Skip to main content
Erschienen in: Diabetology & Metabolic Syndrome 1/2014

Open Access 01.12.2014 | Research

Ramipril inhibits AGE-RAGE-induced matrix metalloproteinase-2 activation in experimental diabetic nephropathy

verfasst von: Kei Fukami, Sho-ichi Yamagishi, Melinda T Coughlan, Brooke E Harcourt, Phillip Kantharidis, Vicki Thallas-Bonke, Seiya Okuda, Mark E Cooper, Josephine M Forbes

Erschienen in: Diabetology & Metabolic Syndrome | Ausgabe 1/2014

Abstract

Background

Advanced glycation end products (AGE)-receptor for AGE (RAGE) axis and renin-angiotensin system (RAS) play a role in diabetic nephropathy (DN). Matrix metalloproteinase-2 (MMP-2) activation also contributes to DN. However, the pathological interaction among AGE-RAGE, RAS and MMP-2 in DN remains unknown. We examined here the involvement of AGE and RAS in MMP-2 activation in streptozotocin (STZ)-induced diabetic rats and in AGE-exposed rat renal proximal tubular cells (RPTCs).

Methods

Experimental diabetes was induced in 6-week-old male Sprague–Dawley (SD) rats by intravenous injection of STZ. Diabetic rats received ramipril (3 mg/kg body weight/day) or vehicle for 32 weeks. AGE-modified rat serum albumin (AGE-RSA) or RSA was intraperitoneally administrated to 6-week-old male SD rats for 16 weeks. RPTCs were stimulated with 100 μg/ml AGE-modified bovine serum albumin (AGE-BSA) or BSA in the presence or absence of 10-7 M ramiprilat, an inhibitor of angiotensin-converting enzyme or 100 nM BAY11-7082, an IκB-α phosphorylation inhibitor.

Results

AGE and RAGE expression levels and MMP-2 activity in the tubules of diabetic rats was significantly increased in association with increased albuminuria, all of which were blocked by ramipril. AGE infusion induced tubular MMP-2 activation and RAGE gene expression in SD rats. Ramiprilat or BAY11-7082 inhibited the AGE-induced MMP-2 activation or reactive oxygen species generation in RPTCs. Angiotensin II increased MMP-2 gene expression in RPTCs, which was blocked by BAY11-7082.

Conclusions

Our present study suggests the involvement of AGE-RAGE-induced, RAS-mediated MMP-2 activation in experimental DN. Blockade of AGE-RAGE axis by ramipril may protect against DN partly via suppression of MMP-2.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1758-5996-6-86) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that there are no conflicts of interest.

Authors’ contributions

MTC, BEH, PK and VTB kindly managed diabetic and AGE-infused rats and measured urinary albumin excretion and AGE concentrations, and supported western blotting and real-time PCR. SY, SO, MEC and JMF supervised our experiments and the manuscript. All authors have approved the final version of the manuscript.
Abkürzungen
DN
Diabetic nephropathy
ECM
Extracellular matrix
AGE
Advanced glycation end products
RAGE
Receptor for AGE
MMPs
Matrix metalloproteinases
RAS
Renin-angiotensin system
ACE
Angiotensin-converting enzyme
Ang II
Angiotensin II
AT1R
Ang II type-1 receptor
STZ
Streptozotocin
ROS
Reactive oxygen species
rat RPTCs
Renal proximal tubular cells
SD
Sprague–Dawley
Ctrl
Control
SBP
Systolic blood pressure
GFR
Glomerular filtration rate
UAE
Urinary albumin excretion
ELISA
Enzyme-linked immunosorbent assay
AGE-BSA
AGE-modified bovine serum albumin
PBS
Phosphate buffer saline
CML
Carboxymethyllysine
EDTA
Ethylenediaminetetraacetic acid
SDS-PAGE
Sodium dodecyl sulfate-polyacrylamide gel electrophoresis
RT-PCR
Reverse-transcription polymerase chain reaction
HbA1c
Glycated hemoglobin.

Introduction

Diabetic nephropathy (DN) is a leading cause of end-stage renal disease, which could account for disability and high mortality rate in patients with diabetes [1, 2]. The development and progression of DN are characterized by glomerular hypertrophy and inflammatory cell infiltration, followed by extracellular matrix (ECM) accumulation in mesangial area and an increased albumin excretion rate [3]. Diabetic nephropathy ultimately progresses glomerular sclerosis associated with renal dysfunction [4]. However, it has recently been recognized that changes within tubulointerstitium are more important than glomerulopathy in terms of renal dysfunction in DN [5, 6].
Reducing sugars can react non-enzymatically with the amino groups of proteins to initiate a complex series of rearrangements and dehydrations, and then to produce a class of irreversibly cross-linked moieties termed advanced glycation end products (AGE) [79]. The formation and accumulation of AGE in various tissues have been shown to progress at an accelerated rate under hyperglycemic conditions [1012]. There is accumulating evidence that AGE and receptor for AGE (RAGE) interaction induces oxidative stress generation and subsequently evokes inflammatory reactions, thereby causing progressive alteration in renal architecture and loss of renal function in diabetes [1315].
Matrix metalloproteinases (MMPs) are a family of zinc-dependent endopeptidases comprising more than 20 members that can degrade numerous types of ECM components [16]. Among various MMPs, MMP-2 has attracted great attention because it can degrade type-I and -IV collagen and laminin, major components of tubular basement membrane and interstitium [17, 18]. Recently, we have found that serum MMP-2 levels are one of the independent determinants of proteinuria in patients with chronic kidney disease [19]. Further, plasma MMP-2 levels and its activity are significantly higher in type-1 diabetic patients compared with control subjects [20], and urinary MMP-2 values are positively associated with renal hyperfiltration and albuminuria in these diabetic patients as well [20]. In addition, in animal models, MMP-2 has been shown to induce renal tubular cell epithelial-mesenchymal transformation, which could cause tubulointerstitial fibrosis in diabetic nephropathy [21, 22]. These observations suggest the involvement of MMP-2 activation in albuminuria and tubulointerstitial injury of diabetic nephropathy [23].
There is accumulating evidence to show the active participation of renin-angiotensin system (RAS) in DN as well [24]. Indeed, inhibition of RAS by angiotensin-converting enzyme (ACE) inhibitor or angiotensin II (Ang II) type-1 receptor (AT1R) blocker has been shown to suppress the development and progression of nephropathy in both type-1 and type-2 diabetic subjects [25, 26]. Further, losartan, an AT1R blocker, improves renal outcome in patients with type-2 diabetes [27]. Moreover, we have previously found that RAS blockers could inhibit the AGE-elicited mesangial cell hypertrophy, proximal tubular cell injury, and podocyte DNA damage and detachment in vitro [2830]. However, the involvement of AGE and RAS in MMP-2 activation in DN remains unknown. Therefore, we first examined the effects of ramipril, an inhibitor of ACE on MMP-2 activity, AGE and RAGE expression in renal tubules of streptozotocin (STZ)-induced diabetic rats. Then we investigated whether AGE injection could stimulate RAGE gene expression and MMP-2 activity in tublules of normal non-diabetic rats. We further studied the effects of ramiprilat, an active metabolite of ramipril, on MMP-2 activity and reactive oxygen species (ROS) generation in AGE-exposed rat renal proximal tubular cells (RPTCs).

Methods

Experimental animal models

Experimental diabetes was induced in 6-week-old male Sprague–Dawley (SD) rats (200–250 g) by intravenous injection of STZ (50 mg/kg body weight) in sodium citrate buffer pH 4.5, following an overnight fast [31]. Rats with plasma glucose concentrations in excess of 15 mmol/L were included in this study. Vehicle-injected control (Ctrl) animals (n = 15) were followed concurrently. Diabetic rats were randomized into two groups and followed for 32 weeks; one group (n = 15) received a vehicle, and the other an ACE inhibitor, ramipril (1 mg/kg body weight/day in drinking water; generously provided by Sanofi, Bridgewater, NJ) for 32 weeks (n = 16) [32]. Two units of ultralente insulin (Ultratard HM, Novo Industries, Bagsvaerd, Denmark) were administrated daily to diabetic animals to prevent ketoacidosis and avoid death. In addition, non-diabetic normal male SD rats (6-week-old) were infused intraperitoneally with AGE-modified rat serum albumin (AGE-RSA) (n = 10) or RSA (n = 9) at a dose of 20 mg/kg body weight/day for 16 weeks by an osmotic pump (Alzet osmotic pumps, model 1004, Cupertino, CA, USA).
Systolic blood pressure (SBP) was measured by tail-cuff plethysmography as described previously [33]. Glomerular filtration rate (GFR) was evaluated by 99Tc-DTPA, and urinary albumin excretion (UAE) levels by an enzyme-linked immunosorbent assay (ELISA) kit (Bethyl Laboratories, Montgomery, TX, USA). Other clinical valuables were measured as described previously [31]. All animal procedures were in accordance with guidelines set by the Baker IDI Heart and Diabetes Institute Ethics Committee and the National Health and Medical Research Council of Australia.

Preparation of AGE-RSA and AGE-modified bovine serum albumin (AGE-BSA)

AGE-RSA and AGE-BSA were prepared by incubating RSA or BSA (Fraction V, Sigma Chemical Co, St. Louis, MO, USA) with 0.5 M D-glucose in PBS at 37°C for 3 months as previously described [34]. After sterilization using 0.2 μm micropore filters, unincorporated glucose was removed by dialysis against phosphate buffer saline (PBS) at 4°C for 48 hr. Samples were passed through Detoxigel column (Pierce Biotechnology Inc., Rockford, IL, USA) in order to remove endotoxin. Preparations were tested for endotoxin using Limulus Amebocyte Lysate validity testing (AMS Laboratories, Sydney, Australia); no endotoxin was detected. Finally, the solution was filtered through 0.2 μm micropore filter in sterile conditions, and percentage of lysine modifications and carboxymethyllysine (CML) moieties were determined by Selective Ion Monitoring Gas chromatography–mass spectrometry as previously described [35]. Control non-glycated RSA or BSA was incubated in the same conditions except for the absence of glucose.

Isolation of renal tubules from kidneys

The cortical tissue was minced and gently pushed through a 250 μm steinless steel mesh with 0.9% sodium chloride solution, and sieved stepwise through 125 μm and 75 μm meshes. The tubules were collected on the 125 μm mesh as described previously [36]. Tubular structures were confirmed by light microscopy. After centrifugation, the tubules were re-suspended in the lysis buffer (50 mM Tris–HCl, 150 mM NaCl, 0.02% sodium azide, 0.1% SDS, 1% Nonidet P-40, 0.5% sodium deoxycholate) containing a complete protease inhibitor cocktail (Roche Molecular Biochemicals, Mannheim, Germany) without ethylenediaminetetraacetic acid (EDTA). Then the samples were sonicated and the protein concentration was measured using a BCA protein assay kit (Pierce Biotechnology Inc.).

Gelatin zymography

Twenty μg protein isolated from renal tubules or cell culture media were separated by 8% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) containing 1 mg/ml gelatin (Labchem, Auburn, NSW, Australia), and active MMP-2 levels were evaluated by zymography as previously described [14].

Measurement of AGE levels in tubules

AGE levels in the isolated tubules were measured with CML ELISA system as previously described [37]. In brief, 1 μg of samples diluted in 50 mM carbonate buffer (pH 9.6) (1:800) or standards were added to a microtitre plate (Nunc-Immuno MaxiSorp, Nunc, Kamstrup, Roskilde, Denmark) and incubated overnight at 4°C. After washing three times with PBS (pH 7.4) containing 0.1% Tween-20, wells were blocked for 1 hr with PBS containing 1% BSA, and then 100 μl of a rabbit polyclonal anti-CML antibody (5 μg/ml) was added [38]. After 1 hr incubation and washing, 100 μl of 0.2 μg/ml goat-anti-rabbit IgG biotinylated antibody (Dako Corporation, Carpinteria, CA, USA) was added to each well. After 1 hr of shaking, wells were washed and 100 μl of streptavidin horseradish peroxidase (Dako Corporation) was added for 30 min. The wells were washed and 100 μl of Tetramethylbenzidine (Sigma Chemical Co.) was added, the reaction being terminated after 15 min using 100 μl 1.8 M H2SO4. The absorbance was quantitated using a microtitre plate reader at 450 nm (EMax, Molecular Devices Corporation, Sunnyvale, CA, USA).

Western blot analysis

Ten μg protein isolated from renal tubules was separated by 10% SDS-PAGE electrophoresis, and then transferred to a polyvinylidene fluoride membrane (Hybond P; Amersham, Buckinghamshire, UK). Membranes were incubated with mouse anti-RAGE (1:1000) or rabbit anti-α-tubulin (1:1000) antibodies overnight, and then the secondary antibody horseradish peroxidase-conjugated mouse or rabbit IgG for 1 hr. Bound antibodies were detected by reaction with an enhanced chemiluminescence kit (Pierce Biotechnology Inc.). All antibodies were obtained from Chemicon (Santa Cruz, SA, USA).

Quantitative real-time reverse-transcription polymerase chain reaction (RT-PCR)

About 4 μg of total RNA extracted from each kidney cortex or RPTCs were used to synthesize cDNA with the SuperScript First-Strand Synthesis System for RT-PCR (Invitrogen, Carlsband, CA, USA) as previously described [35]. Quantitative real-time RT-PCR was performed using Assay-on-Demand and TaqMan 5 fluorogenic nuclease chemistry (Applied Biosystems, Foster City, CA, USA) according to the supplier’s recommendation. The forward, reverse primers and specific probes for rat RAGE and MMP-2 genes were 5′-TCCTGGTGGGACCGTGAC-3′, 5′-GGGTGTGCCATCTTTTATCCA-3′, and FAM5′-TGTGCCATCTCTGC-3′-MGB, and 5′-GCCCCTATCTACACCTACACCAA-3′, 5′-TGGATCCCCTTGATGTCATCA-3′, and FAM-5′-AACTTCCGATTATCCC-3′-MGB, respectively. TaqMan Ribosomal RNA Control Reagents (18S) was used as an endogenous control (Applied Biosystems).

Isolation and characterization of RPTCs

RPTCs were isolated from rat kidney as previously described [39]. In brief, kidneys were removed from male SD rats (200–250 g), and renal cortex was minced and centrifuged at 250 g, 4°C for 5 min. Final pellets were digested in Dulbecco’s Modified Eagle’s Medium/Ham’s F12 (1:1) containing 1 mg/mL collagenase (Type-2, Worthington Biochemical Corporation, Lakewood, NJ, USA) for 30 min at 37°C with constant agitation. This suspension was filtered through a 75 μm pore size metal sieve to remove glomeruli, and re-suspended in a 50% Percoll solution (Sigma-Aldrich Chemie GmbH, Buchs, Switzerland) followed by centrifugation at 26,500 g at 4°C for 30 min. The lowest band was retrieved as it is enriched for proximal tubule fragments at a purity of greater than 98% as described previously [40]. RPTCs were characterized as cobble stone-like appearance and immunocytochemical characteristics with positive staining for cytokeratin and P. vulgaris lectin in the absence of CD90.1 (thy1.1) using confocal microscopy. Treatments with AGE-BSA, Ang II (Sigma Chemical Co.), an inhibitor of ACE, ramiprilat, or an inhibitor of IκB-α phosphorylation, BAY11-7082 ((E)-3-(4-methylphenylsulfonyl)-2-propenenitrile) (Biomol international, Lp, USA) [41] were carried out in Minimum Essential Medium containing D-valine instead of L-valine without fetal calf serum in humidified 5% CO2/95% air atmosphere at 37°C.

Intracellular ROS generation

RPTCs were treated with 100 μg/ml AGE-BSA or BSA in the presence or absence of 10-7 M ramiprilat for 24 hr, and then intracellular ROS generation was measured using the fluorescent probe 5,6-chloromethyl- 2′,7′-dichlorohydrofluorescein diacetate (Molecular Probes, Eugene, OR, USA) as described previously [28].

Statistical analysis

Results are expressed as mean ± standard error. Data for albuminuria were not normally distributed, therefore analyzed as logarithmic transformation. One-way ANOVA followed by the Tukey-test or unpaired t-test was performed for statistical comparisons; p < 0.05 was considered significant. All statistical analyses were performed with SPSS system (Ver. 20, SPSS, Chicago, IL, USA).

Results

Characteristics of animals

Compared with non-diabetic Ctrl rats, plasma glucose, glycated hemoglobin (HbA1c), SBP, kidney-to-body weight ratio, GFR and UAE levels were significantly higher in diabetic rats (p < 0.001) (Table 1). SBP was partially, but not significantly, decreased after ramipril treatment (Table 1). Furthermore, although plasma glucose, HbA1c or GFR levels were not affected by the treatment with ramipril, it significantly reduced the UAE levels in diabetic rats (p < 0.001) (Table 1).
Table 1
Characteristics of animals
 
Ctrl
DM
DM + ramipril
RSA
AGE-RSA
Number
15
15
16
9
10
Plasma glucose (mmol/L)
6.9 ± 095
33.2 ± 2.7*
33.1 ± 3.5*
7.9 ± 1.0
7.5 ± 0.5
%HbA1c (%)
5.5 ± 0.7
18.3 ± 2.6*
19.3 ± 2.7*
4.1 ± 1.5
3.9 ± 0.8
Systolic BP (mmHg)
114 ± 9
134 ± 13*
122 ± 8#
121 ± 4
118 ± 7
KW/BW ratio
5.3 ± 0.6
10.9 ± 1.5*
11.3 ± 1.1*
5.4 ± 0.3
5.4 ± 0.4
eGFR (ml/min)
6.7 ± 1.1
11.2 ± 1.4*
12.0 ± 1.7*
4.3 ± 1.0
4.4 ± 0.6
UAE (mg/24 h)
5.6 ± 4.3
53.9 ± 51.8*
13.0 ± 6.8$
1.0 ± 0.4
1.3 ± 0.5
Data are mean ± SEM. *p < 0.001 vs Ctrl, #p < 0.05 vs Ctrl, $p < 0.001 vs DM.
Abbreviation: Ctrl control, DM diabetes mellitus, RSA rat serum albumin, AGE-RSA advanced glycation end product-modified RSA, HbA1c glycated hemoglobin, BP blood pressure, KW/BW ratio kidney-to-body weight ratio, GFR glomerular filtration rate, UAE urinary albumin excretion.
Compared with RSA treatment, AGE-RSA injection tended to increase UAE levels, but the effects were modest, not significant (p = 0.13) (Table 1). In addition, AGE-RSA injection did not affect any clinical parameters in SD rats compared with RSA infusion.

Ramipril decreased MMP-2 activity, AGE and RAGE expression in the tubules of diabetic rats

As shown in Figure 1A-C, compared with Ctrl rats, MMP-2 activity, AGE and RAGE expression levels in the tubules of diabetic rats was significantly increased, all of which were inhibited by the treatment with ramipril.

AGE-RSA infusion increased tubular MMP-2 activity and RAGE gene expression in SD rats

We next investigated whether AGE-RSA could directly induce MMP-2 activation in tubules. We evaluated renal rather than tubular RAGE gene expression in the present study because (1) tubules and interstitium make up approximately 80-90% of the renal volume and (2) isolation of tubules from the kidney cortex might affect mRNA stability and expression level [42]. As shown in Figure 1D and E, compared with RSA-infused rats, renal RAGE gene expression and tubular MMP-2 activity were significantly increased in AGE-RSA-infused rats.

Ramiprilat suppressed the AGE-elicited MMP-2 activation and ROS generation in RPTCs

We examined whether and how AGE could induce MMP-2 activation in vitro. Since AGE or Ang II exert various biological effects via activation of redox-sensitive transcription factor NF-κB, we studied the effects of BAY11-7082, an inhibitor of NF-κB activation on AGE- or Ang II-induced MMP-2 gene expression and activity in PRTCs. Treatment with 100 μg/ml AGE-BSA significantly increased active MMP-2 production by RPTCs, which was completely prevented by 10-7 M ramiprilat or 100 nM BAY11-7082 (Figure 2A and B). Ramiprilat completely blocked the AGE-induced ROS generation in RPTCs, whereas 10-6 M Ang II significantly increased MMP-2 gene expression, which was also inhibited by BAY11-7082 (Figure 2C and D).

Discussion

In the present study, we demonstrated that (1) MMP-2 activity, AGE and RAGE expression levels were significantly increased in renal tubules of diabetic rats, all of which were blocked by the treatment with ramipril; (2) UAE, a prognostic marker of DN, was increased in diabetic rats, which was also inhibited by ramipril; (3) AGE injection significantly increased renal RAGE gene expression and MMP-2 activity in the tubules of normal SD rats; (4) AGE treatment enhanced MMP-2 activity and ROS generation in RPTCs, which were also suppressed by ramiprilat; and (5) inhibition of NF-κB activation by BAY11-7082 blocked the MMP-2 activity or gene expression in AGE- or Ang II-exposed RPTCs, respectively.
In this study, ramipril decreased the AGE accumulation and RAGE expression in the renal tubules of diabetic rats. We have previously shown that there could exist the crosstalk between AGE-RAGE axis and RAS in the pathogenesis of DN [14]. Indeed, irbesartan, an AT1R blocker, inhibited the AGE-induced apoptotic cell death and inflammatory and fibrotic reactions in human PTCs by reducing ROS generation via suppression of RAGE expression [29] (AGE-RAGE → RAS). Furthermore, AGE elicited mesangial cell hypertrophy by inducing Ang II production, which was also blocked by candesartan, other type of AT1R blocker [28] (AGE-RAGE → RAS). In addition, Ang II infusion increased RAGE expression in retinas via ROS generation [43] (RAS → AGE-RAGE). Consistent with these findings, treatment with low-dose valsartan, an AT1R antagonist, decreased serum AGE levels in association with reduced oxidative stress generation in type-2 diabetic patients [44] (RAS → AGE-RAGE). Therefore, ramipril could inhibit the AGE-RAGE system by suppression the RAS (Figure 3). Engagement of RAGE with AGE stimulates ROS generation, which could promote the formation and accumulation of AGE and subsequent RAGE expression, thereby making a vicious cycle between RAGE-downstream signaling pathways and AGE formation in a variety of cells [45, 46]. Therefore, it is also conceivable that ramipril could break the vicious cycle between AGE and RAGE downstream pathway by blocking the crosstalk between the RAS and AGE-RAGE via inhibition of ROS generation (Figure 3). In our in vitro experiment, ramiprilat completely inhibited AGE-elicited ROS generation in RPTCs, which could support our speculation.
In our study, we demonstrated that RAS inhibition by ramipril suppressed diabetes or AGE-induced MMP-2 activation in vivo and vitro. It has been reported that AGE-RAGE interaction induces MMP-2 expression and activation in several cell lines [47, 48]. The interaction of AGE-modified amyloid β and RAGE has been shown to induce MMP-2 expression and vascular inflammatory stress in brain endothelial cells [47]. MMP-2 activity in gingival extracts from diabetic mice was significantly increased, which was inhibited by the administration with soluble RAGE, acting as a decoy receptor for AGE [48]. Further, increased vascular ROS generation and NF-κB activation also promote MMP-2 activity in rat model of renovascular hypertension [49]. In our present study, AGE-induced MMP-2 activation was inhibited by the blockade of NF-κB, thus suggesting that AGE-RAGE-mediated ROS generation might induce MMP-2 activation via NF-κB. In addition, Ang II-induced MMP-2 expression was completely suppressed by the inhibition of NF-κB in RPTCs as well. These observations suggest that AGE-RAGE axis might stimulate Ang II generation, which could evoke the ROS-NF-κB signaling pathway, thereby being implicated in the tubular MMP-2 activation in diabetes.
It has been shown that decreased glomerular MMP-2 activity is associated with increased mesangial ECM accumulation and glomerular sclerosis in DN [50]. However, there is still controversy about the role of MMP-2 in DN. It is reported that the increased MMP-2 activation in tubules was involved in the development and progression of tubulointerstitial injury [51]. Furthermore, we have recently found that serum MMP-2 levels were positively associated with proteinuria and inversely correlated with estimated GFR in patients with chronic kidney disease [19]. These observations further support the concept that ramipril might inhibit AGE-RAGE axis, which could slow down the development and progression of tubulointerstitial injury by reducing albuminuria in DN via suppression of MMP-2.

Limitations

We performed experiments in Figure 2A and B, separately. This is a reason why two gels looked slightly different and the fold increase in active MMP-2 after AGE is lower in Figure 2B in respect to Figure 2A. However, there was no statistically significant difference of the increase of active MMP-2 after AGE treatment between Figure 2A and B experiments (p = 0.11). Although the increase of ROS generation in RPTCs stimulated with AGE versus BSA was modest, we found here that (1) ramipril significantly suppressed the AGE-induced MMP-2 activation and ROS generation in RPTCs and (2) inhibition of NF-κB by BAY11-7082 completely inhibited the increase of active MMP-2 in AGE-exposed cells. These findings suggest the biological significance of modest ROS generation in the AGE-signaling.

Conclusions

In this study, we found that MMP-2 activity, AGE and RAGE expressions were increased in renal tubules of type 1 diabetic rats, all of which were blocked by the treatment with ramipril. Further, AGE infusion induced tubular MMP-2 activation and RAGE gene expression in rats. Ramiprilat inhibited the AGE-induced MMP-2 activation or reactive oxygen species generation in RPTCs. These findings suggest the involvement of AGE-RAGE-induced, RAS-mediated MMP-2 activation in experimental DN. Blockade of AGE-RAGE axis by ramipril may protect against DN partly via suppression of MMP-2.

Acknowledgement

The authors thank Prof. Suzanne Thorpe, University of South Carolina, for the analysis of AGE moieties in RSA and BSA preparations. This work was supported by the Juvenile Diabetes Research Foundation (JDRF) and the National Health and Medical Research Council of Australia. Josephine Forbes is an NHMRC fellow.
This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​4.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Competing interests

The authors declare that there are no conflicts of interest.

Authors’ contributions

MTC, BEH, PK and VTB kindly managed diabetic and AGE-infused rats and measured urinary albumin excretion and AGE concentrations, and supported western blotting and real-time PCR. SY, SO, MEC and JMF supervised our experiments and the manuscript. All authors have approved the final version of the manuscript.
Anhänge

Authors’ original submitted files for images

Literatur
1.
Zurück zum Zitat Krolewski AS, Warram JH, Valsania P, Martin BC, Laffel LM, Christlieb AR: Evolving natural history of coronary artery disease in diabetes mellitus. Am J Med. 1991, 90 (2A): 56S-61S.CrossRefPubMed Krolewski AS, Warram JH, Valsania P, Martin BC, Laffel LM, Christlieb AR: Evolving natural history of coronary artery disease in diabetes mellitus. Am J Med. 1991, 90 (2A): 56S-61S.CrossRefPubMed
2.
Zurück zum Zitat Maisonneuve P, Agodoa L, Gellert R, Stewart JH, Buccianti G, Lowenfels AB, Wolfe RA, Jones E, Disney AP, Briggs D, McCredie M, Boyle P: Distribution of primary renal diseases leading to end-stage renal failure in the United States, Europe, and Australia/New Zealand: results from an international comparative study. Am J Kidney Dis. 2000, 35 (1): 157-165.CrossRefPubMed Maisonneuve P, Agodoa L, Gellert R, Stewart JH, Buccianti G, Lowenfels AB, Wolfe RA, Jones E, Disney AP, Briggs D, McCredie M, Boyle P: Distribution of primary renal diseases leading to end-stage renal failure in the United States, Europe, and Australia/New Zealand: results from an international comparative study. Am J Kidney Dis. 2000, 35 (1): 157-165.CrossRefPubMed
3.
Zurück zum Zitat Mauer SM, Lane P, Hattori M, Fioretto P, Steffes MW: Renal structure and function in insulin-dependent diabetes mellitus and type I membranoproliferative glomerulonephritis in humans. J Am Soc Nephrol. 1992, 2 (10 Suppl): S181-S184.PubMed Mauer SM, Lane P, Hattori M, Fioretto P, Steffes MW: Renal structure and function in insulin-dependent diabetes mellitus and type I membranoproliferative glomerulonephritis in humans. J Am Soc Nephrol. 1992, 2 (10 Suppl): S181-S184.PubMed
4.
Zurück zum Zitat Sharma K, Ziyadeh FN: Hyperglycemia and diabetic kidney disease. The case for transforming growth factor-beta as a key mediator. Diabetes. 1995, 44 (10): 1139-1146.CrossRefPubMed Sharma K, Ziyadeh FN: Hyperglycemia and diabetic kidney disease. The case for transforming growth factor-beta as a key mediator. Diabetes. 1995, 44 (10): 1139-1146.CrossRefPubMed
5.
Zurück zum Zitat Taft JL, Nolan CJ, Yeung SP, Hewitson TD, Martin FI: Clinical and histological correlations of decline in renal function in diabetic patients with proteinuria. Diabetes. 1994, 43 (8): 1046-1051.CrossRefPubMed Taft JL, Nolan CJ, Yeung SP, Hewitson TD, Martin FI: Clinical and histological correlations of decline in renal function in diabetic patients with proteinuria. Diabetes. 1994, 43 (8): 1046-1051.CrossRefPubMed
6.
Zurück zum Zitat Ziyadeh FN, Goldfarb S: The renal tubulointerstitium in diabetes mellitus. Kidney Int. 1991, 39 (3): 464-475.CrossRefPubMed Ziyadeh FN, Goldfarb S: The renal tubulointerstitium in diabetes mellitus. Kidney Int. 1991, 39 (3): 464-475.CrossRefPubMed
7.
Zurück zum Zitat Brownlee M, Cerami A, Vlassara H: Advanced glycosylation end products in tissue and the biochemical basis of diabetic complications. N Engl J Med. 1988, 318 (20): 1315-1321.CrossRefPubMed Brownlee M, Cerami A, Vlassara H: Advanced glycosylation end products in tissue and the biochemical basis of diabetic complications. N Engl J Med. 1988, 318 (20): 1315-1321.CrossRefPubMed
8.
Zurück zum Zitat Grandhee SK, Monnier VM: Mechanism of formation of the Maillard protein cross-link pentosidine. Glucose, fructose, and ascorbate as pentosidine precursors. J Biol Chem. 1991, 266 (18): 11649-11653.PubMed Grandhee SK, Monnier VM: Mechanism of formation of the Maillard protein cross-link pentosidine. Glucose, fructose, and ascorbate as pentosidine precursors. J Biol Chem. 1991, 266 (18): 11649-11653.PubMed
9.
Zurück zum Zitat Dyer DG, Blackledge JA, Thorpe SR, Baynes JW: Formation of pentosidine during nonenzymatic browning of proteins by glucose. Identification of glucose and other carbohydrates as possible precursors of pentosidine in vivo. J Biol Chem. 1991, 266 (18): 11654-11660.PubMed Dyer DG, Blackledge JA, Thorpe SR, Baynes JW: Formation of pentosidine during nonenzymatic browning of proteins by glucose. Identification of glucose and other carbohydrates as possible precursors of pentosidine in vivo. J Biol Chem. 1991, 266 (18): 11654-11660.PubMed
10.
Zurück zum Zitat Genuth S, Sun W, Cleary P, Sell DR, Dahms W, Malone J, Sivitz W, Monnier VM, Group DSCAS: Glycation and carboxymethyllysine levels in skin collagen predict the risk of future 10-year progression of diabetic retinopathy and nephropathy in the diabetes control and complications trial and epidemiology of diabetes interventions and complications participants with type 1 diabetes. Diabetes. 2005, 54 (11): 3103-3111.PubMedCentralCrossRefPubMed Genuth S, Sun W, Cleary P, Sell DR, Dahms W, Malone J, Sivitz W, Monnier VM, Group DSCAS: Glycation and carboxymethyllysine levels in skin collagen predict the risk of future 10-year progression of diabetic retinopathy and nephropathy in the diabetes control and complications trial and epidemiology of diabetes interventions and complications participants with type 1 diabetes. Diabetes. 2005, 54 (11): 3103-3111.PubMedCentralCrossRefPubMed
11.
Zurück zum Zitat Thomas MC, Forbes JM, Cooper ME: Advanced glycation end products and diabetic nephropathy. Am J Ther. 2005, 12 (6): 562-572.CrossRefPubMed Thomas MC, Forbes JM, Cooper ME: Advanced glycation end products and diabetic nephropathy. Am J Ther. 2005, 12 (6): 562-572.CrossRefPubMed
12.
Zurück zum Zitat Nożyński J, Zakliczyński M, Konecka-Mrowka D, Zielinska T, Zakliczynska H, Nikiel B, Mlynarczyk-Liszka J, Mrowka A, Zembala-Nozynska E, Pijet M, Rdzanowska K, Lange D, Przybylski R, Zembala M: Advanced glycation end product accumulation in the cardiomyocytes of heart failure patients with and without diabetes. Ann Transplant. 2012, 17 (2): 53-61.CrossRefPubMed Nożyński J, Zakliczyński M, Konecka-Mrowka D, Zielinska T, Zakliczynska H, Nikiel B, Mlynarczyk-Liszka J, Mrowka A, Zembala-Nozynska E, Pijet M, Rdzanowska K, Lange D, Przybylski R, Zembala M: Advanced glycation end product accumulation in the cardiomyocytes of heart failure patients with and without diabetes. Ann Transplant. 2012, 17 (2): 53-61.CrossRefPubMed
13.
Zurück zum Zitat Beisswenger PJ, Drummond KS, Nelson RG, Howell SK, Szwergold BS, Mauer M: Susceptibility to diabetic nephropathy is related to dicarbonyl and oxidative stress. Diabetes. 2005, 54 (11): 3274-3281.CrossRefPubMed Beisswenger PJ, Drummond KS, Nelson RG, Howell SK, Szwergold BS, Mauer M: Susceptibility to diabetic nephropathy is related to dicarbonyl and oxidative stress. Diabetes. 2005, 54 (11): 3274-3281.CrossRefPubMed
14.
Zurück zum Zitat Fukami K, Yamagishi S, Ueda S, Okuda S: Role of AGEs in diabetic nephropathy. Curr Pharm Des. 2008, 14 (10): 946-952.CrossRefPubMed Fukami K, Yamagishi S, Ueda S, Okuda S: Role of AGEs in diabetic nephropathy. Curr Pharm Des. 2008, 14 (10): 946-952.CrossRefPubMed
15.
Zurück zum Zitat Coughlan MT, Thorburn DR, Penfold SA, Laskowski A, Harcourt BE, Sourris KC, Tan AL, Fukami K, Thallas-Bonke V, Nawroth PP, Brownlee M, Bierhaus A, Cooper ME, Forbes JM: RAGE-induced cytosolic ROS promote mitochondrial superoxide generation in diabetes. J Am Soc Nephrol. 2009, 20 (4): 742-752.PubMedCentralCrossRefPubMed Coughlan MT, Thorburn DR, Penfold SA, Laskowski A, Harcourt BE, Sourris KC, Tan AL, Fukami K, Thallas-Bonke V, Nawroth PP, Brownlee M, Bierhaus A, Cooper ME, Forbes JM: RAGE-induced cytosolic ROS promote mitochondrial superoxide generation in diabetes. J Am Soc Nephrol. 2009, 20 (4): 742-752.PubMedCentralCrossRefPubMed
16.
Zurück zum Zitat Mason RM, Wahab NA: Extracellular matrix metabolism in diabetic nephropathy. J Am Soc Nephrol. 2003, 14 (5): 1358-1373.CrossRefPubMed Mason RM, Wahab NA: Extracellular matrix metabolism in diabetic nephropathy. J Am Soc Nephrol. 2003, 14 (5): 1358-1373.CrossRefPubMed
17.
Zurück zum Zitat Karagiannis ED, Popel AS: A theoretical model of type I collagen proteolysis by matrix metalloproteinase (MMP) 2 and membrane type 1 MMP in the presence of tissue inhibitor of metalloproteinase 2. J Biol Chem. 2004, 279 (37): 39105-39114.CrossRefPubMed Karagiannis ED, Popel AS: A theoretical model of type I collagen proteolysis by matrix metalloproteinase (MMP) 2 and membrane type 1 MMP in the presence of tissue inhibitor of metalloproteinase 2. J Biol Chem. 2004, 279 (37): 39105-39114.CrossRefPubMed
18.
Zurück zum Zitat Lenz O, Elliot SJ, Stetler-Stevenson WG: Matrix metalloproteinases in renal development and disease. J Am Soc Nephrol. 2000, 11 (3): 574-581.PubMed Lenz O, Elliot SJ, Stetler-Stevenson WG: Matrix metalloproteinases in renal development and disease. J Am Soc Nephrol. 2000, 11 (3): 574-581.PubMed
19.
Zurück zum Zitat Nagano M, Fukami K, Yamagishi S, Ueda S, Kaida Y, Matsumoto T, Yoshimura J, Hazama T, Takamiya Y, Kusumoto T, Gohara S, Tanaka H, Adachi H, Okuda S: Circulating matrix metalloproteinase-2 is an independent correlate of proteinuria in patients with chronic kidney disease. Am J Nephrol. 2009, 29 (2): 109-115.CrossRefPubMed Nagano M, Fukami K, Yamagishi S, Ueda S, Kaida Y, Matsumoto T, Yoshimura J, Hazama T, Takamiya Y, Kusumoto T, Gohara S, Tanaka H, Adachi H, Okuda S: Circulating matrix metalloproteinase-2 is an independent correlate of proteinuria in patients with chronic kidney disease. Am J Nephrol. 2009, 29 (2): 109-115.CrossRefPubMed
20.
Zurück zum Zitat Thrailkill KM, Bunn RC, Moreau CS, Cockrell GE, Simpson PM, Coleman HN, Frindik JP, Kemp SF, Fowlkes JL: Matrix metalloproteinase-2 dysregulation in type 1 diabetes. Diabetes Care. 2007, 30 (9): 2321-2326.PubMedCentralCrossRefPubMed Thrailkill KM, Bunn RC, Moreau CS, Cockrell GE, Simpson PM, Coleman HN, Frindik JP, Kemp SF, Fowlkes JL: Matrix metalloproteinase-2 dysregulation in type 1 diabetes. Diabetes Care. 2007, 30 (9): 2321-2326.PubMedCentralCrossRefPubMed
21.
Zurück zum Zitat Cheng S, Lovett DH: Gelatinase A (MMP-2) is necessary and sufficient for renal tubular cell epithelial-mesenchymal transformation. Am J Pathol. 2003, 162 (6): 1937-1949.PubMedCentralCrossRefPubMed Cheng S, Lovett DH: Gelatinase A (MMP-2) is necessary and sufficient for renal tubular cell epithelial-mesenchymal transformation. Am J Pathol. 2003, 162 (6): 1937-1949.PubMedCentralCrossRefPubMed
22.
Zurück zum Zitat Essawy M, Soylemezoglu O, Muchaneta-Kubara EC, Shortland J, Brown CB, el Nahas AM: Myofibroblasts and the progression of diabetic nephropathy. Nephrol Dial Transplant. 1997, 12 (1): 43-50.CrossRefPubMed Essawy M, Soylemezoglu O, Muchaneta-Kubara EC, Shortland J, Brown CB, el Nahas AM: Myofibroblasts and the progression of diabetic nephropathy. Nephrol Dial Transplant. 1997, 12 (1): 43-50.CrossRefPubMed
23.
Zurück zum Zitat Zandi-Nejad K, Eddy AA, Glassock RJ, Brenner BM: Why is proteinuria an ominous biomarker of progressive kidney disease?. Kidney Int Suppl. 2004, 92: S76-S89.CrossRefPubMed Zandi-Nejad K, Eddy AA, Glassock RJ, Brenner BM: Why is proteinuria an ominous biomarker of progressive kidney disease?. Kidney Int Suppl. 2004, 92: S76-S89.CrossRefPubMed
24.
Zurück zum Zitat Fukami K, Yamagishi S, Ueda S, Okuda S: Novel therapeutic targets for diabetic nephropathy. Endocr Metab Immune Disord Drug Targets. 2007, 7 (2): 83-92.CrossRefPubMed Fukami K, Yamagishi S, Ueda S, Okuda S: Novel therapeutic targets for diabetic nephropathy. Endocr Metab Immune Disord Drug Targets. 2007, 7 (2): 83-92.CrossRefPubMed
25.
Zurück zum Zitat Lewis EJ, Hunsicker LG, Bain RP, Rohde RD: The effect of angiotensin-converting-enzyme inhibition on diabetic nephropathy. The Collaborative Study Group. N Engl J Med. 1993, 329 (20): 1456-1462.CrossRefPubMed Lewis EJ, Hunsicker LG, Bain RP, Rohde RD: The effect of angiotensin-converting-enzyme inhibition on diabetic nephropathy. The Collaborative Study Group. N Engl J Med. 1993, 329 (20): 1456-1462.CrossRefPubMed
26.
Zurück zum Zitat Haller H, Ito S, Izzo JL, Januszewicz A, Katayama S, Menne J, Mimran A, Rabelink TJ, Ritz E, Ruilope LM, Rump LC, Viberti G: Olmesartan for the delay or prevention of microalbuminuria in type 2 diabetes. N Engl J Med. 2011, 364 (10): 907-917.CrossRefPubMed Haller H, Ito S, Izzo JL, Januszewicz A, Katayama S, Menne J, Mimran A, Rabelink TJ, Ritz E, Ruilope LM, Rump LC, Viberti G: Olmesartan for the delay or prevention of microalbuminuria in type 2 diabetes. N Engl J Med. 2011, 364 (10): 907-917.CrossRefPubMed
27.
Zurück zum Zitat Brenner BM, Cooper ME, de Zeeuw D, Keane WF, Mitch WE, Parving HH, Remuzzi G, Snapinn SM, Zhang Z, Shahinfar S: Effects of losartan on renal and cardiovascular outcomes in patients with type 2 diabetes and nephropathy. N Engl J Med. 2001, 345 (12): 861-869.CrossRefPubMed Brenner BM, Cooper ME, de Zeeuw D, Keane WF, Mitch WE, Parving HH, Remuzzi G, Snapinn SM, Zhang Z, Shahinfar S: Effects of losartan on renal and cardiovascular outcomes in patients with type 2 diabetes and nephropathy. N Engl J Med. 2001, 345 (12): 861-869.CrossRefPubMed
28.
Zurück zum Zitat Fukami K, Ueda S, Yamagishi S, Kato S, Inagaki Y, Takeuchi M, Motomiya Y, Bucala R, Iida S, Tamaki K, Imaizumi T, Cooper ME, Okuda S: AGEs activate mesangial TGF-beta-Smad signaling via an angiotensin II type I receptor interaction. Kidney Int. 2004, 66 (6): 2137-2147.CrossRefPubMed Fukami K, Ueda S, Yamagishi S, Kato S, Inagaki Y, Takeuchi M, Motomiya Y, Bucala R, Iida S, Tamaki K, Imaizumi T, Cooper ME, Okuda S: AGEs activate mesangial TGF-beta-Smad signaling via an angiotensin II type I receptor interaction. Kidney Int. 2004, 66 (6): 2137-2147.CrossRefPubMed
29.
Zurück zum Zitat Matsui T, Yamagishi S, Takeuchi M, Ueda S, Fukami K, Okuda S: Irbesartan inhibits advanced glycation end product (AGE)-induced proximal tubular cell injury in vitro by suppressing receptor for AGEs (RAGE) expression. Pharmacol Res. 2010, 61 (1): 34-39.CrossRefPubMed Matsui T, Yamagishi S, Takeuchi M, Ueda S, Fukami K, Okuda S: Irbesartan inhibits advanced glycation end product (AGE)-induced proximal tubular cell injury in vitro by suppressing receptor for AGEs (RAGE) expression. Pharmacol Res. 2010, 61 (1): 34-39.CrossRefPubMed
30.
Zurück zum Zitat Fukami K, Yamagishi S, Kaifu K, Matsui T, Kaida Y, Ueda S, Takeuchi M, Asanuma K, Okuda S: Telmisartan inhibits AGE-induced podocyte damage and detachment. Microvasc Res. 2013, 88: 79-83.CrossRefPubMed Fukami K, Yamagishi S, Kaifu K, Matsui T, Kaida Y, Ueda S, Takeuchi M, Asanuma K, Okuda S: Telmisartan inhibits AGE-induced podocyte damage and detachment. Microvasc Res. 2013, 88: 79-83.CrossRefPubMed
31.
Zurück zum Zitat Forbes JM, Thallas V, Thomas MC, Founds HW, Burns WC, Jerums G, Cooper ME: The breakdown of preexisting advanced glycation end products is associated with reduced renal fibrosis in experimental diabetes. Faseb J. 2003, 17 (12): 1762-1764.PubMed Forbes JM, Thallas V, Thomas MC, Founds HW, Burns WC, Jerums G, Cooper ME: The breakdown of preexisting advanced glycation end products is associated with reduced renal fibrosis in experimental diabetes. Faseb J. 2003, 17 (12): 1762-1764.PubMed
32.
Zurück zum Zitat Forbes JM, Cooper ME, Thallas V, Burns WC, Thomas MC, Brammar GC, Lee F, Grant SL, Burrell LA, Jerums G, Osicka TM: Reduction of the accumulation of advanced glycation end products by ACE inhibition in experimental diabetic nephropathy. Diabetes. 2002, 51 (11): 3274-3282.CrossRefPubMed Forbes JM, Cooper ME, Thallas V, Burns WC, Thomas MC, Brammar GC, Lee F, Grant SL, Burrell LA, Jerums G, Osicka TM: Reduction of the accumulation of advanced glycation end products by ACE inhibition in experimental diabetic nephropathy. Diabetes. 2002, 51 (11): 3274-3282.CrossRefPubMed
33.
Zurück zum Zitat Bunag RD: Validation in awake rats of a tail-cuff method for measuring systolic pressure. J Appl Physiol. 1973, 34 (2): 279-282.PubMed Bunag RD: Validation in awake rats of a tail-cuff method for measuring systolic pressure. J Appl Physiol. 1973, 34 (2): 279-282.PubMed
34.
Zurück zum Zitat Oldfield MD, Bach LA, Forbes JM, Nikolic-Paterson D, McRobert A, Thallas V, Atkins RC, Osicka T, Jerums G, Cooper ME: Advanced glycation end products cause epithelial-myofibroblast transdifferentiation via the receptor for advanced glycation end products (RAGE). J Clin Invest. 2001, 108 (12): 1853-1863.PubMedCentralCrossRefPubMed Oldfield MD, Bach LA, Forbes JM, Nikolic-Paterson D, McRobert A, Thallas V, Atkins RC, Osicka T, Jerums G, Cooper ME: Advanced glycation end products cause epithelial-myofibroblast transdifferentiation via the receptor for advanced glycation end products (RAGE). J Clin Invest. 2001, 108 (12): 1853-1863.PubMedCentralCrossRefPubMed
35.
Zurück zum Zitat Forbes JM, Thorpe SR, Thallas-Bonke V, Pete J, Thomas MC, Deemer ER, Bassal S, El-Osta A, Long DM, Panagiotopoulos S, Jerums G, Osicka TM, Cooper ME: Modulation of soluble receptor for advanced glycation end products by angiotensin-converting enzyme-1 inhibition in diabetic nephropathy. J Am Soc Nephrol. 2005, 16 (8): 2363-2372.CrossRefPubMed Forbes JM, Thorpe SR, Thallas-Bonke V, Pete J, Thomas MC, Deemer ER, Bassal S, El-Osta A, Long DM, Panagiotopoulos S, Jerums G, Osicka TM, Cooper ME: Modulation of soluble receptor for advanced glycation end products by angiotensin-converting enzyme-1 inhibition in diabetic nephropathy. J Am Soc Nephrol. 2005, 16 (8): 2363-2372.CrossRefPubMed
37.
Zurück zum Zitat Coughlan MT, Thallas-Bonke V, Pete J, Long DM, Gasser A, Tong DC, Arnstein M, Thorpe SR, Cooper ME, Forbes JM: Combination therapy with the advanced glycation end product cross-link breaker, alagebrium, and angiotensin converting enzyme inhibitors in diabetes: synergy or redundancy?. Endocrinology. 2007, 148 (2): 886-895.CrossRefPubMed Coughlan MT, Thallas-Bonke V, Pete J, Long DM, Gasser A, Tong DC, Arnstein M, Thorpe SR, Cooper ME, Forbes JM: Combination therapy with the advanced glycation end product cross-link breaker, alagebrium, and angiotensin converting enzyme inhibitors in diabetes: synergy or redundancy?. Endocrinology. 2007, 148 (2): 886-895.CrossRefPubMed
38.
Zurück zum Zitat Forbes JM, Soulis T, Thallas V, Panagiotopoulos S, Long DM, Vasan S, Wagle D, Jerums G, Cooper ME: Renoprotective effects of a novel inhibitor of advanced glycation. Diabetologia. 2001, 44 (1): 108-114.CrossRefPubMed Forbes JM, Soulis T, Thallas V, Panagiotopoulos S, Long DM, Vasan S, Wagle D, Jerums G, Cooper ME: Renoprotective effects of a novel inhibitor of advanced glycation. Diabetologia. 2001, 44 (1): 108-114.CrossRefPubMed
39.
Zurück zum Zitat Lewis MP, Fine LG, Norman JT: Pexicrine effects of basement membrane components on paracrine signaling by renal tubular cells. Kidney Int. 1996, 49 (1): 48-58.CrossRefPubMed Lewis MP, Fine LG, Norman JT: Pexicrine effects of basement membrane components on paracrine signaling by renal tubular cells. Kidney Int. 1996, 49 (1): 48-58.CrossRefPubMed
40.
Zurück zum Zitat Vinay P, Gougoux A, Lemieux G: Isolation of a pure suspension of rat proximal tubules. Am J Physiol. 1981, 241 (4): F403-F411.PubMed Vinay P, Gougoux A, Lemieux G: Isolation of a pure suspension of rat proximal tubules. Am J Physiol. 1981, 241 (4): F403-F411.PubMed
41.
Zurück zum Zitat Pierce JW, Schoenleber R, Jesmok G, Best J, Moore SA, Collins T, Gerritsen ME: Novel inhibitors of cytokine-induced IkappaBalpha phosphorylation and endothelial cell adhesion molecule expression show anti-inflammatory effects in vivo. J Biol Chem. 1997, 272 (34): 21096-21103.CrossRefPubMed Pierce JW, Schoenleber R, Jesmok G, Best J, Moore SA, Collins T, Gerritsen ME: Novel inhibitors of cytokine-induced IkappaBalpha phosphorylation and endothelial cell adhesion molecule expression show anti-inflammatory effects in vivo. J Biol Chem. 1997, 272 (34): 21096-21103.CrossRefPubMed
42.
Zurück zum Zitat Van der Hauwaert C, Savary G, Gnemmi V, Glowacki F, Pottier N, Bouillez A, Maboudou P, Zini L, Leroy X, Cauffiez C, Perrais M, Aubert S: Isolation and characterization of a primary proximal tubular epithelial cell model from human kidney by CD10/CD13 double labeling. PLoS One. 2013, 8 (6): e66750-PubMedCentralCrossRefPubMed Van der Hauwaert C, Savary G, Gnemmi V, Glowacki F, Pottier N, Bouillez A, Maboudou P, Zini L, Leroy X, Cauffiez C, Perrais M, Aubert S: Isolation and characterization of a primary proximal tubular epithelial cell model from human kidney by CD10/CD13 double labeling. PLoS One. 2013, 8 (6): e66750-PubMedCentralCrossRefPubMed
43.
Zurück zum Zitat Yamagishi S, Takeuchi M, Matsui T, Nakamura K, Imaizumi T, Inoue H: Angiotensin II augments advanced glycation end product-induced pericyte apoptosis through RAGE overexpression. FEBS Lett. 2005, 579 (20): 4265-4270.CrossRefPubMed Yamagishi S, Takeuchi M, Matsui T, Nakamura K, Imaizumi T, Inoue H: Angiotensin II augments advanced glycation end product-induced pericyte apoptosis through RAGE overexpression. FEBS Lett. 2005, 579 (20): 4265-4270.CrossRefPubMed
44.
Zurück zum Zitat Saisho Y, Komiya N, Hirose H: Effect of valsartan, an angiotensin II receptor blocker, on markers of oxidation and glycation in Japanese type 2 diabetic subjects: blood pressure-independent effect of valsartan. Diabetes Res Clin Pract. 2006, 74 (2): 201-203.CrossRefPubMed Saisho Y, Komiya N, Hirose H: Effect of valsartan, an angiotensin II receptor blocker, on markers of oxidation and glycation in Japanese type 2 diabetic subjects: blood pressure-independent effect of valsartan. Diabetes Res Clin Pract. 2006, 74 (2): 201-203.CrossRefPubMed
45.
Zurück zum Zitat Yamagishi S, Imaizumi T: Diabetic vascular complications: pathophysiology, biochemical basis and potential therapeutic strategy. Curr Pharm Des. 2005, 11 (18): 2279-2299.CrossRefPubMed Yamagishi S, Imaizumi T: Diabetic vascular complications: pathophysiology, biochemical basis and potential therapeutic strategy. Curr Pharm Des. 2005, 11 (18): 2279-2299.CrossRefPubMed
46.
Zurück zum Zitat Soro-Paavonen A, Watson AM, Li J, Paavonen K, Koitka A, Calkin AC, Barit D, Coughlan MT, Drew BG, Lancaster GI, Thomas M, Forbes JM, Nawroth PP, Bierhaus A, Cooper ME, Jandeleit-Dahm KA: Receptor for advanced glycation end products (RAGE) deficiency attenuates the development of atherosclerosis in diabetes. Diabetes. 2008, 57 (9): 2461-2469.PubMedCentralCrossRefPubMed Soro-Paavonen A, Watson AM, Li J, Paavonen K, Koitka A, Calkin AC, Barit D, Coughlan MT, Drew BG, Lancaster GI, Thomas M, Forbes JM, Nawroth PP, Bierhaus A, Cooper ME, Jandeleit-Dahm KA: Receptor for advanced glycation end products (RAGE) deficiency attenuates the development of atherosclerosis in diabetes. Diabetes. 2008, 57 (9): 2461-2469.PubMedCentralCrossRefPubMed
47.
Zurück zum Zitat Du H, Li P, Wang J, Qing X, Li W: The interaction of amyloid β and the receptor for advanced glycation endproducts induces matrix metalloproteinase-2 expression in brain endothelial cells. Cell Mol Neurobiol. 2012, 32 (1): 141-147.CrossRefPubMed Du H, Li P, Wang J, Qing X, Li W: The interaction of amyloid β and the receptor for advanced glycation endproducts induces matrix metalloproteinase-2 expression in brain endothelial cells. Cell Mol Neurobiol. 2012, 32 (1): 141-147.CrossRefPubMed
48.
Zurück zum Zitat Lalla E, Lamster IB, Feit M, Huang L, Spessot A, Qu W, Kislinger T, Lu Y, Stern DM, Schmidt AM: Blockade of RAGE suppresses periodontitis-associated bone loss in diabetic mice. J Clin Invest. 2000, 105 (8): 1117-1124.PubMedCentralCrossRefPubMed Lalla E, Lamster IB, Feit M, Huang L, Spessot A, Qu W, Kislinger T, Lu Y, Stern DM, Schmidt AM: Blockade of RAGE suppresses periodontitis-associated bone loss in diabetic mice. J Clin Invest. 2000, 105 (8): 1117-1124.PubMedCentralCrossRefPubMed
49.
Zurück zum Zitat Cau SB, Guimaraes DA, Rizzi E, Ceron CS, Souza LL, Tirapelli CR, Gerlach RF, Tanus-Santos JE: Pyrrolidine dithiocarbamate down-regulates vascular matrix metalloproteinases and ameliorates vascular dysfunction and remodelling in renovascular hypertension. Br J Pharmacol. 2011, 164 (2): 372-381.PubMedCentralCrossRefPubMed Cau SB, Guimaraes DA, Rizzi E, Ceron CS, Souza LL, Tirapelli CR, Gerlach RF, Tanus-Santos JE: Pyrrolidine dithiocarbamate down-regulates vascular matrix metalloproteinases and ameliorates vascular dysfunction and remodelling in renovascular hypertension. Br J Pharmacol. 2011, 164 (2): 372-381.PubMedCentralCrossRefPubMed
50.
Zurück zum Zitat Del Prete D, Anglani F, Forino M, Ceol M, Fioretto P, Nosadini R, Baggio B, Gambaro G: Down-regulation of glomerular matrix metalloproteinase-2 gene in human NIDDM. Diabetologia. 1997, 40 (12): 1449-1454.CrossRefPubMed Del Prete D, Anglani F, Forino M, Ceol M, Fioretto P, Nosadini R, Baggio B, Gambaro G: Down-regulation of glomerular matrix metalloproteinase-2 gene in human NIDDM. Diabetologia. 1997, 40 (12): 1449-1454.CrossRefPubMed
51.
Zurück zum Zitat Iwazu Y, Muto S, Hirahara I, Fujisawa G, Takeda S, Kusano E: Matrix metalloproteinase 2 induces epithelial-mesenchymal transition in proximal tubules from the luminal side and progresses fibrosis in mineralocorticoid/salt-induced hypertensive rats. J Hypertens. 2011, 29 (12): 2440-2453.CrossRefPubMed Iwazu Y, Muto S, Hirahara I, Fujisawa G, Takeda S, Kusano E: Matrix metalloproteinase 2 induces epithelial-mesenchymal transition in proximal tubules from the luminal side and progresses fibrosis in mineralocorticoid/salt-induced hypertensive rats. J Hypertens. 2011, 29 (12): 2440-2453.CrossRefPubMed
Metadaten
Titel
Ramipril inhibits AGE-RAGE-induced matrix metalloproteinase-2 activation in experimental diabetic nephropathy
verfasst von
Kei Fukami
Sho-ichi Yamagishi
Melinda T Coughlan
Brooke E Harcourt
Phillip Kantharidis
Vicki Thallas-Bonke
Seiya Okuda
Mark E Cooper
Josephine M Forbes
Publikationsdatum
01.12.2014
Verlag
BioMed Central
Erschienen in
Diabetology & Metabolic Syndrome / Ausgabe 1/2014
Elektronische ISSN: 1758-5996
DOI
https://doi.org/10.1186/1758-5996-6-86

Weitere Artikel der Ausgabe 1/2014

Diabetology & Metabolic Syndrome 1/2014 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

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