Open Access

Modified Huangqi Chifeng decoction inhibits excessive autophagy to protect against Doxorubicin‑induced nephrotic syndrome in rats via the PI3K/mTOR signaling pathway

  • Authors:
    • Zi‑Kai Yu
    • Bin Yang
    • Yu Zhang
    • Liu‑Sheng Li
    • Jin‑Ning Zhao
    • Wei Hao
  • View Affiliations

  • Published online on: July 20, 2018     https://doi.org/10.3892/etm.2018.6492
  • Pages: 2490-2498
  • Copyright: © Yu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The aim of the present study was to investigate whether modified Huangqi Chifeng decoction (MHCD) could be an effective treatment against Doxorubicin‑induced nephrosis in rats and whether it regulates autophagy via the phosphoinositide‑3 kinase/mammalian target of rapamycin (PI3K/mTOR) signaling pathway. A total of 40 male Sprague‑Dawley rats were randomly divided into blank, model, telmisartan and MHCD groups. The rat model of nephrosis was induced by intragastric administration of Doxorubicin for 8 weeks. Rats were housed in metabolic cages and urine was collected once every 2 weeks to measure 24‑h protein levels. Blood samples were obtained from the abdominal aorta and levels of albumin (ALB), total cholesterol (TCH), triacylglyceride (TG) and serum creatinine (Scr) were assessed. Renal pathological changes were examined using hematoxylin‑eosin, Masson's trichome and periodic acid‑Schiff staining. Podocytes and autophagosomes were observed using an electron microscope. The expression and distribution of microtubule‑associated proteins 1A/1B light chain 3B (LC3), LC3‑I, LC3‑II, beclin‑1, PI3K and mTOR were determined using immunohistochemistry and western blotting. At weeks 6 and 8, 24‑h proteinuria significantly decreased in the MHCD group compared with the model group (P<0.05). Compared with the model group, the MHCD group exhibited significantly reduced levels of TG, TCH and Scr, as well as significantly increased ALB levels (P<0.05). MHCD was demonstrated to prevent glomerular and podocyte injury. The number of autophagosomes was significantly decreased and the expression of beclin‑1, LC3, LC3‑I and LC3‑II was inhibited following MHCD treatment compared with the model group (P<0.05). MHCD treatment significantly increased the expression of PI3K and mTOR in Doxorubicin nephrotic rats compared with the model group (P<0.05). In conclusion, MHCD was demonstrated to ameliorate proteinuria and protect against glomerular and podocyte injury by inhibiting excessive autophagy via the PI3K/mTOR signaling pathway.

Introduction

Chronic kidney disease (CKD) is defined as kidney damage or a glomerular filtration rate <60 ml/min/1.73 m2 for ≥3 months, irrespective of the cause (1). A total of ~500 million adults are diagnosed with CKD all over the world, whichhas become a global public health problem due to its high prevalence and the accompanying risk of end-stage renal disease (2). The Global Burden of Disease Study 2013 reported that, over the past 23 years, CKD is the most increased non-communicable cause of mortality (3). Angiotensin converting enzyme inhibitors and angiotensin receptor blockers have been used to treat CKD; however, these agents do not completely prevent the progression of CKD (4). In China, traditional Chinese medicine (TCM) has been widely used to treat CKD (5,6). Modified Huangqi Chifeng decoction (MHCD) has previously been reported to be an effective treatment for CKD (7). However, the underlying mechanisms of MHCD in CKD remain to be elucidated.

The role of autophagy in CKD has previously been studied and it was reported that kidney cells differentiate by acquiring specialized membranous components (8). However, the phenomenon of autophagy in the kidney was not clearly defined at that time. Over the past few decades, autophagy in CKD has attracted increased attention. Several studies have demonstrated that autophagy is a major protective mechanism against podocyte aging and glomerular injury in rat kidney cells, suggesting that autophagy serves a role in ameliorating human glomerular disease and aging-associated loss of renal function (913). It is therefore important to elucidate the relevant molecular mechanisms associated with autophagy in CKD.

A previous study by our group demonstrated that MHCD attenuated renal fibrosis by inhibiting excessive autophagy and upregulating basal autophagy in rats with Doxorubicin-induced nephropathy (14), which is an experimental model of progressive kidney disease (15,16). In the present study, Doxorubicin-induced nephrotic rats with proteinuria were used as an experimental model. The aim of the present study was to investigate whether MHCD was able to regulate autophagy in rats to minimize podocyte and glomerular injuries via the phosphoinositide-3 kinase/mammalian target of rapamycin (PI3K/mTOR) signaling pathway.

Materials and methods

Drugs and antibodies

MHCD, comprised of 30 g Sheng Huang-qi (Astragalus membranaceus Bge), 20 g Qian Shi (Euryale ferox Salish), 20 g Jin Ying-zi (Rosae Laevigatae Michx), 10 g Chi Shao (Paeonia lactiflora Pall), 10 g Fang Feng (Saposhnikovia divaricata Schischk), 10 g Di Long (Pheretima vulgaris Chen) and 10 g Bai Hua-she-she-cao (Hedyotis diffusa Wild), was provided by the Pharmacy Department of Xiyuan Hospital of China Academy of Chinese Medical Sciences. The herbs were initially soaked in water for 1 h at room temperature, followed by 1 h of decoction at 100°C. The extracts harvested from the decoction were vacuum dried. Water-soluble extracts of MHCD were dissolved in double-distilled water (ddH2O). Telmisartan (Micardis; 80 mg/pill) was purchased from Boehringer Ingelheim International GmbH (Ingelheim am Rhein, Germany). Doxorubicin hydrochloride for injection (instant; also known as Doxorubicin) was purchased from Pfizer Italia Srl (Rome, Italy). Anti-beclin1 (1:1,000; cat. no. ab210498), anti-LC3 (1:1,000; cat. no. ab48394), anti-PI3K (1:1,000; cat. no. ab86714) and anti-mTOR (1:1,000; cat. no. ab2732) primary antibodies were purchased from Abcam (Cambridge, MA, USA). The secondary antibodies used were part of a general-purpose two-step immunohistochemical kit (cat. no. PV-6000; ZSGB Biological Technology; OriGene Technologies, Inc., Rockville, MD, USA). The DAB kit was purchased from ZSGB Biological Technology (OriGene Technologies, Inc.).

Animal grouping and treatment

A total of 40 male Sprague-Dawley rats (weight, 220±18 g; age, 2–3 months) were purchased from Beijing HFK Bioscience Co., Ltd. (Beijing, China). The rats were housed in humidity-controlled rooms (60±10%) at 24±1°C with a 12-h light/dark cycle and free access to standard food and tap water. All rats were housed in metabolic cages and acclimated to laboratory conditions for 7 days, following which they were randomly divided into either the blank (n=10) or Doxorubicin-induced nephrosis (n=30) group. The Doxorubicin-induced nephrosis group was treated once with Doxorubicin at a dose of 6.2 mg/kg, which was injected into a tail vein, whereas the blank group was treated once with normal saline by intravenous injection into a tail vein. The Doxorubicin-induced nephrosis group was further divided into the model (n=10), telmisartan (n=10) and MHCD (n=10) groups. After 2 weeks, rats in the blank and model groups received normal saline, (0.1 ml/10 g) the telmisartan group received telmisartan (8.33 mg/kg) and the MHCD group received MHCD (11.46 g/kg) by intragastric administration once a day for 6 weeks. All drugs were diluted with distilled water and the dosages were evaluated by body surface coefficient conversion between humans and rats. Urine was collected from rats in the metabolic cages to determine the 24-h protein level. Urine collection was performed once every 2 weeks. Blood was obtained from the abdominal aorta following intragastric administration at week 8 to determine albumin (ALB), total cholesterol (TCH), triacylglyceride (TG) and serum creatinine (Scr) levels using a Roche Cobas-800 automatic biochemical analyzer (Roche Diagnositics GmbH, Mannheim, Germany). The rats were then sacrificed and the kidneys were harvested. The Animal Care and Use Committee of Xiyuan Hospital of China Academy of Chinese Medical Sciences approved the experimental protocol.

Histopathological analysis

Sections of cortical tissues from the right kidney were fixed in 10% buffered formalin at room temperature for 48 h, embedded in paraffin and sliced to 3 µm thick sections. The sections were stained at room temperature with hematoxylin-eosin (HE) for 3 min, Masson's trichome for 30 sec-5 min and periodic acid-Schiff (PAS) for 30 sec-5 min, following which they were visualized using light microscopy (magnification, ×400). Other sections of the right kidneys were fixed in 2.5% glutaraldehyde at 4°C for 24 h, washed with phosphate-buffered saline and sliced to 70–90 nm. Following staining with osmium tetroxide and lead citrate at room temperature for 5–10 min, the ultrastructure of the kidneys was observed under a Hitachi H-600 transmission electron microscope (magnification, ×8,000 and ×20,500; Hitachi, Ltd., Tokyo, Japan).

Immunohistochemistry

To evaluate the expression and distribution of microtubule-associated proteins 1A/1B light chain 3B (LC3), beclin-1, mTOR and PI3K, the paraffin-embedded renal cortical sections were dewaxed with xylene and rehydrated with a descending alcohol series. Sections were incubated with 3% hydrogen peroxide (H2O2) and washed with PBS, following which they were incubated with primary antibodies (1:200) at 4°C overnight and washed with PBS. Next, the sections were incubated with the secondary antibodies (1:2,500) and washed with PBS. Sections were subsequently stained with DAB at room temperature for 10 sec-2 min, washed with PBS, dehydrated with a descending alcohol series, permeabilized with xylene, mounted and viewed using light microscopy (magnification, ×400). The expression of proteins was demonstrated by the ratio of integral optical density (IOD). IOD=average optical density × positive area.

Western blotting

To evaluate the expression and distribution of LC3-I, LC3-II, beclin-1, mTOR and PI3K protein, total protein was extracted using radioimmunoprecipitation assay buffer, following which the protein concentration was determined using a BCA Protein assay kit (cat. no. 23225; Bio-Rad Laboratories Inc., Hercules, CA, USA). Proteins were added to protein sample buffer, boiled in water for 5 min and separated by SDS-PAGE. Proteins were transferred onto polyvinylidene fluoride membranes and Blocked at 4°C overnight with Tris-buffered saline/Tween 20 (TBST) containing 5% non-fat dried milk. Membranes were incubated with anti-beclin1 (1:1,000), anti-LC3 (1:1,000), anti-PI3K (1:1,000), anti-mTOR (1:1,000) and anti-GAPDH antibodies (1:2,500; cat. no. sc-365062; Santa Cruz Biotechnology, Inc., Dallas, TX, USA) primary antibodies, at 4°C overnight. The membranes were rinsed three times with PBS Tween20 and then incubated with the secondary antibodies (1:2,500) for 2 h at room temperature. The protein bands were visualized using an enhanced chemiluminescent kit (EMD Millipore, Billerica, MA, USA). ImageJ software (Version 4.0; National Institutes of Health, Bethesda, MD, USA) was used to analyze the grayscale values of each group and GAPDH was used as the internal reference.

Statistical analysis

Statistical analysis was performed using one-way analysis of variance followed by a post-hoc Bonferroni test with SPSS software 20.0 (IBM Corp., Armonk, NY, USA). Data are expressed as the mean ± standard deviation and P<0.05 was considered to indicate a statistically significant difference.

Results

General conditions

The model group rats were in emotional distress and exhibited diet reduction, messy hair, rough tail, diarrhea and a difference in color and luster of their hair after 3 days (data not shown). Two rats exhibited a slight ulcer in their tails (data not shown). Compared with the blank group, Doxorubicin-induced nephrotic rats exhibited mild edema in the testicles (data not shown). The rats in the blank group exhibited free movement, a normal diet, smooth body hair and no diarrhea or other abnormal reactions (data not shown). No significant differences in the weight were observed among the blank (509±8 g), model (496±30 g), Telmisartan (500±32 g) and MHCD group (498±29 g) at week 8.

MHCD ameliorates proteinuria

As demonstrated in Fig. 1, proteinuria in Doxorubicin-induced nephrotic rats increased to peak levels at week 6. Proteinuria was higher at weeks 2, 4 (both P<0.05), 6 and 8 (both P<0.01) in the model group compared with the blank group, indicating the successful establishment of the model. Proteinuria in the MHCD and telmisartan groups increased at weeks 2, 4 and 6 compared with the blank group (all P<0.05). However, at weeks 6 and 8, 24-h proteinuria significantly decreased in the telmisartan and MHCD groups compared with the model group (P<0.05). However, no significant differences were observed between the telmisartan and MHCD groups. At week 8, no significant differences were observed between the MHCD and blank groups. These results indicate that MHCD ameliorates proteinuria in Doxorubicin-induced nephrotic rats.

MHCD ameliorates the levels of ALB, Scr, TG and TCH

As demonstrated in Fig. 2A, ALB levels in the model group decreased significantly compared with the blank group (P<0.05). Compared with the model group, the MHCD and telmisartan groups exhibited significantly increased levels of ALB (both P<0.05). Scr levels in the model group increased significantly compared with the blank group (P<0.05; Fig. 2B). Compared with the model group, the MHCD and telmisartan groups exhibited significantly decreased levels of Scr (both P<0.05). Furthermore, levels of TG and TCH in the model group increased significantly compared with the blank group (P<0.05; Fig. 2C). Compared with the model group, TG and TCH were significantly decreased in the MHCD and telmisartan groups (both P<0.05). These results indicate that MHCD ameliorates Doxorubicin-induced changes in ALB, Scr, TG and TCH levels in rats. Long-term proteinuria causes hyperlipidemia due to dysfunctional hepatic lipid protein synthesis (17). MHCD may improve lipid levels by reducing albuminuria in Doxorubicin-induced nephrotic rats. However, it is unclear whether MHCD can directly improve lipid levels.

MHCD prevents glomerular and podocyte injury

As demonstrated in Fig. 3, renal pathological changes were examined using HE, Masson's trichome and PAS staining. Compared with the blank group, the model group had a greater number of proliferative mesangial cells, increased extracellular matrix deposition, a thickened glomerular basement membrane and disorderly arranged tubular cells (Fig. 3). Compared with the model group, telmisartan and MHCD treatments ameliorated the Doxorubicin-induced renal pathological changes in rats. These results demonstrate the protective effect of MHCD on the kidneys of Doxorubicin-induced nephrotic rats.

As demonstrated in Fig. 4, the blank group exhibited normal morphology. The podocytes in the model group were flattened or fused and an exposed basement membrane and increased number of proliferating mesangial cells were observed. In addition, lipid vacuoles were observed in a number of endothelial cells. Compared with the model group, telmisartan and MHCD treatments alleviated the degree of podocyte foot process fusion. Additionally, only a small number of swollen endothelial cells were observed following MHCD treatment, further supporting the findings.

MHCD inhibits excessive autophagy

Autophagy is a highly conserved cellular process that is important in anaphase cells, including neurocytes and podocytes (18,19). Autophagy in podocytes was observed using transmission electron microscopy. Podocytes in the blank group exhibited a normal nucleus, mitochondria, endoplasmic reticulum and Golgi apparatus (Fig. 5), whereas podocytes in the model group had a marked increase in the number of autophagosomes, dilated endoplasmic reticula and swollen mitochondria. A lower number of autophagosomes were observed in the MHCD and telmisartan groups compared with the model group. These results demonstrate that MHCD inhibits excessive autophagy in rats with Doxorubicin-induced nephrosis.

To assess whether autophagy is affected by MHCD, the expression of beclin-1 and LC3 were measured using immunohistochemistry (Fig. 6A) and western blotting (Fig. 7A). Beclin-1 (Figs. 6B and 7B), LC3 (Fig. 6C), LC3-I and LC3-II (Fig. 7B; all P<0.05) expression was significantly higher in the model group compared with the blank group. These increases were significantly inhibited following treatment with MHCD or telmisartan (all P<0.05).

MHCD inhibits excessive autophagy by inducing the PI3K/mTOR signaling pathway

Excessive autophagy leads to cell death and it has been demonstrated that the PI3K/mTOR signaling pathway serves a critical role in regulating this process (20,21). To assess whether MHCD inhibits excessive autophagy via the PI3K/mTOR signaling pathway, PI3K and mTOR expression were measured by western blotting (Fig. 7A) and immunohistochemistry (Fig. 8A). As presented in Figs. 7B, 8B and C, the expression of PI3K and mTOR was significantly lower in the model group compared with the blank group (all P<0.05). When Doxorubicin-induced nephrotic rats were treated with MHCD or telmisartan, PI3K and mTOR expression increased significantly compared with the model group (all P<0.05). However, as presented in Fig. 6, autophagic activity in the model group was significantly higher compared with the blank group, indicating that the inhibition of the PI3K/mTOR signaling pathway resulted in excessive autophagy. These results indicate that MHCD inhibits excessive autophagy by inducing the PI3K/mTOR signaling pathway.

Discussion

The aim of the present study was to investigate whether the regulatory effects of MHCD in autophagy are mediated via the PI3K/mTOR signaling pathway. It was revealed that the protective effects of MHCD in Doxorubicin-induced nephrotic rats are achieved by suppressing excessive autophagy via activation of the PI3K/mTOR pathway.

Proteinuria, hypoalbuminemia and hyperlipidemia are typical manifestations of Doxorubicin-induced nephrosis in rats (22) and these characteristics were observed in rats in the present study. Histological changes, including focal segmental glomerularsclerosis, tubulointerstitial inflammation, fibrosis and podocyte fusion, typical of Doxorubicin-induced nephropathy, were also observed in the present study (23). In TCM, MHCD has been demonstrated to reduce urinary protein and cholesterol levels, increase serum albumin levels and prevent glomerular and podocyte injuries in Doxorubicin-induced nephrotic rats (24). These results may be used to develop novel methods for delaying glomerulosclerosis using TCM.

Autophagy is a process by which cellular components are recycled (25) and injured organelles and proteins are removed (26). Autophagy comprises mechanistically distinct steps, including the induction, identification and selection of cargo, the formation of vesicles, autophagosome vacuole fusion, the breakdown of cargo and the release of degradation products into the cytoplasm (27). The formation of autophagosomes serves an important role in autophagy and requires the recruitment of ubiquitin-like-conjugating enzyme ATG (Atg) (28). LC3 has a molecular mass of ~17 kDa and is a mammalian ortholog of yeast Atg8 (29). Previous studies have demonstrated that LC3 is recruited into autophagosomal membranes, indicating that LC3 is a marker for autophagy (30,31). Beclin-1, another key regulator of autophagy, is the mammalian ortholog of yeast Atg6 (32,33). Beclin-1 has been demonstrated to induce autophagy via regulating PI3K VPS34 (Vps-34) to promote the formation of beclin 1-Vps34-Vps15 core complexes (34). As such, LC3 and beclin-1 are effective biomarkers for monitoring autophagy. Cells typically trigger autophagy to reduce cellular damage following infection, ischemia, starvation or growth factor deficiency (35). Autophagy, which occurs under basal conditions, serves an important role in cell growth, development and homeostasis by maintaining a balance between the synthesis and subsequent recycling of cellular products (36). However, excessive or sustained autophagy triggers non-apoptotic programmed cell death due to excessive self-digestion and degradation of essential cellular constituents (20,37,38). It has been reported that excessive autophagy serves a role in kidney disease (39); furthermore, excessive autophagy has been demonstrated to result in cell death (40). The results of the present study are in agreement with previous reports, indicating that MHCD attenuates excessive autophagy in the kidney by attenuating LC3 and beclin-1 overexpression and upregulating basal autophagy to protect Doxorubicin-induced nephrotic rats.

The specific mechanism by which MHCD regulates autophagy has not previously been elucidated, however it has been reported that PI3K/mTOR pathway activation is involved in autophagy (41,42). Based on this, the effect of MHCD on PI3K/mTOR pathway activation was investigated. PI3K is a family of lipid kinases (43) and mTOR is a highly conserved serine/threonine kinase (44). In the present study, the expression of PI3K and mTOR in the renal cortical tissues of Doxorubicin-induced nephrotic rats was examined. The results revealed that MHCD activates the PI3K/mTOR signaling pathway. Luo et al (45) reported that excessive autophagy resulted in delayed cell death due to inhibition of the PI3K/mTOR signaling pathway. Furthermore, excessive autophagy is often observed when the PI3K/mTOR signaling pathway is chemically blocked (46). A previous study revealed that autophagy was one of the main mechanisms of cell death when the PI3K/mTOR signaling pathway is inhibited (47). Wang et al (48) revealed that PI3K/Akt signaling was associated with the protection of neurons via autophagy inhibition. Xing et al (49) reported that mTOR upregulation inhibited autophagy. The results of the present study revealed that MHCD attenuates excessive autophagy and activates the PI3K/mTOR signaling pathway in Doxorubicin-induced renal injury. This suggests that MHCD attenuates excessive autophagy by inhibiting the PI3K/mTOR signaling pathway.

Podocyte activity has previously been assessed in vitro using CCK-8 and it was revealed that Doxorubicin promoted autophagy compared with the autophagy inhibitor 3-methyladenine (50). The aim of the present study was to assess whether MHCD inhibited excessive autophagy caused by Doxorubicin only; no other agents were assessed. In future studies additional autophagy inhibitors and PI3K/mTOR signaling pathway inhibitors should be assessed in vivo to confirm the results of the present study. Furthermore, autophagy is a complicated process; elucidating the specific effects of individual MHCD components in autophagy requires the synthesis of specific small molecule compounds for validation.

In the present study, it rats with untreated Doxorubicin-induced nephrosis exhibited a marked and sustained increase in proteinuria until week 6. In contrast, proteinuria in MHCD- and telmisartan-treated rats increased only slightly between weeks 2 and 4. Future studies should include a group of individuals treated with MHCD for 2 weeks prior to Doxorubicin treatment to further demonstrate that MHCD has protective roles in Doxorubicin-induced nephrotic rats. In conclusion, the results of the present study suggest that MHCD ameliorates proteinuria, increases ALB levels and decreases Scr, TG and TCH levels. Additionally, it protects against glomerular and podocyte injuries by inhibiting excessive autophagy via the PI3K/mTOR signaling pathway and upregulating basal autophagy.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Subjective Selected Subjects Program of China Academy of Chinese Medical Sciences (grant no. ZZ0708105).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

YZ conceived and designed the animal experiments and helped to draft the paper. ZKY, BY, LSL, JNZ and WH performed the experiments. ZKY and BY analyzed the data and wrote the paper.

Ethics approval and consent to participate

The Animal Care and Use Committee of Xiyuan Hospital of China Academy of Chinese Medical Sciences (Beijing, China) approved the experimental protocol.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

MHCD

modified Huangqi Chifeng decoction

ALB

albumin

TCH

total cholesterol

TG

triacylglyceride

Scr

serum creatinine

LC3

microtubule-associated proteins 1A/1B light chain 3B

PI3K

phosphoinositide-3 kinase

mTOR

mammalian target of rapamycin

CKD

chronic kidney disease

TCM

traditional Chinese medicine

PAS

periodic acid Schiff

References

1 

Kirsztajn GM, Filho NS, Draibe SA, Netto MV, Thomé FS, Souza E and Bastos MG: Fast reading of the KDIGO 2012: Guidelines for evaluation and management of chronic kidneydisease in clinical practice. J Bras Nefrol. 36:63–73. 2014.(In Portuguese). View Article : Google Scholar : PubMed/NCBI

2 

Mills KT, Xu Y, Zhang W, Bundy JD, Chen CS, Kelly TN, Chen J and He J: A systematic analysis of worldwide population-based data on the global burden of chronic kidney disease in 2010. Kidney Int. 88:950–957. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Carney EF: Epidemiology: Global burden of disease study 2013 reports that disability caused by CKD is increasing worldwide. Nat Rev Nephrol. 11:4462015. View Article : Google Scholar : PubMed/NCBI

4 

Barnett AH, Bain SC, Bouter P, Karlberg B, Madsbad S, Jervell J and Mustonen J; Diabetics Exposed to Telmisartan and Enalapril Study Group, : Angiotensin-receptor blockade versus converting-enzyme inhibition in type 2 diabetes and nephropathy. N Engl J Med. 351:1952–1961. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Gong XZ, Zhou LF, Wang Q, Tang XC, Qian YR, Wang YR, Lu L and Zhou JJ: Effect of Chuanhuang No.1 recipe on renal function and micro-inflammation in phase 3 chronic kidney disease patients. Zhongguo Zhong Xi Yi Jie He Za Zhi. 35:137–141. 2015.(In Chinese). PubMed/NCBI

6 

Dong F, Cheng J, Lin S, Hu Z, Chen G and He L: The clinical research on serum cystatin-C alteration on stage II chronic kidney disease with gubenquduyishen decoction treatment. J Ethnopharmacol. 131:581–584. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Zhang Yu: Clinical observation on 50 cases of chronic nephritis proteinuria treated with modified Huangqi Chifeng Decoction. Chin Med Guides. 36:137–138. 2007.(In Chinese).

8 

Clark SL Jr: Cellular differentiation in the kidneys of newborn mice studied with the electron microscope. J Biophys Biochem Cytol. 3:349–362. 1957. View Article : Google Scholar : PubMed/NCBI

9 

Berkenstam A, Ahlberg J and Glaumann H: Isolation and characterization of autophagic vacuoles from rat kidney cortex. Virchows Arch B Cell Pathol Incl Mol Pathol. 44:275–286. 1983. View Article : Google Scholar : PubMed/NCBI

10 

Asanuma K, Tanida I, Shirato I, Ueno T, Takahara H, Nishitani T, Kominami E and Tomino Y: MAP-LC3, a promising autophagosomal marker, is processed during the differentiation and recovery of podocytes from PAN nephrosis. FASEB J. 17:1165–1167. 2003. View Article : Google Scholar : PubMed/NCBI

11 

Mizushima N, Yamamoto A, Matsui M, Yoshimori T and Ohsumi Y: In vivo analysis of autophagy in response to nutrient starvation using transgenic mice expressing a fluorescent autophagosome marker. Mol Biol Cell. 15:1101–1104. 2004. View Article : Google Scholar : PubMed/NCBI

12 

Hartleben B, Gödel M, Meyer-Schwesinger C, Liu S, Ulrich T, Köbler S, Wiech T, Grahammer F, Arnold SJ, Lindenmeyer MT, et al: Autophagy influences glomerular disease susceptibility and maintains podocyte homeostasis in aging mice. J Clin Invest. 120:1084–1096. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Zeng C, Fan Y, Wu J, Shi S, Chen Z, Zhong Y, Zhang C, Zen K and Liu Z: Podocyte autophagic activity plays a protective role in renal injury and delays the progression of podocytopathies. J Pathol. 234:203–213. 2014.PubMed/NCBI

14 

Yu Z, Yang B and Zhang Y: Protective effect of modified Huangqi Chifeng decoction on Doxorubicin Nephrosis (AN) rats kidney and its regulated effect on its autophagy level. Zhongguo Zhong Yi Ji Chu Yi Xue Za Zhi. 23:477–479+563. 2017.(In Chinese).

15 

Faleiros CM, Francescato HD, Papoti M, Chaves L, Silva CG, Costa RS and Coimbra TM: Effects of previous physical training on adriamycin nephropathy and its relationship with endothelial lesions and angiogenesis in the renal cortex. Life Sci. 169:43–51. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Lee VW and Harris DC: Adriamycin nephropathy: A model of focal segmental glomerulosclerosis. Nephrology (Carlton). 16:30–38. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Rahman M, Yang W, Akkina S, Alper A, Anderson AH, Appel LJ, He J, Raj DS, Schelling J, Strauss L, et al: Relation of serum lipids and lipoproteins with progression of CKD: The CRIC study. Clin J Am Soc Nephrol. 9:1190–1198. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Liu N, Xu L, Shi Y and Zhuang S: Podocyte autophagy: A potential therapeutic target to prevent the progression of diabetic nephropathy. J Diabetes Res. 2017:35602382017. View Article : Google Scholar : PubMed/NCBI

19 

Mijaljica D, Prescott M and Devenish RJ: The intriguing life of autophagosomes. Int J Mol Sci. 13:3618–3635. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Stern ST, Adiseshaiah PP and Crist RM: Autophagy and lysosomal dysfunction as emerging mechanisms of nanomaterial toxicity. Part Fibre Toxicol. 9:202012. View Article : Google Scholar : PubMed/NCBI

21 

Guan H, Piao H, Qian Z, Zhou X, Sun Y, Gao C, Li S and Piao F: 2,5-Hexanedione induces autophagic death of VSC4.1 cells via a PI3K/Akt/mTOR pathway. Mol Biosyst. 13:1993–2005. 2017. View Article : Google Scholar : PubMed/NCBI

22 

Wang Z, Wang ZG and Liu Z: Changes of glomerular fixed anionic charge sites in adriamycin nephrosis in rats. Chin Med J (Engl). 104:128–131. 1991.PubMed/NCBI

23 

Bertani T, Poggi A, Pozzoni R, Delaini F, Sacchi G, Thoua Y, Mecca G, Remuzzi G and Donati MB: Adriamycin-induced nephrotic syndrome in rats: Sequence of pathologic events. Lab Invest. 46:16–23. 1982.PubMed/NCBI

24 

Wang YL, Zhang Y, Wang HX and Hao W: Impact of jiaweihuangqichifeng decoction on proteinuria, renal cortex SOD and renal cortex MDA in nephrotic rats induced by adriamycin. Chin J Basic Med Trad Chin Med. 17:505–507. 2011.(In Chinese).

25 

Costas MA and Rubio MF: Autophagy: A strategy for cell survival. Medicina (B Aires). 77:314–320. 2017.(In Spanish). PubMed/NCBI

26 

Yao ST and Qin SC: The relationship of autophagy with endoplasmic reticulum stress and its role in pathogenesis, prevention and therapy of atherosclerosis. Sheng Li Xue Bao. 69:515–521. 2017.(In Chinese). PubMed/NCBI

27 

He C and Klionsky DJ: Regulation mechanisms and signaling pathways of autophagy. Annu Rev Genet. 43:67–93. 2009. View Article : Google Scholar : PubMed/NCBI

28 

Davis S, Wang J and Ferro-Novick S: Crosstalk between the secretory and autophagy pathways regulates autophagosome formation. Dev Cell. 41:23–32. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Tanida I, Ueno T and Kominami E: LC3 and autophagy. Methods Mol Biol. 445:77–88. 2008. View Article : Google Scholar : PubMed/NCBI

30 

Shibata M, Yoshimura K, Tamura H, Ueno T, Nishimura T, Inoue T, Sasaki M, Koike M, Arai H, Kominami E and Uchiyama Y: LC3, a microtubule-associated protein1A/B light chain3, is involved in cytoplasmic lipid droplet formation. Biochem Biophys Res Commun. 393:274–279. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Tanida I, Ueno T and Kominami E: LC3 conjugation system in mammalian autophagy. Int J Biochem Cell Biol. 36:2503–2518. 2004. View Article : Google Scholar : PubMed/NCBI

32 

Glover K, Li Y, Mukhopadhyay S, Leuthner Z, Chakravarthy S, Colbert CL and Sinha SC: Structural transitions in conserved, ordered Beclin 1 domains essential to regulating autophagy. J Biol Chem. 292:16235–16248. 2017. View Article : Google Scholar : PubMed/NCBI

33 

Mei Y, Glover K, Su M and Sinha SC: Conformational flexibility of BECN1: Essential to its key role in autophagy and beyond. Protein Sci. 25:1767–1785. 2016. View Article : Google Scholar : PubMed/NCBI

34 

Kang R, Zeh HJ, Lotze MT and Tang D: The Beclin 1 network regulates autophagy and apoptosis. Cell Death Differ. 18:571–580. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Yin Z, Pascual C and Klionsky DJ: Autophagy: Machinery and regulation. Microb Cell. 3:588–596. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Ravikumar B, Sarkar S, Davies JE, Futter M, Garcia-Arencibia M, Green-Thompson ZW, Jimenez-Sanchez M, Korolchuk VI, Lichtenberg M, Luo S, et al: Regulation of mammalian autophagy in physiology and pathophysiology. Physiol Rev. 90:1383–1435. 2010. View Article : Google Scholar : PubMed/NCBI

37 

Martinet W, Agostinis P, Vanhoecke B, Dewaele M and De Meyer GR: Autophagy in disease: A double-edged sword with therapeutic potential. Clin Sci (Lond). 116:697–712. 2009. View Article : Google Scholar : PubMed/NCBI

38 

Kroemer G and Jäättelä M: Lysosomes and autophagy in cell death control. Nat Rev Cancer. 5:886–897. 2005. View Article : Google Scholar : PubMed/NCBI

39 

Johnson-Lyles DN, Peifley K, Lockett S, Neun BW, Hansen M, Clogston J, Stern ST and McNeil SE: Fullerenol cytotoxicity in kidney cells is associated with cytoskeleton disruption, autophagic vacuole accumulation, and mitochondrial dysfunction. Toxicol Appl Pharmacol. 248:249–258. 2010. View Article : Google Scholar : PubMed/NCBI

40 

Klionsky DJ: Autophagy: From phenomenology to molecular understanding in less than a decade. Nat Rev Mol Cell Biol. 11:931–937. 2007. View Article : Google Scholar

41 

Peng Y, Qiu L, Xu D, Zhang L, Yu H, Ding Y, Deng L and Lin J: M4IDP, a zoledronic acid derivative, induces G1 arrest, apoptosis and autophagy in HCT116 colon carcinoma cells via blocking PI3K/Akt/mTOR pathway. Life Sci. 185:63–72. 2017. View Article : Google Scholar : PubMed/NCBI

42 

Wang S, Li J, Du Y, Xu Y, Wang Y, Zhang Z, Xu Z, Zeng Y, Mao X and Cao B: The class I PI3K inhibitor S14161 induces autophagy in malignant blood cells by modulating the Beclin 1/Vps34 complex. J Pharmacol Sci. 134:197–202. 2017. View Article : Google Scholar : PubMed/NCBI

43 

De Santis MC, Sala V, Martini M, Ferrero GB and Hirsch E: PI3K signaling in tissue hyper-proliferation: From overgrowth syndromes to kidney cysts. Cancers (Basel). 9(pii): E302017. View Article : Google Scholar : PubMed/NCBI

44 

Reilly R, Mroz MS, Dempsey E, Wynne K, Keely SJ, McKone EF, Hiebel C, Behl C and Coppinger JA: Targeting the PI3K/Akt/mTOR signalling pathway in cystic fibrosis. Sci Rep. 7:76422017. View Article : Google Scholar : PubMed/NCBI

45 

Luo T, Liu G, Ma H, Lu B, Xu H, Wang Y, Wu J, Ge P and Liang J: Inhibition of autophagy via activation of PI3K/Akt pathway contributes to the protection of ginsenoside Rb1 against neuronal death caused by ischemic insults. Int J Mol Sci. 15:15426–15442. 2014. View Article : Google Scholar : PubMed/NCBI

46 

Oh I, Cho H and Lee Y, Cheon M, Park D and Lee Y: Blockage of autophagy rescues the dual PI3K/mTOR Inhibitor BEZ235-induced growth inhibition of colorectal cancer cells. Dev Reprod. 20:1–10. 2016. View Article : Google Scholar : PubMed/NCBI

47 

Echeverry N, Ziltener G, Barbone D, Weder W, Stahel RA, Broaddus VC and Felley-Bosco E: Inhibition of autophagy sensitizes malignant pleural mesothelioma cells to dual PI3K/mTOR inhibitors. Cell Death Dis. 6:e17572015. View Article : Google Scholar : PubMed/NCBI

48 

Wang Y, Wang W, Li D, Li M, Wang P, Wen J, Liang M, Su B and Yin Y: IGF-1 alleviates NMDA-induced excitotoxicity in cultured hippocampal neurons against autophagy via the NR2B/PI3K-AKT-mTOR pathway. J Cell Physiol. 229:1618–1629. 2014. View Article : Google Scholar : PubMed/NCBI

49 

Xing Y, Liu YX, Liu X, Wang SL, Li P, Lin XH, Sui CL, Xu C, Qi B and Tong Q: Effects of Gui Zhu Yi Kun formula on the P53/AMPK pathway of autophagy in granulosa cells of rats with polycystic ovary syndrome. Exp Ther Med. 13:3567–3573. 2017. View Article : Google Scholar : PubMed/NCBI

50 

Li S: The effect of MHCD on proteinuria and its cellular molecular mechanism [D]. Beijing Zhong Yi Yao Da Xue. 2016.(In Chinese).

Related Articles

Journal Cover

September-2018
Volume 16 Issue 3

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yu ZK, Yang B, Zhang Y, Li LS, Zhao JN and Hao W: Modified Huangqi Chifeng decoction inhibits excessive autophagy to protect against Doxorubicin‑induced nephrotic syndrome in rats via the PI3K/mTOR signaling pathway. Exp Ther Med 16: 2490-2498, 2018
APA
Yu, Z., Yang, B., Zhang, Y., Li, L., Zhao, J., & Hao, W. (2018). Modified Huangqi Chifeng decoction inhibits excessive autophagy to protect against Doxorubicin‑induced nephrotic syndrome in rats via the PI3K/mTOR signaling pathway. Experimental and Therapeutic Medicine, 16, 2490-2498. https://doi.org/10.3892/etm.2018.6492
MLA
Yu, Z., Yang, B., Zhang, Y., Li, L., Zhao, J., Hao, W."Modified Huangqi Chifeng decoction inhibits excessive autophagy to protect against Doxorubicin‑induced nephrotic syndrome in rats via the PI3K/mTOR signaling pathway". Experimental and Therapeutic Medicine 16.3 (2018): 2490-2498.
Chicago
Yu, Z., Yang, B., Zhang, Y., Li, L., Zhao, J., Hao, W."Modified Huangqi Chifeng decoction inhibits excessive autophagy to protect against Doxorubicin‑induced nephrotic syndrome in rats via the PI3K/mTOR signaling pathway". Experimental and Therapeutic Medicine 16, no. 3 (2018): 2490-2498. https://doi.org/10.3892/etm.2018.6492