Apelin‑13 ameliorates metabolic and cardiovascular disorders in a rat model of type 2 diabetes with a high‑fat diet

  • Authors:
    • Meng Li
    • Huijuan Fang
    • Jian Hu
  • View Affiliations

  • Published online on: October 29, 2018     https://doi.org/10.3892/mmr.2018.9607
  • Pages: 5784-5790
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Apelin has been reported to be associated with multiple physiological processes in the cardiovascular system. The aim of the present study was to investigate the effects of Apelin‑13 administration on cardiac function, hyperglycemia, insulin resistance (IR), dyslipidemia, endothelial function, inflammation and glucose metabolism in type 2 diabetic Goto‑Kakizaki (GK) rats, and compare the protective effects of Apelin‑13 with metformin or atorvastatin. In the present study, type 2 diabetes was induced in male Goto‑Kakizaki (GK) rats fed with high‑fat diet (HFD). Simultaneously, the rats were treated with metformin (350 mg/kg/d, by gavage), atorvastatin (50 mg/kg/d, by gavage) or Apelin‑13 (200 µg/kg/d, intraperitoneal injection) once daily for 4 consecutive weeks. Hemodynamic parameters were examined by RM6240BD multi‑channel physiological signal monitoring. Fasting plasma glucose (FPG), fasting insulin (FINS), homeostasis model assessment for insulin resistance (HOMA‑IR), total cholesterol (TC), triglyceride (TG), high density lipoprotein‑cholesterol (HDL‑C), low density lipoprotein‑cholesterol (LDL‑C), endothelin‑1 (ET‑1), nitric oxide (NO), constitutive nitric oxide synthase (cNOS) activity, tumor necrosis factor‑α (TNF‑α), leptin and Apelin‑12 levels were measured. Western blotting was performed to determine the levels of Apelin‑12, glucose transporter 4 (GLUT4) and phosphorylated (p)‑5'adenosine monophosphate‑activated protein kinase (AMPK) α2. It was demonstrated that Apelin‑13 decreased heart rate, left ventricular end‑diastolic pressure, FPG, FINS, HOMA‑IR, TC, TG, LDL‑C, ET‑1, TNF‑α and leptin, whereas it increased the rise and fall of maximum rate of left ventricular pressure, HDL‑C, NO, cNOS activity and Apelin‑12 compared with the GK‑HFD group. In addition, GLUT4 and p‑AMPKα2 levels in myocardial tissues were elevated by administration of Apelin‑13. This protective effect of Apelin‑13 was comparable to that of metformin or atorvastatin. Overall, the present study demonstrated that administration ofApelin‑13 may be a promising therapeutic agent for the treatment of type 2 diabetes and metabolic syndrome.

Introduction

Diabetes, characterized by hyperglycemia, is one of the most common chronic diseases (1). Statistical analyses have suggested that 415 million people worldwide suffer from diabetes, and the number of patients will increase to 642 million by 2040 (2,3). A total of >90% of all diabetics are diagnosed with type 2 diabetes (4). Type 2 diabetes is characterized by inadequate insulin secretion from dysfunctional β cells and insulin resistance (IR) (4,5). Previous evidence has revealed that diabetes is a predominant risk factor for cardiovascular disease (CVD) (6). Type 2 diabetes is one of the most prevalent diseases in developing and developed countries and is more susceptible to the occurrence of CVD than type 1 diabetes (7,8). Metabolic syndrome, defined as the aggregation of three or more metabolic disorders including obesity, dyslipidemia, hyperglycemia and hypertension, may also increase the risk of type 2 diabetes and CVD (9,10). Currently, the discovery of novel therapeutic agents is still of primary concern for the treatment of type 2 diabetes.

Apelin, an endogenous ligand for angiotensin II protein J (APJ), was discovered in bovine stomach tissue by Tatemoto et al in 1998 (11). Apelin, a 77-amino acid prepropeptide, can be cleaved into active formsincluding Apelin-12, −13, −17 and −36 (12). Apelin is expressed in human plasma, kidney, heart, liver, brain, adipose tissue, gastrointestinal tract and endothelium (13). Apelin/APJ is associated with multiple physiological processes in the cardiovascular system, including enhancement of cardiac contractility, relaxation of blood vessels, and regulation of blood pressure and insulin sensitivity (14,15). Metformin is one of the most widely used drugs in the treatment of type 2 diabetes (16). Atorvastatin has been reported to improve endothelial dysfunction (17). However, whether Apelin-13 has protective effects in high-fat diet (HFD)-induced type 2 diabetes in Goto-Kakizaki (GK) rats remains unclear.

The present study investigated the effects of Apelin-13 administration on cardiac function, hyperglycemia, IR, dyslipidemia, endothelial function, inflammation and glucose metabolism in type 2 diabetic GK rats.

Materials and methods

Animals and grouping

A total of 32 specific-pathogen-free (SPF), male, Goto-Kakizaki (GK) rats (12-weeks-old; 240–280 g) and a total of 8 non-diabetic, male, Wistar rats (12-weeks-old; 240–280 g) were purchased from Shanghai SLAC Laboratory Animal (Shanghai, China). The experiments were performed in accordance with the Guide for the Care and Use of Laboratory Animals and were approved by the Animal Care and Use Committee of China Medical University (Shenyang, China). Ethical clearance was obtained from the Institutional Animal Care and Use Committee (approval no. 2015052R). The animals were maintained under SPF conditions (a 12-h light/dark cycle; temperature, 21±2°C; humidity, 60±10%) with access to food and water ad libitum.

Following an adaptive feeding for 1 week, the animals were divided into 5 groups (n=8 rats/group): i) Control, ii) GK-HFD, iii) Metformin, iv) Atorvastatin and v) Apelin-13. Non-diabetic Wistar rats fed with a standard chow and treated with distilled water by gavage were used as the control rats. The GK rats in the GK-HFD, Metformin, Atorvastatin or Apelin-13 group were fed with a high-fat diet (66.5% standard chow, 10% lard, 20% sucrose, 2.5% cholesterol and 1% pig bile salt) and given distilled water, metformin (350 mg/kg/d, by gavage; Sino-American Shanghai Squibb Pharmaceuticals Ltd., Shanghai, China), atorvastatin (50 mg/kg/d, by gavage; Beijing Jialin Pharmaceutical Co., Ltd., Beijing, China) or Apelin-13 (200 µg/kg/d, intraperitoneal injection; Anaspec Inc., Fremont, CA, USA) once daily for 4 weeks simultaneously.

Hemodynamic parameters

Following 4 weeks of treatment, the rats underwent a 12 h starvation period. Then, the rats were anesthetized with 3% pentobarbital sodium (35 mg/kg; Sinopharm Group Co., Ltd., Shanghai, China) and hemodynamic parameters were monitored using RM6240BD multi-channel physiological signal monitor (Chengdu Instrument Factory, Chengdu, China), including heart rate, left ventricular end diastolic pressure (LVEDP), the maximum rate of left ventricular pressure fall (-dP/dtmax) and maximum rate of left ventricular pressure rise (+dP/dtmax).

Assessment of biochemical parameters in serum

Fasting venous blood samples were obtained from each rat and serum was obtainedby centrifugation at 1,550 × g for 10 min at 4°C. Subsequently, serum levels of fasting insulin (FINS; cat. no. F01PZA), endothelin-1 (ET-1; cat. no. D11PZA) and leptin (cat. no. C16PDA) were measured using commercial kits obtained from Beijing North Institute of Biological Technology (Beijing, China). Tumor necrosis factor-α (TNF-α; cat. no. XFFM1870) level in serum was examined using a commercial kit from Shanghai Xinfan Biotechnology Co., Ltd. (Shanghai, China). Nitric oxide (NO; cat. no. A012-1) levels and the activity of constitutive nitric oxide synthase (cNOS; cat. no. A014-1-1) were measured using kits obtained from Nanjing Jiancheng Bioengineering Institute (Nanjing, China). All procedures using commercial kits were conducted according to the manufacturer's protocol. Total cholesterol (TC), triglyceride (TG), high density lipoprotein-cholesterol (HDL-C), low density lipoprotein-cholesterol (LDL-C) levels were determined using a Beckman 700 automatic biochemical analyzer (Beckman Coulter, Inc., Brea, CA, USA).

Determination of fasting plasma glucose (FPG)

The animals were deprived of food for 12 h and blood samples were obtained from the tail vein. The levels of FPG were measured using the ACCU-CHEK Active Glucose Monitoring System (Roche Diagnostics GmbH, Mannheim, Germany) once weekly.

Measurement of homeostasis model assessment for insulin resistance (HOMA-IR)

Homeostasis model assessment for insulin resistance (HOMA-IR) was calculated using the following formula: FPGxFINS/22.5.

Apelin-12 expression by ELISA

Apelin-12 expression levels in serum, myocardial tissues and aortic tissues were examined by ELISA according to the manufacturer's protocol (cat. no. EK-057-23; Beijing Shengke Boyuan Biotechnology Co., Ltd., Beijing, China).

Reverse transcription-semi-quantitative polymerase chain reaction (RT-sqPCR) analysis

Myocardial or aortic tissues were lysed with TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA) and liquid nitrogen. Total RNAs were isolated from the tissues and RNA concentration was determined by measuring optical density260. Subsequently, total RNAs were reverse-transcribed into cDNAs and PCR was performed in a final 25 µl volume using a RT-PCR kit (Takara Biotechnology Co., Ltd., Dalian, China). The PCR conditions were: 95°C for 5 min; 35 cycles at 94°C for 30 sec, 59°C for 30 sec and 72°C for 40 sec, with a final extension at 72°C for 5 min. The PCR products were run on 1.2% agarose gels and stained with 0.5 µg/ml ethidium bromide (Amresco, LLC, Solon, OH, USA). The optical densities of the bands were quantified using a Gel Documentation system (GDS-8000; UVP, LLC, Phoenix, AZ, USA). β-actin was used as an internal control. The relative gene expression level was normalized to β-actin levels. All primers were synthesized by Beijing SBS Genetech Co., Ltd (Beijing, China) and the primer sequences were as follows: Forward, 5′-TGCTCTGGCTCTCCTTGACT-3′ and reverse, 5′-ATGGGTCCCTTATGGGAGAG-3′ for Apelin-12; forward, 5′-ATCTGGACCCACACCTTC-3′ and reverse, 5′-AGCCAGGTCCAGACGCA-3′ for β-actin.

Western blotting

Myocardial tissues were lysed using radioimmunoprecipitation assay lysis buffer, and homogenized using an ultrasonic homogenizer UP200S (Hielscher, Teltow, Germany). Following incubation on ice for 20 min, the tissue homogenates were centrifuged at 4°C at 20,000 × g for 10 min and the supernatant was harvested. Protein concentration was determined using the bicinchoninic acid assay method. Proteins were then separated by 10% SDS-PAGE and transferred onto nitrocellulose membranes (GE Healthcare, Chicago, IL, USA). Following blocking with 5% non-fat milk at room temperature for 1 h, the membranes were incubated at 4°C overnight with primary antibodies against Apelin-12 (1:1,000; cat. no. orb364270; Biorbyt LLC, San Francisco, CA, USA), glucose transporter (GLUT) 4 (1:2,000; cat. no. PA5-19621; Invitrogen; Thermo Fisher Scientific, Inc.) and phosphorylated (p)-5′adenosine monophosphate-activated protein kinase (AMPK; 1:1,000; cat. no. 18167-1-AP; Wuhan Sanying Biotechnology, Wuhan, China), followed by incubation with horseradish peroxidase-conjugated secondary antibody (1:3,000; cat. no. bs-40296G-HRP; BIOSS, Beijing, China) at room temperature for 1–2 h. Subsequently, protein bands were developed using Super ECL Plus Detection Reagent (cat. no. C05-07004; BIOSS) and quantified using Scion Image software version 1.6.3 (Scion Corporation, Frederick, MD, USA). β-actin (1:5,000; cat. no. bsm-33036M; BIOSS) served as an internal control.

Statistical analysis

Data were expressed as the means ± standard deviation. Statistical analyses were performed using GraphPad Prism version 7.0 (GraphPad Software, Inc., La Jolla, CA, USA) with one-way analysis of variance followed by Newman-keuls post-hoc test. P<0.05 was considered to indicate astatistically significant difference.

Results

Treatment with metformin, atorvastatin and Apelin-13 improves cardiac function

The results demonstrated that heart rate (Fig. 1A) and LVEDP (Fig. 1B) in the GK-HFD group were significantly increased compared with control group, whereas +dP/dtmax (Fig. 1C) and -dP/dtmax (Fig. 1D) were decreased compared with control. There was a significant decrease in heart rate and LVEDP in the Atorvastatin and Apelin-13 groups compared with the GK-HFD group, and an increase in +dP/dtmax and -dP/dtmax. Treatment with metformin significantly decreased LVEDP compared with the GK-HFD group, whereas no statistically significant differences were observed for heart rate and ±dP/dtmax.

Treatment with metformin, atorvastatin and Apelin-13 improves insulin resistance

Compared with the control group, the rats in the GK-HFD group had significantly increased levels of FPG (Fig. 2A), FINS (Fig. 2B) and HOMA-IR (Fig. 2C). However, metformin, atorvastatin and Apelin-13 treatment lowered the levels of FPG, FINS and HOMA-IR compared with the GK-HFD group.

Treatment with metformin, atorvastatin and Apelin-13 improves lipid metabolism

The present study then evaluated the effect of metformin, atorvastatin and Apelin-13 on serum levels of TC, TG, LDL-C and HDL-C in rats. The GK-HFD group demonstrated markedly increased levels of TC (Fig. 3A), TG (Fig. 3B) and LDL-C in serum (Fig. 3C) and significantly decreased HDL-C (Fig. 3D) compared with the control group. However, treatment with metformin, atorvastatin and Apelin-13 decreased serum levels of TC, TG and LDL-C and increased HDL-C in GK-HFD rats.

Effect of metformin, atorvastatin and Apelin-13 treatment on ET-1, NO and cNOS

The ET-1 level in serum (Fig. 4A) was significantly increased in the GK-HFD group compared with the control group, however was significantly decreased in the Metformin, Atorvastatin and Apelin-13 groups, compared with GK-HFD group. In addition, the GK-HFD group exhibited a significant decrease in serum NO level (Fig. 4B) and a reduction in cNOS activity (Fig. 4C). However, metformin, atorvastatin and Apelin-13 administration elevated NO level and cNOS activity.

Effect of metformin, atorvastatin and Apelin-13 treatment on serum TNF-α and leptin

Compared with the control rats, GK-HFD rats exhibited a significant increase in TNF-α (Fig. 5A) and leptin (Fig. 5B) in serum. However, the serum levels of TNF-α and leptin in the Metformin, Atorvastatin and Apelin-13 groups were significantly decreased, compared with the GK-HFD group.

Effect of metformin, atorvastatin and Apelin-13 treatment on Apelin-12 expression

Following this, the expression levels of Apelin-12 in serum, myocardial tissues and aortic tissues were measured. As presented in Fig. 6, the levels of Apelin-12 in myocardial tissues (Fig. 6A, D and F), aortic tissues (Fig. 6B and E) and serum (Fig. 6C) in the GK-HFD group were significantly decreased compared with the control group. However, treatment with metformin, atorvastatin and Apelin-13 significantly induced the expression of Apelin-12 in serum, myocardial tissues and aortic tissues, compared with the GK-HFD group.

Effect of metformin, atorvastatin and Apelin-13 treatment GLUT4 and p-AMPKα2

The levels of GLUT4 and p-AMPKα2 (Fig. 6F) in the myocardial tissues of GK-HFD rats were significantly decreased compared with control group. However, metformin and Apelin-13 injections markedly elevated GLUT4 and p-AMPKα2 levels, compared with the GK-HFD group. Atorvastatin treatment resulted in slight increases in GLUT4 and p-AMPKα2 levels; however, the differences were not statistically significant.

Discussion

The GK rat may be used as a genetic animal model of type 2 diabetes (18). In the present study, type 2 diabetes was induced in GK rats. The effects of metformin, atorvastatin and Apelin-13 on cardiac function, hyperglycemia, IR, dyslipidemia, endothelial function, inflammation and glucose metabolism in GK-HFD rats were investigated.

Hemodynamic indices, including HR, LVEDP, +dP/dtmax and -dP/dtmax, are often used to evaluate cardiac function (19). +dP/dtmax indicates systolic cardiac function, whereas LVEDP and -dP/dtmax indicate diastolic cardiac function (20). The results demonstrated that Apelin-13 decreased heart rate and LVEDP, and increased +dP/dtmax and -dP/dtmax, indicating the improvement of LV systolic and diastolic function in GK rats fed with HFD.

IR and high FPG are important risk factors for type 2 diabetes (21,22). HOMA-IR, calculated from FPG and FINS, is commonly used as a primary index for IR evaluation in the prevention of diabetes and screening of high-risk groups (23). The HOMA-IR was used to determine IR on the basis of fasted insulin and glucose levels. Elevated HOMA-IR has a positive impact on the development of type 2 diabetes in patients with impaired insulin secretion (24). In the present study, it was demonstrated that Apelin-13 significantly reduced the elevated FPG, FINS and HOMA-IR. The results suggested that Apelin-13 improved IR in the rat model of type 2 diabetes.

Dyslipidemia, characterized by increases in TC, TG and LDL-C, and decreases in HDL-C, is a common disorder in type 2 diabetics (25,26). TC and TG, two predominant types of lipids in plasma, are predictors of the balance of lipid metabolism (27). Elevated plasma HDL-C results in a cardioprotective effect, whereas higher LDL-C levels are considered an atherogenic factor (28). In the present study, it was demonstrated that Apelin-13 decreased TC, TG and LDL-C, and increased HDL-C concentration in serum, which indicated that Apelin-13 improved dyslipidemia in a rat model of type 2 diabetes.

Endothelial dysfunction has a vital role in the progression of diabetic vasculopathy and hypertension (29). It has been reported that ET-1 is overproduced in animal models of diabetes and patients (30). ET-1 is primarily produced in the endothelium, cardiomyocytes, vascular smooth muscle cells, fibroblasts, leukocytes and macrophages (31). ET-1, which is a potent peptide vasoconstrictor with proinflammatory and profibrotic properties, participates in the development of diabetic vasculopathy via regulation of vascular homeostasis (30,32,33). NO is important in various physiological processes. cNOS produces a small amount of NO and has been reported to be associated with β-cell dysfunction during the development of type 2 diabetes (34). In addition, a previous study revealed that overproduction of ET-1 may contribute to endothelial dysfunction by inhibiting NO secretion (31). The results of the present study demonstrated that Apelin-13 injection resulted in decreased levels of ET-1 and increased NO serum levels and cNOS activity. These results suggested that Apelin-13 alleviated endothelial dysfunction by regulating the imbalance of ET-1 secretion and NO production.

Diabetes is an inflammatory disease in which the levels of pro-inflammatory cytokines, including TNF-α, are elevated in the serum of patients (35,36). A previous study indicated that high levels of TNF-α is a crucial risk factor for diabetes (37). Furthermore, TNF-α is an important indicator of insulin resistance in obesity and may serve as a target for improving obesity-induced insulin resistance in patients with type 2 diabetes (38). Leptin, first discovered by Zhang et al in 1994, is a hormone that is secreted from adipose tissues and circulates in the blood (39,40). Leptin controls body weight and adipose tissue mass via regulation of energy homeostasis (41). Patients with obesity and type 2 diabetes usually have an increased plasma level of leptin and leptin resistance results in a failure to improve hyperglycemia (42). In addition, elevated leptin levels in plasma are correlated with IR, independent of insulin sensitivity and obesity (41). It was demonstrated that Apelin-13 injection reduced the increased levels of TNF-α and leptin induced by diabetes. These results suggested that Apelin-13 may alleviate diabetic disorders via inhibition of TNF-α and leptin secretion.

A previous study suggested that the circulating levels of Apelin are decreased in patients with type 2 diabetes (43). Apelin, associated with glucose uptake and IR, may promote the translocation of GLUT4 from the cytoplasm to the plasma membrane (44). GLUT4 is primarily present in cardiac, adipose and skeletal tissues, and is an insulin-regulated glucose transporter (45). Glucose is transported across the cell membrane via glucose transporters (GLUTs). GLUT4 is reported to be decreased in diabetic patients, which leads to a decrease in the uptake/utilization of glucose; whereas, cardiac contractility and metabolism are improved when GLUT4 is upregulated (46). AMPK, a conserved serine/threonine protein kinase, acts as a target for metabolic syndrome prevention (47). In the present study, it was demonstrated that Apelin-13 elevated Apelin-12 expression in serum, myocardial tissues and aortic tissues and resulted in increases in myocardial GLUT4 and p-AMPKα2 levels. These results indicated that Apelin-13 may enhance glucose metabolism and activate the AMPK signaling pathway.

In conclusion, the results of the present study demonstrated that Apelin-13 exerted beneficial effects on cardiac function, hyperglycemia, IR, dyslipidemia, endothelial function, inflammation and glucose metabolism, via upregulation of Apelin-12 and activation of the AMPK signaling pathway in type 2 diabetes. This protective effect of Apelin was comparable to that of metformin or atorvastatin. These findings indicate that Apelin-13 may be a potential therapeutic agent for the treatment of type 2 diabetes.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

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

Authors' contributions

ML and JH conceived the study and designed the experiments. ML and HJF performed the experiments and analyzed the data. ML and HJF provided reagents, materials and analysis tools. ML and JH wrote and revised the manuscript.

Ethics approval and consent to participate

The experiments were performed in accordance with the Guide for the Care and Use of Laboratory Animals and were approved by the Animal Care and Use Committee of China Medical University (Shenyang, China). Ethical clearance was obtained from the Institutional Animal Care and Use Committee (approval no. 2015052R).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Yakaryilmaz FD and Ozturk ZA: Treatment of type 2 diabetes mellitus in the elderly. World J Diabetes. 8:278–285. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Koch A, Grammatikos G, Trautmann S, Schreiber Y, Thomas D, Bruns F, Pfeilschifter J, Badenhoop K and Penna-Martinez M: Vitamin D supplementation enhances c18(dihydro)ceramide levels in type 2 diabetes patients. Int J Mol Sci. 18:E15322017. View Article : Google Scholar : PubMed/NCBI

3 

Bilir B, Bilir Ekiz B, Yilmaz I, Atile Soysal N, Yildirim T, Kara SP, Gumustas SA, Orhan AE and Aydin M: Association of apelin, endoglin and endocan with diabetic peripheral neuropathy in type 2 diabetic patients. Eur Rev Med Pharmacol Sci. 20:892–898. 2016.PubMed/NCBI

4 

Peiro C, Lorenzo O, Carraro R and Sanchez-Ferrer CF: IL-1β inhibition in cardiovascular complications associated to diabetes mellitus. Front Pharmacol. 8:3632017. View Article : Google Scholar : PubMed/NCBI

5 

Dunn JS, Mlynarski WM, Pezzolesi MG, Borowiec M, Powers C, Krolewski AS and Doria A: Examination of PPP1R3B as a candidate gene for the type 2 diabetes and MODY loci on chromosome 8p23. Ann Hum Genet. 70:587–593. 2006. View Article : Google Scholar : PubMed/NCBI

6 

Tun NN, Arunagirinathan G, Munshi SK and Pappachan JM: Diabetes mellitus and stroke: A clinical update. World J Diabetes. 8:235–248. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Shukla SK, Liu W, Sikder K, Addya S, Sarkar A, Wei Y and Rafiq K: HMGCS2 is a key ketogenic enzyme potentially involved in type 1 diabetes with high cardiovascular risk. Sci Rep. 7:45902017. View Article : Google Scholar : PubMed/NCBI

8 

Naqvi S, Naveed S, Ali Z, Ahmad SM, Khan Asadullah R, Raj H, Shariff S, Rupareliya C, Zahra F and Khan S: Correlation between glycated hemoglobin and triglyceride level in type 2 diabetes mellitus. Cureus. 9:e13472017.PubMed/NCBI

9 

Kupelian V, Hayes FJ, Link CL, Rosen R and McKinlay JB: Inverse association of testosterone and the metabolic syndrome in men is consistent across race and ethnic groups. J Clin Endocrinol Metab. 93:3403–3410. 2008. View Article : Google Scholar : PubMed/NCBI

10 

Saibandith B, Spencer JPE, Rowland IR and Commane DM: Olive polyphenols and the metabolic syndrome. Molecules. 22:E10822017. View Article : Google Scholar : PubMed/NCBI

11 

Tatemoto K, Hosoya M, Habata Y, Fujii R, Kakegawa T, Zou MX, Kawamata Y, Fukusumi S, Hinuma S, Kitada C, et al: Isolation and characterization of a novel endogenous peptide ligand for the human APJ receptor. Biochem Biophys Res Commun. 251:471–476. 1998. View Article : Google Scholar : PubMed/NCBI

12 

Cirillo P, Ziviello F, Pellegrino G, Conte S, Cimmino G, Giaquinto A, Pacifico F, Leonardi A, Golino P and Trimarco B: The adipokine apelin-13 induces expression of prothrombotic tissue factor. Thromb Haemost. 113:363–372. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Folino A, Montarolo PG, Samaja M and Rastaldo R: Effects of apelin on the cardiovascular system. Heart Fail Rev. 20:505–518. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Zhou Q, Cao J and Chen L: Apelin/APJ system: A novel therapeutic target for oxidative stress-related inflammatory diseases (Review). Int J Mol Med. 37:1159–1169. 2016. View Article : Google Scholar : PubMed/NCBI

15 

Huang S, Chen L, Lu L and Li L: The apelin-APJ axis: A novel potential therapeutic target for organ fibrosis. Clin Chim Acta. 456:81–88. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Wang YW, He SJ, Feng X, Cheng J, Luo YT, Tian L and Huang Q: Metformin: A review of its potential indications. Drug Des Devel Ther. 11:2421–2429. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Sena CM, Matafome P, Louro T, Nunes E and Seica RM: Effects of atorvastatin and insulin in vascular dysfunction associated with type 2 diabetes. Physiol Res. 63:189–197. 2014.PubMed/NCBI

18 

Howarth FC, Qureshi MA, Sobhy ZH, Parekh K, Yammahi SR, Adrian TE and Adeghate E: Structural lesions and changing pattern of expression of genes encoding cardiac muscle proteins are associated with ventricular myocyte dysfunction in type 2 diabetic goto-kakizaki rats fed a high-fat diet. Exp Physiol. 96:765–777. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Liu Y, Qi H, Wang Y, Wu M, Cao Y, Huang W, Li L, Ji Z and Sun H: Allicin protects against myocardial apoptosis and fibrosis in streptozotocin-induced diabetic rats. Phytomedicine. 19:693–698. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Wichi R, Malfitano C, Rosa K, De Souza SB, Salemi V, Mostarda C, De Angelis K and Irigoyen MC: Noninvasive and invasive evaluation of cardiac dysfunction in experimental diabetes in rodents. Cardiovasc Diabetol. 6:142007. View Article : Google Scholar : PubMed/NCBI

21 

Er LK, Wu S, Chou HH, Hsu LA, Teng MS, Sun YC and Ko YL: Triglyceride glucose-body mass index is a simple and clinically useful surrogate marker for insulin resistance in nondiabetic individuals. PLoS One. 11:e01497312016. View Article : Google Scholar : PubMed/NCBI

22 

Relimpio F: ‘The relative contributions of insulin resistance and beta-cell dysfunction to the pathophysiology of Type 2 diabetes’ by kahn se. Diabetologia. 46:17072003. View Article : Google Scholar : PubMed/NCBI

23 

Tang Q, Li X, Song P and Xu L: Optimal cut-off values for the homeostasis model assessment of insulin resistance (HOMA-IR) and pre-diabetes screening: Developments in research and prospects for the future. Drug Discov Ther. 9:380–385. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Morimoto A, Tatsumi Y, Soyano F, Miyamatsu N, Sonoda N, Godai K, Ohno Y, Noda M and Deura K: Increase in homeostasis model assessment of insulin resistance (HOMA-IR) had a strong impact on the development of type 2 diabetes in Japanese individuals with impaired insulin secretion: The Saku study. PLoS One. 9:e1058272014. View Article : Google Scholar : PubMed/NCBI

25 

Zhu XW, Deng FY and Lei SF: Meta-analysis of atherogenic index of plasma and other lipid parameters in relation to risk of type 2 diabetes mellitus. Prim Care Diabetes. 9:60–67. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Mooradian AD: Dyslipidemia in type 2 diabetes mellitus. Nat Clin Pract Endocrinol Metab. 5:150–159. 2009.PubMed/NCBI

27 

Tian L and Fu M: The relationship between high density lipoprotein subclass profile and plasma lipids concentrations. Lipids Health Dis. 9:1182010. View Article : Google Scholar : PubMed/NCBI

28 

Tian L, Liu Y, Qin Y, Long S, Xu Y and Fu M: Association of the low-density lipoprotein cholesterol/high-density lipoprotein cholesterol ratio and concentrations of plasma lipids with high-density lipoprotein subclass distribution in the Chinese population. Lipids Health Dis. 9:692010. View Article : Google Scholar : PubMed/NCBI

29 

Wong WT, Wong SL, Tian XY and Huang Y: Endothelial dysfunction: The common consequence in diabetes and hypertension. J Cardiovasc Pharmacol. 55:300–307. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Matsumoto T, Lopes RA, Taguchi K, Kobayashi T and Tostes RC: Linking the beneficial effects of current therapeutic approaches in diabetes to the vascular endothelin system. Life Sci. 118:129–135. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Kalani M: The importance of endothelin-1 for microvascular dysfunction in diabetes. Vasc Health Risk Manag. 4:1061–1068. 2008. View Article : Google Scholar : PubMed/NCBI

32 

Finch J and Conklin DJ: Air pollution-induced vascular dysfunction: Potential role of endothelin-1 (ET-1) system. Cardiovasc Toxicol. 16:260–275. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Ergul A: Endothelin-1 and diabetic complications: Focus on the vasculature. Pharmacol Res. 63:477–482. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Kaneko Kurohane Y and Ishikawa T: Dual role of nitric oxide in pancreatic β-cells. J Pharmacol Sci. 123:295–300. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Xie W and Du L: Diabetes is an inflammatory disease: Evidence from traditional chinese medicines. Diabetes Obes Metab. 13:289–301. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Ziamajidi N, Nasiri A, Abbasalipourkabir R and Moheb Sadeghi S: Effects of garlic extract on TNF-α expression and oxidative stress status in the kidneys of rats with STZ + nicotinamide-induced diabetes. Pharm Biol. 55:526–531. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Zhao L, Sun T and Wang L: Chitosan oligosaccharide improves the therapeutic efficacy of sitagliptin for the therapy of chinese elderly patients with type 2 diabetes mellitus. Ther Clin Risk Manag. 13:739–750. 2017. View Article : Google Scholar : PubMed/NCBI

38 

Liu Y, Wang X, Zhao Y, Zhao P, Wang L, Zhai Q, Zhang X, Tian W, Xiang X and Li T: Upregulation of tumor necrosis factor-α-induced protein 8-like 2 mrna is negatively correlated with serum concentrations of tumor necrosis factor-α and interleukin 6 in type 2 diabetes mellitus. J Diabetes Res. 2017:48023192017. View Article : Google Scholar : PubMed/NCBI

39 

Zhang Y, Proenca R, Maffei M, Barone M, Leopold L and Friedman JM: Positional cloning of the mouse obese gene and its human homologue. Nature. 372:425–432. 1994. View Article : Google Scholar : PubMed/NCBI

40 

Hosoi T and Ozawa K: Possible pharmacological approach targeting endoplasmic reticulum stress to ameliorate leptin resistance in obesity. Front Endocrinol (Lausanne). 7:592016.PubMed/NCBI

41 

Jacobo-Cejudo MG, Valdes-Ramos R, Guadarrama-Lopez AL, Pardo-Morales RV, Martinez-Carrillo BE and Harbige LS: Effect of n-3 polyunsaturated fatty acid supplementation on metabolic and inflammatory biomarkers in type 2 diabetes mellitus patients. Nutrients. 9:E5732017. View Article : Google Scholar : PubMed/NCBI

42 

Catalan V, Gomez-Ambrosi J, Rodriguez A, Salvador J and Fruhbeck G: Adipokines in the treatment of diabetes mellitus and obesity. Expert Opin Pharmacother. 10:239–254. 2009. View Article : Google Scholar : PubMed/NCBI

43 

Erdem G, Dogru T, Tasci I, Sonmez A and Tapan S: Low plasma apelin levels in newly diagnosed type 2 diabetes mellitus. Exp Clin Endocrinol Diabetes. 116:289–292. 2008. View Article : Google Scholar : PubMed/NCBI

44 

Zhu S, Sun F, Li W, Cao Y, Wang C, Wang Y, Liang D, Zhang R, Zhang S, Wang H and Cao F: Apelin stimulates glucose uptake through the PI3K/Akt pathway and improves insulin resistance in 3T3-L1 adipocytes. Mol Cell Biochem. 353:305–313. 2011. View Article : Google Scholar : PubMed/NCBI

45 

Shen X, Zhou N, Mi L, Hu Z, Wang L, Liu X and Zhang S: Phloretin exerts hypoglycemic effect in streptozotocin-induced diabetic rats and improves insulin resistance in vitro. Drug Des Devel Ther. 11:313–324. 2017. View Article : Google Scholar : PubMed/NCBI

46 

Williams LJ, Nye BG and Wende AR: Diabetes-related cardiac dysfunction. Endocrinol Metab (Seoul). 32:171–179. 2017. View Article : Google Scholar : PubMed/NCBI

47 

Kang OH, Shon MY, Kong R, Seo YS, Zhou T, Kim DY, Kim YS and Kwon DY: Anti-diabetic effect of black ginseng extract by augmentation of AMPK protein activity and upregulation of GLUT2 and GLUT4 expression in db/db mice. BMC Complement Altern Med. 17:3412017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2018
Volume 18 Issue 6

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
Li M, Fang H and Hu J: Apelin‑13 ameliorates metabolic and cardiovascular disorders in a rat model of type 2 diabetes with a high‑fat diet. Mol Med Rep 18: 5784-5790, 2018
APA
Li, M., Fang, H., & Hu, J. (2018). Apelin‑13 ameliorates metabolic and cardiovascular disorders in a rat model of type 2 diabetes with a high‑fat diet. Molecular Medicine Reports, 18, 5784-5790. https://doi.org/10.3892/mmr.2018.9607
MLA
Li, M., Fang, H., Hu, J."Apelin‑13 ameliorates metabolic and cardiovascular disorders in a rat model of type 2 diabetes with a high‑fat diet". Molecular Medicine Reports 18.6 (2018): 5784-5790.
Chicago
Li, M., Fang, H., Hu, J."Apelin‑13 ameliorates metabolic and cardiovascular disorders in a rat model of type 2 diabetes with a high‑fat diet". Molecular Medicine Reports 18, no. 6 (2018): 5784-5790. https://doi.org/10.3892/mmr.2018.9607