atrophy and interstitial fibrosis were significantly diminished in EPL-/- and L-/- compared to WT at day 12 after UUO. CONCLUSION Following UUO, selectins mediate macrophage infiltration into the obstructed kidney, which in turn may induce tubular apoptosis, tubular atrophy and interstitial fibrosis.
Collapse
Affiliation(s)
- Bärbel Lange-Sperandio
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia 22908-0386, USA
| | | | | | | |
Collapse
|
89
|
Gerth JH, Kriegsmann J, Trinh TT, Stahl RAK, Wendt T, Sommer M, Stein G, Wolf G. Induction of p27KIP1 after unilateral ureteral obstruction is independent of angiotensin II. Kidney Int 2002; 61:68-79. [PMID: 11786086 DOI: 10.1046/j.1523-1755.2002.00111.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Unilateral ureteral obstruction (UUO) is characterized by proliferation of tubular and interstitial cells, and infiltration of the renal parenchyma with macrophages/monocytes. These alterations lead ultimately to tubulointerstitial fibrosis and tubular atrophy. Some of these changes are caused by an activated renin-angiotensin system (RAS). We have previously demonstrated that angiotensin II induces the expression of the cell cycle inhibitor p27KIP1 in cultured tubular cells. The current study tested the hypothesis that interference with the RAS may modulate renal expression of p27KIP1 after UUO. METHODS The ureter of the left kidney of Sprague-Dawley rats was ligated. Sham-operated animals served as controls. Rats were randomized in four groups and received one of the following: no therapy, enalapril, losartan, or triple therapy (hydralazine, reserpine, hydrochlorothiazide). Kidneys were removed and cortical protein lysates were prepared for the detection of p27KIP1 by Western blotting. Immunohistochemistry was performed for p27KIP1, PCNA, ED-1, and alpha-smooth muscle actin. Apoptosis was quantified by TUNEL-staining. RESULTS p27KIP1 expression as detected by Western blotting reached a maximum 10 days after UUO. Tubular and interstitial cells contributed to this increase in p27KIP1 expression whereas the number of glomerular p27KIP1 positive cell did not change. p27KIP1-positive cells were macrophages/monocytes (positive ED-1 staining) or had the characteristics of myofibroblasts (positive alpha-smooth muscle actin staining). Tubular and interstitial proliferation [proliferating cell nuclear antigen (PCNA)-positive staining] and apoptosis [terminal deoxy transferase uridine triphosphate nick end labeling (TUNEL) staining] also was increased after UUO. However, individual cells stained either positive for p27KIP1 or PCNA, but not both. Although enalapril and losartan reduced the number of macrophages/monocytes and attenuated the degree of tubular and interstitial apoptosis, these drugs did not influence p27KIP1 expression. There was no change in the number of p27KIP1-positive cells in the contralateral kidney undergoing hypertrophy. CONCLUSION Induction of p27KIP1 in this model represents an endogenous response to likely limit proliferation that is independent of angiotensin II. Since there was no close correlation between apoptosis and p27KIP1 expression, it may be that the overall number of p27KIP1 expressing cells sets a general restriction point for apoptosis rather than defines an individual level of cell fate.
Collapse
Affiliation(s)
- Jens H Gerth
- Klinik für Innere Medizin IV der Universität Jena, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
90
|
Terada Y, Hanada S, Nakao A, Kuwahara M, Sasaki S, Marumo F. Gene transfer of Smad7 using electroporation of adenovirus prevents renal fibrosis in post-obstructed kidney. Kidney Int 2002; 61:S94-8. [PMID: 11841620 DOI: 10.1046/j.1523-1755.2002.0610s1094.x] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Unilateral ureteral obstruction (UUO) leads to interstitial fibrosis of the obstructed kidney, and TGF-beta is considered to play an important role in this fibrotic process. Smad7 has been recently identified as an antagonist of TGF-beta signaling. To investigate whether this novel molecule can be exploited for therapy of renal fibrosis, we determined the effect of exogenous Smad7, introduced by a recombinant adenovirus vector combined with in vivo electroporation (EP), on UUO-induced renal fibrosis in rats. METHODS A model of UUO was made in SD rats by ligating their left ureters. The next day, the rats were divided into four groups and adenovirus was injected into the extended pelvic space (two groups received AdCMV-LacZ and two groups received AdCMV-Smad7). Then, EP was performed in one group of AdCMV-LacZ-injected rats and one group of AdCMV-Smad7-injected rats. The renal tissues were obtained 3, 5, 10, and 14 days after the UUO operation. We detected the efficiency of transgene by immunoblots of renal cortical and medullary tissues and immunohistochemical studies for Smad7 and FLAG (the FLAG gene was introduced in the AdCMV-Smad7 as a marker). The renal fibrosis was monitored by histological scoring of Masson stainings. RESULTS In immunoblotting, both Smad7 and FLAG were clearly detected in the renal medullary tissue of the rats given AdCMV-Smad7 with EP. In contrast, immunoblots of renal cortical tissue did not demonstrate positive bands. In immunohistological study, Smad7 was stained in the renal medulla in the rats given AdCMV-Smad7 with EP. In the rats given AdCMV-Smad7 without EP, only a weak signal was detected in renal medullary tissue. The rats given AdCMV-Smad7 with EP demonstrated significantly more suppression of renal fibrosis than rats treated with AdCMV-LacZ. The rats treated with AdCMV-Smad7 without EP did not demonstrate significant suppression of renal fibrosis. CONCLUSION These data indicate that gene transfer of Smad7 prevents UUO-induced renal fibrosis, suggesting that Smad7 may be applicable for the treatment of renal fibrosis. In vivo electroporation of adenovirus may be a powerful tool for gene delivery in renal tissue.
Collapse
Affiliation(s)
- Yoshio Terada
- Homeostasis Medicine and Nephrology, Tokyo Medical and Dental University and Allergy Research Center, Juntendo University, School of Medicine, Tokyo, Japan.
| | | | | | | | | | | |
Collapse
|
91
|
Sakharova OV, Taal MW, Brenner BM. Pathogenesis of diabetic nephropathy: focus on transforming growth factor-beta and connective tissue growth factor. Curr Opin Nephrol Hypertens 2001; 10:727-38. [PMID: 11706299 DOI: 10.1097/00041552-200111000-00001] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Although considerable improvement in the prognosis of diabetic nephropathy has been achieved in recent years due to intensive insulin and angiotensin-converting enzyme inhibitor treatment, these approaches do not provide complete protection against progression of diabetic nephropathy. An urgent need for additional novel therapies to prevent or further slow the progression of diabetic nephropathy motivated us to provide an up-to-date review with particular emphasis on the potential role of two growth factors--transforming growth factor-beta and connective tissue growth factor--in the pathogenesis of diabetic nephropathy. The most intensively studied to date, transforming growth factor-beta appears to play a central role in the pathogenesis of diabetic nephropathy. Recently, attention has focused on connective tissue growth factor, which mimics the biological activity of transforming growth factor-beta in profibrotic tissue formation. Thus, acting as a downstream mediator of the profibrotic activity of transforming growth factor-beta, connective tissue growth factor may constitute a more specific target for future antifibrotic therapies.
Collapse
Affiliation(s)
- O V Sakharova
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA.
| | | | | |
Collapse
|
92
|
Ohishi M, Ueda M, Tanabe S, Han YS, Mendelsohn FA, Zhuo JL. Increased angiotensin-converting enzyme and type 1 angiotensin receptors in cortical vasculature and tubulointerstitium of chronically rejected human kidney allografts. Nephrology (Carlton) 2001. [DOI: 10.1046/j.1440-1797.2001.00045.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
93
|
Ayan S, Roth JA, Freeman MR, Bride SH, Peters CA. Partial ureteral obstruction dysregulates the renal renin-angiotensin system in the fetal sheep kidney. Urology 2001; 58:301-6. [PMID: 11489731 DOI: 10.1016/s0090-4295(01)01156-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
OBJECTIVES To investigate whether partial ureteral obstruction (PUO) in the fetus induces dysregulation of the renin-angiotensin system (RAS) and of transforming growth factor-beta 1 (TGF-beta1) and tissue inhibitors of metalloproteinase (TIMP1) expression. Previous studies have indicated that renal and urinary tract development depend on an intact renal RAS. Fetal urinary obstruction is distinct from postnatal obstruction. It has been suggested in postnatal animal studies that dysregulation of the RAS, and subsequent increased expression of TGF-beta1 and TIMP1, leads to changes in extracellular matrix composition. METHODS Bilateral PUO was created in 4 fetal sheep. Seven animals (four obstructed and three controls) were killed at birth and their kidneys removed. Semiquantitative reverse transcriptase-polymerase chain reaction was used to quantify the levels of renin, angiotensinogen, angiotensin receptor type 1 (AT1 receptor), angiotensin receptor type 2 (AT2 receptor), TGF-beta1, and TIMP1. These messages were normalized to glyceraldehyde-3-phosphate dehydrogenase mRNA. RESULTS All obstructed animals had moderate to severe hydronephrosis with enlarged kidneys (mean weight 22.0 g versus 9.4 g for the control animals; P <0.05). The increase in the levels of renin, angiotensinogen, AT1 receptor, TGF-beta1, and TIMP1 mRNA was significant in the PUO group compared with the control group (P <0.05). AT2 receptor levels did not increase, but the AT1/AT2 mRNA ratio was significantly increased over normal (P <0.005). Also, a significant linear correlation was found between the increased renal weight and increased TGF-beta1 mRNA levels (P <0.005). CONCLUSIONS Our findings suggest that fetal PUO can cause upregulation of the renal RAS and increased expression of TGF-beta1 and TIMP1, which may alter the balance between the generation and degradation of the extracellular matrix. The coordinate increases in renin, angiotensinogen, and AT1 receptor mRNA levels in chronic fetal PUO may represent a maladaptive response that contributes to interstitial fibrosis and prolonged vasoconstriction. RAS components and growth factors, particularly TGF-beta1, may be considered relevant targets in the prevention and treatment of congenital obstructive nephropathy.
Collapse
Affiliation(s)
- S Ayan
- Department of Urology, Children's Hospital, Boston, Massachusetts 02115, USA
| | | | | | | | | |
Collapse
|
94
|
Guo G, Morrissey J, McCracken R, Tolley T, Liapis H, Klahr S. Contributions of angiotensin II and tumor necrosis factor-alpha to the development of renal fibrosis. Am J Physiol Renal Physiol 2001; 280:F777-85. [PMID: 11292619 DOI: 10.1152/ajprenal.2001.280.5.f777] [Citation(s) in RCA: 101] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Angiotensin II upregulates tumor necrosis factor-alpha (TNF-alpha) in the rat kidney with unilateral ureteral obstruction (UUO). In a mouse model of UUO, we found that tubulointerstitial fibrosis is blunted when the TNF-alpha receptor, TNFR1, is functionally knocked out. In this study, we used mutant mice with UUO in which the angiotensin II receptor AT(1a) or the TNF-alpha receptors TNFR1 and TNFR2 were knocked out to elucidate interactions between the two systems. The contribution of both systems to renal fibrosis was assessed by treating TNFR1/TNFR2-double knockout (KO) mice with an angiotensin-converting enzyme inhibitor, enalapril. The increased interstitial volume (Vv(int)) in the C57BI/6 wild-type mouse was decreased in the AT(1a) KO from 32.8 +/- 4.0 to 21.0 +/- 3.7% (P < 0.005) or in the TNFR1/TNFR2 KO to 22.3 +/- 2.1% (P < 0.005). The Vv(int) of the TNFR1/TNFR2 KO was further decreased to 15.2 +/- 3.7% (P < 0.01) by enalapril compared with no treatment. The induction of TNF-alpha mRNA and transforming growth factor-beta1 (TGF-beta1) mRNA in the kidney with UUO was significantly blunted in the AT(1a) or TNFR1/TNFR2 KO mice compared with the wild-type mice. Treatment of the TNFR1/TNFR2 KO mouse with enalapril reduced both TNF-alpha and TGF-beta1 mRNA and their proteins to near normal levels. Also, alpha-smooth muscle actin expression and myofibroblast proliferation were significantly inhibited in the AT(1a) or TNFR1/TNFR2 KO mice, and they were further inhibited in enalapril-treated TNFR1/TNFR2 KO mice. Incapacitating the angiotensin II or the TNF-alpha systems individually leads to partial blunting of fibrosis. Incapacitating both systems, by using a combination of genetic and pharmacological means, further inhibited interstitial fibrosis and tubule atrophy in obstructive nephropathy.
Collapse
MESH Headings
- Actins/biosynthesis
- Angiotensin II/physiology
- Angiotensin-Converting Enzyme Inhibitors/pharmacology
- Animals
- Antigens, CD/genetics
- Collagen/metabolism
- Enalapril/pharmacology
- Enzyme-Linked Immunosorbent Assay
- Fibrosis
- Kidney/pathology
- Kidney/ultrastructure
- Lymphotoxin-alpha/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Microscopy, Electron
- Muscle, Smooth/metabolism
- RNA, Messenger/biosynthesis
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor, Type I
- Receptors, Tumor Necrosis Factor, Type II
- Tumor Necrosis Factor-alpha/physiology
- Ureteral Obstruction/pathology
Collapse
Affiliation(s)
- G Guo
- Departments of Internal Medicine, Washington University School of Medicine at Barnes-Jewish Hospital, 216 S. Kingshighway Blvd., St. Louis, Missouri 63110-1092, USA
| | | | | | | | | | | |
Collapse
|
95
|
Miyajima A, Chen J, Poppas DP, Vaughan ED, Felsen D. Role of nitric oxide in renal tubular apoptosis of unilateral ureteral obstruction. Kidney Int 2001; 59:1290-303. [PMID: 11260390 DOI: 10.1046/j.1523-1755.2001.0590041290.x] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND The obstructed kidney in unilateral ureteral obstruction (UUO) is characterized by renal atrophy and tissue loss, which is mediated by renal tubular apoptosis. We sought to determine whether NO is involved in renal tubular apoptosis in vitro and in vivo. METHODS Rat renal tubular epithelial cells (NRK-52E) were subjected to mechanical stretch, and apoptosis and cell size were analyzed by flow cytometry. Furthermore, we studied UUO in mice lacking the gene for inducible nitric oxide synthase (iNOS-/-) and their wild-type littermates. Tubular apoptosis and proliferation were detected by immunostaining. NOS activity and NOS expression were assessed by a citrulline assay and Western blot, respectively. RESULTS Stretching-induced apoptosis in NRK-52E, which was reduced when NO was increased; conversely, stretch-induced apoptosis was increased when a NOS inhibitor was added to the cells. Stretched cells are larger and more apoptotic than unstretched cells. In UUO, the obstructed kidney of iNOS-/- mice exhibited more apoptotic renal tubules than the wild-type mice through 14 days of UUO. The obstructed kidney of iNOS-/- mice at day 3 showed more proliferative tubules compared with wild type. The obstructed kidney of wild-type mice exhibited higher total NOS activity until day 7 after UUO compared with iNOS-/- mice. However, the obstructed kidney of day 14 wild-type mice exhibited significantly lower iNOS activity and protein compared with the day 0 kidney. CONCLUSION These results suggest that mechanical stretch is related to renal tubular apoptosis and that NO plays a protective role in this system in UUO.
Collapse
Affiliation(s)
- A Miyajima
- Center for Pediatric Urology and Laboratory for Minimally Invasive Urologic Surgery, Department of Urology, New York Presbyterian Hospital, Weill Medical College of Cornell University, New York, New York 10021, USA
| | | | | | | | | |
Collapse
|
96
|
Satoh M, Kashihara N, Yamasaki Y, Maruyama K, Okamoto K, Maeshima Y, Sugiyama H, Sugaya T, Murakami K, Makino H. Renal interstitial fibrosis is reduced in angiotensin II type 1a receptor-deficient mice. J Am Soc Nephrol 2001; 12:317-325. [PMID: 11158221 DOI: 10.1681/asn.v122317] [Citation(s) in RCA: 114] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Unilateral ureteral obstruction (UUO) results in tubulointerstitial fibrosis of the affected kidney by stimulating the renin-angiotensin system. This study established a UUO model in angiotensin type 1a receptor (AT1a) deficient (mutant) mice to elucidate the role of angiotensin II through AT1a on the fibrosis of the obstructed kidney (OBK). The relative volume of the tubulointerstitium was measured by an image analyzer; deposition of collagen types III and IV and monocyte/macrophage infiltration were histologically examined using specific antibodies. Also determined were the mRNA levels of transforming growth factor-beta by Northern blot analysis. Nuclear factor-kappaB activity was assessed by gel shift assay. UUO in wild mice resulted in a marked expansion of relative volume of the tubulointerstitium, together with increased deposition of collagen types III and IV and number of infiltrated monocytes/macrophages in the interstitium, relative to sham-operated mice. In comparison, these changes were significantly lower in mutant mice with UUO. The mRNA level of transforming growth factor-beta was significantly higher in the OBK of wild mice with UUO compared with sham-operated mice. In contrast, the increase in mRNA level in the OBK of mutant mice was significantly less than in wild mice. Finally, UUO resulted in activation of nuclear factor-kappaB in wild mice but was inhibited in the OBK of mutant mice. The results provide direct evidence that angiotensin II acting via the AT1a plays a pivotal role in the development of tubulointerstitial fibrosis in UUO.
Collapse
Affiliation(s)
- Minoru Satoh
- Department of Medicine III, Okayama University Medical School, Okayama, Japan
| | - Naoki Kashihara
- Division of Nephrology, Department of Internal Medicine, Kawasaki Medical School, Kurashiki, Japan
| | - Yasushi Yamasaki
- Department of Medicine III, Okayama University Medical School, Okayama, Japan
| | - Keisuke Maruyama
- Department of Medicine III, Okayama University Medical School, Okayama, Japan
| | - Kazunori Okamoto
- Department of Medicine III, Okayama University Medical School, Okayama, Japan
| | - Youhei Maeshima
- Department of Medicine III, Okayama University Medical School, Okayama, Japan
| | - Hitoshi Sugiyama
- Department of Medicine III, Okayama University Medical School, Okayama, Japan
| | - Takeshi Sugaya
- Discovery Research Laboratory, Tanabe Seiyaku Co., Ltd., Osaka, Japan
| | - Kazuo Murakami
- Institute of Applied Biochemistry, Tsukuba Advanced Research Alliance, University of Tsukuba, Ibaragi, Japan
| | - Hirofumi Makino
- Department of Medicine III, Okayama University Medical School, Okayama, Japan
| |
Collapse
|
97
|
Iwano M, Fischer A, Okada H, Plieth D, Xue C, Danoff TM, Neilson EG. Conditional abatement of tissue fibrosis using nucleoside analogs to selectively corrupt DNA replication in transgenic fibroblasts. Mol Ther 2001; 3:149-59. [PMID: 11237671 DOI: 10.1006/mthe.2000.0251] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Progressive tissue fibrosis can compromise epithelial function resulting in organ failure. Appreciating evidence suggests that fibroblasts provide fibrogenic collagens during such injury. We further tested this notion by attempting to reduce the physiologic consequences of organ fibrosis through the selective killing of fibroblasts at sites of injury. Here, we report the conditional reduction of tissue fibroblasts using the coding sequence for herpesvirus thymidine kinase (DeltaTK) put under the control of a cell-specific promoter from the gene encoding fibroblast-specific protein 1 (FSP1). Transgenic fibroblasts from mice carrying FSP1.DeltaTK minigenes expressed thymidine kinase concordantly with native FSP1 and, compared to transgenic epithelium, were selectively susceptible to the lethal effects of nucleoside analogs either in culture or during experimental renal fibrosis. The numbers of fibroblasts in fibrogenic kidney tissue were reduced on exposure to nucleoside analogs as was the degree of type I collagen deposition and the extent of fibrosis. Fibroblast reduction following the stress of DNA chain termination highlights the important contribution of cell division during fibrogenesis. Our findings convey a proof of principle regarding the importance of FSP1(+) fibroblasts in fibrosis as well as providing a new approach to treating the relentless scarification of tissue.
Collapse
Affiliation(s)
- M Iwano
- Division of Nephrology and Hypertension, Vanderbilt University, Nashville, Tennessee 37232, USA
| | | | | | | | | | | | | |
Collapse
|
98
|
|
99
|
DUSSAULE JEANCLAUDE, THARAUX PIERRELOUIS, BOFFA JEANJACQUES, FAKHOURI FADI, ARDAILLOU RAYMOND, CHATZIANTONIOU CHRISTOS. Mechanisms Mediating the Renal Profibrotic Actions of Vasoactive Peptides in Transgenic Mice. J Am Soc Nephrol 2000. [DOI: 10.1681/asn.v11suppl_2s124] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Abstract. Transgenic mice are useful tools to investigate the mechanisms of the renal profibrotic actions of endothelin and angiotensin II. The overexpression of angiotensinogen and renin genes induces renal sclerosis independently of changes in systemic hemodynamics. The same results are observed when the endothelin-1 gene is overexpressed. Transgenic mice harboring the luciferase gene, under the control of the collagen I α2 chain promoter, and made hypertensive by induction of a nitric oxide (NO) deficiency have been studied. In this strain of mice, luciferase activity is an early index of renal and vascular fibrosis. Luciferase activity was increased in preglomerular arterioles and glomeruli when mice were treated with Nω-nitro-L-arginine methyl ester, an inhibitor of NO synthases. Bosentan (an endothelin receptor antagonist) was as efficient as losartan (an AT1 receptor antagonist) in preventing renal fibrosis, although it did not decrease BP. In short-term experiments, angiotensin II produced an increase in luciferase activity that was entirely prevented by losartan but also by bosentan. It can be concluded that, during chronic inhibition of NO, the collagen I gene is activated, which contributes to the development of nephroangiosclerosis and glomerulosclerosis. Angiotensin II plays a major role in this fibrogenic process, and its effect is at least partly independent of systemic hemodynamics and mediated by the profibrotic action of endothelin-1.
Collapse
|
100
|
Chevalier RL, Thornhill BA, Chang AY. Unilateral ureteral obstruction in neonatal rats leads to renal insufficiency in adulthood. Kidney Int 2000; 58:1987-95. [PMID: 11044219 DOI: 10.1111/j.1523-1755.2000.00371.x] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Although unilateral ureteropelvic junction obstruction is the most common cause of congenital obstructive nephropathy in infants and children, management remains controversial, and follow-up after pyeloplasty is generally limited to the pediatric ages. We have developed a model of temporary unilateral ureteral obstruction (UUO) in the neonatal rat: One month following the relief of five-day UUO, the glomerular filtration rate (GFR) of the postobstructed kidney was normal despite a 40% reduction in the number of glomeruli and residual vascular, glomerular, tubular, and interstitial injury. METHODS To determine whether hyperfiltration and residual injury of remaining nephrons leads to progression of renal insufficiency in later life, 31 rats were sham operated or subjected to left UUO at one day of age, with relief of UUO five days later, and were studied at one year of age. GFR was measured by inulin clearance, and the number of glomeruli, tubular atrophy, glomerular sclerosis, and interstitial fibrosis were measured by histomorphometry in sham, obstructed (UUO), and intact opposite kidneys. Intrarenal macrophages and alpha-smooth muscle actin were identified by immunohistochemistry. RESULTS Despite relief of UUO, ultimate growth of the postobstructed kidney was impaired. The number of glomeruli was reduced by 40%, and GFR was decreased by 80%. However, despite significant compensatory growth of the opposite kidney, there was no compensatory increase in GFR, and proteinuria was increased. Moreover, glomerular sclerosis, tubular atrophy, macrophage infiltration, and interstitial fibrosis were significantly increased not only in the postobstructed kidney, but also in the opposite kidney. CONCLUSIONS Although GFR is initially maintained following relief of five-day UUO in the neonatal rat, there is eventual profound loss of function of the postobstructed and opposite kidneys because of progressive tubulointerstitial and glomerular damage. These findings suggest that despite normal postoperative GFR in infancy, children undergoing pyeloplasty for ureteropelvic junction obstruction should be followed into adulthood. Elucidation of the cellular response to temporary UUO may lead to improved methods to assess renal growth, injury, and functional reserve in patients with congenital obstructive nephropathy.
Collapse
Affiliation(s)
- R L Chevalier
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia 22908, USA.
| | | | | |
Collapse
|