Open Access

Sericin enhances the insulin‑PI3K/AKT signaling pathway in the liver of a type 2 diabetes rat model

  • Authors:
    • Chengjun Song
    • Donghui Liu
    • Songhe Yang
    • Luyang Cheng
    • Enhong Xing
    • Zhihong Chen
  • View Affiliations

  • Published online on: August 17, 2018     https://doi.org/10.3892/etm.2018.6615
  • Pages: 3345-3352
  • Copyright: © Song 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 current study was to investigate the regulatory effect of sericin on the hepatic insulin‑­phosphoinositide 3‑kinase (PI3K)/protein kinase B (AKT) signaling pathway in a type 2 diabetes rat model. Male Sprague Dawley rats were randomly divided into four groups: Control group, diabetic model group, high‑dose sericin group and low‑dose sericin group, with 12 rats in each group. Fasting blood glucose was detected by the glucose oxidase method, and hepatic glycogen was determined by periodic acid‑Schiff staining. The morphology of the liver was observed by hematoxylin and eosin staining. Immunohistochemical staining, western blotting and reverse transcription‑quantitative polymerase chain reaction were used to determine the protein and mRNA expression levels of insulin receptor (IR), IR substrate‑1 (IRS‑1), PI3K and AKT. Compared with the control group, the blood glucose of the diabetic model group was significantly increased (P<0.05). The glycogen content and the expression levels of IR, IRS‑1, PI3K and AKT in the diabetic model group were significantly lower (P<0.05), and the liver morphological structure of the diabetic model group exhibited obvious pathological changes compared with the control group. Compared with the diabetic model group, the blood glucose of the high‑ and low‑dose sericin groups was significantly reduced, while the glycogen content and the expression levels of IR, IRS‑1, PI3K and AKT in the sericin treatment groups were significantly increased (P<0.05). Additionally, the liver pathological changes of high‑dose and low‑dose sericin groups were markedly reduced. Sericin may enhance the signaling transduction effect of insulin by upregulating the expression levels of key factors (IR, IRS‑1, PI3K and AKT) in the liver insulin‑PI3K/AKT signaling pathway, thus promoting glucose transport and liver glycogen synthesis, and further reducing blood glucose.

Introduction

Type 2 diabetes is primarily characterized by insulin resistance, and one of the important causes of insulin resistance is insulin signal transduction disorder (1,2). Insulin is the only hormone in the body that is able to lower blood glucose level. It first binds to the insulin receptor (IR) on the cell membrane and then activates the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) or Ras/Raf/mitogen-activated protein kinase signaling pathway (3). The PI3K/AKT signaling pathway is the primary pathway of insulin signaling transduction, through which insulin regulates glucose uptake, glycogen synthesis and degradation (4).

In the liver, insulin binds to the α subunit of IR on liver cells, and then activates IR substrate (IRS). IRS then binds to p85, the regulatory subunit of PI3K, and activates p110, the catalytic subunit of PI3K. The activated PI3K produces the second messengers Phosphatidylinositol (3,4)-trisphosphate [PtdIns(3,4)P2] and PtdIns(3,4,5)P3, which promote the activation of AKT (5). Activated AKT regulates the process of carbohydrate metabolism in hepatocytes via the following pathways: First, activated AKT promotes the transfer of glucose transporter 4 to cell membranes, which facilitates the transportation of extracellular glucose into the cells (6). Second, activated AKT mediates glycogen synthesis through glycogen synthase kinase 3 (GSK3) (7). Third, activated AKT inhibits hepatocyte gluconeogenesis through peroxisome proliferator-activated receptor-γ coactivator-α (8). Therefore, the insulin-PI3K/AKT signaling pathway serves an important function in the regulation of carbohydrate metabolism in liver.

At present, chemical drugs, including metformin, are primarily used for the clinical treatment of diabetes (9). Blood glucose level is significantly reduced by these drugs, but insulin resistance is not effectively improved. In addition, long-term use of these drugs can cause a variety of serious side effects, leading to liver and kidney damage (10). Therefore, it is necessary to develop natural drugs with reliable hypoglycemic effects and less toxic side effects.

Sericin is a water-soluble protein of the silkworm cocoon, which is composed of 18 different amino acids (11). The content of serine, aspartic acid, threonine and other polar amino acids (which are rich in hydroxyl, amino and carboxyl groups) in sericin is >70%, therefore sericin possesses good water solubility (12). Sericin has been used in wound healing, tissue regeneration, drug delivery and biomedicine (11,13). Studies have indicated that the addition of sericin to the daily diet may reduce the intestinal intake of cholesterol, triglycerides and other lipids, as well as improve the body's tolerance to glucose (14,15). Previous studies by our group indicated that sericin could effectively reduce blood glucose level in a streptozotocin-induced type 2 diabetic rat model (16,17). However, the hypoglycemic mechanism of sericin remains unclear.

In the current study, a type 2 diabetes rat model was established by administration with high-fat and high-glucose diet combined with streptozotocin intraperitoneal injection. The expression levels of IR, IRS-1, PI3K and AKT in the rat liver were analyzed to investigate whether sericin exerts a hypoglycemic effect via the insulin-PI3K/AKT signaling pathway. The current study may provide an experimental basis for the use of sericin in the treatment of diabetes in the clinic.

Materials and methods

Animals, grouping and sampling

A total of 48 specific-pathogen-free 6-week-old male Sprague-Dawley rats (purchased from Beijing Vital River Laboratory Animal Technology Co., Ltd., Beijing, China) were used in the current study. The rats were housed with a natural light cycle at 18–26°C and relative humidity of 40–70%. They were randomly divided into four groups: Control group, diabetic model group, high-dose sericin group and low-dose sericin group, with 12 rats in each group. The type 2 diabetes rat model was established as follows: by high-fat and high-glucose diet combined with streptozotocin (35 mg/kg, twice) intraperitoneal injection. The rats were fed with high-fat and high-glucose diets as previously described (18) for 4 weeks and streptozotocin (35 mg/kg) was administered twice in succession by intraperitoneal injection. After 72 h, blood glucose was measured and rats with a fasting blood glucose of 11.1 mmol/l were deemed to have diabetes (17,19). Model rats were fed with high-fat and high-glucose diet for another 3 weeks and the diet was changed to normal diet when they were fed with sericin. The control group was fed with normal diet throughout the experiment. For the high- and low-dose sericin groups, the rats were continuously administered with 2.4 and 1.8 g/kg/day sericin for 35 days, respectively, while the control rats were administered with the same volume of saline for 35 days. All animal experiments were approved by the Ethics Committee of Chengde Medical University (Chengde, China) and conducted according to the ethical guidelines of Chengde Medical University.

The rats in each group were fasted for 12 h following treatment and anesthetized by intraperitoneal injection of 10% chloral hydrate (300 mg/kg body weight) (2022). Then, the thoracic cavity was opened and venous blood samples were collected from the right atrium of the rats. During blood sample collection, rats were in deep anesthesia and no signs of peritonitis were observed. Then, the rats were sacrificed by decapitation and the left hepatic lobe was collected following opening of the abdominal cavity. The blood was centrifuged at 800 × g for 20 min at 4°C, and the serum was isolated. The liver tissues were kept in liquid nitrogen until further analysis.

Determination of glucose level

The blood glucose level of rats in each group was measured using a glucose oxidase method (23) on a Beckman Coulter AU5800 Clinical Chemistry Analyzer (Beckman Coulter, Inc., Brea, CA, USA).

Hematoxylin and eosin (H&E) staining

The liver morphology of each group was observed under an Olympus BH-2 light microscope (Olympus Corporation, Tokyo, Japan) following H&E staining. Briefly, the liver tissues were fixed with Bouins fixative [a mixture of picric acid saturated liquid (1.22%), formaldehyde and glacial acetic acid at a ratio of 15:5:1] for 24 h at room temperature and embedded in paraffin, and continuously sliced into 5-µm-thick sections. At room temperature, the sections were stained with hematoxylin for 7 min, differentiated with 1% HCl in 70% alcohol for 5 sec, stained with eosin for 1 min and dehydrated in a serial ethanol solution of increasing concentrations. Then, the sections were deparaffinized with xylene and sealed.

Periodic acid-Schiff staining

The hepatic glycogen content was determined by periodic acid-Schiff staining. The liver tissues were fixed as above described and embedded in paraffin, and continuously sliced into 5-µm-thick sections. The sections were oxidized with periodic acid for 10 min and stained with Schiff reagent for 15 min at room temperature. Cell nuclei were re-stained with hematoxylin for 5 min at room temperature. Then, sections were deparaffinized with xylene and sealed. Cells with red or magenta particles in the cytoplasm were defined as positive for glycogen. Tissue sections digested with amylase were used as negative controls.

For the quantification of glycogen content, six rat livers were randomly selected from each group, three sections were selected from each rat liver, and three views were observed in each section. The liver lobules with intact tissue structure were selected for observation using an light microscope (BH-2; Olympus Corporation, Tokyo, Japan; magnification, ×200). Image-Pro Plus 6.0 image analysis software (Media Cybernetics, Inc., Rockville, MD, USA) was used to calculate the integral optical density of glycogen in hepatocyte cytoplasm, and the mean value was determined as the glycogen content.

Immunohistochemical staining

Protein expression of IR, IRS-1, PI3K and AKT in the liver was analyzed. Briefly, the sections were dewaxed and incubated in 3% H2O2-methanol at 37°C for 30 min. Following antigen retrieval by microwaving, the sections were blocked with 10% goat serum (OriGene Technologies, Inc., Beijing, China) at 37°C for 30 min. The sections were then incubated with primary antibodies against IR (1:100; cat. no. ab131238), IRS-1 (1:100; cat. no. ab131487; both Abcam, Cambridge, MA USA), PI3K (1:100; cat. no. 611398; BD Biosciences, San Jose, CA, USA) and AKT (1:200; cat. no. ab179463; Abcam) at 4°C overnight. Next, the sections were incubated with goat anti-rabbit IgG secondary antibodies, which was provided by the rabbit streptavidin-biotin assay system (cat. no. SP-9001; Zhongshan Jinqiao Biotechnology Co., Ltd., Beijing, China) according to the manufacturer's protocol, and then counterstained with DAB for 5–8 min at room temperature. Cell nuclei were re-stained with hematoxylin for 10 min at room temperature. PBS was used to substitute the primary antibody as the negative control. Cells with brownish yellow and/or brown particles were defined as positive staining.

For quantification, six rat livers were randomly selected from each group, three sections were selected from each rat liver, and three views were observed in each section. The liver lobules with intact tissue structure were selected for observation by an Olympus BH-2 microscope (magnification, ×200). Image-Pro Plus 6.0 image analysis software was used to calculate the integral optical density of each protein, and the mean value was determined as the corresponding protein expression level.

Western blot analysis

Total protein was extracted by RIPA Tissue/Cell Lysates (Beijing Solarbio & Technology Co., Ltd., Beijing, China) from 100 mg liver tissues, and the protein concentration was determined using a BCA protein kit (Kangwei Shiji Biotechnology Co., Ltd., Beijing, China). Proteins (108 µg/lane) were separated by 10% SDS-PAGE and transferred on to a PVDF membrane. Following blocking with 5% skim milk overnight at 4°C, the membrane was incubated with primary antibodies [IR, IRS-1, AKT (all 1:1,000), PI3K (1:500) and β-actin (1:1,000; cat no. AF7018; Affinity Biosciences, Cincinnati, OH, USA)] at room temperature for 2 h. Then, the membrane was incubated with goat anti-rabbit IgG (1:5,000; cat. no. 074-1506) or goat anti-mouse IgG (1:5,000; cat. no. 074-1806; both KPL, Inc., Gaithersberg, MD, USA) for a further 1.5 h at room temperature. The membrane was developed with Super ECL Plus ultra-sensitive luminescent liquid (Applygen Technologies, Inc., Beijing, China). The protein bands were analyzed with Quantity One v4.6.2 software (Bio-Rad Laboratories, Inc., Hercules, CA, USA). The density of each target band was calculated relative to the β-actin band to determine relative expression levels.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA was extracted from 100 mg liver tissues with a TaKaRa MiniBEST Universal RNA Extraction kit (cat. no. 9767) and reverse transcribed into cDNA using a PrimeScript™ RT Master mix (cat. no. RR036A; both Takara Biotechnology Co., Ltd., Dalian, China). The reverse transcription protocol was as follows: 37°C for 15 min and 85°C for 5 sec. The sample was put on ice and the obtained cDNA was stored at −20°C. Then, the mRNA expression of IR, IRS-1, PI3K, AKT and GAPDH was determined by qPCR with a SYBR Premix Ex Taq™ II kit (cat. no. RR820A; Takara Biotechnology Co., Ltd.). The primer sequences are listed in Table I. The PCR procedure was as follows: Pre-denaturation at 98°C for 1 min, followed by 40 cycles of denaturation at 98°C for 7 sec, annealing and polymerization at 60°C for 30 sec, then final polymerization at 60°C for 5 min. A relative standard curve method (24) was used to quantify the mRNA and the relative mRNA expression level of each target gene was determined relative to the corresponding GAPDH.

Table I.

Primer sequences used for quantitative polymerase chain reaction analysis.

Table I.

Primer sequences used for quantitative polymerase chain reaction analysis.

GenePrimer sequence (5′-3′)
IRF: TCATGGATGGAGGCTATCTGGA
R: TCCTTGAGCAGGTTGACGATTTC
IRS-1F: AAGCACCTATGCCAGCATCAAC
R: GAGGATTGCTGAGGTCATTTAGGTC
PI3KF: CCAGAAGAAGGGACAGTGGTATG
R: TCGTAGCCAATCAGGGAGGT
AKTF: ATGGACTTCCGGTCAGGTTCA
R: GCCCTTGCCCAGTAGCTTCA
GAPDHF: GGCACAGTCAAGGCTGAGAATG
R: ATGGTGGTGAAGACGCCAGTA

[i] IR, insulin receptor; IRS-1, IR substrate-1; PI3K, phosphoinositide 3-kinase; AKT, protein kinase B.

Statistical analysis

Statistical analysis was performed with the statistical software SPSS 20.0 (IBM Corp., Armonk, NY, USA). All data are expressed as mean ± standard deviation. One-way analysis of variance was used to compare multiple groups, followed by Tukey's post hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

Morphological changes of liver following sericin treatment

To observe the effect of sericin on the liver morphology of type 2 diabetic rats, H&E staining was performed. In the control group, the structure of the hepatic lobules was clear and the hepatocytes were arranged in a cord-like manner around the central vein (Fig. 1). The cell nuclei were large and round, located in the center of cells, and the cytoplasm was stained uniformly. The liver sinus was clear. In the diabetic model group, the hepatocytes were basically arranged in a cord-like manner around the central vein, but the liver cells were swollen, the volume increased, and obvious vacuolar structure appeared in the cytoplasm. A number of the liver cells exhibited soluble necrosis, and the hepatic sinus exhibited stenosis or atresia. Compared with the diabetic model group, the pathological changes of the rat liver in the high- and low-dose sericin groups were markedly reduced (Fig. 1). Rat liver lobular structures in these two sericin treatment groups were clear, and the hepatocytes were arranged in a cord-like manner around the central vein, with large round nuclei in the center of the cells. A small number of hepatocytes exhibited vacuolar structure in the cytoplasm, and the liver sinus was clear. This indicates that sericin may improve the liver morphological structure of type 2 diabetic rats.

Glycogen content in liver following sericin treatment

To determine the effect of sericin on the glycogen content in type 2 diabetic rat livers, periodic acid-Schiff staining was performed. Hepatic glycogen positive expression was observed in the liver sections of all groups, indicated by red and purple particles in the cytoplasm. As indicated in Fig. 2A, the number of positive cells in the control group was high, and the staining was dark purple. In the diabetic model group, there were fewer positively stained cells, and the staining was a lighter reddish color. In the high-dose sericin group, there was a higher number of positively stained cells compared with the low-dose sericin group, and the staining was darker. As indicated in Fig. 2B, compared with the control group, the glycogen content in the rat liver of the diabetic model group was significantly decreased (P<0.05). Compared with the diabetic model group, the glycogen content in the liver of the high- and low-dose sericin groups was significantly increased (P<0.01). Furthermore, the glycogen content in the rat liver of the high-dose sericin group was significantly higher compared with the low-dose sericin group (P<0.01). This indicates that sericin may significantly increase the liver glycogen content of type 2 diabetic rats.

Blood glucose levels following sericin treatment

To determine the effect of sericin on the blood glucose level of the type 2 diabetic rats, the rat blood glucose level was measured. As indicated in Fig. 2C, the blood glucose level in the diabetic model group was significantly increased compared with the control group (P<0.05). Compared with the diabetic model group, the blood glucose levels of the rats in the high- and low-dose sericin groups were significantly decreased (P<0.01). However, there was no significant difference in the blood glucose levels between the high- and how-dose sericin groups. This indicates that sericin may significantly decrease the blood glucose levels of type 2 diabetic rats.

Expression of associated factors in the PI3K/AKT signaling pathway

The protein levels of IR, IRS-1, PI3K, and AKT were detected with immunohistochemical staining and western blot analysis. Compared with the control group, the protein expression levels of IR, IRS-1, PI3K and AKT were significantly decreased in the diabetic model group (P<0.05; Figs. 3 and 4). Compared with the diabetic model group, the expression levels of these proteins in the liver of the high- and low-dose sericin groups were significantly increased (P<0.05; Figs. 3 and 4). Furthermore, the expression levels of AKT (Fig. 3) and IR (Figs. 3 and 4) in the high-dose sericin group were significantly higher compared with the low-dose sericin group (P<0.05).

To further verify these results, RT-qPCR was conducted to detect the mRNA levels of IR, IRS-1, PI3K and AKT. As indicated in Fig. 5, the mRNA levels of IR, IRS-1, PI3K and AKT in the diabetic model group were significantly lower compared with the control group (P<0.05). However, following sericin treatment in the high- and how-dose sericin groups, mRNA expression was significantly higher compared with the diabetic model group (P<0.05). No significant difference in mRNA levels of these genes was identified between the high- and low-dose sericin groups.

In summary, these results indicate that sericin may promote the insulin/PI3K/AKT signaling pathway in type 2 diabetic rat liver by upregulating IR, IRS-1, PI3K and AKT expression, and thus may reduce blood glucose levels.

Discussion

Type 2 diabetes is a chronic metabolic disease characterized by insulin resistance. On a global scale, the incidence of type 2 diabetes is increasing year by year, with faster growth rates in developing countries (25,26). The expected number of patients with type 2 diabetes in 2030 is 552 million (27). At present, diabetes has become the third most prevalent non-communicable disease that threatens human life and health after cardiovascular disease and cancer (28). Therefore, it is of great importance to explore the pathogenesis of type 2 diabetes and search for effective and economical treatments.

Insulin resistance is the initiating factor for the development of type 2 diabetes. Researchers have identified that the PI3K/AKT signaling pathway is closely associated with insulin resistance-associated diseases, including diabetes and obesity (29,30). Furthermore, the PI3K/AKT signaling pathway is one of the key pathways by which insulin regulates blood glucose balance. Reduced expression of PI3K regulatory subunit p85, as well as functional defects of PI3K regulatory subunit p85 and catalytic subunit p110, may lead to glucose and lipid metabolism disorder (31,32) which further indicates the important role of this pathway in regulating glucose and lipid metabolism.

Hepatic insulin resistance also serves a key function in the pathophysiology of diabetes. It has been reported that feeding with high-fat diet and alcohol simultaneously could induce rat liver insulin resistance through inhibition of mRNA and protein expression levels of IRS-1 and PI3K (33). Rats with non-alcoholic liver disease also present typical symptoms of fatty liver, insulin resistance and glucose metabolism disorder, and silinin, a hepatoprotective agent, may attenuate hepatic steatosis and insulin resistance caused by non-alcoholic fatty liver via the IRS-1/PI3K/AKT pathway (34). The results of the current study also suggest that there may be associations among the hepatic insulin PI3K/AKT signaling pathway, hepatic insulin resistance and diabetes mellitus.

IR is an important mediator of the insulin PI3K/AKT signaling pathway, and its downregulation or mutation may lead to reduced insulin sensitivity and insulin resistance (35,36). Consistent with this, the current study identified that IR protein and mRNA levels in the type 2 diabetic model rat liver were significantly reduced. Furthermore, IRS-1, PI3K and AKT exhibited the same trend as IR. A decrease in AKT level could decrease the release and transport of glucose transporter in the vesicles (37). A decrease in AKT could also interfere with GSK3-mediated hepatic glycogen synthesis, leading to a significant decrease in hepatic glycogen content in diabetic model rats. AKT also reduces the inhibition of gluconeogenesis (7). These three effects of AKT may finally lead to a significant increase in blood glucose levels and insulin resistance in the diabetic rats.

Currently, oral hypoglycemic drugs are primarily used for the treatment of diabetes. Although various types of hypoglycemic drugs may effectively control blood sugar and delay the progression of the disease and complications, the majority of drugs exert side effects to varying degrees, including ineffective improvement of insulin resistance or islet cell protection, and drug resistance (38). Therefore, in recent years, researchers have turned their attention to natural substances that exert hypoglycemic effects, so as to obtain a drug that has good absorption and efficacy without obvious side effects. The ‘Compendium of Materia Medica’ records that cocoons may have effects on diabetes (39). Sericin used in the current study is a natural protein in silkworm cocoons, which is coated on the outside of silk fibroin, accounting for approximately 25% of the cocoon. Sericin is usually discarded when reeling. However, cocoons soaked in boiling water have long been used to control blood glucose in Chinese folk medicine. Modern research indicates that sericin is a water-soluble protein consisting of 18 amino acids. It is biocompatible, biodegradable and non-toxic to humans, and has no obvious side effects (40). Our previous studies also identified that sericin could effectively reduce blood glucose level (16,17). However, it is not yet clear whether the PI3K/AKT signaling pathway is involved in the effect of sericin on blood glucose.

The current study observed the effect of sericin on associated factors of the PI3K/AKT signaling pathway, including IR, IRS-1, PI3K and AKT, in the liver of type 2 diabetic rats, and investigated whether sericin could reduce blood glucose and improve insulin resistance. Following administration of sericin in diabetic model rats, the expression levels of IR, IRS-1, PI3K and AKT in the model rat liver significantly increased, the blood glucose level significantly reduced and the hepatic glycogen content significantly increased.

In conclusion, the current findings demonstrate that sericin is able to increase the expression of IR, IRS-1, PI3K and AKT in the liver of diabetic rats at the gene and protein level. Sericin may regulate the PI3K/AKT signaling pathway in diabetes to enhance the transduction effect of insulin signal and promote the synthesis of hepatic glycogen. This may be the mechanism by which sericin is able to lower blood glucose and improve insulin resistance.

Acknowledgements

Not applicable.

Funding

The current study was supported by the National Natural Science Foundation of China (grant no. 81441133) and the Natural Science Foundation of Hebei Province (grant no. H2013406096).

Availability of data and materials

All data generated or analyzed in the present study are included in this published article.

Authors' contributions

CS performed all experiments. DL fed the animals, collected the samples and participated in the blood glucose test. SY participated in the immunohistochemistry experiments. LC helped with the hematoxylin and eosin staining and glycogen staining. EX participated in the polymerase chain reaction analysis. ZC designed and directed the study.

Ethics approval and consent to participate

All animal experiments were approved by the Ethics Committee of Chengde Medical University.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Chattopadhyay T, Singh RR, Gupta S and Surolia A: Bone morphogenetic protein-7 (BMP-7) augments insulin sensitivity in mice with type II diabetes mellitus by potentiating PI3K/AKT pathway. Biofactors. 43:195–209. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Chaudhury A, Duvoor C, Reddy Dendi VS, Kraleti S, Chada A, Ravilla R, Marco A, Shekhawat NS, Montales MT, Kuriakose K, et al: Clinical review of antidiabetic drugs: Implications for type 2 diabetes mellitus management. Front Endocrinol (Lausanne). 8:62017.PubMed/NCBI

3 

Horita S, Nakamura M, Suzuki M, Satoh N, Suzuki A and Seki G: Selective insulin resistance in the kidney. Biomed Res Int. 2016:58251702016. View Article : Google Scholar : PubMed/NCBI

4 

Gao YF, Zhang MN, Wang TX, Wu TC, Ai RD and Zhang ZS: Hypoglycemic effect of D-chiro-inositol in type 2 diabetes mellitus rats through the PI3K/Akt signaling pathway. Mol Cell Endocrinol. 433:26–34. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Hou XL, Wang WQ, Shi CY, Tong Q and Fang JG: Research progress in pharmacological effects of dihydromyricelin. Chinese Traditional and Herbal Drugs. 46:603–609. 2015.

6 

Gandhi GR, Jothi G, Antony PJ, Balakrishna K, Paulraj MG, Ignacimuthu S, Stalin A and Al-Dhabi NA: Gallic acid attenuates high-fat diet fed-streptozotocin-induced insulin resistance via partial agonism of PPARgamma in experimental type 2 diabetic rats and enhances glucose uptake through translocation and activation of GLUT4 in PI3K/p-Akt signaling pathway. Eur J Pharmacol. 745:201–216. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Liu TY, Shi CX, Gao R, Sun HJ, Xiong XQ, Ding L, Chen Q, Li YH, Wang JJ, Kang YM and Zhu GQ: Irisin inhibits hepatic gluconeogenesis and increases glycogen synthesis via the PI3K/Akt pathway in type 2 diabetic mice and hepatocytes. Clin Sci (Lond). 129:839–850. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Chang S: Progress in studying the relationship between PDK/Akt signal access and insulin resistance. Zhong Yi Yao Dao Bao. 14:113–116. 2008.

9 

Ahrén B, Masmiquel L, Kumar H, Sargin M, Karsbøl JD, Jacobsen SH and Chow F: Efficacy and safety of once-weekly semaglutide versus once-daily sitagliptin as an add-on to metformin, thiazolidinediones, or both, in patients with type 2 diabetes (SUSTAIN 2): A 56-week, double-blind, phase 3a, randomised trial. Lancet Diabetes Endocrinol. 5:341–354. 2017. View Article : Google Scholar : PubMed/NCBI

10 

Type 2 diabetes and metformin. First choice for monotherapy: Weak evidence of efficacy but well-known and acceptable adverse effects. Prescrire Int. 23:269–272. 2014.PubMed/NCBI

11 

Chen H, Zhu LJ, Min SJ and Hu GL: Structure, property and utilization of silk sericin. Journal of Functional Polymers. 14:344–347. 2001.

12 

Takasu Y, Yamada H, Tamura T, Sezutsu H, Mita K and Tsubouchi K: Identification and characterization of a novel sericin gene expressed in the anterior middle silk gland of the silkworm Bombyx mori. Insect Biochem Mol Biol. 37:1234–1240. 2007. View Article : Google Scholar : PubMed/NCBI

13 

Aramwit P, Siritientong T and Srichana T: Potential applications of silk sericin, a natural protein from textile industry by-products. Waste Manag Res. 30:217–224. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Okazaki Y, Kakehi S, Xu Y, Tsujimoto K, Sasaki M, Ogawa H and Kato N: Consumption of sericin reduces serum lipids, ameliorates glucose tolerance and elevates serum adiponectin in rats fed a high-fat diet. Biosci Biotechnol Biochem. 74:1534–1538. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Limpeanchob N, Trisat K, Duangjai A, Tiyaboonchai W, Pongcharoen S and Sutheerawattananonda M: Sericin reduces serum cholesterol in rats and cholesterol uptake into Caco-2 cells. J Agric Food Chem. 58:12519–12522. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Chen Z, He Y, Song C, Dong Z, Su Z and Xue J: Sericin can reduce hippocampal neuronal apoptosis by activating the Akt signal transduction pathway in a rat model of diabetes mellitus. Neural Regen Res. 7:197–201. 2012.PubMed/NCBI

17 

Song CJ, Yang ZJ, Tang QF and Chen ZH: Effects of sericin on the testicular growth hormone/insulin-like growth factor-1 axis in a rat model of type 2 diabetes. Int J Clin Exp Med. 8:10411–10419. 2015.PubMed/NCBI

18 

Hao WJ, Li JY, Jiang H, Li YH, Shen Z, Hou JJ, Xiong J and Li XR: Advantages of purified high-fat and high-glucose diet. Wei Sheng Yan Jiu. 46:143–147. 2017.

19 

Li N, Liu Q, Li XJ, Bai XH, Liu YY, Jin ZY, Jing YX, Yan ZY and Chen JX: Establishment and evaluation of a rat model of type 2 diabetes associated with depression. Zhongguo Ying Yong Sheng Li Xue Za. 31:23–26. 2015.(In Chinese).

20 

Santos EL, Dias BH, Andrade AC, Pascoal AM, Vasconcelos Filho FE, Medeiros Fd and Guimarães SB: Effects of acupuncture and electroacupuncture on estradiol-induced inflammation and oxidative stress in health rodents. Acta Cir Bras. 28:582–588. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Silachev DN, Usatikova EA, Pevzner IB, Zorova LD, Babenko VA, Gulyaev MV, Pirogov YA, Plotnikov EY and Zorov DB: Effect of anesthetics on efficiency of remote ischemic preconditioning. Biochemistry. Biokhimiia (Mosc). 82:1006–1016. 2017. View Article : Google Scholar

22 

Vollmar B, Janata J, Yamauchi JI and Menger MD: Attenuation of microvascular reperfusion injury in rat pancreas transplantation by L-arginine. Transplantation. 67:950–955. 1999. View Article : Google Scholar : PubMed/NCBI

23 

Kura RR, Kilari EK and Shaik M: Influence of aprepitant on the pharmacodynamics and pharmacokinetics of gliclazide in rats and rabbits. PeerJ. 6:e47982018. View Article : Google Scholar : PubMed/NCBI

24 

Wang NN, Liu F, Xu X, Zhang SW, Nie R, Yin DD, Liu GQ and Wang GJ: Establishment and preliminary application of fluorescence quantitative RT-PCR detection method for tambus virus. Zhong Guo Shou Yi Ke Xue. 45:15–19. 2015.

25 

Chahardah-Cherik SM, Gheibizadeh MP, Jahani SP and Cheraghian BP: The relationship between health literacy and health promoting behaviors in patients with type 2 diabetes. Int J Commun Based Nurs Midwifery. 6:65–75. 2018.

26 

Papier K, D'Este C, Bain C, Banwell C, Seubsman S, Sleigh A and Jordan S: Consumption of sugar-sweetened beverages and type 2 diabetes incidence in Thai adults: Results from an 8-year prospective study. Nutr Diabetes. 7:e2832017. View Article : Google Scholar : PubMed/NCBI

27 

Whiting DR, Guariguata L, Weil C and Shaw J: IDF diabetes atlas: Global estimates of the prevalence of diabetes for 2011 and 2030. Diabetes Res Clin Pract. 94:311–321. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Gautam S and Banerjee M: The macrophage Ox-LDL receptor, CD36 and its association with type II diabetes mellitus. Mol Genet Metab. 102:389–398. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Hu X, Wang S, Xu J, Wang DB, Chen Y and Yang GZ: Triterpenoid saponins from Stauntonia chinensis ameliorate insulin resistance via the AMP-activated protein kinase and IR/IRS-1/PI3K/Akt pathways in insulin-resistant HepG2 cells. Int J Mol Sci. 15:10446–10458. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Lu J, Chen LN and Ma X: PI3K/Akt signaling pathway and insulin resistance related diseases. Guo Wai Yi Xue (Yi Xue Di Li Fen Ce). 33:127–131. 2012.(In Chinese).

31 

Winnay JN, Dirice E, Liew CW, Kulkarni RN and Kahn CR: p85α deficiency protects β-cells from endoplasmic reticulum stress-induced apoptosis. Proc Natl Acad Sci USA. 111:1192–1197. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Nelson VL, Jiang YP, Dickman KG, Ballou LM and Lin RZ: Adipose tissue insulin resistance due to loss of PI3K p110alpha leads to decreased energy expenditure and obesity. Am J Physiol Endocrinol Metab. 306:E1205–E1216. 2014. View Article : Google Scholar : PubMed/NCBI

33 

Qu W, Hao L, Chen Y and Zhou S: Effect of chronic ethanol intake on insulin receptor, insulin receptor subsrate-1 and phosphoinositide 3-kinase mRNA expression in skeletal muscle of rats. Wei Sheng Yan Jiu. 36:172–175. 2007.(In Chinese). PubMed/NCBI

34 

Zhang Y, Hai J, Cao M, Zhang Y, Pei S, Wang J and Zhang Q: Silibinin ameliorates steatosis and insulin resistance during non-alcoholic fatty liver disease development partly through targeting IRS-1/PI3K/Akt pathway. Int Immunopharmacol. 17:714–720. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Wu QP, Xiao C, Yang XB and Zhang JM: Hypoglycemic effects of components extracted from edible and medicinal fungi and their mechanisms of action. Acta Edulis Fungi. 16:80–86. 2009.

36 

Tao MX, Wang F, Liu J, Cheng GY and Jin BQ: Hypoglycemic Effect of Pleurotus citrinopileats Polysaccharide. Food Sci. 30:227–230. 2009.

37 

Kuai M, Li Y, Sun X, Ma Z, Lin C, Jing Y, Lu Y, Chen Q, Wu X, Kong X and Bian H: A novel formula Sang-Tong-Jian improves glycometabolism and ameliorates insulin resistance by activating PI3K/AKT pathway in type 2 diabetic KKAy mice. Biomed Pharmacother. 84:1585–1594. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Bodmer M, Meier C, Krähenbühl S, Jick SS and Meier CR: Metformin, sulfonylureas, or other antidiabetes drugs and the risk of lactic acidosis or hypoglycemia: A nested case-control analysis. Diabetes Care. 31:2086–2091. 2008. View Article : Google Scholar : PubMed/NCBI

39 

Li SZ: Compendium of Materia Medica. Beijing: People's Health Publishing House; pp. 1–1052. 1985

40 

Zhang HP, Zhu LJ and Hu H: Studies on the properties of L-asparaginase immobilized on sericin protein powder. Bulletin of Sericulture. 34:16–19. 2003.

Related Articles

Journal Cover

October-2018
Volume 16 Issue 4

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
Song C, Liu D, Yang S, Cheng L, Xing E and Chen Z: Sericin enhances the insulin‑PI3K/AKT signaling pathway in the liver of a type 2 diabetes rat model. Exp Ther Med 16: 3345-3352, 2018
APA
Song, C., Liu, D., Yang, S., Cheng, L., Xing, E., & Chen, Z. (2018). Sericin enhances the insulin‑PI3K/AKT signaling pathway in the liver of a type 2 diabetes rat model. Experimental and Therapeutic Medicine, 16, 3345-3352. https://doi.org/10.3892/etm.2018.6615
MLA
Song, C., Liu, D., Yang, S., Cheng, L., Xing, E., Chen, Z."Sericin enhances the insulin‑PI3K/AKT signaling pathway in the liver of a type 2 diabetes rat model". Experimental and Therapeutic Medicine 16.4 (2018): 3345-3352.
Chicago
Song, C., Liu, D., Yang, S., Cheng, L., Xing, E., Chen, Z."Sericin enhances the insulin‑PI3K/AKT signaling pathway in the liver of a type 2 diabetes rat model". Experimental and Therapeutic Medicine 16, no. 4 (2018): 3345-3352. https://doi.org/10.3892/etm.2018.6615