Infusion of adipose‑derived mesenchymal stem cells inhibits skeletal muscle mitsugumin 53 elevation and thereby alleviates insulin resistance in type 2 diabetic rats

  • Authors:
    • Zihui Deng
    • Huiyan Xu
    • Jinying Zhang
    • Chen Yang
    • Liyuan Jin
    • Jiejie Liu
    • Haijing Song
    • Guanghui Chen
    • Weidong Han
    • Yiling Si
  • View Affiliations

  • Published online on: April 19, 2018     https://doi.org/10.3892/mmr.2018.8901
  • Pages: 8466-8474
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

It is widely accepted that infusion of mesenchymal stem cells (MSCs) ameliorates hyperglycemia by alleviating insulin resistance in rats with type 2 diabetes mellitus (T2D). However, the detailed underlying mechanisms are not clearly defined. Mitsugumin 53 (MG53) is an E3 ligase that has recently been implicated in the aggravation of insulin resistance by promoting the ubiquitinoylation of insulin receptor substrate‑1 (IRS‑1) in skeletal muscles. It was therefore hypothesized that MG53 may be involved in MSC‑mediated therapeutic effects on insulin resistance. To test this hypothesis, in the present study, T2D rat models were induced by a high‑fat diet combined with streptozotocin administration and MSC infusion was performed four times (once every 2 weeks for 8 weeks). The therapeutic effects of MSC infusion on insulin resistance were evaluated and the effect on the expression of MG53 and insulin receptor signaling elements in skeletal muscle was also investigated by immunofluorescence staining and western blotting. The results demonstrated that MSC infusion ameliorated hyperglycemia and insulin resistance in T2D rats. Furthermore, MSC infusion inhibited MG53 elevation and reversed the decreases in glucose transporter type 4, insulin receptor, IRS‑1 and phosphorylated‑AKT levels in the skeletal muscle of T2D rats. These results indicated that MSC infusion has therapeutic effects in rats and that MG53 in skeletal muscle may be a promising novel therapeutic target protein for MSC‑mediated amelioration of insulin resistance in T2D.

Introduction

Type 2 diabetes mellitus (T2D) is a major metabolic disease and is a risk to human health worldwide (1,2). T2D is characterized by reduced sensitivity of insulin receptors in target organs and absolutely or relatively insufficient secretion of insulin (3,4). These characteristics result in persistent hyperglycemia, which affects the heart, blood vessels, eyes, kidney and nerves, in addition to the wound-healing process, in diabetic patients (510). Although drugs or exogenous insulin administration can ameliorate hyperglycemia, the amelioration of peripheral insulin resistance in target tissues is not very effective.

Currently, cell-based therapy is being explored as a potential treatment strategy for diabetes (11,12). Mesenchymal stem cells (MSCs) possess various properties, including differentiation potential, local microenvironment modulatory and immunoregulatory effects, and the capacity to secrete various factors (13). These properties of MSCs make them excellent candidates for diabetes management. To date, numerous studies on diabetic animal models have demonstrated that infusion of MSCs ameliorates hyperglycemia (1417). In addition, the majority of registered clinical trials on MSC-treated type 1 and/or type 2 diabetes in phase I/II have indicated that MSC administration exerts promising therapeutic effects in diabetic volunteers (18). Furthermore, our previous study demonstrated that MSCs alleviated hyperglycemia and insulin resistance by reversing the reduced expression of glucose transporter type 4 (Glut4) and insulin receptor substrate-1 (IRS-1), as well as AKT phosphorylation, in peripheral insulin target tissues of T2D rats, including the skeletal muscle, adipose and liver (19). These results concerning the beneficial effect of MSC infusion in alleviating insulin resistance have been widely verified by different research institutions (14,2022). However, the precise underlying mechanisms require further investigation.

Insulin resistance is a key pathogenic factor that presents in several metabolic disorders, including obesity and T2D. In the peripheral insulin target tissues, skeletal muscle accounts for 70–90% of insulin-stimulated glucose metabolism (23). The insulin resistance of skeletal muscle has been the focus of numerous studies worldwide. Recent studies have demonstrated that the muscle-specific TRIM family protein mitsugumin 53 (MG53; also termed TRIM72) is implicated in insulin resistance (2427). In addition to acting as a key component of plasma membrane repair during normal cellular physiology (28,29), MG53 is also an E3 ligase that interacts with IRS-1 (24,25,27,29). Certain studies have demonstrated that MG53 expression is elevated in the skeletal muscle of rodents and humans with insulin resistance or metabolic disorders (24,27). In addition, it has been indicated that elevated MG53 in skeletal muscle may interact with and ubiquitinated IRS-1, thereby disrupting insulin signaling and inducing insulin resistance or metabolic disorders (24,27). Notably, the role of MG53 in metabolic disorders is controversial, as other studies have indicated that muscle samples derived from human diabetic patients and mice with insulin resistance exhibit normal expression of MG53 (25,26). Nevertheless, we recently reported that MG53 was elevated in the cardiac muscle of rats with diabetic cardiomyopathy (30). Therefore, the expression of MG53 in skeletal muscle during T2D should be investigated in greater detail. In addition, the aforementioned studies (19,24,27) identified that MSC infusion and MG53 may exert their effects via certain common molecules in the insulin signaling pathway, including IRS-1, AKT phosphorylation and Glut4, and our previous results indicated that MSC infusion inhibited MG53 elevation in the cardiac muscle of T2D rats (30). Therefore, it was hypothesized that MG53 in skeletal muscle may be a promising novel therapeutic target protein for MSC-mediated amelioration of insulin resistance in T2D.

To test this hypothesis and investigate the specific therapeutic mechanisms or targets involved in the beneficial effects of MSC infusion, a T2D rat model was generated and MSC infusion performed. The effects of MSC infusion on hyperglycemia, insulin resistance and MG53 expression in skeletal muscle, in addition to the expression of proteins associated with insulin signaling, were investigated. The results demonstrated that MSC infusion ameliorated hyperglycemia through improving insulin resistance. The underlying mechanisms may include the inhibition of MG53 elevation and reduced degradation of IRS-1 and phosphorylated-AKT (p-AKT) in skeletal muscle.

Materials and methods

Animals

A total of 40 adult (aged 8 weeks; weight, 210±12 g) and 20 immature (aged 4 weeks; weight, 80–100 g) male Sprague-Dawley rats were supplied by the Experimental Animal Center of the Chinese PLA General Hospital (Beijing, China). All animal experiments were conducted in accordance with the National Institutes of Health Guide for the Care and Use of Laboratory Animals (Beijing, China) and were approved by the Animal Care and Use Committee of the Chinese PLA General Hospital. Animals were maintained in a room with filtered air and a 40–70% relative humidity, with 12 h light/dark cycle and an ambient temperature of 22–25°C. Unless required to fast, animals had free access to food and water. At the end of experiments, rats were anesthetized with pentobarbital sodium intraperitoneally (60 mg/kg) and sacrificed by cervical dislocation.

Adipose-derived MSC isolation, culture and identification

Adipose-derived MSCs were isolated and purified from immature rats as described previously (17,19). Briefly, rats were anesthetized with pentobarbital sodium intraperitoneally (60 mg/kg) and sacrificed by cervical dislocation; adipose tissue isolated from the groin was digested using 0.05% trypsin and 0.1% collagen I. Following filtration and centrifugation at 600 × g for 10 min at room temperature, cells were cultured in low-glucose Dulbecco's modified Eagle's medium (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 10% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.), penicillin (80 U/ml) and streptomycin (0.2 mg/ml) at 37°C in an atmosphere of 5% CO2 and a relative humidity of ~100%. MSCs were identified as previously described (17,19). In order to perform surface immunophenotype analysis, third passage (P3) MSCs were used for flow cytometry. Once an ~80% confluence was reached, cells were collected and counted, then cells were randomly divided into six groups (one group per antibody), each containing 1×106 cells. Cells were then washed with PBS and incubated for 15 min at room temperature in the dark with the following antibodies: Allophycocyanin-conjugated CD90 (1:20; cat. no. 561409; BD Biosciences, San Jose, CA, USA), R-phycoerythrin-conjugated CD54 (1:20; cat. no. 554970; BD Biosciences), fluorescein isothiocyanate (FITC)-conjugated CD44 (1:20; cat. no. 550974; BD Biosciences), FITC-conjugated CD34 (1:20; cat. no. 11-0341-82; eBioscience; Thermo Fisher Scientific, Inc.), FITC-conjugated CD11b (1:20; cat. no. 554982; BD Biosciences) and FITC-conjugated CD45 (1:20; cat. no. 554877; BD Biosciences). Following incubation, cells were washed with PBS and then subjected to flow cytometry analysis, which was performed using a BD Accuri C6 software system (version 1.0.264.21; BD Biosciences).

In order to perform differentiation potential analysis, P3 MSCs were cultured in a six-well plate at a density of 104 cells/well at 37°C in an atmosphere of 5% CO2 and a relative humidity of ~100%. Once cells had reached a confluency of ~70 or ~100% for osteogenic or adipogenic differentiation, respectively; the medium was replaced with SD rat MSCs adipogenic (cat. no. RASMD-90031) or osteogenic (cat. no. RASMD-90021) differentiation medium (Cyagen Biosciences Inc., Guangzhou, China) and following this, the cells were subsequently cultured at 37°C in an atmosphere of 5% CO2 and a relative humidity of ~100% for 2 weeks. Following a fixation using 4% paraformaldehyde at room temperature for 30 min, adipogenic differentiation was identified by staining with 0.5% Oil red O (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) at room temperature for 1 h, and osteogenic differentiation was identified via staining with 0.1% alizarin red S (pH 4.2; Sigma-Aldrich; Merck KGaA) at room temperature for 30 min. The cells were observed using an inverted microscope. The freshly harvested, early passage (P4) MSCs were used in all subsequent experiments.

Induction and treatment of T2D rat models

A high-fat diet (HFD) combined with streptozotocin (STZ) injection-induced T2D model was established as previously described (19). Briefly, adult (8 weeks old) rats were provided with a HFD (40% fat, 41% carbohydrate and 19% protein) for 8 weeks. Subsequently, a low dose of STZ (20 mg/kg) was administrated intraperitoneally. At 1 week following STZ administration, the fasting blood-glucose (FBG) and refeeding blood-glucose levels were measured. In addition, 5 normal chow diet-induced rats and 15 HFD+STZ-induced rats were randomly chosen for oral glucose tolerance tests (OGTTs) and insulin tolerance tests (IPITTs) to verify the T2D model as previously described (19). For FBG, OGTTs and IPITTs, rats were fasted overnight prior to measurements or procedures. The verified T2D model rats were divided into a T2D group and a MSC-treated group (n=15 each). Each rat in the MSC-treated group was infused with 2×106 MSCs suspended in 0.3 ml physiological saline via the tail vein once every 2 weeks, for a total of four infusions. Rats in the normal group (n=10) were provided with a normal chow diet and only infused with 0.3 ml physiological saline.

Determination of the effects of infused MSCs on hyperglycemia and insulin resistance in T2D rats

At 24 h (refed) and 48 h (fasted) following each MSC injection, blood glucose levels were detected with a glucometer (Accu-Chek Advantage Meter; Roche Diagnostics GmbH, Mannheim, Germany), and levels were monitored throughout the whole experiment. At 1 week following the completion of the final MSC infusion, 5 rats from each group were random chosen for the OGTTs and IPITTs assessment again. Blood samples were obtained from rats by squeezing the caudal vein. Following this, serum was isolated from whole blood samples via centrifugation at 800 × g for 10 min at 4°C. The serum insulin (FINS) levels were measured using an ELISA assay kit (cat. no. EZRMI-13K; EMD Millipore, Billerica, MA, USA) 1 week following the final injection of MSCs. Additionally, at 1 week after the final MSC infusion, 5 rats from each group were random chosen for the hyperinsulinemic-euglycemic clamp studies to measure insulin sensitivity, as previously described (19). Briefly, rats were fasted overnight and 8 mU/kg/min insulin (Novo Nordisk Ltd., Bagsvaerd, Denmark) was intravenously administered. Blood glucose was monitored at 5 min intervals and exogenous glucose infusion rates (GIRs) were assessed until a steady blood glucose level was achieved. In addition, the homeostatic model assessment (HOMA) was used to assess changes in insulin resistance (HOMA-IR) and pancreatic β-cell function (HOMA-β, HBCI), calculated according to the following equations: HOMA-IR=(FBGⅹFINS)/22.5 and HOMA-β=(20ⅹFINS)/(FBG-3.5) (19).

Immunofluorescence staining

Following the final hyperglycemia/insulin resistance readings that had been taken 1 week following final MSCs injection, rats were sacrificed and muscle samples were obtained. The localization of MG53 protein in rat tibialis anterior muscle was detected by immunofluorescence staining with 5-µm-thick frozen sections, which were permeabilized at room temperature for 15 min using 0.25% TritonX-100 (cat. no. 30632-2I; FARCO Chemical Supplies, Hong Kong, SAR, China) diluted in PBS. Following this, sections were incubated with a 1X blocking buffer (cat. no. 12411; Cell Signaling Technology, Inc., Danvers, MA, USA) at 37°C for 1 h, and then initially incubated with rabbit polyclonal anti-MG53/TRIM72 antibody diluted in PBS (8 µg/ml; cat. no. ab118651; Abcam, Cambridge, UK) overnight at 4°C, followed by incubation for 1 h at 37°C with anti-rabbit IgG fragment antibody (1:200; Alexa Fluor 555-conjugated, red; cat. no. 4413; Cell Signaling Technology, Inc.). Nuclei were stained with DAPI (cat. no. H-1200; Vector Laboratories, Inc., Burlingame, CA, USA) for 15 min at room temperature. Negative controls were processed simultaneously by replacing the antibodies with PBS. The sections were examined by confocal microscopy using a Zeiss 780 system (Zeiss AG, Oberkochen, Germany).

Western blot analysis

Following the final hyperglycemia/insulin resistance readings that had been taken 1 week following final MSCs injection, rats were sacrificed and muscle samples were obtained. Whole cell lysate from the skeletal muscle was extracted using radioimmunoprecipitation assay lysis buffer (Santa Cruz Biotechnology, Inc., Dallas, TX, USA) containing 1% protease inhibitor cocktail. The protein concentration of lysate was then determined using bicinchoninic acid assay (Applygen Technologies Inc., Beijing China). For electrophoresis, a total of 30 µg protein was loaded onto an 8 or 10% SDS-PAGE gel. Following transfer to polyvinylidene difluoride membranes, the membranes were blocked in 10% non-fat milk in TBS/Tween-20 (0.2%) for 1 h at 37°C and then incubated with rabbit antibodies against MG53/TRIM72 (1:500; cat. no. ab118651; Abcam), Glut4 (1:500; cat. no. sc-7938; Santa Cruz Biotechnology, Inc.), Na+K+ATPase (1:100,000; cat. no. ab76020; Abcam), insulin receptor (1:2,000; cat. no. ab131238; Abcam), IRS-1 (1:500; cat. no. ab131487; Abcam), p-AKT (1:500; cat. no. 9271; Cell Signaling Technology, Inc.), AKT (1:1,000; cat. no. 9272; Cell Signaling Technology, Inc.) and β-actin (1:2,000; cat. no. sc-1616-R; Santa Cruz Biotechnology, Inc.) diluted in TBS/Tween-20 overnight at 4°C. Following three washes in TBS-Tween-20, the membranes were incubated with horseradish peroxidase-conjugated goat anti-rabbit IgG secondary antibodies (cat. no. TA130023; OriGene Technologies, Inc., Beijing, China) at a dilution of 1:2,000 in TBS-Tween-20 for 40 min at 37°C. The immunoreactive bands were visualized using a western blotting luminol reagent (cat. no. TA100016; OriGene Technologies, Inc.) and captured on X-ray film. Densitometric analysis was performed using a Gel-Pro Analyzer 3.0 (Media Cybernetics, Inc., Rockville, MD, USA).

Statistical analysis

SPSS software version 19.0 (IBM Corp., Armonk, NY, USA) was used to process the data. All experiments were performed at least five times independently. Data are presented as the mean ± standard deviation. Student's t-test or one-way analysis of variance with Tukey's post-hoc analysis was used when data passed the test for normality and equal variance. P<0.05 was considered to indicate a statistically significant difference.

Results

Development and validation of the T2D rat model

Rats were fed a HFD or normal chow diet for 8 weeks. No significant differences were observed in the blood glucose level between the two groups during this 8-week period (Fig. 1A and B). However, HFD induced a higher body weight compared with the normal chow diet (Fig. 1C). At 1 week following STZ administration in HFD rats, blood glucose levels significantly increased by 4–5 fold compared with normal control rats (Fig. 1A and B). In addition, the results of OGTT and IPITT experiments indicated a notable decrease in glucose tolerance and insulin sensitivity compared with normal control rats (Fig. 1D and E). These results confirmed that the T2D rat model had been established successfully.

MSC infusion alleviates hyperglycemia in T2D rats

Infused MSCs were identified in advance by their phenotypes and the potential for differentiating into adipocytes and osteoblasts (Fig. 2). For immunological phenotypes, the cells were positive for CD90, CD54 and CD44, and negative for CD34, CD11b and CD45 (Fig. 2A). Successful adipogenic and osteogenic differentiation was confirmed by oil red O and alizarin red staining, respectively (Fig. 2B). Each rat in the MSC-treated group was infused with MSCs four times (Fig. 3A). In different stage phases, untreated T2D rats demonstrated notable hyperglycemia and decreased body weight (Fig. 3). However, following the four continuous infusions of MSCs, the blood glucose demonstrated a marked decrease, almost returning to normal levels (Fig. 3B and C), and the body weight was significantly increased (Fig. 3D), compared with T2D rats without treatment. These results indicated that continuous infusion of MSCs improved the long-term regulation of blood glucose levels in T2D rats.

MSC infusion improves glucose metabolism and insulin sensitivity in T2D rats

To investigate the effects of MSC infusion on hyperglycemia alleviation in T2D rats, glucose metabolism and insulin sensitivity were compared, and hyperinsulinemic-euglycemic clamp studies performed. As demonstrated in Fig. 4, the results of OGTTs (Fig. 4A) and IPTTs (Fig. 4B) revealed a significant deterioration in glucose metabolism and insulin sensitivity in T2D rats, compared with normal control rats. In addition, the GIR was also significantly decreased in T2D rats compared with normal control rats (Fig. 5A). Consistent with hyperglycemia alleviation, MSC-treated rats demonstrated a significant improvement in glucose metabolism and were more sensitive to insulin compared with untreated T2D rats (Figs. 4 and 5A). The serum insulin levels were significantly increased in the T2D and MSC-treated T2D groups compared with the normal control group; however, no significant difference was observed between the T2D and MSC-treated T2D groups (Fig. 5B). Nevertheless, the HOMA results (IR and HBCI) indicated that insulin resistance and pancreatic β-cell function were notably improved in MSC-treated T2D rats (Fig. 5C and D). These results demonstrated that MSC-mediated hyperglycemia alleviation may be associated with improvements in target tissue insulin sensitivity.

MSC infusion inhibits MG53 elevation in the skeletal muscle of T2D rats

To further investigate the potential mechanisms of MSC-induced alleviation of insulin resistance and the potential association with MG53, MG53 protein expression in the skeletal muscle of T2D rats with or without MSC administration was assessed by immunofluorescence staining and western blotting. Immunofluorescence results demonstrated that MG53 expression was markedly elevated in the skeletal muscle of T2D rats compared with normal control rats, but MSC infusion markedly inhibited this MG53 elevation in T2D rats (Fig. 6A). The results were further confirmed by western blot analysis (Fig. 6B). These results were consistent with the amelioration of hyperglycemia and insulin sensitivity by MSC infusion, which indicates that MG53 in skeletal muscle may be a potential therapeutic target in the treatment of T2D with MSCs.

Insulin signaling elements in skeletal muscle are restored by MSC infusion in T2D rats

The IRS family of proteins serves a central role in insulin signal transduction. Studies have demonstrated that elevated MG53 in skeletal muscle may ubiquitinate IRS-1 and subsequently decrease AKT phosphorylation, which is considered to be one important mechanism of insulin resistance in T2D or metabolic disorders (24,27). Thus, in the present study, the protein expression levels of insulin receptor, IRS-1 and p-AKT in skeletal muscle were analyzed to further confirm whether skeletal muscle MG53 may be a potential therapeutic target during the treatment of T2D by MSC infusion. As demonstrated in Fig. 7A-D, consistent with the alleviation of insulin resistance and inhibition of MG53 elevation, reductions in feeding-induced expression of insulin receptor, IRS-1 and p-AKT in skeletal muscle were markedly restored in T2D rats by MSC infusion. In addition, the decrease in the expression of Glut4 in the skeletal muscle of T2D rats was also restored following MSC infusion. Therefore, these results indicate that MSC infusion in T2D may inhibit MG53 elevation, subsequently inhibiting insulin signaling element degradation and alleviating insulin resistance.

Discussion

Numerous studies and clinical trials have demonstrated that MSC infusion is able to alleviate hyperglycemia in diabetes mellitus and that MSCs are potential candidates for the treatment of T2D (11,14,18,19). Despite extensive research in this field, the specific underlying mechanisms remain poorly understood. Our previous study demonstrated that infusion of MSCs contributed to ameliorating hyperglycemia by improving peripheral insulin sensitivity in rats with T2D (19). These findings have been confirmed a number of times by different institutes (14,2022). However, the precise mechanisms remain unclear.

The mechanism of insulin resistance is complex. In previous publications, it has been emphasized that defects in the glucose signaling pathway are a major obstacle, which typically manifest as decreased expression of Glut4 and defective Glut4 traffic to the surface membrane, and disruption of phosphatidylinositol 3-kinase (PI3K)-AKT phosphorylation (31,32). Our previous study demonstrated that infusion of MSCs contributed to improving peripheral insulin sensitivity in rats with T2D by reversing the reduced expression of Glut4 and IRS-1, as well as AKT phosphorylation, in peripheral insulin target tissues, including skeletal muscle, adipose and liver tissues (19). A recent study indicated a novel mechanism by which MSCs alleviated insulin resistance, which involved the regulation of M2 macrophage polarization and promotion of interleukin-6 production in adipose tissue (33). Another study indicated that MSCs improved hyperglycemia by regulating hepatic glucose metabolism in an AMP-activated protein kinase signaling pathway-dependent manner (17). However, in the peripheral insulin target tissues, skeletal muscle accounts for ~70–90% of insulin-stimulated glucose disposal, which serves an important role in the modulation of insulin resistance (23). Previous studies have demonstrated that muscle-specific MG53 may be implicated in insulin resistance (2427). Other studies have demonstrated that in addition to acting as a key component of plasma membrane repair during normal cellular physiology (28,29), MG53 is elevated in the skeletal muscle of insulin resistance or metabolic disorder models, and that elevated MG53 may interact with and ubiquitinate IRS-1, thereby disrupting insulin signaling (24,27). By contrast, certain studies have indicated that muscle samples originating from human patients with diabetes and mice with insulin resistance did not exhibit an abnormal expression of MG53 (25,26). The key point of contention is whether MG53 expression is elevated in skeletal muscle in rodent or human metabolic disorders. Thus, further investigation is required to investigate MG53 expression and its effects on insulin resistance. We recently reported that MG53 was elevated in cardiac muscle in diabetic cardiomyopathy in rats (30). The present study indicated that MG53 was also elevated in the skeletal muscle of T2D rats. Therefore, it is hypothesized that MSC-mediated MG53 reduction in skeletal muscle may be a novel mechanism for alleviating insulin resistance. Until now, the association between MSCs and skeletal muscle MG53 expression, and the associated effects on insulin signaling, were unknown.

The results of the current study indicated that MSC infusion inhibited MG53 elevation in the skeletal muscle of T2D rats. These results are consistent with the hyperglycemia and insulin resistance alleviation effects of MSCs also observed in the present study. Thus, MG53 in skeletal muscle may be a promising novel therapeutic target protein of MSCs during their alleviation of insulin resistance in T2D. To further verify this hypothesis, the protein levels of insulin receptor, IRS-1 and p-AKT, components of the insulin signaling pathway, were analyzed by western blotting. Notably, consistent with the alleviation of insulin resistance and inhibition of MG53 elevation, the decreased expression of insulin receptor, IRS-1 and p-AKT in the skeletal muscle of T2D rats was markedly restored by MSC infusion. The IRS family of proteins (IRS-1-4) are important components in insulin signal transduction. Studies of single-gene knockout mice have demonstrated that the roles of IRS-1 and IRS-2 may be more distinctive and partially overlapping, while IRS-3 and IRS-4 do not appear to be as important in terms of the effects of insulin on glucose homeostasis (34,35). However, double knockout of IRS-1 and IRS-3 has been reported to induce severe phenotypes of diabetes, indicating a strong compensatory role of IRS-1 and IRS-3; and Laustsen et al (34) also demonstrated that the major factor in the development of this diabetic phenotype was the deficiency of IRS-1 and IRS-3 in adipose tissue and the associated decreased level of adipose-derived leptin. As IRS-3 is reported to be most abundant in adipocytes, with its mRNA also detected in the liver, heart, lungs and kidneys (34), the IRS-1 expression in skeletal muscle may be more important in regulating insulin resistance. MG53 is a muscle-specific E3-ligase that has been reported to ubiquitinate IRS-1 and subsequently inactivate the downstream PI3K-AKT signaling pathway to impair glucose homeostasis in skeletal muscle (25,27). Thus, MSC infusion may inhibit MG53 elevation and subsequently restore insulin receptor, IRS-1 and feeding-induced p-AKT levels in the skeletal muscle of T2D rats. This may explain why MSC infusion has been demonstrated to alleviate insulin resistance.

Furthermore, in the present study, the decreased expression of Glut4 in the skeletal muscle of T2D rats was also restored following MSC infusion. Previous studies have demonstrated that decreased levels of Glut4 are implicated in insulin resistance, acting as an obstacle for glucose disposal (31,32). Therefore, the results of the present study indicate that MSC infusion in T2D may inhibit MG53 elevation in skeletal muscle, subsequently inhibiting insulin signaling element degradation and alleviating insulin resistance. However, certain limitations remain that should be addressed in future studies, including the specific mechanisms involved in the negative regulation of MG53 expression by MSCs.

In conclusion, the results of the present study demonstrated that MSC infusion may ameliorate hyperglycemia by alleviating insulin resistance. The specific mechanisms involved may include inhibiting the elevation of skeletal muscle MG53 and the subsequent degradation of IRS-1 and p-AKT in skeletal muscle. These findings indicate that MG53 may be a potential therapeutic target in the treatment of T2D with MSCs.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant no. 81471052).

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

ZD, HX, WH and GC designed and conducted experiments, performed data analyses and contributed to the writing of the manuscript. JZ, CY, LJ, JL and HS conducted the experiments and performed data analyses. YS designed and conducted experiments, performed data analyses and revised the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

All animal experiments were conducted in accordance with the National Institutes of Health Guide for the Care and Use of Laboratory Animals and were approved by the Animal Care and Use Committee of the Chinese PLA General Hospital (Beijing, China).

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Chen L, Magliano DJ and Zimmet PZ: The world wide epidemiology of type 2 diabetes mellitus-present and future perspectives. Nat Rew Endocrinol. 8:228–236. 2011. View Article : Google Scholar

2 

Shaw JE, Sicree RA and Zimmet PZ: Global estimates of the prevalence of diabetes for 2010 and 2030. Diabetes Res Clin Pract. 87:4–14. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Fonseca VA: Defining and characterizing the progression of type 2 diabetes. Diabetes Care. 32 Suppl 2:S151–S156. 2009. View Article : Google Scholar : PubMed/NCBI

4 

Stumvoll M, Goldstein BJ and van Haeften TW: Type 2 diabetes: Principles of pathogenesis and therapy. Lancet. 365:1333–1346. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Russell ND and Cooper ME: 50 years forward: Mechanisms of hyperglycemia driven diabetic complications. Diabetologia. 58:1708–1714. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Wang ZV and Hill JA: Diabetic cardiomyopathy: Catabolism driving metabolism. Circulation. 131:771–773. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Shi Y and Vanhoutte PM: Macro- and microvascular endothelial dysfunction in diabetes. J Diaberes. 9:434–449. 2017. View Article : Google Scholar

8 

Shih KC, Lam KS and Tong L: A systematic review on the impact of diabetes mellitus on the ocular surface. Nutr Diabetes. 7:e2512017. View Article : Google Scholar : PubMed/NCBI

9 

Lim AKh: Diabetic nephropathy-complications and treatment. Int J Nephrol Renovasc Dis. 7:361–381. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Volmer-Thole M and Lobmann R: Neuropathy and diabetic foot syndrome. Int J Mol Sci. 17:pii: E917. 2016. View Article : Google Scholar : PubMed/NCBI

11 

Lilly MA, Davis MF, Fabie JE, Terhune EB and Gallicano GI: Current stem cell based therapies in diabetes. Am J Stem Cells. 5:87–98. 2016.PubMed/NCBI

12 

Liu X, Wang Y, Li Y and Pei X: Research status and prospect of stem cells in the treatment of diabetes mellitus. Sci China Life Sci. 56:306–312. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Orbay H, Tobita M and Mizuno H: Mesenchymal stem cells isolated from adipose and other tissues: Basic biological properties and clinical applications. Stem Cells Int. 2012:4617182012. View Article : Google Scholar : PubMed/NCBI

14 

Cao M, Pan Q, Dong H, Yuan X, Li Y, Sun Z, Dong X and Wang H: Adipose-derived mesenchymal stem cells improve glucose homeostasis in hight-fat diet-induced obese mice. Stem Cell Res The. 6:2082015. View Article : Google Scholar

15 

Hao H, Liu J, Shen J, Zhao Y, Liu H, Hou Q, Tong C, Ti D, Dong L, Cheng Y, et al: Multiple intravenous infusion of bone marrow mesenchymal stem cells reverse hyperglycemia in experimental type 2 diabetes rats. Biochem Biophys Res Commun. 436:418–423. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Aali E, Mirzamohammadi S, Ghaznavi H, Madjd Z, Larijani B, Rayegan S and Sharifi AM: A comparative study of mesenchymal stem cell transplantation with its paracrine effect on control of hyperglycemia in type 1 diabetic rats. J Diabetes Metab Disord. 13:762014. View Article : Google Scholar : PubMed/NCBI

17 

Xie M, Hao HJ, Cheng Y, Xie ZY, Yin YQ, Zhang Q, Gao JQ, Liu HY, Mu YM and Han WD: Adipose-derived mesenchymal cells ameliorate hyperglycemia through regulating hepatic glucose metabolism in type 2 diabetic rats. Biochem Biophys Res Commun. 483:435–441. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Cheng SK, Park EY, Pehar A, Rooney AC and Gallicano GI: Current progress of human trials using stem cell therapy as a treatment for diabetes mellitus. Am J Stem Cells. 5:74–86. 2016.PubMed/NCBI

19 

Si Y, Zhao Y, Hao H, Liu J, Guo Y, Mu Y, Shen J, Cheng Y, Fu X and Han W: Infusion of mesenchymal stem cells ameliorates hyperglycemia in type 2 diabetes rats: Identification of a novel role in improving insulin sensitivity. Diabetes. 61:1616–1625. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Pde Bueno G, Yochite JN, Derigge-Pisani GF, de Farias Malmegrim KC, de Avó LR, Voltarelli JC and Leal ÂM: Metabolic and pancreatic effects of bone marrow mesenchymal stem cells transplantation in mice fed high-fat diet. PLoS One. 10:e01243692015. View Article : Google Scholar : PubMed/NCBI

21 

Hughey CC, Ma L, James FD, Bracy DP, Wang Z, Wasserman DH, Rottman JN, Hittel DS and Shearer J: Mesenchymal stem cell transplantation for the infracted heart: Therapeutic potential for insulin resistance beyond the heart. Cardiovasc Diabetol. 12:1282013. View Article : Google Scholar : PubMed/NCBI

22 

Shree N and Bhonde RR: Conditioned media from adipose tissue derived mesenchymal stem cells reverse insulin resistance in cellular models. J Cell Biochem. 118:2037–2043. 2017. View Article : Google Scholar : PubMed/NCBI

23 

Shulman GI, Rothman DL, Jue T, Stein P, DeFronzo RA and Shulman RG: Quantitation of muscle glycogen synthesis in normal subjects and subjects with non-insulin-dependent diabetes by 13C nuclear magnetic resonance spectroscopy. N Engl J Med. 322:223–228. 1990. View Article : Google Scholar : PubMed/NCBI

24 

Song R, Peng W, Zhang Y, Lv F, Wu HK, Guo J, Cao Y, Pi Y, Zhang X, Jin L, et al: Central role of E3 ubiquitin ligase MG53 in insulin resistance and metabolic disorders. Nature. 494:375–379. 2013. View Article : Google Scholar : PubMed/NCBI

25 

Yi JS, Park JS, Ham YM, Nguyen N, Lee NR, Hong J, Kim BW, Lee H, Lee CS, Jeong BC, et al: MG53-induced IRS-1 ubiquitination negatively regulates skeletal myogenesis and insulin signaling. Nat Commun. 4:23542013. View Article : Google Scholar : PubMed/NCBI

26 

Ma H, Liu J, Bian Z, Cui Y, Zhou X, Zhou X, Zhang B, Adesanya TM, Yi F, Park KH, et al: Effects of metabolic syndrome on mitsugumin 53 expression and function. PLoS One. 10:e01241282015. View Article : Google Scholar : PubMed/NCBI

27 

Lee H, Park JJ, Nguyen N, Park JS, Hong J, Kim SH, Song WY, Kim HJ, Choi K, Cho S, et al: MG53-IRS-1 (Mitsugumin 53-insulin receptor substrate-1) interaction disruptor sensitizes insulin signaling in skeletal muscle. J Biol Chem. 291:26627–26635. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Cai C, Masumiya H, Weisleder N, Matsuda N, Nishi M, Hwang M, Ko JK, Lin P, Thornton A, Zhao X, et al: MG53 nucleates assembly of cell membrane repair machinery. Nat Cell Biol. 11:56–64. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Tan T, Ko YG and Ma J: Dual function of MG53 in membrane repair and insulin signaling. BMB Rep. 49:414–423. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Yang C, Deng ZH, Chen S, Chen S, Zhang JY, Jin LY, Si YL and Chen GH: Adipose-derived mesenchymal stem cells alleviating heart dysfunction through suppressing MG53 protein in rat model of diabetic cardiomyopathy. Int J Clin Exp Pathol. 10:4009–4022. 2017.

31 

Rea S and James DE: Moving GLUT4: The biogenesis and trafficking of GLUT4 storage vesicles. Diabetes. 46:1667–1677. 1997. View Article : Google Scholar : PubMed/NCBI

32 

Sylow L, Kleinert M, Pehmøller C, Prats C, Chiu TT, Klip A, Richter EA and Jensen TE: Akt and Rac1 signaling are jointly required for insulin-stimulated glucose uptake in skeletal muscle and downregulated in insulin resistance. Cell Signal. 26:323–331. 2014. View Article : Google Scholar : PubMed/NCBI

33 

Xie Z, Hao H, Tong C, Cheng Y, Liu J, Pang Y, Si Y, Guo Y, Zang L, Mu Y and Han W: Human umbilical cord-derived mesenchymal stem cells elicit macrophages into an anti-inflammatory phenotype to alleviate insulin resistance in type 2 diabetic rats. Stem Cells. 34:627–639. 2016. View Article : Google Scholar : PubMed/NCBI

34 

Laustsen PG, Michael MD, Crute BE, Cohen SE, Ueki K, Kulkarni RN, Keller SR, Lienhard GE and Kahn CR: Lipoatrophic diabetes in Irs1(−/−)/Irs3(−/−) double knockout mice. Genes Dev. 16:3213–3222. 2002. View Article : Google Scholar : PubMed/NCBI

35 

Bunner AE, Chandrasekera PC and Barnard ND: Knockout mouse models of insulin signaling: Relevance past and future. Word J Diabetes. 5:146–159. 2014. View Article : Google Scholar

Related Articles

Journal Cover

June-2018
Volume 17 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
Deng Z, Xu H, Zhang J, Yang C, Jin L, Liu J, Song H, Chen G, Han W, Si Y, Si Y, et al: Infusion of adipose‑derived mesenchymal stem cells inhibits skeletal muscle mitsugumin 53 elevation and thereby alleviates insulin resistance in type 2 diabetic rats. Mol Med Rep 17: 8466-8474, 2018
APA
Deng, Z., Xu, H., Zhang, J., Yang, C., Jin, L., Liu, J. ... Si, Y. (2018). Infusion of adipose‑derived mesenchymal stem cells inhibits skeletal muscle mitsugumin 53 elevation and thereby alleviates insulin resistance in type 2 diabetic rats. Molecular Medicine Reports, 17, 8466-8474. https://doi.org/10.3892/mmr.2018.8901
MLA
Deng, Z., Xu, H., Zhang, J., Yang, C., Jin, L., Liu, J., Song, H., Chen, G., Han, W., Si, Y."Infusion of adipose‑derived mesenchymal stem cells inhibits skeletal muscle mitsugumin 53 elevation and thereby alleviates insulin resistance in type 2 diabetic rats". Molecular Medicine Reports 17.6 (2018): 8466-8474.
Chicago
Deng, Z., Xu, H., Zhang, J., Yang, C., Jin, L., Liu, J., Song, H., Chen, G., Han, W., Si, Y."Infusion of adipose‑derived mesenchymal stem cells inhibits skeletal muscle mitsugumin 53 elevation and thereby alleviates insulin resistance in type 2 diabetic rats". Molecular Medicine Reports 17, no. 6 (2018): 8466-8474. https://doi.org/10.3892/mmr.2018.8901