Skip to main content
Erschienen in: BMC Nephrology 1/2017

Open Access 01.12.2017 | Research article

Intermedin attenuates renal fibrosis by induction of heme oxygenase-1 in rats with unilateral ureteral obstruction

verfasst von: Xi Qiao, Lihua Wang, Yanhong Wang, Xiaole Su, Yufeng Qiao, Yun Fan, Zhiqiang Peng

Erschienen in: BMC Nephrology | Ausgabe 1/2017

Abstract

Background

Intermedin [IMD, adrenomedullin-2 (ADM-2)] attenuates renal fibrosis by inhibition of oxidative stress. However, the precise mechanisms remain unknown. Heme oxygenase-1 (HO-1), an antioxidant agent, is associated with antifibrogenic effects. ADM is known to induce HO-1. Whether IMD has any effect on HO-1 is unclear. Herein, we determined whether the antifibrotic properties of IMD are mediated by induction of HO-1.

Methods

Renal fibrosis was induced by unilateral ureteral obstruction (UUO) performed on male Wistar rats. Rat proximal tubular epithelial cell line (NRK-52E) was exposed to rhTGF-β1 (10 ng/ml) to establish an in vitro model of epithelial-mesenchymal transition (EMT). IMD was over-expressed in vivo and in vitro using the vector pcDNA3.1-IMD. Zinc protoporphyrin (ZnPP) was used to block HO-1 enzymatic activity. IMD effects on HO-1 expression in the obstructed kidney of UUO rat and in TGF-β1-stimulated NRK-52E were analyzed by real-time RT-PCR, Western blotting or immunohistochemistry. HO activity in the obstructed kidney, contralateral kidney of UUO rat and NRK-52E was examined by measuring bilirubin production. Renal fibrosis was determined by Masson trichrome staining and collagen I expression. Macrophage infiltration and IL-6 expression were evaluated using immunohistochemical analysis. In vivo and in vitro EMT was assessed by measuring α-smooth muscle actin (α-SMA) and E-cadherin expression using Western blotting or immunofluorescence, respectively.

Results

HO-1 expression and HO activity were increased in IMD-treated UUO kidneys or NRK-52E. The obstructed kidneys of UUO rats demonstrated significant interstitial fibrosis on day 7 after operation. In contrast, kidneys that were treated with IMD gene transfer exhibited minimal interstitial fibrosis. The obstructed kidneys of UUO rats also had greater macrophage infiltration and IL-6 expression. IMD restrained infiltration of macrophages and expression of IL-6 in UUO kidneys. The degree of EMT was extensive in obstructed kidneys of UUO rats as indicated by decreased expression of E-cadherin and increased expression of α-SMA. In vitro studies using NRK-52E confirmed these observations. EMT was suppressed by IMD gene delivery. However, all of the above beneficial effects of IMD were eliminated by ZnPP, an inhibitor of HO enzyme activity.

Conclusion

This study demonstrates that IMD attenuates renal fibrosis by induction of HO-1.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s12882-017-0659-6) contains supplementary material, which is available to authorized users.
Abkürzungen
ADM
Adrenomedullin
cDNA
Complementary DNA
CKD
Chronic kidney disease
Col-I
Collagen I
ECM
Extracellular matrix
EMT
Epithelial-mesenchymal transition
ESRD
End stage renal diseases
HO
Heme oxygenase
IMD
Intermedin
ROS
Reactive oxygen species
SD
Standard deviation
UUO
Unilateral ureteral obstruction
ZnPP
Zinc protoporphyrin
α-SMA
α-smooth muscle actin

Background

Chronic kidney disease (CKD) is a major health problem and its incidence and prevalence are increasing worldwide [1]. Regardless of disease etiology, fibrosis is a final common pathogenic process for CKD leading to end stage renal diseases (ESRD) [2]. Renal fibrosis is characterized by excessive accumulation of extracellular matrix (ECM) in the tubulointerstitium. Reactive oxygen species (ROS) play an important pathogenic role in the development of renal fibrosis by inducing epithelial-mesenchymal transition (EMT) of tubular epithelial cells and by inducing macrophage infiltration [35]. Intermedin [IMD, also known as adrenomedullin-2 (ADM-2)] is a novel member of the calcitonin/calcitonin gene-related peptide family [6]. Our previous study demonstrated that kidney-specific IMD gene transfer significantly ameliorated unilateral ureteral obstruction (UUO)-mediated fibronectin up-regulation and the fibrotic changes, and the anti-fibrotic effect is achieved by inhibition of oxidative stress [7]. However, the precise mechanisms remain unknown.
Heme oxygenase (HO) is the rate-limiting enzyme in the degradation of heme [8]. It converts heme to biliverdin via a reaction that produces carbon monoxide and liberates iron [9]. There are two isozymes for HO, an inducible form (HO-1) and a constitutive one (HO-2) [10]. HO-1 is strongly induced by oxidative stress and shows cytoprotective effects by the anti-inflammatory, anti-apoptotic, and anti-proliferative actions [11]. Its expression is upregulated in renal fibrosis [12], and furthermore, recent study demonstrated that induction of HO-1 prevented renal fibrosis induced by UUO [13].
ADM is reported to induce HO-1 gene expression in rat vascular smooth muscle cells [14]. Whether IMD has any effect on HO-1 has not been investigated yet. Since IMD and ADM have similar biological actions, we hypothesized that IMD can also induce HO-1 expression and that may explain its anti-oxidant and anti-fibrotic effects. To test this hypothesis, we investigated the effects of IMD overexpression on HO-1 expression and HO activity in the obstructed kidney of UUO rat and in rhTGF-β1-stimulated rat renal proximal tubular epithelial cells (NRK-52E). To evaluate the role of HO-1 induction in the protective effects of IMD on renal fibrosis, zinc protoporphyrin (ZnPP), an HO activity inhibitor [15], was used. Furthermore, NRK-52E were analyzed for the effect of IMD on EMT.

Methods

Animals and UUO-induced renal fibrosis model

Ethics Committee for Animals of Shanxi Medical University approved all animal protocols. Male Wistar rats (180 to 200 g) were purchased from the Experimental Animal Center of Shanxi Medical University (Taiyuan, China) and maintained in a specific pathogen-free environment in our facility. All animal were fed with standard chow and had free access to water. Renal fibrosis was induced by ligation of left ureter, UUO, as we previously reported [7]. Briefly, rats were anaesthetized, laparotomy performed, and the left ureter identified and ligated at two points along the ureter. Sham-operated rats underwent the same surgical procedure except for the ureter ligation.

Ultrasound-mediated gene delivery into the kidney

The eukaryotic expression plasmid pcDNA3.1-IMD containing fulllength complementary DNA (cDNA) sequence of rat IMD was successfully constructed in our previous study [16]. Before the ureter was obstructed, pcDNA3.1-IMD plasmid or control empty vector was transfected into the left kidney via the renal artery using an ultrasound-microbubble mediated system as we previously described [16]. Rats were killed at 7 days after UUO. The transfection rate was detected by quantitative RT-PCR and Western blot analysis, respectively.

Experiment design

Animal study

Rats were randomly divided into the following five groups: sham, UUO, UUO + empty plasmid, UUO + IMD and UUO + IMD + ZnPP. Rats in UUO + empty plasmid, UUO + IMD and UUO + IMD + ZnPP groups were treated with ultrasound as described above before UUO operation. In UUO + IMD + ZnPP group, ZnPP (50 μg/kg body wt, Sigma-Aldrich, St. Louis, MO, prepared in the dark) was administered intraperitoneally with a single dose 48 h before UUO operation as previously reported [17]. Animals were killed at 7 days after UUO. Obstructed kidneys and contralateral kidneys were harvested for further analysis.

Cell culture and treatments

Rat proximal tubular epithelial cell line (NRK-52E) was obtained from the Cell Bank of the Chinese Academy of Sciences (Shanghai, China). Cells were cultured in DMEM/Ham’s F-12 (2:1) containing with 10% fetal bovine serum and incubated at 37 °C, room air with 5% CO2. Cells were then cultured in fibronectin pre-coated flasks until they reached approximately 80% confluence. The over-expression of IMD in the NRK-52E cells that were transfected with pcDNA3.1-IMD was accomplished according to the methods described in our previous study [18]. The model of EMT in the NRK-52E cells was established by stimulating with recombinant human TGF-β1 (rhTGF-β1, 10 ng/ml, Gibco) for 72 h. The cells were assigned to 1 of 5 groups: control (untreated NRK-52E cells); TGF-β1 (NRK-52E cells were incubated with rhTGF-β1); TGF-β1 + empty vector (NRK-52E cells were transfected with the empty plasmid pcDNA3.1 and incubated with rhTGF-β1); TGF-β1 + IMD (NRK-52E cells were transfected with pcDNA3.1-IMD and incubated with rhTGF-β1); and TGF-β1 + IMD+ ZnPP [NRK-52E cells were transfected with pcDNA3.1-IMD and incubated with rhTGF-β1 plus 2 μM ZnPP (prepared in the dark)].

Quantitative RT-PCR

Total renal RNA was extracted with Trizol reagent (Invitrogen, Carlsbad, CA, USA), and then subjected to RT using a first-strand cDNA synthesis system (TAKARA, Dalian, China). Quantitative RT-PCR amplifications were performed in triplicate using the SYBR Green I assay and were carried out using Strategene M3000 Sequence Detection System (Stratagene, Santa Clara, CA, USA). Specific primers were used for IMD (sense: 5′-GGCCCAGTTGCTGATGGT-3′ and antisense: 5′-TGCCCGGGAGCAGGTA-3′), HO-1 (sense: 5′-AGGTGCACATCCGTGCAGAG -3′ and antisense: 5′-CTTCCAGGGCCGTATAGATATGGTA-3′) and β-actin (sense: 5′-CCCATCTATGAGGGTTACGC-3′ and antisense: 5′-TTTAATGTCACGCAC GATTTC-3′). The reaction was carried out in a 96-well plate in 25 μL reactions containing 2 × SYBR Green Master mix (TAKARA, Dalian, China)—2 pmol each of sense and antisense primer and the conditions were 95 °C for 2 min, followed by 95 °C for 10 s, 60 °C for 15 s, for 40 cycles. In each assay, a standard curve was determined concurrently with examined samples. Gene expression was quantified using a medication of the 2-ΔΔct method. The amount of PCR products was normalized to the level of β-actin to determine the relative expression ratio for each mRNA.

Western blot analysis

Analysis was carried out as previous described [16]. Immunoblot analysis was performed with rabbit polyclonal anti-IMD (Beijing Biosynthesis Biotechnology, Beijing, China), goat polyclonal anti-HO-1 (Santa Cruz Biotechnology), mouse monoclonal anti-collagen I (Col-I, Santa Cruz Biotechnology), mouse monoclonal anti-α-SMA (Sigma-Aldrich), or rabbit moloclonal anti-E-cadherin (Abcam, Cambridge, UK) and then peroxidase conjugated AffiniPure secondary antibodies (Santa Cruz Biotechnology), respectively. The ECL Western blotting system (Santa Cruz Biotechnology) was used for detection. Rabbit polyclonal anti-β-actin (Santa Cruz Biotechnology) was used as the control for each sample.

HO enzymatic activity assay

HO activity was estimated by determining the level of bilirubin generated in isolated microsomes as described earlier [19]. In brief, homogenized samples from the obstructed kidneys, contralateral kidneys or cultured NRK-52E cells were incubated in medium consisting of heme (0.25 mmol/L), rat liver cytosol (5 mg/mL) as a source of biliverdin reductase, MgCl2(1 mmol/L), glucose-6-phosphate dehydrogenase (1 unit), glucose-6-phosphate (0.8 mmol/L) and NADPH (0.8 mmol/L) in 0.5 mL of 0.1 mol/L phosphate buffer saline (pH 7.4) for 1 h at 37 °C in the dark. Reactions were terminated by ice and then the bilirubin product was extracted with chloroform. Absorbance was measured using a spectrometer at wavelengths 464 and 530 nm. HO activity was calculated by the difference in absorbance between the two wavelengths, and expressed as picomoles of bilirubin per milligram of protein per h.

Immunohistochemical analysis

Rat kidneys were fixed in 10% formaldehyde and embedded in paraffin. Paraffin sections (4 μm) were mounted on glass slides, deparaffinized in xylene, and rehydrated in ethanol with increasing concentrations of water. The rehydrated sections were pretreated with 3% H2O2 for 10 min at room temperature to block the endogenous peroxidase. After boiling in antigen retrieval solution (1 mmol/L tris-HCl, 0.1 mmol/L EDTA, pH = 8.0) for 10 min at high power in a microwave oven, the sections were incubated overnight at 4 °C with primary goat polyclonal anti-HO-1 (1:200, Santa Cruz Biotechnology), mouse monoclonal anti-ED-1 (1:200, Santa Cruz Biotechnology) or mouse moloclonal anti-IL-6 (1:200, Abcam, Cambridge, UK). This was followed by biotinylated secondary antibodies (Santa Cruz Biotechnology) and finally by avidin conjugated horseradish peroxidase. All slides were counterstained with haematoxylin. HO-1 or IL-6 positive stained areas were assessed and expressed as integrated optical density (IOD) and area. Interstitial macrophages were expressed as the percentage of ED-1 positive interstitial area. Three sections of each rat kidney were measured, and 10 random fields were chosen and calculated under magnification of 400×. The IOD and positive area were acquired by the Image-Pro Plus 6.0 program (Media Cybernetics, Bethesda, MD, USA).

Histological examinations

Renal histological changes were assessed at days 7 after UUO. Paraffin-embedded transverse kidney slices were sectioned at 4 μm. For analyzing the degree of tubulointerstitial collagen deposition, sections were stained with Masson trichrome. Twenty cortical tubulointerstitial fields that were randomly selected at ×200 magnifications were assessed in each rat, and the density of trichrome positive signals was analyzed using Image-Pro Plus 6.0 program (Media Cybernetics, Bethesda, MD, USA). Briefly, the tonality of the fibrosis area (blue) was determined with control sections as reference. The number of pixels with the predetermined color of tone was counted in each section, and then automatically converted into dimensions.

Immunofluorescence

NRK-52E monolayers were fixed in methanol (20 min, −20 °C), and blocked with 2% bovine serum albumin (BSA) in PBS (1 h, room temperature). Cells were then incubated with primary antibodies against epithelial cell marker rabbit moloclonal anti-E-cadherin (1:50, Abcam, Cambridge, UK) and mesenchymal marker mouse monoclonal anti-α-SMA (1:100; Sigma-Aldrich) with 2% BSA overnight at 4 °C). Secondary fluorescent conjugated anti-rabbit Alexa Fluor®647 (1:400; Invitrogen) was used for E-cadherin and anti-mouse Alexa Fluor®546 (1:400; Invitrogen) was used for α-SMA for 40 min at room temperature in dark. Slides were mounted using ProLong® Gold Antifade reagent with DAPI (Life technologies). Images were photographed using Olympus FV1000 Confocal microscope at ×40 magnification.

Statistical analysis

The data are expressed as means ± standard deviation (SD). Significant differences between groups were assessed by one-way ANOVA with Student-Newman-Keuls post hoc tests. P values <0.05 were considered statistically significant.

Results

IMD is transfected into the kidney or into NRK-52E cells successfully

The transfection efficiency of IMD by ultrasound mediated gene delivery into the Kidney in vivo and by FuGENE HD into NRK-52E cells in vitro was examined by quantitative RT-PCR and Western blot analysis. We showed that after 7 days of transfection, obstructed kidneys from rats treated with pcDNA3.1-IMD plasmid exhibited significant increase in IMD expression compared with that from rats treated with control empty vector (Fig. 1a–c). Similarly, the expression of IMD in NRK-52E cells transfected with pcDNA3.1-IMD plasmid was much high than in controls, indicating that IMD was transfected into the kidney or NRK-52E successfully (Fig. 1d–f).

HO-1 is upregulated and HO activity is increased in the obstructed kidney of UUO rats, and is further induced by IMD gene transfer

HO-1 mRNA and protein expression was very low in the sham-operated kidneys, whereas there was a significant increase in HO-1 mRNA and protein abundance in the obstructed kidney after UUO. IMD gene transfer further increased HO-1 expressions in the obstructed kidneys, while the control empty plasmid had no effect on HO-1 levels. Interestingly, the levels of HO-1 mRNA and protein were also increased in UUO rats with IMD and ZnPP combined treatment (Fig. 2a and b). The result was in line with previous report [17].
Localization of HO-1 protein in the obstructed kidneys was examined by immunohistochemical analysis. We showed that HO-1 immunoreactivity was increased in the obstructed kidneys of UUO rats. Positive staining of HO-1 protein was mainly observed in tubular cells. HO-1 expression was further enhanced by IMD gene delivery. Immunohistochemical studies also demonstrated an upregulation of HO-1 with additional ZnPP treatment. Control empty plasmid did not affect the expression level of HO-1(Fig. 2c).
HO activity was significantly increased in UUO kidneys compared to sham controls. Pretreatment with IMD further elevated HO enzymatic activity in the obstructed kidneys of UUO animals. Administration of ZnPP abrogated IMD action on HO activity. Control empty plasmid had no effect on HO activity (Fig. 2d). We also evaluated the effect of IMD on HO activity of the contralateral kidneys of UUO rats, and didn’t find any difference between IMD treated UUO rats and untreated UUO controls (Additional file 1: Figure S1).

IMD gene delivery induces HO-1 expression and HO activity in proximal tubular epithelial cells

We investigated the effect of IMD on HO-1 expression and HO activity in rhTGF-β1-stimulated rat proximal tubular epithelial cell line NRK-52E. Our results showed that HO-1 expression and HO activity were induced by rhTGF-β1 administration. Overexpression of IMD by gene transfer further increased HO-1 expression and HO activity. ZnPP up-regulated HO-1 expression but reduced HO activity. As a control, empty plasmid had little effect on HO activity (Fig. 3).

IMD reduces renal fibrosis in obstructed kidneys by induction of HO-1

Tubulointerstitial fibrosis was assessed by Masson’s trichrome staining for collagen as we previously described [7]. No changes were detected in the sham group. We showed that tubulointerstitial fibrosis was produced in the obstructed kidney of UUO rats 7 days after operation compared with sham controls. By contrast, IMD gene delivery significantly ameliorated UUO-induced tubulointerstitial fibrosis in the obstructed kidneys. To ascertain the anti-fibrotic role of IMD could be afforded by induction of HO-1, ZnPP, a known inhibitor of HO enzymatic activity, was used. As expected, the suppression effect of tubulointerstitial fibrosis by IMD was blocked by ZnPP. Increased renal fibrosis in UUO was confirmed by Col-I, a major ECM protein, expression. Western blot of Col-I yielded significant increase in Col-I expression in the untreated UUO rats compared with the sham. IMD treatment markedly inhibited the up-regulation of Col-I. ZnPP administration abrogated the protective effect of IMD on Col-I expression. These data demonstrate that the overexpression of IMD ameliorates renal interstitial fibrosis by induction of HO-1 (Fig. 4).

IMD inhibits inflammation in the obstructed kidneys by induction of HO-1

To investigate whether IMD could inhibit renal inflammation, we evaluated macrophage infiltration and IL-6 expression in the obstructed kidneys of UUO rats. Staining for ED-1 revealed the degree of interstitial macrophage infiltration. Few ED-1-positive cells were observed in the interstitium of sham-operated kidneys. The number of ED-1-positive cells in the obstructed kidneys from UUO rats markedly increased, mainly in the periglomerular and peritubular areas. IMD gene transfer significantly inhibited macrophages infiltration, which could subsequently be blocked by the HO-1 inhibitor ZnPP (Fig. 5a and b). Pro-inflammatory cytokine IL-6 expression was elevated in the obstructed kidneys after UUO operation as compared with the sham controls, which was inhibited by IMD gene delivery. ZnPP attenuated the protective effect of IMD on IL-6 expression (Fig. 5c and d).

UUO-induced tubular epithelial cells EMT is inhibited by IMD via induction of HO-1

For determination if IMD could inhibit UUO-induced EMT, we examined α-SMA, a mesenchymal marker, and E-cadherin, an epithelial marker, expressions by Western blot analysis. We showed that α-SMA expression up-regulated significantly, while E-cadherin down-regulated markedly in the obstructed kidneys of untreated UUO rats. IMD inhibited UUO-induced increase of α-SMA expression and decrease of E-cadherin in the obstructed kidney. Inhibition of HO-1 enzyme activity by ZnPP reversed the effects of IMD (Fig. 6).

TGF-β1-mediated proximal tubular epithelial cells EMT is inhibited by IMD via induction of HO-1

To confirm the effect of IMD on EMT, we examined α-SMA/E-cadherin double staining in rat proximal tubular epithelial cell line NRK-52E by immunofluorescence assay. In response to rhTGF-β1, E-cadherin expression decreased, while α-SMA increased. IMD increased E-cadherin expression and suppressed α-SMA expression significantly. The protective effect of IMD was abolished by the addition of ZnPP (Fig. 7).

Discussion

Tubulointerstitial fibrosis is a common pathway of all CKD leading to ESRD. Furthermore, it remains the best predictor of disease progression [20]. In the present study, we analyzed the protective effect of IMD on renal fibrosis in rat UUO model, a well-established in vivo model of renal fibrosis. Our results showed that IMD reduced parameters of fibrosis such as elevated collagen deposition measured by Masson’s trichrome staining, and Col-I production. In addition, IMD inhibited macrophages infiltration, pro-inflammatory cytokine IL-6 expression and EMT in UUO-induced kidneys. In vitro studies demonstrated that TGF-β1-stimulated EMT in proximal tubular epithelial cells was reduced by IMD. Furthermore, all the above beneficial effects of IMD were abolished by inhibition of HO-1 enzyme activity with ZnPP.
It is commonly accepted that ROS have been implicated in the pathophysiology of renal fibrosis [21]. Our previous studies demonstrated that IMD attenuated renal fibrosis by inhibition of oxidative stress [7] and the precise mechanisms warrant further studies. HO is a microsomal enzyme that exists in two isoforms: the inducible HO-1 and the constitutive HO-2. HO-1 is a stress response protein with antioxidative properties [22]. It is reported that induction of HO-1 is an adaptive and protective response against renal fibrosis [17]. Whether IMD has any effect on HO-1 expression and HO activity has not been investigated yet. In this study, we observed a significant amplification of HO-1 mRNA and protein as well as HO activity in IMD-treated animals (Fig. 2a–d). Immunohistology reveals that IMD-induced HO-1 protein expressions were mainly in tubular epithelial cells (Fig. 2c). We reached the same results in TGF-β1-stimulated tubular cells (Fig. 3). Importantly, this upregulation of HO-1 and induction of HO activity by IMD was paralleled with suppression of tubulointerstitial fibrosis (Fig. 4).
To verify if the anti-fibrotic effect of IMD is achieved by induction of HO-1, we used ZnPP. It has been reported that ZnPP inhibited HO-1 enzyme activity, although it increased HO-1 protein and mRNA expression [23]. Although HO-2 activity is also inhibited by ZnPP, the constitutive nature of HO-2 makes it less attractive as a drug target [10]. In line with previous reports, our results indicated that ZnPP markedly decreased HO activity in obstructed kidney and cultured tubular cells. HO activity in contralateral kidney of UUO rat was also inhibited by ZnPP, indicating that the effect of ZnPP on HO activity was systemic. We showed that inhibition of HO enzyme activity abrogated the protective effect of IMD on interstitial fibrosis. Thus, the antifibrotic effect of IMD in our model is dependent on induction of HO-1.
A number of evidence demonstrated that oxidative stress could trigger the inflammatory response in renal fibrosis [24]. Inflammation plays an important role in the development and progression of renal fibrosis [25]. Macrophages, a major type of inflammatory cell, are recruited during tubulointerstitial fibrosis [26, 27]. IL-6 is a well-known pro-inflammatory cytokine that is also correlated with increased fibrosis [28]. In this study, we showed the number of macrophages and expression of IL-6 in the obstructed kidneys of UUO rats was significantly higher than that of the sham-operated rats. Although the recruitment of macrophages may contribute to kidney repair at the early stages, this repair can ultimately cause renal fibrosis [27, 29]. In our study, in line with the infiltration of macrophages and up-regulation of IL-6, renal fibrosis is associated enhanced (Fig. 4). IMD overexpression obviously inhibited the infiltration of macrophages and IL-6 expression following UUO, which effect could subsequently be blocked by the HO-1 inhibitor ZnPP. This indicates that the protection against macrophage infiltration and IL-6 production afforded by IMD was due to induction of HO-1.
EMT plays a pivotal role in the pathogenesis of renal tubulointerstitial fibrosis [30]. EMT is recognized as a loss of epithelial cell phenotype, decrease of intercellular epithelial adhesion molecules, such as E-cadherin, with a concomitant development of mesenchymal phenotype, including expression of α-SMA [31]. In this study, we showed that α-SMA expression significantly increased, while E-cadherin expression markedly decreased, after UUO operation, indicating extensive EMT (Fig. 6). It has been suggested that EMT is mediated, at least in part, through ROS [32]. Thus, IMD, through its antioxidant properties, may reduce EMT. Our results demonstrated that IMD gene delivery attenuated UUO-induced EMT, which was abolished by HO-1 activity inhibition.
For further clarification of the effects of IMD on TGF-β1-mediated EMT in proximal tubules, in vitro studies were performed to eliminate the confounding effects of infiltrating cells. These in vitro studies, using rat proximal tubular cells (NRK-52E), indicated that IMD inhibited TGF-β1-mediated EMT as indicated by up-regulation of E-cadherin and down-regulation of α-SMA measured by immunofluorescence (Fig. 7). ZnPP abrogated the beneficial effects of IMD on EMT. These results, along with the in vivo evidence, demonstrate that IMD inhibits EMT by induction of HO-1.
Although our current study provided significant results, there are still some limitations. The primary one is that we did not use HO-1-deficient mice or HO-1-deficient tubular cells to investigate the effect of IMD on renal fibrosis. ZnPP is commonly used as a HO-1 inhibitor [15], however, its specificity is still a matter of debate [33, 34]. The second limitation is the use of UUO as an animal model CKD. Although UUO is a very well-accepted renal fibrosis model, some experts criticized it as a model of CKD because of the compensation by the normal contralateral kidney that precludes meaningful renal functional measures [35]. More CKD models that provide us with an opportunity to include renal functional study end-points together with quantitative measures of fibrosis severity are needed. The last one is we didn’t investigate the transcription pathways by which IMD activates HO-1. Examining some transcription factors involved in HO-1 modulation are required to provide further mechanistic insight into the mechanism of IMD induced HO-1 activation pathway.

Conclusion

In summary, our study demonstrates that IMD gene transfer significantly attenuates renal fibrosis in parallel with the blockade of inflammation and EMT of tubular cell, and that this is associated with an increased HO-1 expression and HO activity. Blockade of HO enzyme activity by a specific inhibitor completely abrogates the effects of IMD, indicating that the beneficial effects of IMD are achieved by HO-1 induction. The results indicate that induction of HO-1 by IMD may be a potential therapeutic strategy to prevent renal fibrosis in patients with CKD.

Acknowledgements

This study was supported by the National Natural Science Foundation of China (Grant No. 81100531), Shanxi Province Health and family planning commission, Science and Technology Project (Grant No. 201601043) and Shanxi Province Scientific Research Foundation for the Returned Overseas Chinese Scholars (Grant No. 2010–58).

Availability of data and materials

All the dataset on which the conclusion is drawn will be deposited in publicly available repositories.

Ethics approval

All animal procedures were approved and conducted in compliance with the guidelines of Ethics Committee for Animals of Shanxi Medical University. Our manuscript reporting adheres to the ARRIVE guidelines in accordance with BioMed Central editorial policies.
Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. 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.
Literatur
1.
Zurück zum Zitat Komenda P, Ferguson TW, Macdonald K, Rigatto C, Koolage C, Sood MM, Tangri N. Cost-effectiveness of primary screening for CKD: a systematic review. Am J Kidney Dis. 2014;63(5):789–97.CrossRefPubMed Komenda P, Ferguson TW, Macdonald K, Rigatto C, Koolage C, Sood MM, Tangri N. Cost-effectiveness of primary screening for CKD: a systematic review. Am J Kidney Dis. 2014;63(5):789–97.CrossRefPubMed
3.
Zurück zum Zitat Wang H, Chen X, Su Y, Paueksakon P, Hu W, Zhang MZ, Harris RC, Blackwell TS, Zent R, Pozzi A. p47(phox) contributes to albuminuria and kidney fibrosis in mice. Kidney Int. 2015;87(5):948–62.CrossRefPubMedPubMedCentral Wang H, Chen X, Su Y, Paueksakon P, Hu W, Zhang MZ, Harris RC, Blackwell TS, Zent R, Pozzi A. p47(phox) contributes to albuminuria and kidney fibrosis in mice. Kidney Int. 2015;87(5):948–62.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Rhyu DY, Yang Y, Ha H, Lee GT, Song JS, Uh ST, Lee HB. Role of reactive oxygen species in TGF-beta1-induced mitogen-activated protein kinase activation and epithelial-mesenchymal transition in renal tubular epithelial cells. J Am Soc Nephrol. 2005;16(3):667–75.CrossRefPubMed Rhyu DY, Yang Y, Ha H, Lee GT, Song JS, Uh ST, Lee HB. Role of reactive oxygen species in TGF-beta1-induced mitogen-activated protein kinase activation and epithelial-mesenchymal transition in renal tubular epithelial cells. J Am Soc Nephrol. 2005;16(3):667–75.CrossRefPubMed
5.
Zurück zum Zitat Paul-Clark MJ, McMaster SK, Sorrentino R, Sriskandan S, Bailey LK, Moreno L, Ryffel B, Quesniaux VF, Mitchell JA. Toll-like receptor 2 is essential for the sensing of oxidants during inflammation. Am J Respir Crit Care Med. 2009;179(4):299–306.CrossRefPubMed Paul-Clark MJ, McMaster SK, Sorrentino R, Sriskandan S, Bailey LK, Moreno L, Ryffel B, Quesniaux VF, Mitchell JA. Toll-like receptor 2 is essential for the sensing of oxidants during inflammation. Am J Respir Crit Care Med. 2009;179(4):299–306.CrossRefPubMed
6.
Zurück zum Zitat Roh J, Chang CL, Bhalla A, Klein C, Hsu SY. Intermedin is a calcitonin/calcitonin gene-related peptide family peptide acting through the calcitonin receptor-like receptor/receptor activity-modifying protein receptor complexes. J Biol Chem. 2004;279(8):7264–74.CrossRefPubMed Roh J, Chang CL, Bhalla A, Klein C, Hsu SY. Intermedin is a calcitonin/calcitonin gene-related peptide family peptide acting through the calcitonin receptor-like receptor/receptor activity-modifying protein receptor complexes. J Biol Chem. 2004;279(8):7264–74.CrossRefPubMed
7.
Zurück zum Zitat Qiao X, Wang L, Wang Y, Zhao N, Zhang R, Han W, Peng Z. Intermedin is upregulated and attenuates renal fibrosis by inhibition of oxidative stress in rats with unilateral ureteral obstruction. Nephrology. 2015;20(11):820–31.CrossRefPubMed Qiao X, Wang L, Wang Y, Zhao N, Zhang R, Han W, Peng Z. Intermedin is upregulated and attenuates renal fibrosis by inhibition of oxidative stress in rats with unilateral ureteral obstruction. Nephrology. 2015;20(11):820–31.CrossRefPubMed
8.
Zurück zum Zitat Maines MD. The heme oxygenase system: a regulator of second messenger gases. Annu Rev Pharmacol Toxicol. 1997;37:517–54.CrossRefPubMed Maines MD. The heme oxygenase system: a regulator of second messenger gases. Annu Rev Pharmacol Toxicol. 1997;37:517–54.CrossRefPubMed
9.
Zurück zum Zitat Hill-Kapturczak N, Chang SH, Agarwal A. Heme oxygenase and the kidney. DNA Cell Biol. 2002;21(4):307–21.CrossRefPubMed Hill-Kapturczak N, Chang SH, Agarwal A. Heme oxygenase and the kidney. DNA Cell Biol. 2002;21(4):307–21.CrossRefPubMed
10.
Zurück zum Zitat Abraham NG, Junge JM, Drummond GS. Translational significance of heme oxygenase in obesity and metabolic syndrome. Trends Pharmacol Sci. 2016;37(1):17–36.CrossRefPubMed Abraham NG, Junge JM, Drummond GS. Translational significance of heme oxygenase in obesity and metabolic syndrome. Trends Pharmacol Sci. 2016;37(1):17–36.CrossRefPubMed
11.
Zurück zum Zitat Kirkby KA, Adin CA. Products of heme oxygenase and their potential therapeutic applications. Am J Physiol Renal Physiol. 2006;290(3):F563–71.CrossRefPubMed Kirkby KA, Adin CA. Products of heme oxygenase and their potential therapeutic applications. Am J Physiol Renal Physiol. 2006;290(3):F563–71.CrossRefPubMed
12.
Zurück zum Zitat Kawada N, Moriyama T, Ando A, Fukunaga M, Miyata T, Kurokawa K, Imai E, Hori M. Increased oxidative stress in mouse kidneys with unilateral ureteral obstruction. Kidney Int. 1999;56(3):1004–13.CrossRefPubMed Kawada N, Moriyama T, Ando A, Fukunaga M, Miyata T, Kurokawa K, Imai E, Hori M. Increased oxidative stress in mouse kidneys with unilateral ureteral obstruction. Kidney Int. 1999;56(3):1004–13.CrossRefPubMed
13.
Zurück zum Zitat Chen X, Wei SY, Li JS, Zhang QF, Wang YX, Zhao SL, Yu J, Wang C, Qin Y, Wei QJ, et al. Overexpression of heme oxygenase-1 prevents renal interstitial inflammation and fibrosis induced by unilateral ureter obstruction. PLoS One. 2016;11(1):e0147084.CrossRefPubMedPubMedCentral Chen X, Wei SY, Li JS, Zhang QF, Wang YX, Zhao SL, Yu J, Wang C, Qin Y, Wei QJ, et al. Overexpression of heme oxygenase-1 prevents renal interstitial inflammation and fibrosis induced by unilateral ureter obstruction. PLoS One. 2016;11(1):e0147084.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Qi YF, Dong LW, Pan CS, Zhang J, Geng B, Zhao J, Tang CS. Adrenomedullin induces heme oxygenase-1 gene expression and cGMP formation in rat vascular smooth muscle cells. Peptides. 2005;26(7):1257–63.CrossRefPubMed Qi YF, Dong LW, Pan CS, Zhang J, Geng B, Zhao J, Tang CS. Adrenomedullin induces heme oxygenase-1 gene expression and cGMP formation in rat vascular smooth muscle cells. Peptides. 2005;26(7):1257–63.CrossRefPubMed
15.
Zurück zum Zitat Amersi F, Buelow R, Kato H, Ke B, Coito AJ, Shen XD, Zhao D, Zaky J, Melinek J, Lassman CR, et al. Upregulation of heme oxygenase-1 protects genetically fat Zucker rat livers from ischemia/reperfusion injury. J Clin Invest. 1999;104(11):1631–9.CrossRefPubMedPubMedCentral Amersi F, Buelow R, Kato H, Ke B, Coito AJ, Shen XD, Zhao D, Zaky J, Melinek J, Lassman CR, et al. Upregulation of heme oxygenase-1 protects genetically fat Zucker rat livers from ischemia/reperfusion injury. J Clin Invest. 1999;104(11):1631–9.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Qiao X, Li RS, Li H, Zhu GZ, Huang XG, Shao S, Bai B. Intermedin protects against renal ischemia-reperfusion injury by inhibition of oxidative stress. Am J Physiol Renal Physiol. 2013;304(1):F112–9.CrossRefPubMed Qiao X, Li RS, Li H, Zhu GZ, Huang XG, Shao S, Bai B. Intermedin protects against renal ischemia-reperfusion injury by inhibition of oxidative stress. Am J Physiol Renal Physiol. 2013;304(1):F112–9.CrossRefPubMed
17.
Zurück zum Zitat Kim JH, Yang JI, Jung MH, Hwa JS, Kang KR, Park DJ, Roh GS, Cho GJ, Choi WS, Chang SH. Heme oxygenase-1 protects rat kidney from ureteral obstruction via an antiapoptotic pathway. J Am Soc Nephrol. 2006;17(5):1373–81.CrossRefPubMed Kim JH, Yang JI, Jung MH, Hwa JS, Kang KR, Park DJ, Roh GS, Cho GJ, Choi WS, Chang SH. Heme oxygenase-1 protects rat kidney from ureteral obstruction via an antiapoptotic pathway. J Am Soc Nephrol. 2006;17(5):1373–81.CrossRefPubMed
18.
Zurück zum Zitat Wang Y, Tian J, Qiao X, Su X, Mi Y, Zhang R, Li R. Intermedin protects against renal ischemia-reperfusion injury by inhibiting endoplasmic reticulum stress. BMC Nephrol. 2015;16:169.CrossRefPubMedPubMedCentral Wang Y, Tian J, Qiao X, Su X, Mi Y, Zhang R, Li R. Intermedin protects against renal ischemia-reperfusion injury by inhibiting endoplasmic reticulum stress. BMC Nephrol. 2015;16:169.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Zhao W, Huang X, Zhang L, Yang X, Wang L, Chen Y, Wang J, Wu G. Penehyclidine hydrochloride pretreatment ameliorates rhabdomyolysis-induced AKI by activating the Nrf2/HO-1 pathway and alleviating [corrected] endoplasmic reticulum stress in rats. PLoS One. 2016;11(3):e0151158.CrossRefPubMedPubMedCentral Zhao W, Huang X, Zhang L, Yang X, Wang L, Chen Y, Wang J, Wu G. Penehyclidine hydrochloride pretreatment ameliorates rhabdomyolysis-induced AKI by activating the Nrf2/HO-1 pathway and alleviating [corrected] endoplasmic reticulum stress in rats. PLoS One. 2016;11(3):e0151158.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Boor P, Ostendorf T, Floege J. Renal fibrosis: novel insights into mechanisms and therapeutic targets. Nat Rev Nephrol. 2010;6(11):643–56.CrossRefPubMed Boor P, Ostendorf T, Floege J. Renal fibrosis: novel insights into mechanisms and therapeutic targets. Nat Rev Nephrol. 2010;6(11):643–56.CrossRefPubMed
21.
Zurück zum Zitat Singh DK, Winocour P, Farrington K. Oxidative stress in early diabetic nephropathy: fueling the fire. Nat Rev Endocrinol. 2011;7(3):176–84.CrossRefPubMed Singh DK, Winocour P, Farrington K. Oxidative stress in early diabetic nephropathy: fueling the fire. Nat Rev Endocrinol. 2011;7(3):176–84.CrossRefPubMed
22.
Zurück zum Zitat Ho YC, Wu ML, Gung PY, Chen CH, Kuo CC, Yet SF. Heme oxygenase-1 deficiency exacerbates angiotensin II-induced aortic aneurysm in mice. Oncotarget. 2016;7(42):67760-76. Ho YC, Wu ML, Gung PY, Chen CH, Kuo CC, Yet SF. Heme oxygenase-1 deficiency exacerbates angiotensin II-induced aortic aneurysm in mice. Oncotarget. 2016;7(42):67760-76.
23.
Zurück zum Zitat Sardana MK, Kappas A. Dual control mechanism for heme oxygenase: tin(IV)-protoporphyrin potently inhibits enzyme activity while markedly increasing content of enzyme protein in liver. Proc Natl Acad Sci U S A. 1987;84(8):2464–8.CrossRefPubMedPubMedCentral Sardana MK, Kappas A. Dual control mechanism for heme oxygenase: tin(IV)-protoporphyrin potently inhibits enzyme activity while markedly increasing content of enzyme protein in liver. Proc Natl Acad Sci U S A. 1987;84(8):2464–8.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Sun Y, Zhang Y, Zhao D, Ding G, Huang S, Zhang A, Jia Z. Rotenone remarkably attenuates oxidative stress, inflammation, and fibrosis in chronic obstructive uropathy. Mediat Inflamm. 2014;2014:670106. Sun Y, Zhang Y, Zhao D, Ding G, Huang S, Zhang A, Jia Z. Rotenone remarkably attenuates oxidative stress, inflammation, and fibrosis in chronic obstructive uropathy. Mediat Inflamm. 2014;2014:670106.
25.
Zurück zum Zitat Lee SB, Kalluri R. Mechanistic connection between inflammation and fibrosis. Kidney Int Suppl. 2010;119:S22–6.CrossRef Lee SB, Kalluri R. Mechanistic connection between inflammation and fibrosis. Kidney Int Suppl. 2010;119:S22–6.CrossRef
26.
Zurück zum Zitat Klahr S, Morrissey J. Obstructive nephropathy and renal fibrosis. Am J Physiol Renal Physiol. 2002;283(5):F861–75.CrossRefPubMed Klahr S, Morrissey J. Obstructive nephropathy and renal fibrosis. Am J Physiol Renal Physiol. 2002;283(5):F861–75.CrossRefPubMed
27.
Zurück zum Zitat Vernon MA, Mylonas KJ, Hughes J. Macrophages and renal fibrosis. Semin Nephrol. 2010;30(3):302–17.CrossRefPubMed Vernon MA, Mylonas KJ, Hughes J. Macrophages and renal fibrosis. Semin Nephrol. 2010;30(3):302–17.CrossRefPubMed
28.
Zurück zum Zitat Ranganathan P, Jayakumar C, Ramesh G. Proximal tubule-specific overexpression of netrin-1 suppresses acute kidney injury-induced interstitial fibrosis and glomerulosclerosis through suppression of IL-6/STAT3 signaling. Am J Physiol Renal Physiol. 2013;304(8):F1054–65.CrossRefPubMedPubMedCentral Ranganathan P, Jayakumar C, Ramesh G. Proximal tubule-specific overexpression of netrin-1 suppresses acute kidney injury-induced interstitial fibrosis and glomerulosclerosis through suppression of IL-6/STAT3 signaling. Am J Physiol Renal Physiol. 2013;304(8):F1054–65.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Duffield JS. Macrophages in kidney repair and regeneration. J Am Soc Nephrol. 2011;22(2):199–201.CrossRefPubMed Duffield JS. Macrophages in kidney repair and regeneration. J Am Soc Nephrol. 2011;22(2):199–201.CrossRefPubMed
30.
Zurück zum Zitat Kie JH, Kapturczak MH, Traylor A, Agarwal A, Hill-Kapturczak N. Heme oxygenase-1 deficiency promotes epithelial-mesenchymal transition and renal fibrosis. J Am Soc Nephrol. 2008;19(9):1681–91.CrossRefPubMedPubMedCentral Kie JH, Kapturczak MH, Traylor A, Agarwal A, Hill-Kapturczak N. Heme oxygenase-1 deficiency promotes epithelial-mesenchymal transition and renal fibrosis. J Am Soc Nephrol. 2008;19(9):1681–91.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Loeffler I, Wolf G. Epithelial-to-mesenchymal transition in diabetic nephropathy: fact or fiction? Cell. 2015;4(4):631–52.CrossRef Loeffler I, Wolf G. Epithelial-to-mesenchymal transition in diabetic nephropathy: fact or fiction? Cell. 2015;4(4):631–52.CrossRef
32.
Zurück zum Zitat Radisky DC, Kenny PA, Bissell MJ. Fibrosis and cancer: do myofibroblasts come also from epithelial cells via EMT? J Cell Biochem. 2007;101(4):830–9.CrossRefPubMedPubMedCentral Radisky DC, Kenny PA, Bissell MJ. Fibrosis and cancer: do myofibroblasts come also from epithelial cells via EMT? J Cell Biochem. 2007;101(4):830–9.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Zygmunt PM, Hogestatt ED, Grundemar L. Light-dependent effects of zinc protoporphyrin IX on endothelium-dependent relaxation resistant to N omega-nitro-L-arginine. Acta Physiol Scand. 1994;152(2):137–43.CrossRefPubMed Zygmunt PM, Hogestatt ED, Grundemar L. Light-dependent effects of zinc protoporphyrin IX on endothelium-dependent relaxation resistant to N omega-nitro-L-arginine. Acta Physiol Scand. 1994;152(2):137–43.CrossRefPubMed
34.
Zurück zum Zitat Ny L, Andersson KE, Grundemar L. Inhibition by zinc protoporphyrin-IX of receptor-mediated relaxation of the rat aorta in a manner distinct from inhibition of haem oxygenase. Br J Pharmacol. 1995;115(1):186–90.CrossRefPubMedPubMedCentral Ny L, Andersson KE, Grundemar L. Inhibition by zinc protoporphyrin-IX of receptor-mediated relaxation of the rat aorta in a manner distinct from inhibition of haem oxygenase. Br J Pharmacol. 1995;115(1):186–90.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Eddy AA, Lopez-Guisa JM, Okamura DM, Yamaguchi I. Investigating mechanisms of chronic kidney disease in mouse models. Pediatr Nephrol. 2012;27(8):1233–47.CrossRefPubMed Eddy AA, Lopez-Guisa JM, Okamura DM, Yamaguchi I. Investigating mechanisms of chronic kidney disease in mouse models. Pediatr Nephrol. 2012;27(8):1233–47.CrossRefPubMed
Metadaten
Titel
Intermedin attenuates renal fibrosis by induction of heme oxygenase-1 in rats with unilateral ureteral obstruction
verfasst von
Xi Qiao
Lihua Wang
Yanhong Wang
Xiaole Su
Yufeng Qiao
Yun Fan
Zhiqiang Peng
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
BMC Nephrology / Ausgabe 1/2017
Elektronische ISSN: 1471-2369
DOI
https://doi.org/10.1186/s12882-017-0659-6

Weitere Artikel der Ausgabe 1/2017

BMC Nephrology 1/2017 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.