Skip to main content
Erschienen in: BMC Nephrology 1/2013

Open Access 01.12.2013 | Research article

Dipeptidyl peptidase IV inhibitor attenuates kidney injury in rat remnant kidney

verfasst von: Kwon Wook Joo, Sejoong Kim, Shin-young Ahn, Ho Jun Chin, Dong-Wan Chae, Jeonghwan Lee, Jin Suk Han, Ki Young Na

Erschienen in: BMC Nephrology | Ausgabe 1/2013

Abstract

Background

The inhibition of dipeptidyl peptidase (DPP) IV shows protective effects on tissue injury of the heart, lung, and kidney. Forkhead box O (FoxO) transcriptional factors regulate cellular differentiation, growth, survival, the cell cycle, metabolism, and oxidative stress. The aims of this study were to investigate whether the DPP IV inhibitor sitagliptin could attenuate kidney injury and to evaluate the status of FoxO3a signaling in the rat remnant kidney model.

Methods

Rats were received two-step surgery of 5/6 renal mass reduction and fed on an oral dose of 200 mg/kg/day sitagliptin for 8 weeks. Before and after the administration of sitagliptin, physiologic parameters were measured. After 8 weeks of treatment, the kidneys were harvested.

Results

The sitagliptin treatment attenuated renal dysfunction. A histological evaluation revealed that glomerulosclerosis and tubulointerstitial injury were significantly decreased by sitagliptin. Sitagliptin decreased DPP IV activity and increased the renal expression of glucagon-like peptide-1 receptor (GLP-1R). The subtotal nephrectomy led to the activation of phosphatidylinositol 3-kinase (PI3K)-Akt and FoxO3a phosphorylation, whereas sitagliptin treatment reversed these changes, resulting in PI3K-Akt pathway inactivation and FoxO3a dephosphorylation. The renal expression of catalase was increased and the phosphorylation of c-Jun N-terminal kinase (JNK) was decreased by sitagliptin. Sitagliptin treatment reduced apoptosis by decreasing cleaved caspase-3 and −9 and Bax levels and decreased macrophage infiltration.

Conclusions

In rat remnant kidneys, DPP IV inhibitor attenuated renal dysfunction and structural damage. A reduction of apoptosis, inflammation and an increase of antioxidant could be suggested as a renoprotective mechanism together with the activation of FoxO3a signaling. Therefore, DPP IV inhibitors might provide a promising approach for treating CKD, but their application in clinical practice remains to be investigated.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1471-2369-14-98) contains supplementary material, which is available to authorized users.
Kwon Wook Joo, Sejoong Kim contributed equally to this work.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

KWJ, JSH, KYN: participated in research design. KWJ, SK, JL: conducted experiments. SA, HJC, DWC: performed data analysis. KYN: wrote or contributed to the writing of the manuscript. All authors read and approved the final manuscript.

Background

Glucagon-like peptide-1 (GLP-1) is a gut incretin hormone, whose mimetics have been used as a therapeutic agent for type 2 diabetes. It stimulates pancreatic beta cell proliferation and insulin secretion in a glucose-dependent manner [1]. However, this peptide is almost immediately degraded by dipeptidyl peptidase (DPP) IV in the circulation. DPP IV has a wide variety of substrates that have important roles in cell migration and differentiation, glucose regulation, metabolism, and inflammation [2]. Sitagliptin, a highly selective DPP IV inhibitor, is currently used in the treatment of type 2 diabetes patients to improve glucose tolerance by increasing the half-life of GLP-1 and glucose-dependent insulinotropic peptide (GIP) [3].
The GLP-1 receptor (GLP-1R) agonist exendin-4 has been reported to ameliorate diabetic nephropathy in animals [4, 5]. Recently, studies have shown that DPP IV inhibitors attenuate kidney injury in diabetic animal models [6, 7]. In addition to diabetic nephropathy, DPP IV inhibition protected the kidney against ischemia-reperfusion injury (IRI) [8]. Tissue protective effects of GLP-1 activation or DPP IV inhibition have also been demonstrated in other organs, including IRI of the lung during transplantation [911] and the outcome of myocardial infarction [12, 13].
Most cases of chronic kidney disease (CKD) inevitably progress to end-stage renal disease, which has a high associated morbidity and mortality. Although the initiating insult of CKD is variable, the progression of the disease seems to be common to all kidney diseases that involve a vicious cycle of nephron destruction, glomerulosclerosis and tubulointerstitial fibrosis [14]. However, few pharmacologic treatments have been shown to attenuate the progression of CKD. The remnant kidney is a disease model that mimics the progression of CKD in humans. In this model, there is early glomerulosclerosis by week 4, with segmental sclerosis with tubulointerstitial fibrosis by week 8. Animals die of uremia starting at week 12 to week 16 [15]. Therefore, 8 weeks after subtotal nephrectomy must be a proper time to observe renal pathology in this model.
Forkhead box O (FoxO) transcriptional factors regulate various downstream target genes, including those involved in cellular differentiation, growth, survival, the cell cycle, glucose and lipid metabolism, stress, and the detoxification of reactive oxygen species (ROS) [16]. The phosphatidylinositol 3-kinase (PI3K) and serine–threonine kinase Akt/PKB (Akt) pathway regulates FoxO through phosphorylation. The Akt-mediated phosphorylation of FoxO inhibits the activity of FoxO by promoting its interaction with 14-3-3 proteins and its nuclear exportation, and also by inducing its degradation by the proteasome [17]. In the kidney, the FoxO3 transcript is the most abundant among four subfamily members of FoxO proteins [18]. However, there is no study on the status of FoxOs in the remnant kidney model.
Based on the previous reports of tissue protective effects, we hypothesize that DPP IV inhibition could have a positive effect on this animal model of CKD. Due to the diverse regulatory functions of FoxO, our hypothesis is that its signaling might also be modulated by DPP IV inhibition in this model. Therefore, it would be interesting to investigate FoxO signaling in the kidneys. The present study aimed to evaluate whether sitagliptin could attenuate kidney injury in a rat remnant kidney model. Moreover, we investigated the status of FoxO3a signaling after sitagliptin treatment in this model. To do this, we made CKD animal model by two-step surgery of 5/6 renal mass reduction, and then fed these rats on a 200 mg/kg/day of sitagliptin for 8 weeks.

Methods

Animal experiments

All animal procedures were approved by the Institutional Animal Care and Use Committee of the Medical Science Research Institute, Seoul National University Bundang Hospital (BA 1112-095/080-01). Male Sprague–Dawley rats (Orient Bio Inc., Seongnam, South Korea) weighing approximately 200 g were used. The rats (n = 21) were randomly assigned to three groups: sham-operation (sham), subtotal nephrectomy (Nx), and subtotal nephrectomy + sitagliptin treatment (Nx+STG) groups. After a right subcostal incision, the right kidney was exposed and separated from the adrenal gland under anesthesia with enflurane (Choongwae Pharma Corp., Seoul, South Korea). The lower and upper thirds of the right kidney were resected. After 1 week, the left kidney was removed. The rats of the sham group underwent the same incision and manipulation of the left and the right kidneys without tissue destruction. One week after the second surgical intervention, the rats in the Nx+STG group were fed a gelled diet containing 200 mg/kg/day of sitagliptin (MSD Korea, Seoul, South Korea), and the rats in the sham and Nx group were fed same gelled diet without sitagliptin. After 8 weeks of treatment, the animals were anesthetized with enflurane, blood samples were obtained, and the kidneys were collected. One portion of the right kidney was fixed in 10% phosphate-buffered formalin for morphologic and immunohistochemical analyses. The remainder of the right kidney was snap-frozen in liquid nitrogen and stored at −80°C for protein extraction.

Physiologic measurements

Before and after the administration of a gelled diet with or without sitagliptin, the rats were weighed and placed in metabolic cages, and their urine was collected for 24 h. The urine volume was measured. Serum samples were taken from the tail vein. The blood glucose levels were measured by an Accu-check meter (Roche diagnostics, St Louis, MO, USA). BUN and creatinine levels in the serum and urine were measured using an automatic analyzer (ADVIA 1650, Siemens, Berlin, Germany). Creatinine clearance was calculated and adjusted for body weight.

Determination of DPP IV enzymatic activity

DPP IV enzymatic activity was assayed in serum using DPP IV Activity Assay Kit (Biovision, Milpitas, CA, USA). A 50 μl volume of serum was diluted with 48 μl of DPP IV assay buffer and mixed with 2 μl substrate Gly-Pro-7-Amino-4-Methylcoumarin (AMC) and then incubated at 37°C for 30 min. The release of AMC from the substrate was measured with a fluorescence spectrophotometer at 360 nm of excitation and 460 nm of emission.

Renal histologic and immunohistochemical analyses

Tissue for light microscopy and immunoperoxidase staining was fixed in formalin and embedded in paraffin. Three-micrometer sections were stained with hematoxylin and eosin (H&E). Apoptosis was detected with the enzymatic labeling of DNA strand breaks using terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end-labeling (TUNEL). TUNEL staining was performed with a Cell Death Detection kit (Roche, Mannheim, Germany). To reveal the total nuclei, the same slides were stained with 4’,6’-diamidino-2-phenyindole (DAPI) in phosphate-buffered saline. Indirect immunoperoxidase staining with an anti-ED-1 antibody (Serotec, Oxford, UK) was performed.

Quantification of morphologic data

All analyses were performed in a blinded manner. Segmental and complete glomerular sclerosis was analyzed using a semiquantitative scoring system from 0 to 4 (0, no glomerulosclerosis; 1, <25% of glomerular area affected; 2, 25–50% affected; 3, 50–75% affected; and 4, 75–100% affected). At least 30 glomeruli were evaluated under × 400 magnification, and the results were averaged. The tubulointerstitial injury score was estimated based on the number of tubule dilatations, the distortion of the tubular basement membranes, and atrophy from 0 to 3 [0, none (<5%); 1, mild (5–25%); 2, moderate (25–50%); and 3, severe (>50%)]. More than 10 consecutive fields were examined under × 200 magnification, and the results were averaged. TUNEL (+) apoptotic nuclei were counted in more than 20 consecutive fields under × 200 magnification, and the results were averaged. The mean numbers of infiltrating macrophages (ED-1 positive cells) were calculated by averaging the total numbers of positive cells in more than 20 sequentially-selected, 0.25-mm2 grids at × 200 magnification.

Western blot analysis

Whole kidneys were homogenized in lysis buffer (250 mM sucrose, 10 mM triethanolamine, 1 μg/ml leupeptin, and 0.1 mg/ml PMSF titrated to pH 7.6). The total protein concentration was measured using a bicinchoninic acid protein assay reagent kit (Sigma, St. Louis, MO, USA). The samples were run on SDS-polyacrylamide minigels (Bio-Rad Mini Protean III). The proteins were transferred to nitrocellulose membranes by electroelution. The following proteins were detected using specific antibodies: GLP-1R (Abcam, Cambridge, UK), β-actin (Santa Cruz Biotech, Santa Cruz, CA, USA), PI3K (BD Bioscience, Franklin Lakes, NJ, USA), total Akt, phospho-Ser473 Akt, total FoxO3a, phospho-Ser253 FoxO3a, total c-Jun N-terminal kinase (JNK), phospho-Thr183/Tyr185 JNK, caspase-3 (all from Cell Signaling Technology, Danvers, MA, USA), caspase-9 (BD Bioscience), Bax (Santa Cruz Biotech), and catalase (Abcam). After incubation with peroxidase-conjugated secondary antibodies (Pierce no. 31458, Rockford, IL, USA), bands were visualized using an enhanced chemiluminescence substrate (ECLTM RPN 2106; Amersham Pharmacia Biotech, Buckinghamshire, UK) before exposure to X-ray film (Hyperfilm; Amersham Pharmacia Biotech, Buckinghamshire, UK). The band densities were quantified by densitometry (GS-700 Imaging Densitometry, Bio-Rad, Hercules, CA, USA).

Statistical analysis

All of the data are presented as the means ± S.D. The statistical analyses were performed using SPSS (version 15.0. for Windows; SPSS Inc., Chicago, IL, USA). Difference among three groups were tested with one-way analysis of variance (ANOVA), followed by Turkey’s multiple comparison post-test. Statistical significance was indicated by P < 0.05.

Results

Table 1 shows the initial and final physiologic parameters of the animals. At the end of the study, rats that received a nephrectomy had a reduced body weight than the sham-operated rats did. However, sitagliptin treatment did not affect body weight or blood glucose levels in the animals. The BUN level remained increased after nephrectomy but was not significantly influenced by treatment with sitagliptin. Serum creatinine levels were increased in the nephrectomized rats, but they were significantly decreased by sitagliptin treatment. Sitagliptin also significantly improved creatinine clearance in nephrectomized rats. Figure 1 shows a representative histological image of the kidneys from each group of animals. In the nephrectomized rats that received sitagliptin, the severity of glomerulosclerosis was significantly reduced compared with the nephrectomized rats that were untreated (score: 1.88 ± 0.83 vs. 3.20 ± 0.63, P < 0.05). The degree of tubulointerstitial injury was also significantly reduced by sitagliptin in the nephrectomized rats (score: 1.88 ± 0.64 vs. 2.71 ± 0.49, P < 0.05). From these results, sitagliptin attenuated renal dysfunction and improved histological damage induced by subtotal nephrectomy.
Table 1
Physiologic parameters of experimental animals
 
Sham (n=6)
Nx (n=8)
Nx+STG (n=7)
Body weight (g)
   
 Initial
205 ± 8
204 ± 9
199 ± 10
 Final
563 ± 34
493 ± 29*
475 ± 31*
Blood glucose (mg/dl)
   
 Initial
111 ± 11
100 ± 10
111 ± 6
 Final
180 ± 9
184 ± 21
172 ± 21
BUN (mg/dl)
   
 Initial
8.27 ± 2.48
8.63 ± 1.56
9.94 ± 3.41
 Final
12.17 ± 2.50
28.19 ± 3.56*
25.04 ± 7.12*
Cr (mg/dl)
   
 Initial
0.39 ± 0.08
0.42 ± 0.06
0.39 ± 0.11
 Final
0.52 ± 0.04
0.90 ± 0.09*
0.70 ± 0.19*#
ClCr (ml/min/100 g)
   
 Initial
0.62 ± 0.25
0.52 ± 0.09
0.78 ± 0.33
 Final
0.48 ± 0.05
0.31 ± 0.03*
0.43 ± 0.12#
The results are expressed as the mean ± s.d.
*Significantly different with respect to the sham-operated rats; #significantly different with respect to the nephrectomized rats; *# P < 0.05.
Sham, sham operation; Nx, nephrectomy; Nx+STG, nephrectomy and sitagliptin treatment.
Cr, creatinine; ClCr, creatinine clearance normalized to body weight.
There was no difference in DPP IV activity in the nephrectomized rats compared with the sham-operated rats (Figure 2A). An almost complete DPP IV inhibition was observed in serum of sitagliptin-treated rats with less than 2% residual DPP IV activity. The expression of GLP-1R in the kidney was reduced by half after 8 weeks of subtotal nephrectomy (Figure 2B). However, treatment with sitagliptin restored the expression of GLP-1R to the level of sham-operated rats.
We determined the levels of PI3K and phospho-Akt in the kidneys. The levels of PI3K and phospho-Akt in the nephrectomized rats were significantly increased compared to the sham-operated rats, whereas sitagliptin treatment of nephrectomized rats significantly reduced both of these levels (Figure 3). Because PI3K activation and Akt phosphorylation serve as negative regulators of FoxO transcription factors, we next examined the phosphorylation of FoxO3a. Western blotting showed that the phospho-FoxO3a/total FoxO3a ratios were significantly increased in the nephrectomized rats compared to the sham-operated rats (Figure 4). However, the phospho-FoxO3a/total FoxO3a ratios were significantly decreased by sitagliptin treatment. Therefore, sitagliptin restored the inactivation of FoxO3a induced by subtotal nephrectomy.
To investigate whether the status of FoxO3a phosphorylation affected downstream signaling activity, we examined changes in the antioxidant protein catalase. As shown in Figure 5A, the expression of catalase was significantly increased by sitagliptin treatment. Because JNK is activated by oxidative stress, we next examined JNK phosphorylation. However, there was no difference of the phospho-JNK/total JNK ratios in the nephrectomized rats compared with the sham-operated rats (Figure 5B). The phospho-JNK/total JNK ratios were significantly decreased by sitagliptin treatment. From these results, the antioxidant effect of catalase decreased the activity of JNK in the nephrectomized rats after sitagliptin treatment.
To investigate the extent of apoptosis, we examined kidney sections after detecting DNA fragmentation with an in situ TUNEL assay. Scattered and bright nuclei stained by the TUNEL assay were easily detected in the kidneys of nephrectomized rats, but the number of nuclei was significantly decreased in the kidneys of the sitagliptin-treated rats (Figure 6A). Next, we examined changes in the proapoptotic proteins caspase-3, caspase-9, and Bax by western blot analysis. The cleaved subtypes of both caspase-3 and caspase-9, and Bax were increased in the kidneys of nephrectomized rats (Figure 6B). However, treatment with sitagliptin significantly reduced the levels of Bax and cleaved subtypes of both caspase-3 and caspase-9 in the nephrectomized rats. These results indicate that sitagliptin reduces the extent of apoptosis in the kidneys of nephrectomized rats.
Subtotal nephrectomy was associated with macrophage infiltration in the tubulointerstitium, as determined by an increase in ED-1-positive cells (Figure 7). After counting the absolute number of ED-1-positive cells, we observed a marked increase in macrophage infiltration after nephrectomy and a significant reduction in response to sitagliptin treatment. The mean ED-1 score was 94.29 ± 48.51 in nephrectomized rats and 34.33 ± 14.12 in sitagliptin-treated nephrectomized rats.

Discussion

This study demonstrated that sitagliptin treatment after renal mass reduction showed a renoprotective effect. To the best of our knowledge, this report is the first one to show the effects of sitagliptin, a DPP IV inhibitor, on renal damage in the remnant kidney model. Sitagliptin ameliorated renal dysfunction and attenuated glomerular and tubulointerstitial injury in this model. Treatment with sitagliptin was found to exert anti-oxidative, anti-apoptotic, and anti-inflammatory effects in this model, together with the inactivation of the PI3K-Akt pathway and the resulting activation of FoxO3a.
In this study, sitagliptin, anti-diabetic drug, did not reduce blood glucose levels in the nephrectomized rats. Therefore, the renoprotective effect of sitagliptin is irrelevant to the reduction of glycemia. DPP IV inhibition does not cause hypoglycemia in a study in healthy male volunteers [19]. Because the action of GLP-1 on insulin secretion is strictly glucose dependent, the risk of hypoglycemia associated with DPP IV inhibitors is low [20].
In the kidney, GLP-1R is strongly expressed in both the glomeruli and proximal tubules [4, 21]. However, it has been reported that its expression is reduced in diabetic kidneys [4, 7]. In the heart, the GLP-1R mRNA expression was significantly reduced after subtotal nephrectomy [22]. We first found that GLP-1R expression was markedly decreased in the kidney after subtotal nephrectomy. Judging from the complete inhibition of DPP IV activity in sitagliptin-treated rats, we are sure that sitagliptin significantly raised plasma GLP-1 levels. It has been reported that GLP-1 agonist acts a renoprotective role through increasing GLP-1R expression in diabetic kidneys [4]. Therefore, chronic sitagliptin treatment in this study might activate renal GLP-1R through DPP IV inhibition because the protein abundance of GLP-1R was significantly increased in kidney homogenates. The dose of sitagliptin used in this study was far above the dose of anti-diabetic usage. To prove the tissue protective effects of DPP IV inhibition, we determined the dose from previous studies [13, 23].
Currently, various target genes of FoxOs have been identified in insulin-responsive tissues [24, 25]. Therefore, the connection between GLP-1 and FoxO has only been studied in pancreatic beta cells [26]. There are a few studies that have investigated FoxO signaling in the kidney [27, 28]. Our study is the first to examine the association between GLP-1 and FoxO signaling in rat remnant kidneys. We only investigated the status of FoxO3 in this study because it is the most abundant protein among FoxO subfamily members. The activation of PI3K, the phosphorylation of Akt, and the inactivation of FoxO3a were the main pathway in this disease model. Sitagliptin treatment reversed this pathway.
Although the precise mechanism remains to be elucidated, CKD has been to known to be associated with oxidative stress [29, 30]. Oxidative stress can occur either as a result of an increased ROS generation, a depressed antioxidant system or both. Catalase is a peroxidase enzyme that is of the major antioxidant defense systems [31]. However, catalase expression and JNK phosphorylation were not changed in this study. Future studies are needed to address these issues. GLP-1R activation using a GLP-1 analog or DPP IV inhibitor reduced oxidative stress in diabetic nephropathy and renal IRI [4, 5, 7, 8]. The specific mechanism underlying the anti-oxidative effect of GLP-1R activation remains unclear. In this study, we speculate that the underlying mechanism might be the up-regulation of antioxidant catalase by FoxO3a activation through sitagliptin treatment.
An anti-apoptotic effect mediated by GLP-1R has been suggested in various tissues, including pancreatic beta cells [32], neurons [33], and cardiomyocytes [12]. GLP-1R activation also inhibited apoptosis in diabetic retinopathy [34] and diabetic nephropathy [4, 7]. The underlying anti-apoptotic mechanism of GLP-1R has been reported in many in vitro studies [3540]. GLP-1 is capable of inducing downregulation of the pro-apoptotic protein Bax, upregulation of the anti-apoptotic protein Bcl-2, phosphorylation and inactivation of Bad, reducing caspase-3 activity and DNA fragmentation.
Inflammatory cell infiltration induced by subtotal nephrectomy was attenuated by sitagliptin treatment in this study. A GLP-1R agonist showed anti-inflammatory effects in diabetic nephropathy [4, 5]. In kidney IRI, GLP-1R activation using a DPP IV inhibitor ameliorated inflammation [8]. The anti-inflammatory effect of GLP-1R activation was also reported in the animal model of atherosclerosis [41]. Therefore, we speculate that GLP-1R activation by sitagliptin in a CKD animal model showed similar results.
Our study has some limitations. First, we performed the experiments with only three groups of animals without a group of animals with sham-operation and sitagliptin treatment. Due to treatment with a high dose of sitagliptin, we should have included this experimental group to observe any adverse effects in the animals. However, higher doses of sitagliptin than those used in our experiment have been proven to be safe in previous studies [12, 18]. Moreover, our experiment showed no significant effects on body weight gain or the changes in blood glucose levels in the animals. Second, there is insufficient evidence that the beneficial effect of sitagliptin is through the activation of GLP-1R. DPP IV acts on a wide range of substrates. There is a possibility that other target molecules of DPP IV except GLP-1 may exert the renoprotective effects because plasma GLP-1 levels were not measured in this study. Knockout experiments inhibiting GLP-1 or GLP-1R would be required in the future. Third, there is no direct evidence to determine the causal relationship between GLP-1R and FoxO3a signaling. In vitro experiments using renal cells would also be required to study the direct effects of the GLP-1R on the signaling proteins (PI3K, Akt, JNK, and FoxO3a).

Conclusions

In summary, sitagliptin treatment attenuated renal dysfunction and structural damage in a model of renal mass reduction. A reduction of apoptosis, inflammation and an increase of antioxidant could be suggested as a renoprotective mechanism, together with the activation of FoxO3a signaling. Therefore, DPP IV inhibitors might provide a promising approach for treating CKD, but their application in clinical practice remains to be investigated.

Acknowledgements

The authors thank Yun-Mi Lee and Jin Young Yang for their technical assistance. This study was supported by the Seoul National University Bundang Hospital [Grant 06-2012-091 and 02-2011-013].
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

KWJ, JSH, KYN: participated in research design. KWJ, SK, JL: conducted experiments. SA, HJC, DWC: performed data analysis. KYN: wrote or contributed to the writing of the manuscript. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Baggio LL, Drucker DJ: Biology of incretins: GLP-1 and GIP. Gastroenterology. 2007, 132: 2131-2157. 10.1053/j.gastro.2007.03.054.CrossRefPubMed Baggio LL, Drucker DJ: Biology of incretins: GLP-1 and GIP. Gastroenterology. 2007, 132: 2131-2157. 10.1053/j.gastro.2007.03.054.CrossRefPubMed
2.
Zurück zum Zitat Mentlein R: Dipeptidyl-peptidase IV (CD26)–role in the inactivation of regulatory peptides. Regul Pept. 1999, 85: 9-24. 10.1016/S0167-0115(99)00089-0.CrossRefPubMed Mentlein R: Dipeptidyl-peptidase IV (CD26)–role in the inactivation of regulatory peptides. Regul Pept. 1999, 85: 9-24. 10.1016/S0167-0115(99)00089-0.CrossRefPubMed
3.
Zurück zum Zitat Drucker DJ, Nauck MA: The incretin system: glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet. 2006, 368: 1696-1705. 10.1016/S0140-6736(06)69705-5.CrossRefPubMed Drucker DJ, Nauck MA: The incretin system: glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet. 2006, 368: 1696-1705. 10.1016/S0140-6736(06)69705-5.CrossRefPubMed
4.
Zurück zum Zitat Park CW, Kim HW, Ko SH, Lim JH, Ryu GR, Chung HW, Han SW, Shin SJ, Bang BK, Breyer MD, Chang YS: Long-term treatment of glucagon-like peptide-1 analog exendin-4 ameliorates diabetic nephropathy through improving metabolic anomalies in db/db mice. J Am Soc Nephrol. 2007, 18: 1227-1238. 10.1681/ASN.2006070778.CrossRefPubMed Park CW, Kim HW, Ko SH, Lim JH, Ryu GR, Chung HW, Han SW, Shin SJ, Bang BK, Breyer MD, Chang YS: Long-term treatment of glucagon-like peptide-1 analog exendin-4 ameliorates diabetic nephropathy through improving metabolic anomalies in db/db mice. J Am Soc Nephrol. 2007, 18: 1227-1238. 10.1681/ASN.2006070778.CrossRefPubMed
5.
Zurück zum Zitat Kodera R, Shikata K, Kataoka HU, Takatsuka T, Miyamoto S, Sasaki M, Kajitani N, Nishishita S, Sarai K, Hirota D, Sato C, Ogawa D, Makino H: Glucagon-like peptide-1 receptor agonist ameliorates renal injury through its anti-inflammatory action without lowering blood glucose level in a rat model of type 1 diabetes. Diabetologia. 2011, 54: 965-978. 10.1007/s00125-010-2028-x.CrossRefPubMed Kodera R, Shikata K, Kataoka HU, Takatsuka T, Miyamoto S, Sasaki M, Kajitani N, Nishishita S, Sarai K, Hirota D, Sato C, Ogawa D, Makino H: Glucagon-like peptide-1 receptor agonist ameliorates renal injury through its anti-inflammatory action without lowering blood glucose level in a rat model of type 1 diabetes. Diabetologia. 2011, 54: 965-978. 10.1007/s00125-010-2028-x.CrossRefPubMed
6.
Zurück zum Zitat Mega C, de Lemos ET, Vala H, Fernandes R, Oliveira J, Mascarenhas-Melo F, Teixeira F, Reis F: Diabetic nephropathy amelioration by a low-dose sitagliptin in an animal model of type 2 diabetes (Zucker diabetic fatty rat). Exp Diabetes Res. 2011, 10.1155/2011/162092. Mega C, de Lemos ET, Vala H, Fernandes R, Oliveira J, Mascarenhas-Melo F, Teixeira F, Reis F: Diabetic nephropathy amelioration by a low-dose sitagliptin in an animal model of type 2 diabetes (Zucker diabetic fatty rat). Exp Diabetes Res. 2011, 10.1155/2011/162092.
7.
Zurück zum Zitat Liu WJ, Xie SH, Liu YN, Kim W, Jin HY, Park SK, Shao YM, Park TS: Dipeptidyl peptidase IV inhibitor attenuates kidney injury in streptozotocin-induced diabetic rats. J Pharmacol Exp Ther. 2012, 340: 248-255. 10.1124/jpet.111.186866.CrossRefPubMed Liu WJ, Xie SH, Liu YN, Kim W, Jin HY, Park SK, Shao YM, Park TS: Dipeptidyl peptidase IV inhibitor attenuates kidney injury in streptozotocin-induced diabetic rats. J Pharmacol Exp Ther. 2012, 340: 248-255. 10.1124/jpet.111.186866.CrossRefPubMed
8.
Zurück zum Zitat Glorie LL, Verhulst A, Matheeussen V, Baerts L, Magielse J, Hermans N, D'Haese PC, De Meester I, De Beuf A: DPP4 inhibition improves functional outcome after renal ischemia reperfusion injury. Am J Physiol Renal Physiol. 2012, 303: F681-F688. 10.1152/ajprenal.00075.2012.CrossRefPubMed Glorie LL, Verhulst A, Matheeussen V, Baerts L, Magielse J, Hermans N, D'Haese PC, De Meester I, De Beuf A: DPP4 inhibition improves functional outcome after renal ischemia reperfusion injury. Am J Physiol Renal Physiol. 2012, 303: F681-F688. 10.1152/ajprenal.00075.2012.CrossRefPubMed
9.
Zurück zum Zitat Lim SB, Rubinstein I, Sadikot RT, Artwohl JE, Önyüksel H: A novel peptide nanomedicine against acute lung injury: GLP-1 in phospholipid micelles. Pharm Res. 2011, 28: 662-672. 10.1007/s11095-010-0322-4.CrossRefPubMed Lim SB, Rubinstein I, Sadikot RT, Artwohl JE, Önyüksel H: A novel peptide nanomedicine against acute lung injury: GLP-1 in phospholipid micelles. Pharm Res. 2011, 28: 662-672. 10.1007/s11095-010-0322-4.CrossRefPubMed
10.
Zurück zum Zitat Jungraithmayr W, De Meester I, Matheeussen V, Baerts L, Arni S, Weder W: CD26/DPP-4 inhibition recruits regenerative stem cells via stromal cell-derived factor-1 and beneficially influences ischaemia-reperfusion injury in mouse lung transplantation. Eur J Cardiothorac Surg. 2012, 41: 1166-1173. 10.1093/ejcts/ezr180.CrossRefPubMed Jungraithmayr W, De Meester I, Matheeussen V, Baerts L, Arni S, Weder W: CD26/DPP-4 inhibition recruits regenerative stem cells via stromal cell-derived factor-1 and beneficially influences ischaemia-reperfusion injury in mouse lung transplantation. Eur J Cardiothorac Surg. 2012, 41: 1166-1173. 10.1093/ejcts/ezr180.CrossRefPubMed
11.
Zurück zum Zitat Zhai W, Jungraithmayr W, De Meester I, Inci I, Augustyns K, Arni S, Hillinger S, Scharpé S, Weder W, Korom S: Primary graft dysfunction in lung transplantation: the role of CD26/dipeptidylpeptidase IV and vasoactive intestinal peptide. Transplantation. 2009, 87: 1140-1146. 10.1097/TP.0b013e31819e04c3.CrossRefPubMed Zhai W, Jungraithmayr W, De Meester I, Inci I, Augustyns K, Arni S, Hillinger S, Scharpé S, Weder W, Korom S: Primary graft dysfunction in lung transplantation: the role of CD26/dipeptidylpeptidase IV and vasoactive intestinal peptide. Transplantation. 2009, 87: 1140-1146. 10.1097/TP.0b013e31819e04c3.CrossRefPubMed
12.
Zurück zum Zitat Noyan-Ashraf MH, Momen MA, Ban K, Sadi AM, Zhou YQ, Riazi AM, Baggio LL, Henkelman RM, Husain M, Drucker DJ: GLP-1R agonist liraglutide activates cytoprotective pathways and improves outcomes after experimental myocardial infarction in mice. Diabetes. 2009, 58: 975-983. 10.2337/db08-1193.CrossRefPubMedPubMedCentral Noyan-Ashraf MH, Momen MA, Ban K, Sadi AM, Zhou YQ, Riazi AM, Baggio LL, Henkelman RM, Husain M, Drucker DJ: GLP-1R agonist liraglutide activates cytoprotective pathways and improves outcomes after experimental myocardial infarction in mice. Diabetes. 2009, 58: 975-983. 10.2337/db08-1193.CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Sauvé M, Ban K, Momen MA, Zhou YQ, Henkelman RM, Husain M, Drucker DJ: Genetic deletion or pharmacological inhibition of dipeptidyl peptidase-4 improves cardiovascular outcomes after myocardial infarction in mice. Diabetes. 2010, 59: 1063-1073. 10.2337/db09-0955.CrossRefPubMedPubMedCentral Sauvé M, Ban K, Momen MA, Zhou YQ, Henkelman RM, Husain M, Drucker DJ: Genetic deletion or pharmacological inhibition of dipeptidyl peptidase-4 improves cardiovascular outcomes after myocardial infarction in mice. Diabetes. 2010, 59: 1063-1073. 10.2337/db09-0955.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Remuzzi G, Bertani T: Pathophysiology of progressive nephropathies. N Engl J Med. 1998, 339: 1448-1456. 10.1056/NEJM199811123392007.CrossRefPubMed Remuzzi G, Bertani T: Pathophysiology of progressive nephropathies. N Engl J Med. 1998, 339: 1448-1456. 10.1056/NEJM199811123392007.CrossRefPubMed
15.
Zurück zum Zitat Fogo AB: Animal models of FSGS: lessons for pathogenesis and treatment. Semin Nephrol. 2003, 23: 161-171. 10.1053/snep.2003.50015.CrossRefPubMed Fogo AB: Animal models of FSGS: lessons for pathogenesis and treatment. Semin Nephrol. 2003, 23: 161-171. 10.1053/snep.2003.50015.CrossRefPubMed
16.
Zurück zum Zitat Zhang X, Tang N, Hadden TJ, Rishi AK: Akt, FoxO and regulation of apoptosis. Biochim Biophys Acta. 1813, 2011: 1978-1986. Zhang X, Tang N, Hadden TJ, Rishi AK: Akt, FoxO and regulation of apoptosis. Biochim Biophys Acta. 1813, 2011: 1978-1986.
17.
Zurück zum Zitat Tzivion G, Dobson M, Ramakrishnan G: FoxO transcription factors; Regulation by AKT and 14-3-3 proteins. Biochim Biophys Acta. 1813, 2011: 1938-1945. Tzivion G, Dobson M, Ramakrishnan G: FoxO transcription factors; Regulation by AKT and 14-3-3 proteins. Biochim Biophys Acta. 1813, 2011: 1938-1945.
18.
Zurück zum Zitat Jacobs FM, van der Heide LP, Wijchers PJ: FoxO6, a novel member of the FoxO class of transcription factors with distinct shuttling dynamics. J Biol Chem. 2003, 278: 35959-35967. 10.1074/jbc.M302804200.CrossRefPubMed Jacobs FM, van der Heide LP, Wijchers PJ: FoxO6, a novel member of the FoxO class of transcription factors with distinct shuttling dynamics. J Biol Chem. 2003, 278: 35959-35967. 10.1074/jbc.M302804200.CrossRefPubMed
19.
Zurück zum Zitat Bergman AJ, Stevens C, Zhou Y, Yi B, Laethem M, De Smet M, Snyder K, Hilliard D, Tanaka W, Zeng W, Tanen M, Wang AQ, Chen L, Winchell G, Davies MJ, Ramael S, Wagner JA, Herman GA: Pharmacokinetic and pharmacodynamic properties of multiple oral doses of sitagliptin, a dipeptidyl peptidase-IV inhibitor: a double-blind, randomized, placebo-controlled study in healthy male volunteers. Clin Ther. 2006, 28: 55-72. 10.1016/j.clinthera.2006.01.015.CrossRefPubMed Bergman AJ, Stevens C, Zhou Y, Yi B, Laethem M, De Smet M, Snyder K, Hilliard D, Tanaka W, Zeng W, Tanen M, Wang AQ, Chen L, Winchell G, Davies MJ, Ramael S, Wagner JA, Herman GA: Pharmacokinetic and pharmacodynamic properties of multiple oral doses of sitagliptin, a dipeptidyl peptidase-IV inhibitor: a double-blind, randomized, placebo-controlled study in healthy male volunteers. Clin Ther. 2006, 28: 55-72. 10.1016/j.clinthera.2006.01.015.CrossRefPubMed
20.
Zurück zum Zitat Weir GC, Mojsov S, Hendrick GK, Habener JF: Glucagonlike peptide I (7–37) actions on endocrine pancreas. Diabetes. 1989, 38: 338-342. 10.2337/diabetes.38.3.338.CrossRefPubMed Weir GC, Mojsov S, Hendrick GK, Habener JF: Glucagonlike peptide I (7–37) actions on endocrine pancreas. Diabetes. 1989, 38: 338-342. 10.2337/diabetes.38.3.338.CrossRefPubMed
21.
Zurück zum Zitat Schlatter P, Beglinger C, Drewe J, Gutmann H: Glucagon-like peptide 1 receptor expression in primary porcine proximal tubular cells. Regul Pept. 2007, 141: 120-128. 10.1016/j.regpep.2006.12.016.CrossRefPubMed Schlatter P, Beglinger C, Drewe J, Gutmann H: Glucagon-like peptide 1 receptor expression in primary porcine proximal tubular cells. Regul Pept. 2007, 141: 120-128. 10.1016/j.regpep.2006.12.016.CrossRefPubMed
22.
Zurück zum Zitat Chaykovska L, von Websky K, Rahnenführer J, Alter M, Heiden S, Fuchs H, Runge F, Klein T, Hocher B: Effects of DPP-4 inhibitors on the heart in a rat model of uremic cardiomyopathy. PLoS One. 2011, 6: e27861-10.1371/journal.pone.0027861.CrossRefPubMedPubMedCentral Chaykovska L, von Websky K, Rahnenführer J, Alter M, Heiden S, Fuchs H, Runge F, Klein T, Hocher B: Effects of DPP-4 inhibitors on the heart in a rat model of uremic cardiomyopathy. PLoS One. 2011, 6: e27861-10.1371/journal.pone.0027861.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Lim S, Choi SH, Shin H, Cho BJ, Park HS, Ahn BY, Kang SM, Yoon JW, Jang HC, Kim YB, Park KS: Effect of a dipeptidyl peptidase-IV inhibitor, des-fluoro-sitagliptin, on neointimal formation after balloon injury in rats. PLoS One. 2012, 7: e35007-10.1371/journal.pone.0035007.CrossRefPubMedPubMedCentral Lim S, Choi SH, Shin H, Cho BJ, Park HS, Ahn BY, Kang SM, Yoon JW, Jang HC, Kim YB, Park KS: Effect of a dipeptidyl peptidase-IV inhibitor, des-fluoro-sitagliptin, on neointimal formation after balloon injury in rats. PLoS One. 2012, 7: e35007-10.1371/journal.pone.0035007.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Kim JJ, Li P, Huntley J, Chang JP, Arden KC, Olefsky JM: FoxO1 haploinsufficiency protects against high-fat diet-induced insulin resistance with enhanced peroxisome proliferator-activated receptor gamma activation in adipose tissue. Diabetes. 2009, 58: 1275-1282. 10.2337/db08-1001.CrossRefPubMedPubMedCentral Kim JJ, Li P, Huntley J, Chang JP, Arden KC, Olefsky JM: FoxO1 haploinsufficiency protects against high-fat diet-induced insulin resistance with enhanced peroxisome proliferator-activated receptor gamma activation in adipose tissue. Diabetes. 2009, 58: 1275-1282. 10.2337/db08-1001.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Fang CX, Dong F, Thomas DP, Ma H, He L, Ren J: Hypertrophic cardiomyopathy in high-fat diet-induced obesity: role of suppression of forkhead transcription factor and atrophy gene transcription. Am J Physiol Heart Circ Physiol. 2008, 295: H1206-H1215. 10.1152/ajpheart.00319.2008.CrossRefPubMedPubMedCentral Fang CX, Dong F, Thomas DP, Ma H, He L, Ren J: Hypertrophic cardiomyopathy in high-fat diet-induced obesity: role of suppression of forkhead transcription factor and atrophy gene transcription. Am J Physiol Heart Circ Physiol. 2008, 295: H1206-H1215. 10.1152/ajpheart.00319.2008.CrossRefPubMedPubMedCentral
26.
27.
Zurück zum Zitat Percy CJ, Brown L, Power DA, Johnson DW, Gobe GC: Obesity and hypertension have differing oxidant handling molecular pathways in age-related chronic kidney disease. Mech Ageing Dev. 2009, 130: 129-138. 10.1016/j.mad.2008.10.003.CrossRefPubMed Percy CJ, Brown L, Power DA, Johnson DW, Gobe GC: Obesity and hypertension have differing oxidant handling molecular pathways in age-related chronic kidney disease. Mech Ageing Dev. 2009, 130: 129-138. 10.1016/j.mad.2008.10.003.CrossRefPubMed
28.
Zurück zum Zitat Chung HW, Lim JH, Kim MY, Shin SJ, Chung S, Choi BS, Kim HW, Kim YS, Park CW, Chang YS: High-fat diet-induced renal cell apoptosis and oxidative stress in spontaneously hypertensive rat are ameliorated by fenofibrate through the PPARα-FoxO3a-PGC-1α pathway. Nephrol Dial Transplant. 2012, 27: 2213-2225. 10.1093/ndt/gfr613.CrossRefPubMed Chung HW, Lim JH, Kim MY, Shin SJ, Chung S, Choi BS, Kim HW, Kim YS, Park CW, Chang YS: High-fat diet-induced renal cell apoptosis and oxidative stress in spontaneously hypertensive rat are ameliorated by fenofibrate through the PPARα-FoxO3a-PGC-1α pathway. Nephrol Dial Transplant. 2012, 27: 2213-2225. 10.1093/ndt/gfr613.CrossRefPubMed
29.
Zurück zum Zitat Vaziri ND, Oveisi F, Ding Y: Role of increased oxygen free radical activity in the pathogenesis of uremic hypertension. Kidney Int. 1998, 53: 1748-1754. 10.1046/j.1523-1755.1998.00947.x.CrossRefPubMed Vaziri ND, Oveisi F, Ding Y: Role of increased oxygen free radical activity in the pathogenesis of uremic hypertension. Kidney Int. 1998, 53: 1748-1754. 10.1046/j.1523-1755.1998.00947.x.CrossRefPubMed
30.
Zurück zum Zitat Miyata T, Kurokawa K, Van Ypersele De Strihou C: Advanced glycation and lipoxidation end products: role of reactive carbonyl compounds generated during carbohydrate and lipid metabolism. J Am Soc Nephrol. 2000, 11: 1744-1752.PubMed Miyata T, Kurokawa K, Van Ypersele De Strihou C: Advanced glycation and lipoxidation end products: role of reactive carbonyl compounds generated during carbohydrate and lipid metabolism. J Am Soc Nephrol. 2000, 11: 1744-1752.PubMed
31.
Zurück zum Zitat Fridovich I: Superoxide radical and superoxide dismutases. Annu Rev Biochem. 1995, 64: 97-112. 10.1146/annurev.bi.64.070195.000525.CrossRefPubMed Fridovich I: Superoxide radical and superoxide dismutases. Annu Rev Biochem. 1995, 64: 97-112. 10.1146/annurev.bi.64.070195.000525.CrossRefPubMed
32.
Zurück zum Zitat Li Y, Hansotia T, Yusta B, Ris F, Halban PA, Drucker DJ: Glucagon-like peptide-1 receptor signaling modulates beta cell apoptosis. J Biol Chem. 2003, 278: 471-478.CrossRefPubMed Li Y, Hansotia T, Yusta B, Ris F, Halban PA, Drucker DJ: Glucagon-like peptide-1 receptor signaling modulates beta cell apoptosis. J Biol Chem. 2003, 278: 471-478.CrossRefPubMed
33.
Zurück zum Zitat During MJ, Cao L, Zuzga DS, Francis JS, Fitzsimons HL, Jiao X, Bland RJ, Klugmann M, Banks WA, Drucker DJ, Haile CN: Glucagon-like peptide-1 receptor is involved in learning and neuroprotection. Nat Med. 2003, 9: 1173-1179. 10.1038/nm919.CrossRefPubMed During MJ, Cao L, Zuzga DS, Francis JS, Fitzsimons HL, Jiao X, Bland RJ, Klugmann M, Banks WA, Drucker DJ, Haile CN: Glucagon-like peptide-1 receptor is involved in learning and neuroprotection. Nat Med. 2003, 9: 1173-1179. 10.1038/nm919.CrossRefPubMed
34.
Zurück zum Zitat Zhang Y, Wang Q, Zhang J, Lei X, Xu GT, Ye W: Protection of exendin-4 analogue in early experimental diabetic retinopathy. Graefes Arch Clin Exp Ophthalmol. 2009, 247: 699-706. 10.1007/s00417-008-1004-3.CrossRefPubMed Zhang Y, Wang Q, Zhang J, Lei X, Xu GT, Ye W: Protection of exendin-4 analogue in early experimental diabetic retinopathy. Graefes Arch Clin Exp Ophthalmol. 2009, 247: 699-706. 10.1007/s00417-008-1004-3.CrossRefPubMed
35.
Zurück zum Zitat Marzioni M, Alpini G, Saccomanno S, Candelaresi C, Venter J, Rychlicki C, Fava G, Francis H, Trozzi L, Benedetti A: Exendin-4, a glucagon-like peptide 1 receptor agonist, protects cholangiocytes from apoptosis. Gut. 2009, 58: 990-997. 10.1136/gut.2008.150870.CrossRefPubMed Marzioni M, Alpini G, Saccomanno S, Candelaresi C, Venter J, Rychlicki C, Fava G, Francis H, Trozzi L, Benedetti A: Exendin-4, a glucagon-like peptide 1 receptor agonist, protects cholangiocytes from apoptosis. Gut. 2009, 58: 990-997. 10.1136/gut.2008.150870.CrossRefPubMed
36.
Zurück zum Zitat Qin Z, Sun Z, Huang J, Hu Y, Wu Z, Mei B: Mutated recombinant human glucagon-like peptide-1 protects SH-SY5Y cells from apoptosis induced by amyloid-beta peptide (1–42). Neurosci Lett. 2008, 444: 217-221. 10.1016/j.neulet.2008.08.047.CrossRefPubMed Qin Z, Sun Z, Huang J, Hu Y, Wu Z, Mei B: Mutated recombinant human glucagon-like peptide-1 protects SH-SY5Y cells from apoptosis induced by amyloid-beta peptide (1–42). Neurosci Lett. 2008, 444: 217-221. 10.1016/j.neulet.2008.08.047.CrossRefPubMed
37.
Zurück zum Zitat Liu JH, Yin F, Guo LX, Deng XH, Hu YH: Neuroprotection of geniposide against hydrogen peroxide induced PC12 cells injury: involvement of PI3 kinase signal pathway. Acta Pharmacol Sin. 2009, 30: 159-165. 10.1038/aps.2008.25.CrossRefPubMedPubMedCentral Liu JH, Yin F, Guo LX, Deng XH, Hu YH: Neuroprotection of geniposide against hydrogen peroxide induced PC12 cells injury: involvement of PI3 kinase signal pathway. Acta Pharmacol Sin. 2009, 30: 159-165. 10.1038/aps.2008.25.CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat Kimura R, Okouchi M, Fujioka H, Ichiyanagi A, Ryuge F, Mizuno T, Imaeda K, Okayama N, Kamiya Y, Asai K, Joh T: Glucagon-like peptide-1 (GLP-1) protects against methylglyoxal-induced PC12 cell apoptosis through the PI3K/Akt/mTOR/GCLc/redox signaling pathway. Neuroscience. 2009, 162: 1212-1219. 10.1016/j.neuroscience.2009.05.025.CrossRefPubMed Kimura R, Okouchi M, Fujioka H, Ichiyanagi A, Ryuge F, Mizuno T, Imaeda K, Okayama N, Kamiya Y, Asai K, Joh T: Glucagon-like peptide-1 (GLP-1) protects against methylglyoxal-induced PC12 cell apoptosis through the PI3K/Akt/mTOR/GCLc/redox signaling pathway. Neuroscience. 2009, 162: 1212-1219. 10.1016/j.neuroscience.2009.05.025.CrossRefPubMed
39.
Zurück zum Zitat Quoyer J, Longuet C, Broca C, Linck N, Costes S, Varin E, Bockaert J, Bertrand G, Dalle S: GLP-1 mediates antiapoptotic effect by phosphorylating Bad through a beta-arrestin 1-mediated ERK1/2 activation in pancreatic beta-cells. J Biol Chem. 2010, 285: 1989-2002. 10.1074/jbc.M109.067207.CrossRefPubMed Quoyer J, Longuet C, Broca C, Linck N, Costes S, Varin E, Bockaert J, Bertrand G, Dalle S: GLP-1 mediates antiapoptotic effect by phosphorylating Bad through a beta-arrestin 1-mediated ERK1/2 activation in pancreatic beta-cells. J Biol Chem. 2010, 285: 1989-2002. 10.1074/jbc.M109.067207.CrossRefPubMed
40.
Zurück zum Zitat Ravassa S, Zudaire A, Carr RD, Díez J: Antiapoptotic effects of GLP-1 in murine HL-1 cardiomyocytes. Am J Physiol Heart Circ Physiol. 2011, 300: H1361-H1372. 10.1152/ajpheart.00885.2010.CrossRefPubMed Ravassa S, Zudaire A, Carr RD, Díez J: Antiapoptotic effects of GLP-1 in murine HL-1 cardiomyocytes. Am J Physiol Heart Circ Physiol. 2011, 300: H1361-H1372. 10.1152/ajpheart.00885.2010.CrossRefPubMed
41.
Zurück zum Zitat Arakawa M, Mita T, Azuma K, Ebato C, Goto H, Nomiyama T, Fujitani Y, Hirose T, Kawamori R, Watada H: Inhibition of monocyte adhesion to endothelial cells and attenuation of atherosclerotic lesion by a glucagon-like peptide-1 receptor agonist, exendin-4. Diabetes. 2010, 59: 1030-1037. 10.2337/db09-1694.CrossRefPubMedPubMedCentral Arakawa M, Mita T, Azuma K, Ebato C, Goto H, Nomiyama T, Fujitani Y, Hirose T, Kawamori R, Watada H: Inhibition of monocyte adhesion to endothelial cells and attenuation of atherosclerotic lesion by a glucagon-like peptide-1 receptor agonist, exendin-4. Diabetes. 2010, 59: 1030-1037. 10.2337/db09-1694.CrossRefPubMedPubMedCentral
Metadaten
Titel
Dipeptidyl peptidase IV inhibitor attenuates kidney injury in rat remnant kidney
verfasst von
Kwon Wook Joo
Sejoong Kim
Shin-young Ahn
Ho Jun Chin
Dong-Wan Chae
Jeonghwan Lee
Jin Suk Han
Ki Young Na
Publikationsdatum
01.12.2013
Verlag
BioMed Central
Erschienen in
BMC Nephrology / Ausgabe 1/2013
Elektronische ISSN: 1471-2369
DOI
https://doi.org/10.1186/1471-2369-14-98

Weitere Artikel der Ausgabe 1/2013

BMC Nephrology 1/2013 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Mehr Lebenszeit mit Abemaciclib bei fortgeschrittenem Brustkrebs?

24.05.2024 Mammakarzinom Nachrichten

In der MONARCHE-3-Studie lebten Frauen mit fortgeschrittenem Hormonrezeptor-positivem, HER2-negativem Brustkrebs länger, wenn sie zusätzlich zu einem nicht steroidalen Aromatasehemmer mit Abemaciclib behandelt wurden; allerdings verfehlte der numerische Zugewinn die statistische Signifikanz.

ADT zur Radiatio nach Prostatektomie: Wenn, dann wohl länger

24.05.2024 Prostatakarzinom Nachrichten

Welchen Nutzen es trägt, wenn die Strahlentherapie nach radikaler Prostatektomie um eine Androgendeprivation ergänzt wird, hat die RADICALS-HD-Studie untersucht. Nun liegen die Ergebnisse vor. Sie sprechen für länger dauernden Hormonentzug.

„Überwältigende“ Evidenz für Tripeltherapie beim metastasierten Prostata-Ca.

22.05.2024 Prostatakarzinom Nachrichten

Patienten mit metastasiertem hormonsensitivem Prostatakarzinom sollten nicht mehr mit einer alleinigen Androgendeprivationstherapie (ADT) behandelt werden, mahnt ein US-Team nach Sichtung der aktuellen Datenlage. Mit einer Tripeltherapie haben die Betroffenen offenbar die besten Überlebenschancen.

So sicher sind Tattoos: Neue Daten zur Risikobewertung

22.05.2024 Melanom Nachrichten

Das größte medizinische Problem bei Tattoos bleiben allergische Reaktionen. Melanome werden dadurch offensichtlich nicht gefördert, die Farbpigmente könnten aber andere Tumoren begünstigen.

Update Innere Medizin

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