Epigallocatechin‑3‑gallate protects from high glucose induced podocyte apoptosis via suppressing endoplasmic reticulum stress

  • Authors:
    • Chunhong Xiang
    • Xiaoyan Xiao
    • Bei Jiang
    • Mengkun Zhou
    • Yidan Zhang
    • Hui Li
    • Zhao Hu
  • View Affiliations

  • Published online on: August 29, 2017     https://doi.org/10.3892/mmr.2017.7388
  • Pages: 6142-6147
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Podocytes serve a critical role in the development of many glomerular diseases, including diabetic nephropathy (DN). Epigallocatechin‑3‑gallate (EGCG), a predominant polyphenolic component of green tea, has indicated its therapeutic effects in diabetes. In the present study, mouse podocyte cells were cultured in vitro, cell injury was induced by high glucose, and the protective effect of EGCG on cell proliferation and apoptosis and the underlying mechanisms were investigated. The results demonstrated that high glucose significantly inhibited cell proliferation after 48 and 72 h compared with normal glucose and mannitol treatment. EGCG (20 µmol/l) markedly promoted podocyte proliferation after 24, 48 and 72 h incubation with high glucose. Furthermore, high glucose significantly reduced WT‑1 and nephrin expression in podocytes compared with the normal glucose and mannitol groups, while EGCG (20 µmol/l) treatment largely restored their expression. High glucose also significantly increased the apoptotic cell population compared with normal glucose and mannitol groups. However, EGCG combined with high glucose greatly decreased the apoptotic cell number compared with high glucose treatment alone. Furthermore, high glucose treatment was demonstrated to significantly increase glucose‑regulated protein 78 (GRP78), phosphorylated‑ PKR‑like ER kinase (p‑PERK) and caspase‑12 protein expression levels, which is representative of endoplasmic reticulum (ER) stress, compared with the normal glucose and mannitol groups. However, EGCG treatment significantly attenuated GRP78, p‑PERK and caspase‑12 protein expression induced by high glucose. These findings suggested that EGCG serves a protective role in glucose‑induced podocyte apoptosis via suppressing ER stress, and may provide a novel therapeutic strategy to ameliorate the process of DN.

Introduction

Diabetic nephropathy (DN) is one of most serious complications of both type 1 and type 2 diabetes, and frequently results in end-stage renal disease (ESRD), which eventually requires renal replacement therapy or renal transplantation (1). Initiation and development of DN is closely associated with podocyte injury and loss, and involved mechanisms include podocyte apoptosis, detachment, hypertrophy, effacement and loss of foot process proteins (25). Among them, podocyte apoptosis is a hotspot of research. Increasing data has that revealed endoplasmic reticulum (ER) stress serves an important role in the podocyte apoptosis (6,7). It is understood that ER stress is a kind of adaptive response of the cell itself. Under normal physiological conditions, aggregated protein does not amass in cells partially due to the presence of cellular ‘quality control’ mechanisms (8). However, the unfolded protein response (UPR), whose role is restoring ER homeostasis and normal function, will activate a series of signaling molecules and induce the apoptosis cascade when the ER is exposed in a harmful environment for an extended period of time (9,10). The UPR is characterized by the activation of three ER transmembrane effector proteins: PKR-like ER kinase (PERK), inositol requiring enzyme 1 (IRE1) and activating transcription factor-6 (ATF-6) (11). One major pathway of UPR is the suppression of most protein translations through phosphorylation of eukaryotic translation initiation factor 2 subunit a (eIf2a) by PERK (12). Another pathway is the upregulated expression of ER-localized molecular chaperones, such as glucose-regulated protein 78 (GRP78/Bip), GRP94 and other molecular chaperones like heat shock proteins (13). After a series of target protein activation, activation of ER resident protein caspase-12 will eventually lead to the cellular apoptosis (14).

Epigallocatechin-3-gallate (EGCG), the most abundant catechin in green tea, has been demonstrated to exert its anti-inflammatory, antioxidant and antitumor properties in chronic diseases including cancer (15,16), neurodegenerative disease (17), diabetes (18), heart disease (19) and autoimmune arthritis (20). EGCG can prevent DN progression through decreasing reactive oxygen species (ROS) expression (21). However, the concrete mechanism of EGCG on high glucose-induced podocyte injury remains uncertain. The current study aimed to investigate the effects of EGCG on proliferation and apoptosis of mouse podocytes cultured in high glucose, and to provide a theoretical basis of the mechanisms involved in ER stress.

Materials and methods

Cell culture

The conditionally immortalized mouse podocytes donated by Dr Peter Mundel (Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA) were maintained in RPMI1640 medium supplemented with 10% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA), 10 U/ml interferon-γ (PeproTech, Inc., Rocky Hill, NJ, USA) and 100 U/ml antibiotics (penicillin and streptomycin). Frozen podocytes were firstly cultivated at 33°C in a 5% CO2 incubator. Then cells were cultured in medium at 37°C without interferon-γ to induce differentiation for at least 2 weeks. Cells were cultured in serum-free medium for 24 h to synchronize the cell growth before each experiment.

Cell viability assay

Podocytes were seeded at a density of 1×10^4 cells per well in 100 ul RPMI1640 complete medium on 96-well plates for 24 h. Cells were divided into 10 groups as follows: 5.6 mM D-glucose (group N), 5.5 mM D-glucose and 24.5 mM D-mannitol (group M), 30 mM D-glucose (group H), 1 µM EGCG (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) and 30 mM D-glucose (group E1), 10 µM EGCG and 30 mM D-glucose (group E10), 20 µM EGCG and 30 mM D-glucose (group E20), 40 µM EGCG and 30 mM D-glucose (group E40), 60 µM EGCG and 30 mM D-glucose (group E60), 80 µM EGCG and 30 mM D-glucose (group E80), and 100 µM EGCG and 30 mM D-glucose (group E100). Cells were exposed to different conditions of reagents for 24, 48 or 72 h. At each time point, cells were cultured with fresh medium with 10 µl Cell Counting kit-8 (CCK-8; BD Biosciences, Franklin Lakes, NJ, USA) at 37°C for 3 h. Cell proliferation was measured using a microplate reader (Model 680, Bio-Rad Laboratories, Hercules, CA, USA) at a wavelength of 450 nm. Each individual experiment was repeated at least three times.

Podocyte injury and apoptosis

Cells of 4 groups (N, M, H and E20) in the above conditions were used for the following experiments. Mouse podocytes were inoculated at a density of 1×105 cells/well on cover slides in six-well plates. Then cells were washed with phosphate buffer saline (PBS) and incubated with 0.1% Triton X-100 at room temperature for 5 min. Hoechst 33258 (Sigma-Aldrich; Merck KGaA) was added on the cells and incubated in the dark for 20 min. Images were captured under a fluorescence microscope (Olympus DP72, Olympus Corporation, Tokyo, Japan).

Flow cytometry

Cells were selected for planting in the petri dishes with treatment of normal glucose, mannitol, high glucose or high glucose with 20 µmol/l EGCG. Cells were collected after 72 h and the cell concentration was adjusted to 1×10^6 in 500 µl binding buffer (BD Biosciences). Annexin V-fluorescein isothiocyanate (5 µl) and propidium iodide (PI; 10 µl) were added into each sample and incubated at 37°C for 30 min without bright light. The apoptosis data was acquired by a flow cytometer (Epics XL, Beckman Coulter, Inc., Kreefeld, Germany).

Western blotting

Cells were treated with different conditions and then lysed in radioimmunoprecipitation buffer containing protease inhibitor. After centrifugation at 24,000 × g and 4°C for 30 min, supernatants were collected and analyzed for protein concentration with a Bicinchoninic Acid protein assay kit (Beyotime Institute of Biotechnology, Shanghai, China). Total protein (30–50 µg) was diluted in sample buffer and boiled for 5 min for denaturation. Proteins in each sample were separated by 10% SDS-PAGE and transferred to a nitrocellulose filter membrane. After blocking with 5% milk in PBS with Tween-20 for 1 h, the membrane was incubated at 4°C overnight with the following primary antibodies: Anti-GAPDH (1:1,000; cat. no. ab9485), anti-Nephrin (1:1,000; cat. no. ab136894), anti-Wilms tumor protein 1 (WT-1; 1:1,000; cat. no. ab180840), anti-GRP78 (1:2,000; cat. no. ab21685), anti-caspase-12 (1:2,000; cat. no. ab62484) (all from Abcam, Cambridge, UK), anti-PERK (1:1,000; cat. no. sc-13073) and anti-phosphorylated (p)-PERK (1:1,000; cat. no. sc-32577) (both from Santa Cruz Biotechnology, Inc., Dallas, TX, USA). The membranes were washed and incubated with a goat anti-rabbit horseradish peroxidase-conjugated IgG secondary antibody (1:5,000; cat. no. ZB-2301, Origene Technologies, Inc., Rockville, MD, USA) at 24°C for 1 h. Protein expression was detected with a chemiluminescence detection kit (EMD Millipore, Billerica, MA, USA). Data were analyzed using a western-blot analyzer (FluorChen E ProteinSimple Inc., San Jose, CA, USA).

Statistical analysis

All data are expressed as the mean ± standard deviation. Data were analyzed using SPSS 18.0 software (SPSS Inc., Chicago, IL, USA). Data were analyzed by one-way analysis variance followed by Dunnetts multiple comparison test. P<0.05 was considered to indicate a statistically significant difference.

Results

EGCG promotes podocyte proliferation

Cells were cultured with normal glucose (N), D-mannitol (M) and high glucose (H) for 24, 48 and 72 h, respectively. CCK-8 reagent was used to detect podocyte proliferation. Optical density 450 values were used to represent cell proliferation. D-mannitol was used to observe the influence of hypertonicity on cells. As presented in Fig. 1A, there was no difference in cell proliferation among groups when cells were incubated for 24 h (P>0.05). Cell proliferation in H group was significantly decreased compared with N and M groups when cells were incubated for 48 or 72 h (P<0.05). There were no significant differences between N and M group. Interestingly, when EGCG with different concentrations was added to high glucose treated cells, EGCG especially at a concentration of 20 µmol/l (E20) significantly promoted cell proliferation (P<0.01, Fig. 1B). However, its effect gradually decreased with increasing EGCG concentration (Fig. 1B).

EGCG attenuates high glucose-induced podocyte injury

WT-1 and nephrin are well-known podocyte-specific protein markers and thus are used to evaluate podocyte damage. It was demonstrated that both WT-1 and nephrin protein expression levels were decreased in podocytes stimulated with high glucose compared with those with normal glucose or mannitol (P<0.05, Fig. 2). However, this significantly increased following treatment of EGCG (20 µmol/l) and high glucose, compared with high glucose alone (P<0.05; Fig. 2).

EGCG reduces podocyte apoptosis induced by high glucose

Cells under different conditions were cultured for 72 h and cell apoptosis was analyzed by flow cytometry. There was little population of apoptotic cells in either the N (0.36%) and M (0.42%) groups, but the apoptotic population significantly increased in the high glucose group (7.1%) compared with the normal and mannitol groups (P<0.05, Fig. 3A). As predicted, EGCG (20 µmol/l) treatment in high glucose incubated cells significantly reduced the apoptotic cell population percentage (3.24%; P<0.05; Fig. 3A and B).

In order to identify apoptotic cells, Hoechst 33258 was used for staining cell nuclear to detect cell apoptosis. In the H group, there were significantly more typical apoptotic cells, in which the nuclear shape became petals or nucleolus pyknosised, compared with the N and M groups (Fig. 3C). EGCG (20 µmol/l) greatly decreased the number of apoptotic cells treated with high glucose (P<0.05; Fig. 3D).

EGCG attenuates GRP78, p-PERK and caspase-12 expression after high glucose stimulation

To investigate whether EGCG protects against high glucose-induced podocyte injury via ER stress signaling, the expression of ER stress-associated markers, GRP78, p-PERK and caspase-12, were assessed. After high glucose stimulation, GRP78, p-PERK and caspase-12 were significantly increased compared with the normal glucose and mannitol groups (P<0.05; Fig. 4). However, these protein expression levels were markedly attenuated after EGCG (20 µmol/l) treatment (P<0.05; Fig. 4).

Discussion

Podocytes are a type of highly differentiated and non-renewable glomerular epithelial cell (22). It is generally accepted that the loss and apoptosis of podocytes which occur at an early stage of DN are closely associated with its progression, and is a main cause of proteinuria and glomerulosclerosis development (23). Previous studies have demonstrated that EGCG treatment can attenuate ischemia/reperfusion-induced renal dysfunction though inhibition of proinflammatory cytokine cell apoptosis (24), and protect against cisplatin-induced nephrotoxicity via suppression of ER stress-induced (25) and mitochondrial dependent apoptotic pathways (26). The results of the present study demonstrated that EGCG is able to promote high-glucose induced podocyte proliferation and can also suppress high-glucose induced cell apoptosis of podocytes.

The ER serves an important role in adaptation of cell alterations under different conditions. It has been demonstrated that numerous factors, including free oxygen, immoderate nutrients, high glucose and free fatty acids, all can initiate apoptosis and generate ER stress and tissue damage (27). Podocyte loss in DN may be associated with ER stress-induced apoptosis (28,29). High glucose can lead to cell injury and trigger ER stress in podocytes (28). GRP78, the first response protein and one of the main modulators of the UPR, has been generally implicated as a marker for the initiation of ER stress (30,31). GRP78 combines with the N-termini of transmembrane ER proteins to prevent protein aggregation under normal conditions. When unfolded proteins amass, GRP78 is released and the vital transmembrane ER signaling proteins, including PERK, IRE1 and ATF6, are triggered to launch ER stress (31,32). PERK cleaves from GRP78, inducing its autophosphorylation through oligomerization to further phosphorylate the α subunit of translation initiation factor 2 (eIF2α) to trigger ER stress (27). Thus, GRP78 serves a fundamental role in identification of unfolded proteins. Caspase-12, unlike other caspases, is specifically located in the cytoplasm of the ER and can be activated during ER stress but not through mitochondrial signals and other death stimuli (14). Upon caspase-12 activation, it can directly enter cytosol and process with downstream caspase family mainly including caspase-3, which leads to cell apoptosis (31). GRP78 is an inductor of ER stress and caspase-12 is an executor of ER stress-induced apoptosis. The current study indicates that ER stress is activated by high glucose, and protein expression of GRP78, p-PERK and caspase-12 are upregulated. EGCG treatment can reduce high glucose-induced ER stress. All the results suggest that EGCG may protect from podocyte apoptosis via suppression of ER stress pathway. A schematic representation of EGCG protection against podocyte apoptosis from high glucose stimulation, via ER stress signaling, is presented in Fig. 5.

In conclusion, the present study demonstrated that EGCG promotes cell proliferation and apoptosis of high glucose-induced podocytes. Notably, EGCG was observed to suppress high glucose-induced ER stress-induced upregulation of GRP78, p-PERK and caspase-12 protein expression levels. Further study is required to clarify the efficacy of EGCG in the treatment of disease in vivo; however, the results of the present study indicated that it may have potential for the development of a therapeutic drug for DN.

Acknowledgements

The present study was supported by a General Financial Grant from the China Postdoctoral Science Foundation (grant no. 2015M572048), the Natural Science Fund of Shandong Province (grant no. ZR2013HM100 and ZR2014HM037) and the Science & Technology Development Program of Shandong Province (grant no. 201401241).

References

1 

Ilatovskaya DV, Levchenko V, Lowing A, Shuyskiy LS, Palygin O and Staruschenko A: Podocyte injury in diabetic nephropathy: Implications of angiotensin II-dependent activation of TRPC channels. Sci Rep. 5:176372015. View Article : Google Scholar : PubMed/NCBI

2 

Khazim K, Gorin Y, Cavaglieri RC, Abboud HE and Fanti P: The antioxidant silybin prevents high glucose-induced oxidative stress and podocyte injury in vitro and in vivo. Am J Physiol Renal Physiol. 305:F691–F700. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Ma J, Wu H, Zhao CY, Panchapakesan U, Pollock C and Chadban SJ: Requirement for TLR2 in the development of albuminuria, inflammation and fibrosis in experimental diabetic nephropathy. Int J Clin Exp Pathol. 7:481–495. 2014.PubMed/NCBI

4 

Liu BC, Song X, Lu XY, Li DT, Eaton DC, Shen BZ, Li XQ and Ma HP: High glucose induces podocyte apoptosis by stimulating TRPC6 via elevation of reactive oxygen species. Biochim Biophys Acta. 1833:1434–1442. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Brunskill EW, Georgas K, Rumballe B, Little MH and Potter SS: Defining the molecular character of the developing and adult kidney podocyte. PLoS one. 6:e246402011. View Article : Google Scholar : PubMed/NCBI

6 

Cao AL, Wang L, Chen X, Wang YM, Guo HJ, Chu S, Liu C, Zhang XM and Peng W: Ursodeoxycholic acid and 4-phenylbutyrate prevent endoplasmic reticulum stress-induced podocyte apoptosis in diabetic nephropathy. Lab Invest. 96:610–622. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Madhusudhan T, Wang H, Dong W, Ghosh S, Bock F, Thangapandi VR, Ranjan S, Wolter J, Kohli S, Shahzad K, et al: Defective podocyte insulin signalling through p85-XBP1 promotes ATF6-dependent maladaptive ER-stress response in diabetic nephropathy. Nat Commun. 6:64962015. View Article : Google Scholar : PubMed/NCBI

8 

Brown MK and Naidoo N: The endoplasmic reticulum stress response in aging and age-related diseases. Front Physiol. 3:2632012. View Article : Google Scholar : PubMed/NCBI

9 

Ron D and Walter P: Signal integration in the endoplasmic reticulum unfolded protein response. Nat Rev Mol Cell Biol. 8:519–529. 2007. View Article : Google Scholar : PubMed/NCBI

10 

Luo B and Lee AS: The critical roles of endoplasmic reticulum chaperones and unfolded protein response in tumorigenesis and anticancer therapies. Oncogene. 32:805–818. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Okada T, Yoshida H, Akazawa R, Negishi M and Mori K: Distinct roles of activating transcription factor 6 (ATF6) and double-stranded RNA-activated protein kinase-like endoplasmic reticulum kinase (PERK) in transcription during the mammalian unfolded protein response. Biochem J. 366:585–594. 2002. View Article : Google Scholar : PubMed/NCBI

12 

Koumenis C, Naczki C, Koritzinsky M, Rastani S, Diehl A, Sonenberg N, Koromilas A and Wouters BG: Regulation of protein synthesis by hypoxia via activation of the endoplasmic reticulum kinase PERK and phosphorylation of the translation initiation factor eIF2alpha. Mol Cell Biol. 22:7405–7416. 2002. View Article : Google Scholar : PubMed/NCBI

13 

Harding HP, Calfon M, Urano F, Novoa I and Ron D: Transcriptional and translational control in the Mammalian unfolded protein response. Annu Rev Cell Dev Biol. 18:575–599. 2002. View Article : Google Scholar : PubMed/NCBI

14 

Nakagawa T, Zhu H, Morishima N, Li E, Xu J, Yankner BA and Yuan J: Caspase-12 mediates endoplasmic-reticulum-specific apoptosis and cytotoxicity by amyloid-beta. Nature. 403:98–103. 2000. View Article : Google Scholar : PubMed/NCBI

15 

Nowakowska A and Tarasiuk J: Comparative effects of selected plant polyphenols, gallic acid and epigallocatechin gallate, on matrix metalloproteinases activity in multidrug resistant MCF7/DOX breast cancer cells. Acta Biochim Pol. 63:571–577. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Granja A, Pinheiro M and Reis S: Epigallocatechin gallate nanodelivery systems for cancer therapy. Nutrients. 8(pii): E3072016. View Article : Google Scholar : PubMed/NCBI

17 

Ortiz-López L, Márquez-Valadez B, Gómez-Sánchez A, Silva-Lucero MD, Torres-Pérez M, Téllez-Ballesteros RI, Ichwan M, Meraz-Ríos MA, Kempermann G and Ramírez-Rodríguez GB: Green tea compound epigallo-catechin-3-gallate (EGCG) increases neuronal survival in adult hippocampal neurogenesis in vivo and in vitro. Neuroscience. 322:208–220. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Huang SM, Chang YH, Chao YC, Lin JA, Wu CH, Lai CY, Chan KC, Tseng ST and Yen GC: EGCG-rich green tea extract stimulates sRAGE secretion to inhibit S100A12-RAGE axis through ADAM10-mediated ectodomain shedding of extracellular RAGE in type 2 diabetes. Mol Nutr Food Res. 57:2264–2268. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Kim SJ, Li M, Jeong CW, Bae HB, Kwak SH, Lee SH, Lee HJ, Heo BH, Yook KB and Yoo KY: Epigallocatechin-3-gallate, a green tea catechin, protects the heart against regional ischemia-reperfusion injuries through activation of RISK survival pathways in rats. Arch Pharm Res. 37:1079–1085. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Yang EJ, Lee J, Lee SY, Kim EK, Moon YM, Jung YO, Park SH and Cho ML: EGCG attenuates autoimmune arthritis by inhibition of STAT3 and HIF-1alpha with Th17/Treg control. PLoS One. 9:e860622014. View Article : Google Scholar : PubMed/NCBI

21 

Leu JG, Lin CY, Jian JH, Shih CY and Liang YJ: Epigallocatechin-3-gallate combined with alpha lipoic acid attenuates high glucose-induced receptor for advanced glycation end products (RAGE) expression in human embryonic kidney cells. An Acad Bras Cienc. 85:745–752. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Mundel P and Shankland SJ: Podocyte biology and response to injury. J Am Soc Nephrol. 13:3005–3015. 2002. View Article : Google Scholar : PubMed/NCBI

23 

Butt A and Riaz S: Study of protein profiling of human urine in diabetic hypertensive nephropathy versus normal healthy controls. Diabetes Technol Ther. 12:379–386. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Lv J, Feng M, Zhang L, Wan X, Zeng YC, Liang PF and Xu AP: Protective effect of epigallocatechin gallate, a major constituent of green tea, against renal ischemia-reperfusion injury in rats. Int Urol Nephrol. 47:1429–1435. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Chen B, Liu G, Zou P, Li X, Hao Q, Jiang B, Yang X and Hu Z: Epigallocatechin-3-gallate protects against cisplatin-induced nephrotoxicity by inhibiting endoplasmic reticulum stress-induced apoptosis. Exp Biol Med (Maywood). 240:1513–1519. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Zou P, Song J, Jiang B, Pei F, Chen B, Yang X, Liu G and Hu Z: Epigallocatechin-3-gallate protects against cisplatin nephrotoxicity by inhibiting the apoptosis in mouse. Int J Clin Exp Pathol. 7:4607–4616. 2014.PubMed/NCBI

27 

Ozcan U, Cao Q, Yilmaz E, Lee AH, Iwakoshi NN, Ozdelen E, Tuncman G, Görgün C, Glimcher LH and Hotamisligil GS: Endoplasmic reticulum stress links obesity, insulin action and type 2 diabetes. Science. 306:457–461. 2004. View Article : Google Scholar : PubMed/NCBI

28 

Cao Y, Hao Y, Li H, Liu Q, Gao F, Liu W and Duan H: Role of endoplasmic reticulum stress in apoptosis of differentiated mouse podocytes induced by high glucose. Int J Mol Med. 33:809–816. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Sun XY, Qin HJ, Zhang Z, Xu Y, Yang XC, Zhao DM, Li XN and Sun LK: Valproate attenuates diabetic nephropathy through inhibition of endoplasmic reticulum stressinduced apoptosis. Mol Med Rep. 13:661–668. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Groenendyk J, Sreenivasaiah PK, Kim DH, Agellon LB and Michalak M: Biology of endoplasmic reticulum stress in the heart. Circ Res. 107:1185–1197. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Xu C, Bailly-Maitre B and Reed JC: Endoplasmic reticulum stress: Cell life and death decisions. J Clin Invest. 115:2656–2664. 2005. View Article : Google Scholar : PubMed/NCBI

32 

Schroder M and Kaufman RJ: ER stress and the unfolded protein response. Mutat Res. 569:29–63. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2017
Volume 16 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Xiang C, Xiao X, Jiang B, Zhou M, Zhang Y, Li H and Hu Z: Epigallocatechin‑3‑gallate protects from high glucose induced podocyte apoptosis via suppressing endoplasmic reticulum stress. Mol Med Rep 16: 6142-6147, 2017
APA
Xiang, C., Xiao, X., Jiang, B., Zhou, M., Zhang, Y., Li, H., & Hu, Z. (2017). Epigallocatechin‑3‑gallate protects from high glucose induced podocyte apoptosis via suppressing endoplasmic reticulum stress. Molecular Medicine Reports, 16, 6142-6147. https://doi.org/10.3892/mmr.2017.7388
MLA
Xiang, C., Xiao, X., Jiang, B., Zhou, M., Zhang, Y., Li, H., Hu, Z."Epigallocatechin‑3‑gallate protects from high glucose induced podocyte apoptosis via suppressing endoplasmic reticulum stress". Molecular Medicine Reports 16.5 (2017): 6142-6147.
Chicago
Xiang, C., Xiao, X., Jiang, B., Zhou, M., Zhang, Y., Li, H., Hu, Z."Epigallocatechin‑3‑gallate protects from high glucose induced podocyte apoptosis via suppressing endoplasmic reticulum stress". Molecular Medicine Reports 16, no. 5 (2017): 6142-6147. https://doi.org/10.3892/mmr.2017.7388