Angiotensin-(1-7) attenuates damage to podocytes induced by preeclamptic serum through MAPK pathways

  • Authors:
    • Jimei Tian
    • Lihong Zhang
    • Yunjiao Zhou
    • Jing Xiao
    • Sheran Li
    • Yaping Chen
    • Zhongdong Qiao
    • Jianying Niu
    • Yong Gu
  • View Affiliations

  • Published online on: July 29, 2014     https://doi.org/10.3892/ijmm.2014.1870
  • Pages: 1057-1064
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The underlying mechanisms of proteinuria, a main characteristic of preeclampsia (PE), have not yet been fully elucidated. Evidence indicates that the renin-angiotensin system (RAS) is involved in the pathogenesis of this disease, including decreased angiotensin-(1-7) [Ang-(1-7)] levels in the circulation and urine. In the present study, we examined the damage to podocytes induced by preeclamptic serum and the effects of Ang-(1-7) on podocytes treated with preeclamptic serum, as well as the underlying mechanisms. The podocytes were incubated with serum obtained from women with PE or with serum from women with normal pregnancies for different periods of time. Cell viability was determined by CCK-8 assay. The cells were treated with various concentrations of Ang-(1-7) and A779 [an (Ang-(1-7) antagonist]. The effects of Ang-(1-7) on the expression of podocyte-specific proteins [nephrin, Wilms tumor‑1 (WT-1) and podocin] and the phosphorylation of mitogen-activated protein kinases (MAPKs) were investigated by western blot analysis. Changes in F-actin rearrangement were determined by immunofluorescence. Podocyte apoptosis was determined by flow cytometry. The results revealed that in the cultured podocytes incubated with preeclamptic serum, there was a decrease in the expression of podocyte-specific proteins (nephrin and WT-1 but not podocin), a rearrangement of F-actin and apoptosis compared with the control group. However, treatment with Ang-(1-7) attenuated podocyte injury in the preeclamptic group, which may be mediated through the downregulation of MAPK (p38, ERK1/2 and JNK) phosphorylation. Thus, our data suggest that Ang-(1-7) plays a protective role in PE through the downregulation of MAPK phosphorylation.

Introduction

Preeclampsia (PE) is one of the leading causes of maternal and neonatal morbidity and mortality worldwide and is characterized by a new onset of hypertension and proteinuria after 20 weeks gestation (1). Kidney damage plays an important role in the pathogenesis of PE. The rates of complications due to worsening renal function and hypertension are increased among pregnant women with moderate or severe renal insufficiency (2). Great advances have been made in recent years in understanding the pathogenesis of PE (3). However, further studies are required in order to fully elucidate the underlying mechanisms of PE and the development of acute kidney injury and chronic kidney disease in mothers.

Studies have demonstrated that proteinuria in pregnant women with PE is due to direct damage to podocytes (4). Adverse agents that act on podocytes can induce the proliferation, apoptosis and necrosis of podocytes, podocyte detachment from the glomerular basement membrane, as well as a loss of podocyte differentiation markers (5).

The renin-angiotensin-aldosterone system (RAAS), particularly renal RAAS, plays an important role in the development of chronic kidney disease. In recent years, RAS has also been shown to be involved in the pathogenesis of PE in a series of studies and has been shown to induce various characteristics of this disease (6,7). In normal pregnancy (NP), women are normotensive despite the upregulation of RAS components, suggesting that there is a counter-regulatory mechanism to reverse the vasoconstriction and sodium retention induced by angiotensin (Ang) II (8). In this regard, Valdés et al (9) found that plasma levels of angiotensin-(1-7) [Ang-(1-7)] are elevated in normotensive pregnant women, and that concentrations of urinary Ang-(1-7) are also markedly increased throughout pregnancy, indicating that Ang-(1-7) may play a vasodilator role. The heptapeptide, Ang-(1-7), is an important component of RAS and acts as a counter-regulatory peptide in RAS, often balancing the physiological actions of Ang II. It has been shown that Ang-(1-7) binds to the Mas G protein-coupled receptor to exert many of its biological effects (10). Merrill et al (11) found that Ang-(1-7) was significantly decreased in pregnant women with PE compared with women NP. Although the levels of renin, Ang I and Ang II are decreased in pregnant women with PE (12), pregnant women with PE have an increased sensitivity to Ang II (13), indicating an exaggerated pressor response. Thus, we hypothesized that the development of proteinuria in PE may be attributed to the imbalance between the angiotensin-converting enzyme (ACE)-Ang II-AT1R axis and the ACE2-Ang-(1-7)-Mas axis and the decreased Ang-(1-7) expression in the circulation and renal system.

Studies have indicated that Ang-(1-7) plays a protective role possibly through the downregulation of mitogen-activated protein kinase (MAPK) phosphorylation in the kidneys. As previously demonstrated, in cultured rat proximal tubular cells, Ang-(1-7) potently inhibits the Ang II-stimulated phosphorylation of ERK1/2, p38 MAPK and JNK (c-Jun N-terminal kinase), an effect that is reversed by pre-treatment with A779, a potent and selective Ang-(1-7) antagonist (14). In renal epithelial LLC-PK cells, high glucose-stimulated protein synthesis and the phosphorylation of p38 MAPK are also inhibited by Ang-(1-7) (15). Although it appears that podocytes express ACE2 and are able to generate Ang-(1-7) in culture (16), to the best of our knowledge, there is currently no information on the effects of Ang-(1-7) on podocyte signalling (17).

In the present study, we hypothesized that serum from pregnant women with PE induces podocyte injury and apoptosis and that this represents an important mechanism to maintain and aggravate proteinuria. In addition, we corrected Ang-(1-7) deficiency in serum from pregnant women with PE in vitro to explore the protective effects of Ang-(1-7) on podocytes under these conditions and to elucidate the underlying mechanisms.

Materials and methods

Study participants

From March 2011 to January 2012, pregnant women with PE (n=20) and gestational age-matched normotensive pregnant women (n=20) were recruited from the Department of Gynecology and Obstetrics, The Fifth People’s Hospital of Shanghai, Fudan University, Shanghai, China. PE was diagnosed by increased blood pressure (≥140/90 mmHg) and proteinuria (300 mg in one 24-h urine collection or >1+ by dipstick in a random urine analysis) after 20 weeks of gestation in a previously normotensive and non-proteinuric woman. Patients with chronic hypertension, pre-existing proteinuria or renal disease were excluded from this study. The patients enrolled in the present study were all newly admitted inpatients and all samples were collected prior to any medication being administered. The biological study was approved by the Ethics Committee of the Fifth People’s Hospital of Shanghai. After obtaining ethical approval and written informed consent from all participants, blood samples were collected from the pregnant women with PE and women with NP. Maternal age, parity, gestational age at delivery, birth weight, blood pressure values, urinary protein excretion, serum creatinine and serum uric acid, serum urea levels and estimated glomerular filtration rate (eGFR) were recorded for each study participant (Table I).

Table I

Clinical characteristics of study participants.

Table I

Clinical characteristics of study participants.

PE (n=20)NP (n=20)
Maternal age, years30.3±5.4a27.2±2.6
Body mass index (BMI), Kg/m227.7±4.326.7±2.3
Blood pressure, mmHg
 Systolic156.8±18.1b119.3±8.4
 Diastolic97.4±11.8a73.7±7.7
Gestational age delivery, weeks36.7±2.5b40.0±1.0
Birth weight of child, g2713±793.5b3526±360.4
Primigravida1412
Urinary protein1+–3+bNegative
Serum creatinine, μmol/l56.9±9.8b45.2±9.6
Serum uric acid, μmol/l361.5±92.0a289.0±82.4
Serum urea, mmol/l4.5±2.0a2.9±1.2
eGFR (MDRD), ml/min/1.73 m2162.3±42.8120.0±27.4

{ label (or @symbol) needed for fn[@id='tfn1-ijmm-34-04-1057'] } eGFR, estimated glomerular filtration rate; MDRD, modification of diet in renal disease. Values are the means ± SD of 20 pregnant women with preeclampsia (PE) and 20 women with normal pregnancies (NP).

a P<0.05,

b P<0.005 compared to women with NP.

Cell culture

The conditionally immortalized human podocytes that were used for the experiments were kindly provided by academician Zhihong Liu (Research Institute of Nephrology of the Jinling Hospital of Nanjing University School of Medicine, Nanjing, China), and the podocyte cell line of Zhihong Liu was provided by Professor Peter Mundel (Department of Medicine and Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, New York, NY, USA). The human podocytes were cultured as previously described (18). The podocytes were propagated and seeded at 33°C in RPMI-1640 medium containing 10% fetal bovine serum (FBS; Gibco, Carlsbad, CA, USA) and 1X insulin, transferrin and selenium solution (ITS) (Invitrogen, Carlsbad, CA, USA). Type I collagen (Invitrogen) is always used to coat the culture dishes to promote podocyte proliferation. Under these permissive conditions, podocytes are small in size, exhibit a polygonal or ‘cobblestone’ appearance and have a relatively small cytoplasmic volume. Subsequently, the cells were incubated at 37.0°C for 10–14 days where they grew to 80% confluence. Under these growth restrictive conditions, the podocytes undergo growth arrest, increase in size, stop replicating, display a more complex arborized pattern of foot process extensions, and express markers of mature podocytic differentiation comparable with filtration slits in vivo.

Cell viability assay

Cell viability was measured using a Cell Counting Kit-8 (CCK-8) (Dojindo Laboratories, Tokyo, Japan). The podocytes were seeded at a concentration of 105/ml in 96-well plates. After adherence, the cells were incubated with serum from pregnant women with PE, or serum from women with NP (1:10 dilution) for different periods of time (6, 12 and 24 h). When the effects of preeclamptic serum and serum from women with NP on podocyte cell viability were investigated, various concentrations of Ang-(1-7) (10−5, 10−6 and 10−7 mol/l) (Sigma-Aldrich, St. Louis, MO, USA) and A779 (10−5, 10−6 and 10−7 mol/l) (Bachem, Bubendorf, Switzerland) were added to the cells for 12 h. At the end of the treatment period, 10 μl of CCK-8 were added to each well, and the cells were further incubated at 37°C for 2 h. The absorbance of CCK-8 was detected at 450 nm using a microplate reader (680 Enzyme-linked Immunosorbent Monitor; Bio-Rad, Tokyo, Japan).

Western blot analysis

The cells were incubated for 12 h with serum derived from pregnant women with PE or serum from women with NP (1:10 dilution). To verify the concentration of Ang-(1-7) required to exert protective effects on podocytes incubated with serum pregnant women with PE, the cells were stimulated with Ang-(1-7) at concentrations of 10−5, 10−6 and 10−7 mol/l. In some experiments, the cells were pre-exposed to A779 (10−5 mol/l, 30 min). Serum from women with NP with A779 [a potent and selective Ang-(1-7) antagonist (10−5 mol/l)] was used as a control. The cells were washed twice with ice-cold PBS and harvested in a lysis buffer containing a protease inhibitor cocktail. Equal amounts of proteins were run on 10% SDS-PAGE and transferred onto PVDF membranes. The membranes were blocked at room temperature with 5% milk in Tris-buffered saline Tween-20 (TBS-T) for 1 h and then probed at 4°C overnight with the following primary antibodies: anti-podocin and anti-Wilms’ Tumor-1 (WT-1) were from Abcam (Burlingame, CA, USA); anti-nephrin antibody was from Sigma-Aldrich; anti-MAS1 antibody was from Novus Biologicals (Oakville, ON, Canada); anti-GAPDH and anti-p-JNK were from Cell Signaling Technology (Beverly, MA, USA); and anti-p-p38 and anti-p-ERK1/2 were purchased from Epitomics (Burlingame, CA, USA). The membranes were washed with TBS-T and then incubated with a peroxidase-conjugated goat anti-rabbit secondary antibody (Cell Signaling Technology) for 1 h at room temperature. Immunoreactivity was detected using an enhanced chemiluminescence detection system. Exposures were recorded on X-ray film.

Immunofluorescence

The cells were incubated with FBS or serum derived from pregnant women with PE, serum from women with NP (1:10 dilution), or serum derived from pregnant women with PE in the presence of Ang-(1-7) (10−6 mol/l), and the podocytes pre-incubated with A779 (10−5 mol/l, 30 min) were stimulated with serum derived from pregnant women with PE in the presence of Ang-(1-7) (10−6 mol/l). The cells were grown on coverslips, washed twice with PBS, fixed in 4% paraformaldehyde for 30 min, permeabilized using 0.1% Triton X-100 for 15 min and incubated in a blocking buffer (5% BSA in PBS, pH 7.4). The cells were then probed with anti-F-actin antibody (1:400)(Abcam) at room temperature for 2 h. The cells were washed with PBS followed by the addition of secondary antibody (Cell Signaling Technology) for 2 h at room temperature. Coverslips were then mounted onto glass microscope slides with DAPI. The cells were observed and photographed using a laser scanning confocal fluorescence microscope (Leica DM5000; Leica Microsystems, Wetzlar, Germany).

Analysis of apoptosis by flow cytometry

Treatment of the podocytes was carried out as described above. The FITC Annexin V Apoptosis Detection kit was purchased from BD Biosciences (Franklin Lakes, NJ, USA). The cells were washed twice with cold PBS and then suspended in 1X binding buffer at a concentration of 1×106 cells/ml. Subsquently, 100 μl of the solution were transferred to a 5-ml culture tube, and 5 μl of Annexin V and 5 μl propidium iodide (PI) were added; the mixture was gently vortex and incubated for 15 min at room temperture (25°C) in the dark. Subsquently, 400 μl of 1X binding buffer were added to each tube. Flow cytometric analysis was performed within 1 h.

Statistical analyses

Values are shown as the the means ± SD. SPSS (version 16.0) software was used to analyze the data using the Student’s t-test or one-way ANOVA or the rank sum test. P-values <0.05 were considered to indicate a statistically significant difference.

Results

Analysis of podocyte viability

To determine the effects of serum from pregnant women with PE and serum from women with NP on podocytes, cell viability was examined by CCK-8 assay. The results revealed that exposure to serum from pregnant women with PE and from women with NP induced a decrease in cell viability in a time-dependent manner (Fig. 1A). At the same time, we did not observe any significant toxic effects of Ang-(1-7) (10−5, 10−6 and 10−7 mol/l) and A779 (10−5, 10−6 and 10−7 mol/l) on podocytes at 12 h (Fig. 1B). Thus, we opted to use serum from pregnant women with PE and from women with NP with 12 h of incubation in the subsequent experiments to determine the underlying mechanisms.

Expression of Mas G protein-coupled receptor in podocytes

As is already known, there is no evidence of the expression of Mas in human podocytes. Thus, western blot analysis was performed to determine whether human podocytes express Mas, the putative receptor for Ang-(1-7). In 3 separate experiments, the results revealed a single band corresponding to the expected 37-kD product for Mas in human podocytes (Fig. 1C).

Effects of Ang-(1-7) on the expression of nephrin, podocin and WT-1 in podocytes treated with preeclamptic serum

The downregulation of podocyte-specific proteins may be involved in the appearance and development of proteinuria in women with PE. Thus, in this study, the expression of nephrin, podocin and WT-1 was assessed in human cultured podocytes treated with serum from pregnant women with PE and from women with NP. As shown in Fig. 2, the expression of nephrin and WT-1 decreased in the podocytes stimulated with preeclamptic serum compared with the cells incubated with serum from women with NP (nephrin, 0.42±0.04-fold vs. 0.64±0.07-fold of GAPDH, P<0.005; WT-1, 0.28±0.05-fold vs. 0.46±0.09-fold of GAPDH, P<0.05). However, the expression of podocin showed no significant difference among the groups. After the podocytes were treated with preeclamptic serum in the presence of Ang-(1-7) (10−5, 10−6 and 10−7 mol/l) for 12 h, the results revealed that Ang-(1-7) exerted the maximum protective effect at a concentration of 10−6 mol/l, and the expression of nephrin and WT-1 was significantly increased (nephrin, 0.57±0.07-fold vs. 0.42±0.04-fold of GAPDH, P<0.05; WT-1, 0.41±0.04-fold vs. 0.28±0.05-fold of GAPDH, P<0.05) compared with the podocytes incubated only with preeclamptic serum (Fig. 2).

A779 reverses the protective effects of Ang-(1-7) on podocyte injury induced by preeclamptic serum

The selective Mas receptor antagonist, A779 [D-Ala7-Ang-(1-7)] (10−5 mol/l), was used to verify the protective role of Ang-(1-7) in another aspect. The results demonstrated that the addition of A779 reversed the protective effects of Ang-(1-7) on podocyte injury induced by preeclamptic serum. The podocytes were pre-treated with A779 for 30 min and then incubated with preeclamptic serum in the presence of Ang-(1-7) (10−6 mol/l) for 12 h. As shown in Fig. 3, after the addition of A779, the expression of nephrin and WT-1 decreased compared with the addition of Ang-(1-7) only (nephrin, 1.55±0.20-fold vs. 2.1±0.19-fold of GAPDH, P<0.01; WT-1, 0.60±0.01-fold vs. 0.89±0.10-fold of GAPDH, P>0.05). Podocytes incubated with serum from NP in the presence of A779 (10−5 mol/l) served as a control and there was no significant difference compared with the podocytes incubated with serum from NP only; this finding suggests that Ang-(1-7) is required to maintain the increased expression of nephrin and WT-1 in PE.

Effects of Ang-(1-7) on the changes in F-actin in podocytes incubated with preeclamptic serum

Cytoskeletal rearrangement is a crucial early event in the pathophysiology of proteinuria and has been reported to result in foot process effacement (19). As shown in Fig. 4, the expression and orientation of F-actin was changed, including cortical F-actin ring formation and stress fibre attenuation in the podocytes that were treated with preeclamptic serum compared with those of the control group. In addition, this change was attenuated by treatment with Ang-(1-7) (10−6 mol/l) and this protective role was reversed by pre-treatment with A779 (Fig 4).

Effects of Ang-(1-7) (10−6 mol/l) on the apoptosis of podocytes induced by preeclamptic serum

To determine the effects of preeclamptic serum on the apoptosis of podocytes, we examined podocyte apoptosis by flow cytometry. The cells were treated with preeclamptic serum in the presence or absence of Ang-(1-7) (10−6 mol/l) and A779 (10−5 mol/l). Cells incubated with serum from women with NP served as a control. As shown in Fig. 5, the number of Annexin V+/PI (apoptotic) cells was significantly increased in the PE group; there were 8.55±0.68% apoptotic cells in the PE group and 2.36±1.05% apoptotic cells in the NP group (P<0.001). As expected, after the addition of Ang-(1-7) (10−6 mol/l) in the PE group [PE plus Ang-(1-7)], the number of apoptotic cells (4.47±0.73%) was significantly decreased compared with the PE group, and this protective role was reversed by pre-treatment with A779 (7.95±0.99%, P<0.001). A779 had no effect on podocyte apoptosis, as shown in the NP plus A779 group (3.59±0.74%); this finding suggests that Ang-(1-7) is required to prevent the apoptosis of podocytes in PE and that the increased apoptosis in the group treated with serum from women with PE plus Ang-(1-7) and A779 did not result from A779 alone.

Effect of Ang-(1-7) on MAPK (p38, ERK 1/2 and JNK) phosphorylation

As illstrated in Fig. 6, the results from western blot analysis showed that the p38, ERK and JNK phosphorylation levels in the cultured podocytes that were stimulated with preeclamptic serum increased compared with those stimulated with serum from NP (p-p38, 1.69±0.20-fold vs. 1.08±0.16-fold of GAPDH, P<0.005; p-ERK, 1.92±0.35-fold vs. 1.04±0.26-fold of GAPDH, P<0.005; p-JNK, 2.35±0.35-fold vs. 1.79±0.30-fold of GAPDH, P<0.05). The addition of Ang-(1-7) to the cells prevented the preeclamptic serum-induced increase in p-p38, ERK and JNK phosphorylation (p-p38, 1.23±0.18-fold vs. 1.69±0.20-fold of GAPDH, P<0.05; p-ERK, 1.14±0.25-fold vs. 0.87±0.18-fold of GAPDH, P<0.01; p-JNK, 1.90±0.10-fold vs. 2.35±0.35-fold of GAPDH, P<0.05) and the effects of Ang-(1-7) were blocked by the Mas receptor inhibitor, A-779.

Discussion

In the present study, we used a conditionally immortalized differentiated human podocyte cell line to explore the damage sustained by glomerular epithelial cells from preeclamptic serum and found that cultured podocytes underwent functional and structural morphologic changes that resulted from the downregulation of podocyte-specific proteins (nephrin and WT-1), cytoskeletal rearrangement (specialized arrangement of F-actin) and apoptosis. In addition, we determined the role of Ang-(1-7) deficiency under these conditions (PE), and the results revealed that Ang-(1-7) attenuated podocyte injury induced by preeclamptic serum through the downregulation of MAPK phosphorylation.

Podocyte injury is usually characterized by the disappearance or effacement of foot processes leading to proteinuria. The foot processes of neighbouring podocytes form an interdigitating pattern with slits that are bridged by a protein complex known as the slit diaphragm (SD) (20). The downregulation of specialized proteins associated with slit-pores (e.g., nephrin and podocin) and podocyte-specific transcriptional factors (e.g., WT-1) may be involved in the initiation and development of proteinuria in pregnant women with PE. Our data indicated that the incubation of podocytes with preeclamptic serum resulted in the decreased expression of nephrin and WT-1 in podocytes and had no effect on podocin expression; these results are consistent with those of other studies (4). The unique shape of podocytes and the maintenance of the processes characterize the well-developed cytoskeleton (20). Cytoskeletal rearrangement has been suggested to underlie foot process effacement, which is a crucial early event in the pathophysiology of proteinuria. The F-actin cytoskeleton in podocyte foot processes is believed to be dynamically maintained at a steady-state with a low turnover (21). In the present study, the expression and orientation of F-actin was altered in the podocytes that were treated with preeclamptic serum compared with the control group (treated with serum from women with NP), which may contribute to podocyte injury in PE. Podocyte depletion has long been shown to be a typical characteristic of glomerulosclerosis and is now considered a key factor in the progression of renal diseases (22). In this study, we found that the apoptosis of podocytes in the PE group was increased compared to the NP group. Our previous study indicated that the number of urinary podocytes in pregnant women with PE was significantly higher than that in women with gestational hypertension without proteinuria and women with NP (23). These results indicate that the progressive depletion of podocytes in PE is associated with an increase in podocyte apoptosis and the detachment from the glomerular basement membrane.

Treatment with Ang-(1-7) has been shown to reduce proteinuria in stroke-prone spontaneously hypertensive rats (SHRSP) (24), and Ang-(1-7) has been shown to have therapeutic potential for reversing glomerulosclerosis by counteracting the effects of Ang II in a rat model of experimental glomerulonephritis (25). The data from the present study indicate that correcting the Ang-(1-7) deficiency in PE can partly reverse the downregulation of podocyte-specific proteins, cytoskeletal rearrangement and the apoptosis of podocytes.

However, the effects of Ang-(1-7) on the kidneys remain controversial. Shao et al (26) found that exogenous Ang-(1-7) injection did not ameliorate diabetic rat renal injury induced by streptozotocin (STZ), and it accelerated progressive diabetic nephropathy. Many factors result in these inconsistent results, such as different experimental designs and different doses of Ang-(1-7). In this study, the doses of Ang-(1-7) used in the experiments ranged from 10−7 to 10−5 mol/l, and we found that Ang-(1-7) exerted a maximum protective effect on podocyte injury in PE at a concentration of 10−6 mol/l.

In the present study, we elucidated the underlying mechanisms of the protective role of Ang-(1-7) in PE. The results revealed that Ang-(1-7) partly inhibited the phosphorylation of p38, ERK1/2 and JNK in cultured podocytes treated with preeclamptic serum, which was reversed by pre-treatment with A779, suggesting an effect mediated by the binding of Ang-(1-7) to the receptor, Mas. In primary cultures of mouse mesangial cells, Moon et al (27) showed that Ang-(1-7) attenuated AngII-induced MAPK phosphorylation, as well as the expression of transforming growth factor (TGF)-β1, fibronectin and collagen IV. However, Zimpelmann and Burns (28) demonstrated that treatment of a human mesangial cell line with Ang-(1-7) caused a rapid and significant increase in arachidonic acid release, as well as the phosphorylation of MAPKs. Overall, Ang-(1-7)/Mas signaling in the kidneys is complex and more detailed studies at the cellular level are required.

In this study, we determined the role of Ang-(1-7) in the pathogenesis of PE and found that Ang-(1-7) reversed podocyte injury in PE through the activation of the podocyte Mas receptor and inhibition of the MAPK pathway. However, a role for correcting Ang-(1-7) deficiency in this condition as a therapeutic strategy requires further investigation.

Acknowledgements

The present study was supported by grants from the Major State Basic Research Development Program of China (973 Program) (no. 2012CB517700), Phase III of the Nephropathy Discipline Construction of 211 Project of State Education Commission, the Shanghai Biopharmaceutics Major Project (08dz1900603), the Fudan University ‘985’ Hospital Key Discipline Construction Project (2012FDYSXK02), the Shanghai Municipal Science and Technology Project (124119a7802), the Shanghai City Health Bureau Project (20114311) and Minhang District Key Disciplines Construction. We thank academician Zhihong Liu for the donation of the podocyte cells. We thank Professor Zhaoxia Wang for providing technical advice and Peng Fang for technical support.

References

1 

Walker JJ: Pre-eclampsia. Lancet. 356:1260–1265. 2000. View Article : Google Scholar : PubMed/NCBI

2 

Jones DC and Hayslett JP: Outcome of pregnancy in women with moderate or severe renal insufficiency. N Engl J Med. 335:226–232. 1996. View Article : Google Scholar : PubMed/NCBI

3 

Levine RJ, Maynard SE, Qian C, et al: Circulating angiogenic factors and the risk of preeclampsia. N Engl J Med. 350:672–683. 2004. View Article : Google Scholar : PubMed/NCBI

4 

Garovic VD, Wagner SJ, Petrovic LM, et al: Glomerular expression of nephrin and synaptopodin, but not podocin, is decreased in kidney sections from women with preeclampsia. Nephrol Dial Transplant. 22:1136–1143. 2007. View Article : Google Scholar : PubMed/NCBI

5 

Herman-Edelstein M, Weinstein T and Gafter U: TGFβ1-dependent podocyte dysfunction. Curr Opin Nephrol Hypertens. 22:93–99. 2013.

6 

Shah DM: Role of the renin-angiotensin system in the pathogenesis of preeclampsia. Am J Physiol Renal Physiol. 288:F614–F625. 2005. View Article : Google Scholar : PubMed/NCBI

7 

Irani RA and Xia Y: Renin angiotensin signaling in normal pregnancy and preeclampsia. Semin Nephrol. 31:47–58. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Anton L and Brosnihan KB: Systemic and uteroplacental renin--angiotensin system in normal and pre-eclamptic pregnancies. Ther Adv Cardiovasc Dis. 2:349–362. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Valdes G, Germain AM, Corthorn J, et al: Urinary vasodilator and vasoconstrictor angiotensins during menstrual cycle, pregnancy, and lactation. Endocrine. 16:117–122. 2001. View Article : Google Scholar : PubMed/NCBI

10 

Santos RA, Ferreira AJ, Pinheiro SV, Sampaio WO, Touyz R and Campagnole-Santos MJ: Angiotensin-(1-7) and its receptor as a potential targets for new cardiovascular drugs. Expert Opin Investig Drugs. 14:1019–1031. 2005. View Article : Google Scholar : PubMed/NCBI

11 

Merrill DC, Karoly M, Chen K, Ferrario CM and Brosnihan KB: Angiotensin-(1-7) in normal and preeclamptic pregnancy. Endocrine. 18:239–245. 2002. View Article : Google Scholar : PubMed/NCBI

12 

Herse F, Staff AC, Hering L, Muller DN, Luft FC and Dechend R: AT1-receptor autoantibodies and uteroplacental RAS in pregnancy and pre-eclampsia. J Mol Med (Berl). 86:697–703. 2008. View Article : Google Scholar : PubMed/NCBI

13 

Abdul-Karim R and Assalin S: Pressor response to angiotonin in pregnant and nonpregnant women. Am J Obstet Gynecol. 82:246–251. 1961.PubMed/NCBI

14 

Su Z, Zimpelmann J and Burns KD: Angiotensin-(1-7) inhibits angiotensin II-stimulated phosphorylation of MAP kinases in proximal tubular cells. Kidney Int. 69:2212–2218. 2006. View Article : Google Scholar : PubMed/NCBI

15 

Gava E, Samad-Zadeh A, Zimpelmann J, et al: Angiotensin-(1-7) activates a tyrosine phosphatase and inhibits glucose-induced signalling in proximal tubular cells. Nephrol Dial Transplant. 24:1766–1773. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Velez JC, Bland AM, Arthur JM, Raymond JR and Janech MG: Characterization of renin-angiotensin system enzyme activities in cultured mouse podocytes. Am J Physiol Renal Physiol. 293:F398–F407. 2007. View Article : Google Scholar : PubMed/NCBI

17 

Zimmerman D and Burns KD: Angiotensin-(1-7) in kidney disease: a review of the controversies. Clin Sci (Lond). 123:333–346. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Saleem MA, O’Hare MJ, Reiser J, et al: A conditionally immortalized human podocyte cell line demonstrating nephrin and podocin expression. J Am Soc Nephrol. 13:630–638. 2002.PubMed/NCBI

19 

Moller CC, Wei C, Altintas MM, et al: Induction of TRPC6 channel in acquired forms of proteinuric kidney disease. J Am Soc Nephrol. 18:29–36. 2007. View Article : Google Scholar : PubMed/NCBI

20 

Pavenstadt H, Kriz W and Kretzler M: Cell biology of the glomerular podocyte. Physiol Rev. 83:253–307. 2003.

21 

Tryggvason K, Pikkarainen T and Patrakka J: Nck links nephrin to actin in kidney podocytes. Cell. 125:221–224. 2006. View Article : Google Scholar : PubMed/NCBI

22 

Kriz W, Gretz N and Lemley KV: Progression of glomerular diseases: is the podocyte the culprit? Kidney Int. 54:687–697. 1998. View Article : Google Scholar : PubMed/NCBI

23 

Chen G, Zhang L, Jin X, et al: Effects of angiogenic factors, antagonists, and podocyte injury on development of proteinuria in preeclampsia. Reprod Sci. 20:579–588. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Giani JF, Munoz MC, Pons RA, et al: Angiotensin-(1-7) reduces proteinuria and diminishes structural damage in renal tissue of stroke-prone spontaneously hypertensive rats. Am J Physiol Renal Physiol. 300:F272–F282. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Benter IF, Yousif MH, Anim JT, Cojocel C and Diz DI: Angiotensin-(1-7) prevents development of severe hypertension and end-organ damage in spontaneously hypertensive rats treated with L-NAME. Am J Physiol Heart Circ Physiol. 290:H684–H691. 2006. View Article : Google Scholar : PubMed/NCBI

26 

Shao Y, He M, Zhou L, Yao T, Huang Y and Lu LM: Chronic angiotensin (1-7) injection accelerates STZ-induced diabetic renal injury. Acta Pharmacol Sin. 29:829–837. 2008. View Article : Google Scholar : PubMed/NCBI

27 

Moon JY, Tanimoto M, Gohda T, et al: Attenuating effect of angiotensin-(1-7) on angiotensin II-mediated NAD(P)H oxidase activation in type 2 diabetic nephropathy of KK-A(y)/Ta mice. Am J Physiol Renal Physiol. 300:F1271–F1282. 2011. View Article : Google Scholar

28 

Zimpelmann J and Burns KD: Angiotensin-(1-7) activates growth-stimulatory pathways in human mesangial cells. Am J Physiol Renal Physiol. 296:F337–F346. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October 2014
Volume 34 Issue 4

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Tian J, Zhang L, Zhou Y, Xiao J, Li S, Chen Y, Qiao Z, Niu J and Gu Y: Angiotensin-(1-7) attenuates damage to podocytes induced by preeclamptic serum through MAPK pathways. Int J Mol Med 34: 1057-1064, 2014
APA
Tian, J., Zhang, L., Zhou, Y., Xiao, J., Li, S., Chen, Y. ... Gu, Y. (2014). Angiotensin-(1-7) attenuates damage to podocytes induced by preeclamptic serum through MAPK pathways. International Journal of Molecular Medicine, 34, 1057-1064. https://doi.org/10.3892/ijmm.2014.1870
MLA
Tian, J., Zhang, L., Zhou, Y., Xiao, J., Li, S., Chen, Y., Qiao, Z., Niu, J., Gu, Y."Angiotensin-(1-7) attenuates damage to podocytes induced by preeclamptic serum through MAPK pathways". International Journal of Molecular Medicine 34.4 (2014): 1057-1064.
Chicago
Tian, J., Zhang, L., Zhou, Y., Xiao, J., Li, S., Chen, Y., Qiao, Z., Niu, J., Gu, Y."Angiotensin-(1-7) attenuates damage to podocytes induced by preeclamptic serum through MAPK pathways". International Journal of Molecular Medicine 34, no. 4 (2014): 1057-1064. https://doi.org/10.3892/ijmm.2014.1870