You are on page 1of 9

Chemico-Biological Interactions 277 (2017) 110e118

Contents lists available at ScienceDirect

Chemico-Biological Interactions
journal homepage: www.elsevier.com/locate/chembioint

Angiotensin II induces calcium-mediated autophagy in podocytes


through enhancing reactive oxygen species levels
Na Gao a, c, 1, Hui Wang b, d, 1, Hongqiang Yin a, Zhuo Yang a, *
a
School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials Ministry of Education, Nankai University,
Tianjin 300071, PR China
b
College of Life Sciences, Nankai University, Tianjin 300071, PR China
c
Tianjin Medical University Cancer Institute and Hospital, Tianjin 300200, PR China
d
School of Mathematical Sciences, Nankai University, Tianjin 300071, PR China

a r t i c l e i n f o a b s t r a c t

Article history: As well known, abnormalities of Angiotensin II (Ang II) is closely related with glomerular damage. This
Received 1 May 2017 study was to investigate whether Ang II could affect autophagy in podocytes via oxidative stress, and
Received in revised form whether autophagy had a positive role in protecting podocytes impaired by Ang II. 3-(4,5-
25 July 2017
dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay showed that Ang II induced podo-
Accepted 11 September 2017
Available online 12 September 2017
cyte death. The measurements of malondialdehyde (MDA) and H2O2 levels, and flow cytometry assay
revealed that Ang II considerably increased reactive oxygen species (ROS) generation in podocytes.
Meaningfully, treatment with ROS scavenger N-(mercaptopropionyl)-glycine (N-MPG) could inhibit
Keywords:
Ang II
podocyte death and attenuate accumulation of ROS induced by Ang II. The patch-clamp experiments
Podocytes indicated that Ang II increased the current of transient receptor potential canonical 6 (TRPC6). Moreover,
TRPC6 measurement of Fluo-3 image showed that Ang II increased intracellular Ca2þ level, as N-MPG and La3þ
Autophagy impeded Ang II induced Ca2þ influx. Acridine orange staining indicated that Ang II induced accumulation
ROS of acidic vacuoles. Beclin-1 and LC3 are essential for autophagosome formation. Furthermore, as one of
the selective substrates for autophagy, P62 plays a key role in the formation of cytoplasmic proteinaceous
inclusion. Western blot assay presented that Ang II obviously elevated LC3-II/LC3-I ratio and expression
of beclin-1, and reduced expression of P62. Meanwhile, N-MPG expectedly down-regulated autophagy in
Ang II-treated podocytes. Rapamycin can enhance the level of autophagy by inhibiting mTOR, and 3-
methyladenine (3-MA) can inhibit autophagosome formation through blocking class III phosphatidyli-
nositol 3-kinase. MTT assay exhibited that rapamycin significantly enhanced the cell viability, while 3-
MA considerably reduced it in Ang II-treated podocytes. Consequently, this study demonstrated that
Ang II could increase TRPC6 induced Ca2þ influx and enhance autophagy through increasing ROS levels in
podocytes, and autophagy could protect Ang II-treated podocytes. Improving TRPC6 channels and
autophagy may become a new targeted therapy to relieve glomerular damage induced by Ang II.
© 2017 Elsevier B.V. All rights reserved.

1. Introduction glomerular endothelial cells compose glomerular filtration bar-


rier. The foot processes of neighboring podocytes form the slit
Podocytes, the glomerular basement membrane, and diaphragm. Through the slit diaphragm, podocytes constitute a
molecular and electric barrier to maintain the function and
structural integrity of glomerular filtration barrier and prevent
Abbreviations: MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bro-
the protein leakage [1,2]. Under pathological conditions, podo-
mide; 3-MA, 3-methyladenine; Ang II, Angiotensin II; MDA, malondialdehyde; N- cytes become the target cells of injury in several conditions of
MPG, N-(mercaptopropionyl)-glycine; ROS, reactive oxygen species; TRPC6, tran- glomerular injury [3]. Because podocytes are terminally differ-
sient receptor potential canonical 6. entiated cells with limited ability of proliferation, podocyte loss
* Corresponding author. School of Medicine, Nankai University, Tianjin 300071,
becomes an important event in glomerulosclerosis and end-stage
PR China. Tel.: þ86 22 23504364; fax: þ86 22 23502554.
E-mail address: zhuoyang@nankai.edu.cn (Z. Yang). kidney disease [4,5]. Transient receptor potential canonical 6
1
Na Gao and Hui Wang have contributed equally to this work. (TRPC6) channels as Ca2þ and Naþ-permeable channels are highly

http://dx.doi.org/10.1016/j.cbi.2017.09.010
0009-2797/© 2017 Elsevier B.V. All rights reserved.
N. Gao et al. / Chemico-Biological Interactions 277 (2017) 110e118 111

expressed in podocytes and play an important role in the patho- 2.2. Podocyte culture
genesis of renal diseases [6e10].
Angiotensin II (Ang II), as an important active substance of Conditionally immortalized mouse podocyte cell lines were
renin-angiotensin system, combined with angiotensin receptor cultured at 33  C in RPMI 1640 medium containing 10% FBS,
plays some biological roles, such as contraction of blood vessels and 100 mg/ml penicillin plus 100 mg/ml streptomycin (Sigma) and 20
increasing blood pressure [11,12]. Within the glomerulus, Ang II U/ml of IFN-g in a humidified atmosphere of 5% CO2. To induce
decreases the ultrafiltration coefficient, modulates glomerular differentiation, podocytes were cultured at 37  C with a IFN-g-free
capillary permselectivity [13], and affects both inflammatory and medium for two weeks. The conditionally immortalized mouse
fibrotic processes [14]. Ang II can increase intracellular calcium podocyte cell line was established by Prof. Peter Mundel [35].
activity, cause a reactive oxygen species (ROS)-mediated F-actin
cytoskeleton rearrangement, and lead to podocyte injury [15,16]. 2.3. Cell viability of podocytes with Ang II and Ang II þ N-MPG
The oxidative stress is an imbalance between ROS and the cell
antioxidant capacity [17]. The increase of oxidative stress usually The cell viability was assessed with MTT assay, which was based
accompanied with the increase of malondialdehyde (MDA) and on the reduction of the dye MTT to formazan crystals, an insoluble
H2O2 levels. MDA, a by-product of lipid peroxidation, is a sensitive intracellular blue product, by cellular dehydrogenases. Briefly, cells
index of oxidative damage. The concentration of MDA reflects the of 1  106 cells/mL were seeded into each well in 96-well culture
proportional to lipid peroxidation and oxidant stress [18]. H2O2 is plates and incubated with different concentrations of Ang II (0,
naturally produced in organisms as a by-product of oxidative 108, 107 and 106 mol/L) for 36 h. Subsequently, 20 ml MTT (5 mg/
metabolism. The increase of oxidative stress usually accompanied ml) was added to each culture well. Following cultured at 37  C for
with the increase of H2O2 level. 4 h, the medium was removed carefully and 150 ml DMSO was
Autophagy plays an important role in maintaining a well- added in each well to dissolve the formazan product. Finally, for-
controlled balance between anabolism and catabolism, which is mazan absorbance was assessed by a microplate reader (Multiskan
necessary to normal cell growth and development [19]. Under Mk3; Thermo Labsystems Helsinki, Finland) with a wave length of
normal conditions, a basal level of autophagy can maintain pro- 492 nm. Meanwhile, the concentrations of Ang II used in assays of
tein quality control and remove damaged proteins, organelles and oxidative stress and autophagy were based on the results of the
lipids, and avoid harming normal cellular function. Autophagy MTT test. The cell viability was expressed as a percentage of the
also plays an essential role during starvation, cellular differenti- viability in control culture. Cell viability (%) ¼ (the viability of Ang
ation, cell death and aging [20]. It permits cells to eliminate un- II-treated group/the control group) x 100.
wanted or unnecessary proteins and organelles, and to reuse the
components [21]. Furthermore, the autophagy-related proteins, 2.4. Measurement of MDA and H2O2 levels
such as LC3 and Beclin-1, are necessary for autophagosome for-
mation in various type of cells [22e24]. As autophagy inhibitors, The MDA and H2O2 levels in podocytes were evaluated by MDA
3-methyladenine (3-MA) can inhibit autophagosome formation and H2O2 assay kit. The podocytes were incubated in 6-well plates
through blocking class III phosphatidylinositol 3-kinase [25]. It for 24 h for stabilization. Then normal cell culture medium, Ang II
has been reported that autophagy has a protective function (107 mol/L) and Ang II (107 mol/L) þ N-MPG (1 mM) were added
against renal damage induced by hypoxia, ischemia, aging and for 36 h, the levels of MDA and H2O2 were measured according to
anticancer drugs [26e32]. While the mechanisms underlying the the manufacturer instructions. The levels of MDA and H2O2 in
development of glomerular injury are greatly complex [33], the control group were defined as 100%, while the levels of MDA and
activation of the intrarenal renin-angiotensin system may be a H2O2 in the rest groups were expressed as a percentage of the MDA
possible mechanism [34]. The purpose of this study was to assess and H2O2 levels in control group.
whether Ang II can affect TRPC6 channels and autophagy in
podocytes via oxidative stress. Meanwhile, we evaluated whether 2.5. Measurement of ROS generation
autophagy had a positive role in protecting podocytes impaired by
Ang II. It may provide an interesting potential clinical application Dichlorodihydrofluorescein diacetate (DCFH-DA) was used to
for chronic glomerular disease. detect the intracellular ROS level. The cells were plated in 6-well
plates. After reaching 60%e70% confluent, they were treated with
normal cell culture medium, Ang II (107 mol/L) and Ang II
2. Materials and methods (107 mol/L) þ N-MPG (1 mM) for 36 h. After incubation, the cells
were washed with PBS (pH 7.4). Following stained with DCFH-DA
2.1. Materials (Genmed Scientifics Inc., Wilmington, DE, USA) for 20 min at
37  C, the cells were detected and analyzed by flow cytometry. The
RPMI 1640 cell culture medium was purchased from GIBCO level of ROS generation was calculated as follows: the level of ROS
Invitrogen. The mouse recombinant interferon-gamma (IFN-g), (%) ¼ the percentage of DCF-positive cells in M1 region.
Rapamycin, 3-MA, fetal bovine serum (FBS), 3-(4, 5-
dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT), 2.6. Whole-cell patch-clamp recording
N-(mercaptopropionyl)-glycine (N-MPG) and Fluo-3/AM fluores-
cent dye were purchased from Sigma Chemical Co., St Louis, MO. The conventional whole-cell patch-clamp recording was carried
The primary antibodies used in the analysis, anti-P62 and anti- out at room temperature (22e24  C). Experiments were performed
Beclin-1ab were purchased from Cell Signaling Technology, USA. in podocytes attached to glass coverslips and transferred to a
Mouse monoclonal anti-LC3 IgG primary antibody was purchased recording chamber on a stage of Olympus inverted microscope
from MBL and rabbit monoclonal anti-b-actin IgG primary antibody immediately before recording. For whole-cell recording TRPC6
was purchased from Santa Cruz Biotechnology, Inc. CA, USA. The channel currents, the bath solution contained (mM): 130 NaCl, 4
reagent kits using for measurement MDA and H2O2 were purchased KCl, 1 MgCl2, 2 CaCl2 and 10 HEPES, pH 7.3. The pipette solution
from Nanking Jiancheng Institute of Biological Engineering Inc. contained (mM): 130 CsOH, 130 L-aspartic acid, 0.3 CaCl2, 2 MgCl2,
(Nanking, China). 10 HEPES, 10 EGTA, 3 ATP-Mg, 0.2 GTP-Na, pH 7.3. The currents
112 N. Gao et al. / Chemico-Biological Interactions 277 (2017) 110e118

were induced by a 150-ms voltage ramp protocol from 100 mV to MA (10 mM), Ang II (107 mol/L), Ang II (107 mol/L) þ rapamycin
100 mV with a holding potential 60 mV. The patch pipettes had a (10 ng/ml), Ang II (107 mol/L) þ 3-MA (10 mM) for 36 h.
resistance ranged from 3 to 6 MU. The cell capacitance ranged from
100 to 250 pF in individual podocytes. 2.11. Statistical analysis

2.7. Ca2þ level detection The current data were acquired using an EPC10 amplifier (HEKA,
Germany), which connected to a computer and stored on a com-
Podocytes were seeded in 6-well plates and treated with normal puter hard disk using pulse 8.52 software (HEKA) analyzed off-line
cell culture medium, Ang II (107 mol/L), Ang II (107 mol/L) þ N- using the pCLAMP 9.0 and Origin 8.0. All data were presented as
MPG (1 mM) and Ang II (107 mol/L) þ La3þ(100 mM) respectively mean ± S.D. The statistical significance was assessed by one-way
for 36 h. The stock solutions of Fluo-3/AM were diluted with HBSS analysis of variance (ANOVA) and Tukey's multiple comparison
that contained (mM): 138 NaCl, 5.3 KCl, 0.3 NaH2PO4, 0.4 KH2PO4, post-test using the SPSS (16.0) software. MTT assay, Western blot
4.2 NaHCO3, 5.6 D-glucose and 10 HEPES. The podocytes were assay, measurement of MDA and H2O2 were repeated thrice; flow
loaded with 5 mM Fluo-3/AM at 37  C for 30 min. Then, the inves- cytometry was repeated 4 times; whole-cell patch-clamp recording
tigated cells were washed by HBSS, and visualized under a confocal was repeated 6 times. In Ca2þ level detection and acridine orange
laser scanning microscope at room temperature (22e24  C). The staining, we randomly selected 20 cells for analysis of fluorescent
calcium-dependent fluorescence was excited at 488 nm laser line intensity. Significant differences were taken when P < 0.05.
and the fluorescence signal was acquired at 522 nm.
3. Results
2.8. Acridine orange staining
3.1. Cell viability assay
Cellular acidic vesicular organelles were examined by acridine
orange staining. Acridine orange, as a fluorescent molecule, can The viability of differentiated podocytes was determined by
interact with DNA emitting green fluorescence and accumulate in MTT assay. Podocytes were treated with medium containing
acidic organelles in which it becomes protonated forming aggre- different concentrations (0, 108, 107, and 106 mol/L) of Ang II for
gates that emit bright red fluorescence [36]. Podocytes were seeded 36 h. As shown in Fig. 1a, the cell viability was decreased when the
in 6-well plates and treated with normal cell culture medium, Ang concentration increased.
II (107 mol/L) and Ang II (107 mol/L) þ N-MPG (1 mM) for 36 h. In order to detect the effect of Ang II induced oxidative stress on
After treatment, the cells were stained with acridine orange at 37  C cell viability, podocytes were treated with Ang II (107 mol/L) and
for 15 min. After washing with PBS, the cells were immediately Ang II (107 mol/L) þ N-MPG (1 mM) for 36 h. It was found that the
visualized by a Leica TCS SP5 laser-scanning confocal microscope viability of podocytes was statistically reduced to 61.56 ± 9.04% in
for detecting acidic vesicular organelles. 107 mol/L Ang II group (P < 0.01, Fig. 1b). Moreover, the cell
viability of Ang II þ N-MPG group was significantly enhanced to
2.9. Western blot analysis 87.46 ± 6.60% (P < 0.05).

Podocytes were treated with normal cell culture medium, Ang II 3.2. Measurement of MDA and H2O2 levels
(107 mol/L) and Ang II (107 mol/L) þ N-MPG (1 mM) for 36 h. The
method of cell lysates and Western blotting was modified on the The effects of N-MPG on the intracellular MDA and H2O2 levels
basis of previous studies [37]. Podocytes were washed with PBS (pH in Ang II-treated podocytes were measured by using the MDA and
7.4), then harvested and lysed in lysis buffer (50 mM Tris-HCl [pH H2O2 test kits (Fig. 1c and d). It was found that the cellular level of
7.4], 150 mM NaCl, 1% Nonidet P-40, 0.1% sodium dodecyl sulphate MDA was remarkably increased in Ang II group (129.69 ± 7.29%,
[SDS], 0.5% deoxycholic acid sodium salt [DOC]) at 0  C. The lysates P < 0.05, Fig. 1c), and it was significantly decreased in Ang II þ N-
were centrifuged at 12000 rpm at 4  C for 10 min. The supernatant MPG group (108.59 ± 3.65%, P < 0.05). Additionally, it could be seen
was mixed with loading buffer (ratio is 4:1) and boiled at 95e100  C that the cellular level of H2O2 was remarkably increased in Ang II
for 20 min. Total proteins were subjected to electrophoresis in a 10 group (250.88 ± 20.69%) compared to that in control group
%e13% SDS-PAGE gel. After separation on polyacrylamide gel, the (P < 0.01, Fig. 1d). Moreover, it was significantly decreased in Ang
proteins were transferred to polyvinylidene fluoride (PVDF) II þ N-MPG group (184.89 ± 8.90%, P < 0.05).
membranes. The PVDF membranes were blocked with 5% zero fat
milk powder in TBS with 0.05% Tween 20 at room temperature for 3.3. Measurement of ROS generation
1 h. Then the PVDF membranes were incubated with primary an-
tibodies. After washing with TBST, the PVDF membranes were DCFH-DA was used to detect the intracellular ROS level. Fig. 1h
incubated with horseradish peroxidase-labeled secondary anti- showed that the ratio of DCF-positive cells was 4.97 ± 0.88% in
bodies. The blots were developed with a chemiluminescence control group. Furthermore, the DCF-positive cell ration in Ang II
detection kit (Pierce) and exposed to X-ray film (Eastman Kodak, group was significantly higher (21.45 ± 2.23%, P < 0.001) than that
Rochester, NY). Equal protein loading and the protein transfer were in control group. In addition, it was much lower in Ang II þ N-MPG
confirmed by immunoblotting for determination of actin protein group (7.65 ± 0.70%, P < 0.001) compared to that in Ang II group.
using b-actin antibody (1:1000 Santa cruz) on the same Western
blot assay. 3.4. Whole-cell recording of TRPC6 currents

2.10. Cell viability of podocytes with autophagy regulators In order to detect the effect of Ang II induced oxidative stress on
TRPC6 currents, Whole-cell patch-clamp technology was used.
The effects of rapamycin and 3-MA, which were known as Whole-cell TRPC6 currents were evoked by 200 ms voltage ramp
autophagy regulators, were detected by the MTT assay. Following protocol from a holding potential of 60 mV. In this series of ex-
procedures of standard method described above, the cells were periments, the patch was depolarized from 100 to 100 mV. Pre-
treated with normal cell culture medium, rapamycin (10 ng/ml), 3- vious studies have shown that these protocols can isolate
N. Gao et al. / Chemico-Biological Interactions 277 (2017) 110e118 113

Fig. 1. N-MPG attenuated Ang II-induced cell death and oxidative stress in podocytes. a The viability of podocytes was determined by MTT assay. Podocytes were treated with
different concentrations of Ang II (0, 108, 107 and 106 mol/L) (n ¼ 3/group). b N-MPG increased the cell viability of podocytes with Ang II. The cell viability of 107 mol/L Ang II
group and Ang II (107 mol/L) þ N-MPG (1 mM) group was determined by MTT assay (n ¼ 3/group). c The effects of N-MPG on the intracellular MDA levels in 107 mol/L Ang II
treated podocytes were measured by using the MDA test kits (n ¼ 3/group). d The effects of N-MPG on the intracellular H2O2 levels in 107 mol/L Ang II treated podocytes were
measured by using H2O2 test kits (n ¼ 3/group); Measurement of ROS generation in podocytes (e-h, n ¼ 4/group). The cells were treated with different culture medium for 36 h. e
control, f Ang II (107 mol/L), g Ang II (107 mol/L) þ N-MPG (1 mM), h Statistical analysis of flow cytometry. Each data represents the mean ± S.D. *P < 0.05, **P < 0.01, ***P < 0.001
vs. control group; #P < 0.05, ###P < 0.001 vs. Ang II group.

macroscopic TRPC6 currents [38]. Ang II increased the amplitude of 3.5. Ca2þ influx detection
TRPC6 currents, and increased the slope of IeV curve (Fig. 2b). Ang
II increased mean currents at all membrane potentials, especially To evaluate the effect of Ang II induced oxidative stress on the
from 20 to 80 mV (P < 0.01). Ca2þ influx, we used Ca2þ-sensitive dye Fluo-3/AM in podocytes.
114 N. Gao et al. / Chemico-Biological Interactions 277 (2017) 110e118

Fig. 2. N-MPG decreased Ang II-induced currents of TRPC6 channel in podocytes. a Examples of TRPC6 currents evoked from a holding potential of 60 mV in differentiated
podocytes. b IeV curves of control group and Ang II (107 mol/L) group (n ¼ 6/group). c Representative microscopic images of intracellular Ca2þ using Fluo 3/AM staining. Scale bar,
50 mm. d Fluorescent intensity of intracellular Ca2þ in podocytes under different conditions (n ¼ 20/group). Each data represents the mean ± S.D. **P < 0.01, ***P < 0.001 vs. control
group; ###P < 0.001 vs. Ang II group.

The fluorescence of Fluo-3/AM in podocytes in these different were P62 (Fig. 3d), beclin-1 (Fig. 3e) and the ratio of LC3 II/LC3 I
groups were shown in Fig. 2c. Changes of cytosolic calcium were (Fig. 3f). It was shown that the ratio of LC3 II/LC3 I and the level of
visualized by confocal laser scanning microscopy. The results beclin-1 were significantly increased in Ang II group compared
showed that Ang II caused elevation of the intracellular Ca2þ con- with those of control group (P < 0.05), and decreased in Ang II þ N-
centration in podocytes. Clearly, N-MPG and La3þ (TRPC6 blocker) MPG group compared with those of Ang II group (P < 0.05).
could significantly decrease Ang II induced Ca2þ influx (Fig. 2d). Meanwhile, it could be seen that the level of P62 was significantly
decreased in Ang II group compared with that of control group
3.6. Acridine orange staining (P < 0.01) and increased in Ang II þ N-MPG group compared with
that of Ang II group (P < 0.01).
Acridine orange was used to stain cellular acidic vesicular or-
ganelles, which reflected the level of autophagy. As shown in 3.8. Cell viability of podocytes with autophagy regulators
Fig. 3a, green fluorescence and a spot of orange fluorescence were
displayed in control group, and in the Ang II-treated podocytes the In order to confirm the protective effect of Ang II induced
orange fluorescence displayed most. However, orange fluorescence autophagy, rapamycin and 3-MA were used in this study. As shown
of Ang II þ N-MPG-treated podocytes was markedly less than Ang II in Fig. 4, the viability of podocytes was statistically reduced to
-treated podocytes. As shown in Fig. 3b, our data provide evidence 63.42 ± 1.79% in Ang II group (P < 0.001). Meanwhile, the cell
that the intensity of red fluorescence increased when the cells were viability was decreased in rapamycin group (86.40 ± 3.73%) and 3-
treated with Ang II than that in control group (P < 0.001), mean- MA group (87.71 ± 2.90%) than that in control group (P < 0.05).
while, the red fluorescent intensity decreased in Ang II þ N-MPG Moreover, the cell viability was further significantly reduced to
group contrast to Ang II group (P < 0.001). In addition, there was no 47.90 ± 5.40% in Ang II þ 3-MA group (P < 0.01), but risen to
statistical difference of green fluorescent intensity among these 74.82 ± 2.04% in Ang II þ rapamycin group (P < 0.01) compare to
three groups. Ang II group.

3.7. Western blot analysis 4. Discussion

The autophagy levels were assessed by measurements of LC3 II Ang II can contribute to the progression of renal injury through
to LC3 I ratio and beclin-1 and P62 levels. The results were obtained its hemodynamic effects [11], and its direct effects on kidney cells
from the experiments in autophagy molecular markers, which [39]. Ang II is known to induce oxidative stress in a variety of renal
N. Gao et al. / Chemico-Biological Interactions 277 (2017) 110e118 115

Fig. 3. N-MPG decreased Ang II-induced autophagy. a Cellular acidic vesicular organelles were examined by acridine orange staining. Podocytes were treated with Ang II
(107 mol/L) and Ang II (107 mol/L) þ N-MPG (1 mM) for 24 h. When stained with acridine orange, DNA emits green fluorescence and acidic vesicular organelles emit red
fluorescence. Scale bar, 50 mm. b Statistical analysis of relative fluorescent intensity (n ¼ 20/group). c immunoblots from single representative experiments of P62, Beclin-1 and LC3.
d P62 band density was measured (n ¼ 3/group). e Beclin-1 band density was measured (n ¼ 3/group). f LC3-II/LC3-I band density ratio was measured (n ¼ 3/group). Each data
represents the mean ± S.D. *P < 0.05, **P < 0.01, ***P < 0.001 vs. control group; #P < 0.05, ##P < 0.01, ###P < 0.001 vs. Ang II group. (For interpretation of the references to colour in
this figure legend, the reader is referred to the web version of this article.)

that Ang II inhibited the viability of podocytes in a concentration-


dependent manner. Our previous study has shown that Ang II can
increase the generation of ROS in podocytes [42]. Meanwhile, the
viability of podocytes treated with ROS scavenger N-MPG was
obviously increased. MTT results showed that when pretreated
with N-MPG, the cell viability increased from 61.56% to 87.46% in
107 mol/L Ang II-treated group. These results showed that N-MPG
could reduce the cell damage by Ang II (p < 0.05). These results
demonstrated that Ang II might induce the death of podocytes by
enhancement of intracellular ROS generation, while N-MPG could
improve the cellular viability and reduce the damage caused by Ang
II. The MDA and H2O2 levels of podocytes cultured with 107 mol/L
Ang II were obviously increased. In contrast to the Ang II group,
MDA and H2O2 levels of the group treated with N-MPG were
decreased from 129.69% to 108.59% (P < 0.05) and from 250.88% to
184.89%, respectively. Furthermore, flow cytometry assay revealed
that Ang II considerably increased ROS generation of podocytes,
and N-MPG alleviated it. The present study indicated that Ang II
Fig. 4. Rapamycin enhanced and 3-MA inhibited the viability in Ang II-treated
podocytes. The viability of podocytes was determined by MTT assay. Podocytes were was able to inhibit podocyte growth and induce apoptosis with the
treated with rapamycin (10 ng/ml), 3-MA (10 mM), Ang II (107 mol/L), Ang II increased ROS levels, while N-MPG decreased ROS levels in Ang II
(107 mol/L) þ rapamycin (10 ng/ml), Ang II (10-7 mol/L) þ 3-MA (10 mM). Each data treated podocytes.
represents the mean ± S.D. n ¼ 3/group. *P < 0.05, ***P < 0.001 vs. control group; Some study has shown that Ang II increases free cytosolic cal-
##
P < 0.01 vs. Ang II group.
cium in podocytes by calcium releasing from intracellular stores
and an infiux from the extracellular space [43]. In this study, the
cells [40]. On the other hand, autophagy, as one of the major cellular currents of TRPC6 channel were measured by whole-cell patch-
mechanisms, has been related to oxidative stress [41]. In our pre- clamp to detect the effects of Ang II in podocytes. TRPC6 is an
sent study, we found that Ang II could increase TRPC6 currents by essential component of the podocyte foot processes and slit dia-
increasing the level of ROS in podocytes. Moreover, TRPC6 medi- phragm and co-localizes with a number of podocyte structural
ating Ca2þ influx induced protective autophagy in podocytes. It is proteins, including nephrin and podocin [44e47]. It was demon-
indicated that regulation of autophagy might be a positive way to strated that mutation of TRPC6 protein might cause dysfunction of
attenuate the Ang II-induced oxidative damage in podocytes. podocytes and lead to focal segmental glomerulosclerosis
In this study, the viabilities of podocytes incubated with [10,48,49]. Results indicated that Ang II increased the current
different concentrations of Ang II were investigated. It was found amplitude and the slope of IeV curve of TRPC6 channel. Some
116 N. Gao et al. / Chemico-Biological Interactions 277 (2017) 110e118

research found that Ang II let Ca2þ influx increase in podocytes and autophagic process [63e65]. The activation of calmodulin (CaM)
led to depolarization [38,50]. Our previous study has shown that activates CaMKKb, followed by AMPK-dependent inhibition of
Ang II can increase the generation of ROS in podocytes [42]. In order mTOR [66]. Moreover, as a fast and potent intracellular Ca2þ buffer,
to detect the effect of ROS on TRPC6 channel, we measured Ca2þ BAPTA-AM could prevent the induction of autophagy, indicating
influx. It was found that ROS scavenger N-MPG inhibited Ang II the importance of cytosolic Ca2þ [67]. In this study, we found that
induced the increase of intracellular Ca2þconcentration, which Ang II induced ROS could regulate TRPC6 channels to mediate Ca2þ
suggested that ROS could induce the opening of TRPC6 channels. influx. Therefore, we suggested that Ang II induced ROS, then
Results of acridine orange staining showed that Ang II induced induced autophagy in podocytes via activating TRPC6 currents.
accumulation of vesicular organelles, which was attenuated by N- However, there is a significant crosstalk between autophagy and
MPG. The morphological alterations that are suggestive of cell ROS. In that regard, several studies reported that there were several
death like blebbing of the plasma membrane could be seen in the signaling pathways, such as Akt/mTOR pathway and p38-NF-kB
cells undergoing autophagic death [51,52]. The data provided evi- pathway, which played a potential role in the autophagy/ROS
dence that the number of acidic vesicular organelles increased signaling [68,69]. Obviously, a further study is needed to investigate
when the cells were treated with Ang II, which meant activation of any clear signaling network between those. Moreover, in this study,
autophagy process, and N-MPG could inhibit the autophagy. we found that Ang II could induce both cell death and protective
Furthermore, Western blot assay was employed to measure the autophagy in podocytes via ROS generation. It will be interesting to
expression of autophagy-related proteins. During autophagy, sol- investigate the underlying mechanisms of Ang II-induced cell
uble form LC3 (LC3 I) is converted to the lipidated form of LC3 (LC3 death, like necrosis, apoptosis.
II), and the LC3 II/LC3 I ratio is increased. It is important to measure
the level of autophagy by detecting LC3 conversion (LC3 I to LC3 II) 5. Conclusion
[53]. In addition, beclin-1 and P62 are important proteins in auto-
phagy process. Researches have shown that beclin-1 regulates each The present study showed that Ang II could induce oxidative
major step in the autophagic pathways, such as autophagosome stress in podocytes. The increase of ROS level activated autophagy
formation and autophagosome maturation, and overexpression of through enhancing TRPC6 mediated Ca2þ influx. Autophagy could
beclin-1 can up-regulate class III phosphoinositide 3-kinase, and protect against Ang II-induced podocytes death. By regulating
finally lead to autophagy activity. P62 can reflect autolysosomal TRPC6 channels and autophagy may become a new way to relieve
lytic activity and autophagy flux, the expression of this protein is Ang II-induced renal injury. It may become a new targeted therapy
decreased when autophagy is promoted [54]. In this study, Western to slow the development of glomerular injury.
blot results indicated that the ratio of LC3-II/LC3-I and expression
level of beclin-1 were enhanced by Ang II, which could confirm that Competing interests
there was an increasing number of autophagosomes. Interestingly,
Ang II enhanced autophagy in podocytes, and N-MPG down- The authors have declared that no competing interest exists.
regulated autophagy in Ang II-treated podocytes.
Autophagy is a homeostatics cytoprotective mechanism, as Acknowledgments
different types of cells have unequal capacity against stress envi-
ronment [55]. However, a massive and uncontrolled autophagy can This work was supported by grant from the National Natural
result in cell death, which is known as autophagic cell death [56]. To Science Foundation of China (81571804, 81771979) and Tianjin
further elucidate the protective function of autophagy, which could Research Program of Application Foundation and Advanced Tech-
be altered by regulating autophagy level, MTT assay was used to nology (14JCZDJC35000).
measure the effects of autophagy regulators on Ang II-induced cell
death. We employed the rapamycin as inducer and 3-MA as in- References
hibitor of autophagy. Rapamycin can enhance the level of auto-
phagy by inhibiting mTOR, which is a negative regulator of [1] K. Asanuma, P. Mundel, The role of podocytes in glomerular pathobiology,
Clin. Exp. Nephrol. 7 (2003) 255e259.
autophagy induction [57]. As autophagy inhibitor, 3-MA can inhibit
[2] H. Pavenstadt, W. Kriz, M. Kretzler, Cell biology of the glomerular podocyte,
autophagosome formation through blocking class III phosphatidy- Physiol. Rev. 83 (2003) 253e307.
linositol 3-kinase [25]. According to the cell viability analysis, it was [3] D. Kerjaschki, Dysfunctions of cell biological mechanisms of visceral epithelial
found that rapamycin rescued cell death from Ang II, however, 3- cell (podocytes) in glomerular diseases, Kidney Int. 45 (1994) 300e313.
[4] R.C. Wiggins, The spectrum of podocytopathies: a unifying view of glomerular
MA aggravated Ang II-induced cell death. The data clearly diseases, Kidney Int. 71 (2007) 1205e1214.
showed that up-regulated autophagy could reduce Ang II-induced [5] W. Kriz, N. Gretz, K.V. Lemley, Progression of glomerular diseases: is the
cell death. It appears that the induction of autophagy may be a podocyte the culprit? Kidney Int. 54 (1998) 687e697.
[6] J. Abramowitz, L. Birnbaumer, Physiology and pathophysiology of canonical
default mechanism to prevent Ang II-induced podocyte injury. transient receptor potential channels, FASEB J. official Publ. Fed. Am. Soc. Exp.
Oxidative stress is an imbalance between ROS production and Biol. 23 (2009) 297e328.
scavenging, resulting in tissue and cell oxidative damage. During [7] K. Kiselyov, R.L. Patterson, The integrative function of TRPC channels, Front.
Biosci. (Landmark Ed.) 14 (2009) 45e58.
oxidative stress, the ROS level is higher than the capacity of the [8] T.E. Woudenberg-Vrenken, R.J. Bindels, J.G. Hoenderop, The role of transient
antioxidant machinery, thus proteins, lipids and DNA are oxidized. receptor potential channels in kidney disease, Nat. Rev. Nephrol. 5 (2009)
Autophagy, as a highly regulated process, is related to long-lived 441e449.
[9] T. Hofmann, A.G. Obukhov, M. Schaefer, C. Harteneck, T. Gudermann,
proteins and distinct organelles [58,59]. Autophagy involves G. Schultz, Direct activation of human TRPC6 and TRPC3 channels by diac-
sequestration of proteins and organelles in autophagosomes, which ylglycerol, Nature 397 (1999) 259e263.
directs sequestered proteins and organelles to lysosomes [60]. It [10] J. Reiser, K.R. Polu, C.C. Moller, P. Kenlan, M.M. Altintas, C.L. Wei, C. Faul,
S. Herbert, I. Villegas, C. Avila-Casado, M. McGee, H. Sugimoto, D. Brown,
was well known that, there is a close relationship between oxida-
R. Kalluri, P. Mundel, P.L. Smith, D.E. Clapham, M.R. Pollak, TRPC6 is a
tive stress and autophagy. Several studies have shown that oxida- glomerular slit diaphragm-associated channel required for normal renal
tive stress plays an important role in the induction of autophagy function, Nat. Genet. 37 (2005) 739e744.
[58,61,62]. However, the mechanism of ROS induced autophagy is [11] S. Yang, B. Yao, Y. Zhou, H. Yin, M.Z. Zhang, R.C. Harris, Intrarenal dopamine
modulates progressive angiotensin II-mediated renal injury, Am. J. physiology.
not well understood. The majority of research indicated that Ren. physiology 302 (2012) F742eF749.
elevated cytosolic Ca2þ concentrations were shown to promote the [12] V.L. King, V.L. English, K. Bharadwaj, L.A. Cassis, Angiotensin II stimulates
N. Gao et al. / Chemico-Biological Interactions 277 (2017) 110e118 117

sympathetic neurotransmission to adipose tissue, Physiol. Rep. 1 (2013). [39] M. Bhaskaran, K. Reddy, N. Radhakrishanan, N. Franki, G. Ding, P.C. Singhal,
[13] L. Sepehr-Ara, S.A. Mohajeri, M. Mahmoudian, Effect of the two new calcium Angiotensin II induces apoptosis in renal proximal tubular cells, American
channel blockers mebudipine and dibudipine on vascular flow of isolated journal of physiology, Ren. Physiol. 284 (2003) F955eF965.
kidney of normal and diabetic rats, Pathophysiol. official J. Int. Soc. Patho- [40] S. Lodha, D. Dani, R. Mehta, M. Bhaskaran, K. Reddy, G.H. Ding, P.C. Singhal,
physiol./ISP 18 (2011) 175e184. Angiotensin II-induced mesangial cell apoptosis: role of oxidative stress, Mol.
[14] M. Ruiz-Ortega, M. Ruperez, V. Esteban, J. Rodriguez-Vita, E. Sanchez-Lopez, Med. 8 (2002) 830e840.
G. Carvajal, J. Egido, Angiotensin II: a key factor in the inflammatory and [41] D. Gozuacik, A. Kimchi, Autophagy and cell death, Curr. Top. Dev. Biol. 78
fibrotic response in kidney diseases, Nephrol. Dial. Transpl. 21 (2006) 16e20. (2007) 217e245.
[15] S.J. Shankland, The podocyte's response to injury: role in proteinuria and [42] N. Gao, H. Wang, X. Zhang, Z. Yang, The inhibitory effect of angiotensin II on
glomerulosclerosis, Kidney Int. 69 (2006) 2131e2147. BKCa channels in podocytes via oxidative stress, Mol. Cell. Biochem. 398
[16] H.H. Hsu, S. Hoffmann, N. Endlich, A. Velic, A. Schwab, T. Weide, E. Schlatter, (2015) 217e222.
H. Pavenstadt, Mechanisms of angiotensin II signaling on cytoskeleton of [43] T.B. Huber, J. Gloy, A. Henger, P. Schollmeyer, R. Greger, P. Mundel,
podocytes, J. Mol. Med. 86 (2008) 1379e1394. H. Pavenstadt, Catecholamines modulate podocyte function, J. Am. Soc.
[17] M. Ott, V. Gogvadze, S. Orrenius, B. Zhivotovsky, Mitochondria, oxidative Nephrol. JASN 9 (1998) 335e345.
stress and cell death, Apoptosis Int. J. Program. Cell death 12 (2007) 913e922. [44] J.Y. Kim, D. Saffen, Activation of M1 muscarinic acetylcholine receptors
[18] X. Xiao, J. Liu, J. Hu, X. Zhu, H. Yang, C. Wang, Y. Zhang, Protective effects of stimulates the formation of a multiprotein complex centered on TRPC6
protopine on hydrogen peroxide-induced oxidative injury of PC12 cells via channels, J. Biol. Chem. 280 (2005) 32035e32047.
Ca(2þ) antagonism and antioxidant mechanisms, Eur. J. Pharmacol. 591 [45] C.C. Moller, J. Flesche, J. Reiser, Sensitizing the slit diaphragm with TRPC6 ion
(2008) 21e27. channels, J. Am. Soc. Nephrol. JASN 20 (2009) 950e953.
[19] D. Glick, S. Barth, K.F. Macleod, Autophagy: cellular and molecular mecha- [46] C.C. Moller, C. Wei, M.M. Altintas, J. Li, A. Greka, T. Ohse, J.W. Pippin,
nisms, J. Pathol. 221 (2010) 3e12. M.P. Rastaldi, S. Wawersik, S. Schiavi, A. Henger, M. Kretzler, S.J. Shankland,
[20] D.J. Klionsky, S.D. Emr, Autophagy as a regulated pathway of cellular degra- J. Reiser, Induction of TRPC6 channel in acquired forms of proteinuric kidney
dation, Science 290 (2000) 1717e1721. disease, J. Am. Soc. Nephrol. JASN 18 (2007) 29e36.
[21] J. Kim, D.J. Klionsky, Autophagy, cytoplasm-to-vacuole targeting pathway, and [47] A. Dietrich, V. Chubanov, T. Gudermann, Renal TRPathies, J. Am. Soc. Nephrol.
pexophagy in yeast and mammalian cells, Annu. Rev. Biochem. 69 (2000) 21 (2010) 736e744.
303e342. [48] M.P. Winn, P.J. Conlon, K.L. Lynn, M.K. Farrington, T. Creazzo, A.F. Hawkins,
[22] Z. Xie, D.J. Klionsky, Autophagosome formation: core machinery and adap- N. Daskalakis, S.Y. Kwan, S. Ebersviller, J.L. Burchette, M.A. Pericak-Vance,
tations, Nat. Cell Biol. 9 (2007) 1102e1109. D.N. Howel, J.M. Vance, P.B. Rosenberg, A mutation in the TRPC6 cation
[23] R. Singh, Y. Xiang, Y. Wang, K. Baikati, A.M. Cuervo, Y.K. Luu, Y. Tang, channel causes familial focal segmental glomerulosclerosis, Science 308
J.E. Pessin, G.J. Schwartz, M.J. Czaja, Autophagy regulates adipose mass and (2005) 1801e1804.
differentiation in mice, J. Clin. Invest. 119 (2009) 3329e3339. [49] S.F. Heeringa, C.C. Moeller, J.Y. Du, L.X. Yue, B. Hinkes, G. Chernin, C.N. Vlangos,
[24] H. Ugland, S. Naderi, A. Brech, P. Collas, H.K. Blomhoff, cAMP induces auto- P.F. Hoyer, J. Reiser, F. Hildebrandt, A novel TRPC6 mutation that causes
phagy via a novel pathway involving ERK, cyclin E and Beclin 1, Autophagy 7 childhood FSGS, PLoS ONE 4 (2009).
(2011) 1199e1211. [50] T. Nijenhuis, A.J. Sloan, J.G. Hoenderop, J. Flesche, H. van Goor, A.D. Kistler,
[25] P.O. Seglen, P.B. Gordon, 3-Methyladenine: specific inhibitor of autophagic/ M. Bakker, R.J. Bindels, R.A. de Boer, C.C. Moller, I. Hamming, G. Navis,
lysosomal protein degradation in isolated rat hepatocytes, Proc. Natl. Acad. J.F. Wetzels, J.H. Berden, J. Reiser, C. Faul, J. van der Vlag, Angiotensin II con-
Sci. U. S. A. 79 (1982) 1889e1892. tributes to podocyte injury by increasing TRPC6 expression via an NFAT-
[26] B. Hartleben, M. Godel, C. Meyer-Schwesinger, S. Liu, T. Ulrich, S. Kobler, mediated positive feedback signaling pathway, Am. J. Pathol. 179 (2011)
T. Wiech, F. Grahammer, S.J. Arnold, M.T. Lindenmeyer, C.D. Cohen, 1719e1732.
H. Pavenstadt, D. Kerjaschki, N. Mizushima, A.S. Shaw, G. Walz, T.B. Huber, [51] S.A. Teter, D.J. Klionsky, Transport of proteins to the yeast vacuole: autophagy,
Autophagy influences glomerular disease susceptibility and maintains podo- cytoplasm-to-vacuole targeting, and role of the vacuole in degradation,
cyte homeostasis in aging mice, J. Clin. Invest. 120 (2010) 1084e1096. Semin. Cell & Dev. Biol. 11 (2000) 173e179.
[27] S. Kume, T. Uzu, K. Horiike, M. Chin-Kanasaki, K. Isshiki, S. Araki, T. Sugimoto, [52] S. Taguwa, H. Kambara, N. Fujita, T. Noda, T. Yoshimori, K. Koike, K. Moriishi,
M. Haneda, A. Kashiwagi, D. Koya, Calorie restriction enhances cell adaptation Y. Matsuura, Dysfunction of autophagy participates in vacuole formation and
to hypoxia through Sirt1-dependent mitochondrial autophagy in mouse aged cell death in cells replicating hepatitis C virus, J. Virol. 85 (2011)
kidney, J. Clin. Invest. 120 (2010) 1043e1055. 13185e13194.
[28] T. Kimura, Y. Takabatake, A. Takahashi, J.Y. Kaimori, I. Matsui, T. Namba, [53] N. Mizushima, T. Yoshimori, How to interpret LC3 immunoblotting, Auto-
H. Kitamura, F. Niimura, T. Matsusaka, T. Soga, H. Rakugi, Y. Isaka, Autophagy phagy 3 (2007) 542e545.
protects the proximal tubule from degeneration and acute ischemic injury, [54] G. Bjorkoy, T. Lamark, A. Brech, H. Outzen, M. Perander, A. Overvatn,
J. Am. Soc. Nephrol. JASN 22 (2011) 902e913. H. Stenmark, T. Johansen, p62/SQSTM1 forms protein aggregates degraded by
[29] M. Jiang, K. Liu, J. Luo, Z. Dong, Autophagy is a renoprotective mechanism autophagy and has a protective effect on huntingtin-induced cell death, J. Cell
during in vitro hypoxia and in vivo ischemia-reperfusion injury, Am. J. Pathol. Biol. 171 (2005) 603e614.
176 (2010) 1181e1192. [55] E. White, R.S. DiPaola, The double-edged sword of autophagy modulation in
[30] A. Takahashi, T. Kimura, Y. Takabatake, T. Namba, J. Kaimori, H. Kitamura, cancer, Clin. Cancer Res. 15 (2009) 5308e5316.
I. Matsui, F. Niimura, T. Matsusaka, N. Fujita, T. Yoshimori, Y. Isaka, H. Rakugi, [56] F. Scarlatti, R. Granata, A.J. Meijer, P. Codogno, Does autophagy have a license
Autophagy guards against cisplatin-induced acute kidney injury, Am. J. Pathol. to kill mammalian cells? Cell Death Differ. 16 (2009) 12e20.
180 (2012) 517e525. [57] T. Noda, Regulation of autophagy through TORC1 and mTORC1, Biomolecules
[31] S. Periyasamy-Thandavan, M. Jiang, Q. Wei, R. Smith, X.M. Yin, Z. Dong, 7 (2017).
Autophagy is cytoprotective during cisplatin injury of renal proximal tubular [58] A.J. Meijer, P. Codogno, Regulation and role of autophagy in mammalian cells,
cells, Kidney Int. 74 (2008) 631e640. Int. J. Biochem. Cell Biol. 36 (2004) 2445e2462.
[32] C. Yang, V. Kaushal, S.V. Shah, G.P. Kaushal, Autophagy is associated with [59] N. Mizushima, Autophagy: process and function, Genes & Dev. 21 (2007)
apoptosis in cisplatin injury to renal tubular epithelial cells, Am. J. physiology. 2861e2873.
Ren. physiology 294 (2008) F777eF787. [60] Y. Ohsumi, N. Mizushima, Two ubiquitin-like conjugation systems essential
[33] M. Brownlee, Biochemistry and molecular cell biology of diabetic complica- for autophagy, Semin. Cell & Dev. Biol. 15 (2004) 231e236.
tions, Nature 414 (2001) 813e820. [61] Y. Al Dhaheri, S. Attoub, G. Ramadan, K. Arafat, K. Bajbouj, N. Karuvantevida,
[34] H. Kobori, M. Kamiyama, L.M. Harrison-Bernard, L.G. Navar, Cardinal role of S. AbuQamar, A. Eid, R. Iratni, Carnosol induces ROS-mediated beclin1-
the intrarenal renin-angiotensin system in the pathogenesis of diabetic ne- independent autophagy and apoptosis in triple negative breast cancer, PLoS
phropathy, J. Invest. Med. 61 (2013) 256e264. ONE 9 (2014) e109630.
[35] C. Faul, K. Asanuma, E. Yanagida-Asanuma, K. Kim, P. Mundel, Actin up: [62] Q. Zhang, Y. Zhang, P. Zhang, Z. Chao, F. Xia, C. Jiang, X. Zhang, Z. Jiang, H. Liu,
regulation of podocyte structure and function by components of the actin Hexokinase II inhibitor, 3-BrPA induced autophagy by stimulating ROS for-
cytoskeleton, Trends Cell Biol. 17 (2007) 428e437. mation in human breast cancer cells, Genes & cancer 5 (2014) 100e112.
[36] S. Paglin, T. Hollister, T. Delohery, N. Hackett, M. McMahill, E. Sphicas, [63] S.E. Crawford, M.K. Estes, Viroporin-mediated calcium-activated autophagy,
D. Domingo, J. Yahalom, A novel response of cancer cells to radiation involves Autophagy 9 (2013) 797e798.
autophagy and formation of acidic vesicles, Cancer Res. 61 (2001) 439e444. [64] M. Shi, H.N. Wang, S.T. Xie, Y. Luo, C.Y. Sun, X.L. Chen, Y.Z. Zhang, Antimi-
[37] H. Wang, N. Gao, Z. Li, Z. Yang, T. Zhang, Autophagy alleviates melamine- crobial peptaibols, novel suppressors of tumor cells, targeted calcium-
induced cell death in PC12 cells via decreasing ROS level, Mol. Neurobiol. mediated apoptosis and autophagy in human hepatocellular carcinoma
53 (2016) 1718e1729. cells, Mol. cancer 9 (2010) 26.
[38] J. Eckel, P.J. Lavin, E.A. Finch, N. Mukerji, J. Burch, R. Gbadegesin, G. Wu, [65] N.R. Brady, A. Hamacher-Brady, H. Yuan, R.A. Gottlieb, The autophagic
B. Bowling, A. Byrd, G. Hall, M. Sparks, Z.S. Zhang, A. Homstad, L. Barisoni, response to nutrient deprivation in the hl-1 cardiac myocyte is modulated by
L. Birbaumer, P. Rosenberg, M.P. Winn, TRPC6 enhances angiotensin II- Bcl-2 and sarco/endoplasmic reticulum calcium stores, FEBS J. 274 (2007)
induced albuminuria, J. Am. Soc. Nephrol. JASN 22 (2011) 526e535. 3184e3197.
118 N. Gao et al. / Chemico-Biological Interactions 277 (2017) 110e118

[66] J.P. Decuypere, G. Bultynck, J.B. Parys, A dual role for Ca(2þ) in autophagy ROS/akt/p70S6K signaling pathway and enhancing hypoxia-inducible Factor-
regulation, Cell calcium 50 (2011) 242e250. 1alpha protein synthesis, PloS one 9 (2014) e112470.
[67] S.H. Wang, Y.L. Shih, W.C. Ko, Y.H. Wei, C.M. Shih, Cadmium-induced auto- [69] H. He, L.H. Zang, Y.S. Feng, L.X. Chen, N. Kang, S. Tashiro, S. Onodera, F. Qiu,
phagy and apoptosis are mediated by a calcium signaling pathway, Cell. Mol. T. Ikejima, Physalin A induces apoptosis via p53-Noxa-mediated ROS gener-
life Sci. CMLS 65 (2008) 3640e3652. ation, and autophagy plays a protective role against apoptosis through p38-
[68] X. Han, S. Sun, M. Zhao, X. Cheng, G. Chen, S. Lin, Y. Guan, X. Yu, Celastrol NF-kappaB survival pathway in A375-S2 cells, J. Ethnopharmacol. 148
stimulates hypoxia-inducible Factor-1 activity in tumor cells by initiating the (2013) 544e555.

You might also like