Skip to main content

Adipose stromal cells increase insulin sensitivity and decrease liver gluconeogenesis in a mouse model of type 1 diabetes mellitus

Abstract

Background

Diabetic ketoacidosis (DKA) is a serious complication of hyperglycemic emergency caused by insulin deficiency through accelerated liver gluconeogenesis and glycogenolysis. DKA is most common in type 1 diabetes (T1D). Transplantation of islet cells and pancreas is an alternative to insulin injection for treating T1D. However, this alternative is only suitable for some patients. This study investigated the effects and mechanisms of adipose stromal vascular fraction (SVF) cells on liver gluconeogenesis and insulin sensitivity in an insulin-dependent T1D animal model.

Methods

SVF cells were obtained from wild-type inguinal adipose tissue and transplanted into the peritoneal cavity of type I diabetic Akita (Ins2Akita) mice.

Results

We found that transplantation of 5 × 106 SVF cells from wild-type adipose tissue significantly downregulated proinflammatory genes of TNF-α, IL-1β, IL-33, iNOS, and DPP4 in the liver and upregulated anti-inflammatory factors IL-10 and FOXP3 in blood serum and liver tissue 7 days after injection. Moreover, we found that the expression levels of G6pc and Pck1 were significantly decreased in the Akita mice livers. Furthermore, the intraperitoneal insulin tolerance test assay showed that diabetic Akita mice significantly had increased insulin sensitivity, reduced fasting blood glucose, and restored glucose-responsive C-peptide expression compared with the control Akita group. This result was noted 14 days after administration of 5 × 106 or 1 × 107 SVF cells from wild-type adipose tissue into diabetic Akita mice.

Conclusions

Together, these findings suggest that adipose tissue-derived SVF cells could suppress liver inflammation, regulate liver gluconeogenesis, and improve insulin sensitivity in an animal model with T1D. Therefore, adipose SVF cells may be novel cellular therapeutic alternatives to maintain steady liver gluconeogenesis in T1D.

Background

Diabetes mellitus (DM) is a metabolic disorder characterized by an increase in blood glucose levels. Globally, the incidence rate of DM has dramatically increased in all countries, especially those in Africa, Southeast Asia, and the Western Pacific. For 2045, the International Diabetes Federation has warned that there will be an increase of up to 783 million people living with diabetes worldwide [1]. Therefore, it is important to prevent diabetes prevalence and monitor treatment strategies.

DM involves complex chronic-inflammation cascades that result in cellular dysfunction through several underlying processes such as hyperglycemia, insulin resistance, hyperinsulinemia, hyperlipidemia, and hyperhomocysteinemia [2, 3]. Diabetic ketoacidosis (DKA) is a form of diabetic emergency that is characterized mainly by the triad of hyperglycemia, ketosis, and anion-gap metabolic acidosis; therefore, its treatment should be immediate [4]. DKA is a life-threatening complication of DM. Abdominal pain, deep-gasping breathing, increased urination, vomiting, weakness, confusion, and loss of consciousness are the signs and symptoms of DKA [5]. Importantly, DKA usually occurs most commonly in type 1 diabetes (T1D). It may be the initial presentation in approximately 25–40% of patients with T1D.

DKA occurs because the human body lacks insulin, and elevated glucagon levels result in increased blood glucose release by liver gluconeogenesis and glycogenolysis [6]. However, gluconeogenesis or glycogenolysis regulates hepatic glucose production (HGP). Gluconeogenesis generates glucose from noncarbohydrate substrates. Glycogenolysis produces glucose through glycogen breakdown. Both processes contribute to HGP during the fasting period [7]. Hepatic gluconeogenesis is the process of glycogen generation from noncarbohydrate precursors such as lactate, pyruvate, glycerol, and glycogenic amino acids [8, 9]. Gluconeogenesis requires four critical enzymes as part of the reverse reaction of glycolysis. Pyruvate carboxylase transforms from glycolysis into oxaloacetate through decarboxylation catalyzed by phosphoenolpyruvate carboxykinase (PEPCK) to generate phosphoenolpyruvate. Fructose-1,6-bisphosphatase facilitates the conversion of fructose-1,6-bisphosphate to fructose-6-phosphate, while glucose-6-phosphatase (G6Pase) dephosphorylates glucose-6-phosphate (G6P) into glucose [7, 8, 10]. The major contributor to the high blood glucose levels observed in DM is the increased production of hepatic glucose due to enhanced gluconeogenesis [11]. Therefore, inhibition of gluconeogenesis may be the most direct route to decrease glucose production in the liver.

Over the past years, cellular therapy has become more conspicuous in the treatment of metabolic/endocrine-related diseases, degenerative disease neurological disorders, pulmonary dysfunctions, reproductive disorders, skin burns, cardiovascular conditions, and cancers [12, 13]. It is necessary to restore insulin production and glucose-dependent insulin secretion control in order to effectively cure diabetes. Insulin replacement therapy is the current standard of care for T1D; however, it is associated with substantial limitations: the risk of hypoglycemia and inability to prevent long-term complications [14]. Hence, stem cell therapy has emerged as a promising therapeutic approach for T1D because it can restore function of β cells and achieve long-term glycemic control [15]. Despite the fact that the precise therapeutic mechanism of stem cell therapies has not yet been clear, it has been shown that stem cells reduce hepatic fibrosis, accelerate liver regeneration, and rehabilitate liver function in vivo [16,17,18]. However, in clinical applications, their sustained efficacy is still in uncertain. This uncertainty may be due to several factors such as a paucity of donors, organ tissue rejection, surgical complications, reduced graft mass, and high medical costs [19]. Therefore, adipose tissue is considered an abundant source of stem cells. The stromal vascular fraction (SVF) cells are an appealing therapeutic option given that their harvesting methods are safe, fast, and they are available in large quantities from fat tissue. The adipose tissue-derived stromal vascular fraction (SVF) is a heterogeneous cell population with repair, regenerative, and immunomodulatory properties. It contains not only mesenchymal stem cells (CD45-CD31-CD29 + CD44+), but also fibroblasts, endothelial cells (CD45-CD31+), leukocytes, and pericytes [18, 20]. In a previous study using a non-alcoholic steatohepatitis mouse model, no significant changes were observed in the frequency of different cell types in the adipose tissue-derived SVFs [21]. Therefore, most experiments investigating the efficacy of SVF treatment in various models have used cell number as a method of standardization.

Few studies have investigated the role of SVF in liver gluconeogenesis, although various applications of SVF cells have been reported [20, 22]. Strategies that target the stem-like autoimmune progenitor pool present an opportunity for innovative and potent immunotherapeutic interventions in the context of T1D [23]. Therefore, in order to address this important and clinically relevant issue, we investigated the effects of adipose tissue-derived SVFs on liver gluconeogenesis in a mouse model with T1D.

Methods

Animals

The Akita strain is a monogenic model for phenotypes associated with type 1 diabetes. Six to eight weeks of pathogen-free Akita (Akita mutation mutant) (C57BL/6J background) have been purchased from Jackson Laboratory (Bar Harbor, ME) and kept at the Animal Center of Kaohsiung Veterans General Hospital. They were house in individually ventilated cages (IVC) systems with ambient lighting control to provide 12 h light/12 h dark cycles. At 3–4 weeks, the mice received genotyping by PCR for the identification genotype. The Akita mutation causes a single amino acid substitution in the insulin 2 gene that causes incorrect folding of the insulin protein [24]. Heterozygous male mice for this mutation have a progressive loss of β-cell function and develop insulin-dependent diabetes, including hyperglycemia hypoinsulinemia, polydipsia, and polyuria, as early as 4 weeks. All animal experimental procedures were designed, performed, and approved by the Institutional Animal Care and Use Committee (IACUC) of Kaohsiung Veterans General Hospital (IACUC-2408-2607-23112-NSTC). This study has been reported in line with the ARRIVE guidelines 2.0.

Isolation of SVF cells from wild-type mice adipose tissue and treatment

Stromal vascular fraction cells isolated from bilateral inguinal adipose tissue from WT mice at 12 to 16 weeks of age. The adipose tissue were minced into small pieces and then digested with 2 mg/ml collagenase 8 (Sigma-Aldrich, Cat# C2139) in HBSS for 15 min in a 37 °C water bath. Subsequently, cells were then filtered through 100 μm cell strainers and centrifuged at 1700 rpm for 10 min. After centrifugation, the cell pellets were collected and retrieved as SVF cells for the following experiments. Counting the number of SVF cells by Cellometer (Nexcelom Bioscience) and used for this study. Wild type mice received 1 ml PBS intraperitoneal injection (total n = 24). The Akita mice were received once by injection of 5 × 106 or 1 × 107 SVF cells from wild-type adipose tissue or PBS control into peritoneal cavity. Akita mice were randomly divided into three groups (total n = 36): Group I received 1 ml PBS intraperitoneal injection as controls; Group II received 5 × 106 SVF cells intraperitoneal injection; Group III received 1 × 107 SVF cells intraperitoneal injection. The mice were euthanized by CO2 gas overdose inhalation before SVFs harvesting. Otherwise, they received intraperitoneal administration of mixture of ketamine (90 mg/kg) and xylazine (10 mg/kg) at 7 or 14 days after injection. No mortality occurred outside of planned euthanasia or humane endpoints. At the end of the study, the animals were sacrificed and then harvested liver tissue and blood for analyze.

RNA isolation and quantitative real-time polymerase chain reaction (RT-QPCR)

Total RNA was extracted from mouse liver tissue using the RNA Miniprep Purification Kits (GeneMark) and processed according to the manufacturer’s instructions. Total RNA was reverse-transcribed to cDNA as the PCR template using the RT kit (Invitrogen, Carlsbad, CA, Lot# 2234812). The gene expression level was determined using primer pairs (Coralville, Iowa) by real-time PCR. For the real-time PCR assay, 200 ng of the cDNA template was added to 25 µl of the mixture containing 12.5 µl of 2X Fast SYBR Green Master Mix (Applied Biosystems, Cat# 4385612), 1.25 µl of each sense and antisense primer (25 µM) and 8 µl of sterile water. The amplification and detection were performed with an ABI Prism step-one plus sequence detection system (Applied Biosystems, New Jersey, USA). The results were analyzed using a comparative critical threshold (Ct) method in which the amount of target gene was normalized to the amount of endogenous control.

Western blotting analysis

Harvested liver tissues were homogenized in tissue extraction buffer (Sigma), containing a proteinase / phosphatase inhibitor cocktail (Roche). The homogenized samples were subjected to 10% or 8% SDS-PAGE at 50 to 100 V for 2 h and then transferred onto the PVDF membrane. After blocking and washing, the membranes were incubated with primary antibodies including DPP4 (GeneTex), iNOS, FOXP3, phosphor-JNK, phosphor-ERK, and β-actin (Cell signaling). Subsequently, the membranes were washed with TBST buffer followed by incubation with horseradish peroxidase (HRP)-labeled secondary antibodies (Jackson Immuno Research, West Baltimore Pike, PA). The blots were developed in the ImmobilonTW Western Chemiluminescence HRP substrate (Millipore, Corporation, Billerica, MA) and visualized by an enhanced chemiluminescence method.

Enzyme‑linked immunosorbent assay (ELISA)

The mouse ELISA kit (eBioscience) was used for TNF-α and IL-10 assay. Blood was centrifuged at 3000 rpm 4 °C for 10 min and then collected serum for use. The ELISA plates were coated with 100 µl capture antibody at 4 °C overnight. After washing, 200 µl of assay dilution buffer was added to block at room temperature for 1 h. Samples and serial dilutions of standards were added and incubated at room temperature for 2 h. After incubation with the detection antibody, avidin-HRP was added and incubated at room temperature for 30 min. Then, the substrate, 3,3′,5,5′-tetramethylbenzidine (TMB), was added and incubated for 20 min. Finally, 100 µl of stop solution to stop the reaction and then subjected plate to absorbance measurement at 450 nm/570 nm by ELISA reader (BioTek). C-peptide was measured using ELISA (mouse C-peptide Elisa kit; #90050; Crystal Chem) according to the manufacturer’s protocol.

Intraperitoneal insulin tolerance test (IPITT)

Mice were tested in the morning after a fasting period for 16 h. Rapid insulin (0.75 U/kg body weight; Novo Nordisk A/S) was injected intraperitoneally at time zero and tail-blood samples were collected and measured using a glucose meter (Accu-check performa; Roche, Switzerland) at 15, 30, 45, 60, 75, 90 and 120 min after insulin injection.

Statistical analysis

Data were analyzed using the unpaired t test for comparisons between two groups or by one-way analysis of variance (ANOVA) followed by Tukey’s Multiple comparison test for the comparisons between multiple groups. All values in the figures and texts were expressed as mean ± standard error of the mean, and p values less than 0.05 are considered statistically significant.

Results

Transplantation of SVF cells from the adipose tissue of wild-type mice into the peritoneal cavity of Akita mice decreases hyperglycemia-induced inflammation gene expression in the liver

Previous studies have indicated that liver inflammation is an important cause of glucose imbalance in DM [25]. Therefore, we used the heterozygous spontaneous mutation of Ins2 to induce T1D in Akita mice for experimentation. In order to investigate the effects of adipose tissue-derived SVF cells on T1D, we purified SVF cells from the inguinal adipose tissues of wild-type mice and transplanted with a different cell number of 5 × 106 or 1 × 107 of SVF cells into the peritoneal cavity of Akita mice. Previously, we have checked CD8, CD11b, and PDGFRα cells in SVFs across different experiments and found that the ratios of cells numbers of those cells in different SVFs are almost the same [26]. Therefore, we can standardize those cells with total cell numbers. The liver tissue were harvested and evaluated 7 days after cell injection. Our real-time polymerase chain reaction (qPCR) data showed that mRNA expression levels of proinflammatory genes i.e., TNF-α, IL-1β, IL-33, CCL2, iNOS, and DPP4 were significantly increased in Akita mice compared with wild-type mice (Fig. 1). Interestingly, transplantation of 5 × 106 or 1 × 107 SVF cells from the adipose tissue of wild-type mice into the peritoneal cavity of Akita mice significantly downregulated proinflammatory genes of TNF-α, IL-1β, IL-33, and iNOS and DPP4 mRNA expression levels in the liver (Fig. 1). Taken together, these results suggest that Akita mice exhibited an increase in proinflammatory gene expression, whereas transplantation of adipose tissue-derived SVF cells from wild-type mice reduced proinflammatory gene expression in the liver of a mouse model with T1D.

Fig. 1
figure 1

Therapeutic effects of transplantation of 5 × 106 or 1 × 107 adipose tissue-derived stromal vascular fraction (SVF) cells from adipose tissue of wild-type mice in the liver in an Akita mouse model with type 1 diabetes. Adipose tissue-derived SVF cells were harvested from the adipose tissue of wild-type mice and then transplanted with a cell number of 5 × 106 or 1 × 107 into the peritoneal cavity of Akita mice. Subsequently, the liver was excised and the mRNA expression level of TNF-α, IL-1β, IL-33, CCL2, iNOS, and DPP4 were determined by RT-qPCR after cell transplantation on day 7. *p < 0.05; **indicates p < 0.01 as compared with control Akita; n = 5/group

Transplantation of SVF cells from wild-type mice into the peritoneal cavity of Akita mice reduces the protein expression levels of TNF-α, iNOS, and DPP4 in the liver

To determine the effects of adipose SVF cells from wild-type mice on the proinflammatory protein expression levels of TNF-α, iNOS, and DPP4 in the liver of Akita mice, we examined these protein expression levels by enzyme-linked immunosorbent assay (ELISA) and immunoblotting at 7 days after cell transplantation. As shown in our results, Akita mice exhibited a higher expression level of TNF-α compared with wild-type mice (5.09 ± 4.39 vs. 65.57 ± 1.1 pg/ml) (Fig. 2a). Transplantation of 5 × 106 or 1 × 107 SVF cells from the adipose tissue of wild-type mice into the peritoneal cavity of Akita mice significantly decreased the TNF-α expression level (41.48 ± 2.19 vs. 65.57 ± 1.1; 50.66 ± 1.1 vs. 65.57 ± 1.1 pg/ml) in the liver (Fig. 2a). Moreover, immunoblotting demonstrated that the protein expression levels of DPP4 and iNOS were significantly increased in Akita mice compared with those in wild-type mice. After transplantation of 5 × 106 adipose tissue-derived SVF cells from wild-type mice into the peritoneal cavity of Akita mice, DPP4 and iNOS protein expression levels in the liver tissue significantly decreased. Meanwhile, the same transplantation of 1 × 107 adipose tissue-derived SVF cells significantly decreased protein expression levels of DPP4 (Fig. 2). Altogether, these data suggest that Akita mice exhibited heightened levels of proinflammatory markers, such as TNF-α, DPP4, and iNOS, in their liver tissue compared with wild-type mice. Therefore, transplantation of SVF cells from the adipose tissue of wild-type mice into Akita mice is capable of suppressing inflammation in the liver.

Fig. 2
figure 2

Effects of transplantation of stromal vascular fraction (SVF) cells from wild-type adipose tissue into diabetic Akita mice on the protein expression levels of TNF-α, DPP4, and iNOS in the liver. Adipose tissue-derived SVF cells were harvested from the adipose tissue of wild-type mice and then transplanted with a cell number of 5 × 106 or 1 × 107 into the peritoneal cavity of Akita mice. Next, the liver tissue was collected and its protein extracted after cell transplantation for 7 days. The protein extract was then subjected to enzyme-linked immunosorbent assay to detect TNF-α expression (a) or western blotting analysis using antibodies against DPP4, iNOS and β-actin (b). (c) Densitometric analyses of (b) n = 3. *indicates p < 0.05; **indicates p < 0.01 as compared to control Akita

Transplantation of SVF cells from wild-type mice into akita mice induces IL-10 expression in the liver and blood

IL-10 is known to inhibit proinflammatory cytokines and antigen-presenting cells, promote tissue repair mechanisms, and play an important role in restricting excessive inflammatory responses [27, 28]. We investigated IL-10 expression levels in liver and blood of Akita mice after transplantation of 5 × 106 or 1 × 107 SVF cells from wild-type mice for 7 days by qPCR and ELISA, respectively. This step was performed to determine whether IL-10 is involved in the mechanism of SVF cell-induced reduction of liver inflammation in Akita mice. Our data showed that the expression level of IL-10 mRNA was significantly lower in Akita mice than those in wild-type mice (Fig. 3a). Transplantation of 5 × 106 or 1 × 107 SVF cells from wild-type mice into the peritoneal cavity of Akita mice significantly increased IL-10 mRNA expression in the liver (Fig. 3a). Similarly, transplantation of 5 × 106 or 1 × 107 SVF cells from the adipose tissue of wild-type mice into the peritoneal cavity of Akita mice significantly increased blood IL-10 levels (30.8 ± 11.7 vs. 13.2 ± 2.5; 27.4 ± 7.5 vs. 13.2 ± 2.5 pg/ml) at 7 days after cell transplantation (Fig. 3b).

Fig. 3
figure 3

Effects of transplantation of stromal vascular fraction (SVF) cells from wild-type adipose tissue into diabetic Akita mice on IL-10 mRNA expression in the liver and blood IL-10 levels. Adipose tissue-derived SVF cells were extracted from the adipose tissue of wild-type mice and then transplanted with a cell number of 5 × 106 or 1 × 107 into the peritoneal cavity of Akita mice. One week later, liver tissue was harvested and mRNA expression levels were determined by real-time polymerase chain reaction. Circulating blood was collected at 7 days after cell transplantation and then subjected to enzyme-linked immunosorbent assay to detect IL-10 levels. *indicates p < 0.05; **indicates p < 0.01

Transplantation of SVF cells from wild-type mice into akita mice induces anti-inflammatory factor Treg expression in the liver

Induction of IL-10 has been shown to be an important mediator of Treg suppression and promoter of Treg differentiation [29]. Therefore, after proving that our transplantation experiments upregulated IL-10 expression, we performed the same procedure to check for promotion of Treg differentiation in the liver. Based on our results, the expression level of Foxp3 (specification factor of Treg cells) was significantly lower in Akita mice than that in wild-type mice. Transplantation of 5 × 106 or 1 × 107 SVF cells from wild-type mice adipose tissue into the peritoneal cavity of Akita mice significantly increased Foxp3 expression level (Fig. 4a) compared with their control Akita counterparts. Subsequently, we observed that transplantation of 5 × 106 or 1 × 107 SVF cells from wild-type adipose tissues significantly increased the protein expression level of FOXP3 and p-ERK in Akita mice at 7 days after cell transplantation (Fig. 4b and c). Collectively, these data suggest that Akita mice exhibited a decrease in Treg expression, whereas transplantation of 5 × 106 or 1 × 107 adipose tissue-derived SVF cells from wild-type mice enhanced anti-inflammatory mediators, such as IL-10 and Foxp3 expression in the liver to mitigate hyperglycemia-induced inflammation.

Fig. 4
figure 4

Transplantation of stromal vascular fraction (SVF) cells from wild-type adipose tissue into diabetic Akita mice increased Foxp3 expression in the liver. Adipose tissue-derived SVF cells were harvested from the adipose tissue of wild-type mice and then transplanted with a cell number of 5 × 106 or 1 × 107 into the peritoneal cavity of Akita mice. Subsequently, liver tissue was harvested and Foxp3 expression level was elevated at 7 days after cell injection by (a) qPCR and analyzed with western blotting (b) using antibodies against Foxp3, p-ERK, ERK, p-JNK, JNK and β-actin. (c) The ratios of Foxp3/β-actin, p-ERK/ERK and p-JNK/JNK were also calculated. n = 3. *indicates p < 0.05; **indicates p < 0.01

Transplantation of SVF cells from wild-type mice into akita mice suppresses the hepatic gluconeogenesis gene G6pc and Pck1 expression

As indicated by Toda et al., IL-10 signaling is necessary for the normal suppression of gluconeogenic gene expression. Also, physiological concentrations of IL-10 and insulin can suppress glucose production in primary hepatocytes [30]. Inhibition of IL-10 promotes increased expression of inflammatory cytokines, worsens insulin signaling, and activates glucogenic and lipogenic pathways [31]. The gluconeogenic enzymes PEPCK and the catalytic subunit of G6Pase are key factors in hepatic gluconeogenesis [10]. As demonstrated in Fig. 5, the gluconeogenesis process of G6pc and Pck1 mRNA expression level in the liver significantly increased in Akita mice that that with wild-type mice by qPCR. Intriguingly, transplantation of 5 × 106 or 1 × 107 SVF cells from wild-type adipose tissue into diabetic Akita mice significantly reduced the gluconeogenic genes of G6pc and Pck1 expression in the liver compared with the control Akita mice group. These data suggest that Akita mice exhibited an increase in the expression level of gluconeogenic genes such as G6pc and Pck1 in the liver. Transplantation of 5 × 106 or 1 × 107 SVF cells from wild-type adipose tissue attenuates liver gluconeogenesis to become more stable in this mouse model with T1D.

Fig. 5
figure 5

Transplantation of stromal vascular fraction (SVF) cells from wild-type adipose tissue into diabetic Akita mice decreased G6pc and Pck1 expression in the liver. Adipose tissue-derived SVF cells with a cell number of 5 × 106 or 1 × 107 were harvested from the adipose tissue of wild-type mice and then transplanted into the peritoneal cavity of Akita mice. One week later, the expressions of G6pc and Pck1 mRNA in the liver were evaluated by RT-qPCR. *indicates p < 0.05; **indicates p < 0.01 as compared to control Akita; n = 5/group

Transplantation of SVF cells from wild-type mice into akita mice improves insulin sensitivity and regulates blood glucose homeostasis

In this context, transplantation of adipose tissue-derived SVF cells from wild-type mice adipose tissue decreases liver gluconeogenesis in Akita mice with diabetes. Next, we determined the effects of functional alteration on transplantation of 5 × 106 or 1 × 107 SVF cells from wild-type mice adipose tissue into the peritoneal cavity of Akita mice. After 16 h of fasting, blood glucose levels were significantly higher in Akita mice than those in wild-type mice (493.2 ± 65.5 vs. 88 ± 16.3 mg/dL). Importantly, transplantation of 5 × 106 or 1 × 107 adipose tissue-derived SVF cells from wild-type mice into diabetic Akita mice significantly decreased the fasting blood glucose levels at 14 days compared with control Akita mice (270.6 ± 39.3 vs. 493.2 ± 65.5; 346.7 ± 56.1 vs. 493.2 ± 65.5 mg/dL) (Fig. 6a). Furthermore, in order to evaluate the effects of SVF cells on diabetic Akita mice with reduced insulin sensitivity, the intraperitoneal insulin tolerance test assay was performed in Akita mice after transplantation of 5 × 106 or 1 × 107 adipose tissue-derived SVF cells from wild-type mice. We found that Akita mice exhibited a significant decrease in insulin sensitivity compared with wild-type mice. Transplantation of 5 × 106 or 1 × 107 adipose tissue-derived SVF cells from wild-type mice into diabetic Akita mice significantly increased insulin sensitivity compared with control Akita group on day 14 (Fig. 6b). Moreover, we also examined the levels of C-peptide in the blood serum 14 days after cell transplantation. The glucose-responsive level of the C-peptide in Akita mice significantly decreased compared with its wild-type counterpart (4.7 ± 6.3 vs. 1,080.4 ± 314.3 pg/ml). Transplantation of 5 × 106 or 1 × 107 adipose tissue-derived SVF cells from wild-type mice significantly increased the C-peptide level compared with control diabetic Akita mice (298.8 ± 123.9 vs. 4.7 ± 6.3; 340.5 ± 159.5 vs. 4.7 ± 6.3 pg/ml) (Fig. 6c). These data suggest that Akita mice presented high expression levels of fasting blood glucose and decreased insulin sensitivity. At the same time, transplantation of 5 × 106 or 1 × 107 adipose tissue-derived SVF cells from wild-type mice improves insulin sensitivity in diabetic Akita mice and modulates hyperglycemia-induced glucose imbalance.

Fig. 6
figure 6

Effects of transplantation of stromal vascular fraction (SVF) cells from the adipose tissue of wild-type mice into diabetic Akita mice on blood glucose, insulin sensitivity, and C-peptide levels. Adipose tissue-derived SVF cells were harvested from the adipose tissue of wild-type mice and then transplanted with a cell number of 5 × 106 or 1 × 107 into the peritoneal cavity of Akita mice for 2 weeks. Consecutively, after a 16-h fasting period, mice were administered insulin (0.75 mIU/g body weight) for 15 min. Blood glucose levels were measured (a) before and (b) every 15 min up to 2 h after insulin administration using a glucose meter. * and # summarize the significance between Akita-SVF(5 × 106) and Akita, Akita-SVF(1 × 107) and Akita, respectively. (c) The levels of C-peptide in serum were detected by enzyme-linked immunosorbent assay. n = 4/group. *indicates p < 0.05; **indicates p < 0.01; *** indicates p < 0.001

Discussion

T1D is considered an autoimmune disease that selectively attacks insulin-producing pancreatic β cells resulting in insulin production and secretion [32]. Without insulin, glucose homeostasis and energy metabolism balance are completely disrupted. In order for blood glucose homeostasis to be maintained, HGP is regulated by glycogenolysis or gluconeogenesis [7]. Accelerated imbalance hepatic gluconeogenesis of DKA is a medical emergency due to insulin deficiency and is the leading cause of death in patients with T1D [33]. In this study, we demonstrated that transplantation of adipose tissue-derived SVF cells from wild-type mice into the peritoneal cavity of diabetic Akita mice significantly inhibited the most important enzymes G6pc and Pck1 in the gluconeogenesis process and regulated fasting blood glucose levels (Fig. 6a). These data suggested that enhanced gluconeogenesis-induced glucose production in the liver is the major contributor to the high blood glucose levels in DM, and inhibiting gluconeogenesis directly decreases glucose production in the liver after cell transplantation. However, Norlin et al. demonstrated that the AMPK activator O304 improves glucose uptake and utilization in the skeletal muscle even in the absence of insulin and alleviates glycogen accumulation caused by hyperglycemia in both the skeletal muscle and heart in vivo of T2D [34]. Furthermore, the skeletal muscles, adipocytes, cardiovascular cells, and pancreatic beta cells exhibit an intricate relationship in glucose homeostasis and systemic insulin resistance. This is an intriguing topic worthy of further investigation on the transplantation effect of SVF cells.

Toda et al. demonstrated that IL-10 derived from adipose macrophages suppresses hepatic gluconeogenesis and reduces HGP in cooperation with insulin [30]. In contrast, inhibition of IL-10 leads to increased expression of inflammatory cytokine, deteriorated insulin signaling, and activated glucogenic and lipogenic pathways [31]. Endogenous IL-10 is a protective factor against diet-induced insulin resistance in the liver [35]. We demonstrated previously that adipose tissue-derived SVF plasma reverses insulin resistance by increasing the expression of M2 cytokines in adipose tissue from type 2 diabetic mice [22]. We demonstrated in this study that adipose tissue-derived SVF cells from wild-type mice significantly increased IL-10 expression in the liver and blood circulation in an animal model with T1D. Note that there is a higher level of expression of IL-10 in human adipose tissue-derived SVF cells than those in adipose tissue-derived mesenchymal stem cells to inhibit inflammatory cell infiltration and induce regeneration in mice liver cirrhosis [18]. The therapeutic function of SVF cells depends on not only the local effects but also the indirect effects of immune regulatory factors or cytokine secretion through multiple mechanisms. SVF cells have immunomodulatory functions and can inhibit the production of proinflammatory cytokines by secreting the anti-inflammatory factor IL-10 or directly regulating the activity of macrophages and T cells [36]. Furthermore, some studies [37, 38] have indicated that SVF cells increase insulin sensitivity by improving insulin signaling pathways (such as IGF-1 and HGF). SVF cells also improve insulin secretion and glucose regulation by promoting the regeneration and functional recovery of pancreatic β-cells [39]. According to these studies, adipose tissue-derived SVF cells could be a novel strategy to increase anti-inflammatory factor IL-10 and improve hepatic gluconeogenesis balance in hyperglycemia.

For several decades, T1D was believed to be a chronic autoimmune disease due to an immune attack that decreases insulin secretion of islets β cells in the pancreas [40,41,42]. Regulatory T cells (Tregs) also exhibit dysfunction in this autoimmune disorder. Both CD4+ and CD8+ T cells are implicated in the development of T1D because they target several beta cell autoantigens and related peptide epitopes [43,44,45,46]. Moreover, a previous study showed that plasma DPP4 levels and DPP4 mRNA expression in rat tissues increased progressively during the development of streptozotocin-induced T1D [47]. During this process, the control and regulation of local inflammatory cytokine production are likely to be critical factors in determining the outcome of autoimmune progression [46]. Therefore, several immunotherapeutic clinical trials have been completed recently in human T1D, such as monoclonal anti-CD3 (teplizumab and otelixizumab) and anti-CD20 antibodies (rituximab) as well as anticytokines of IL-1β (canakinumab and anakinra) and TNF-α (etanercept). Targeting these factors is likely to preserve the remaining β-cell function, but curative treatments can only be realistically achieved by simultaneously trying to replace the lost part of the β-cell mass (lost during the autoimmune process) [46]. Thus far, immunotherapy alone has proven insufficient to achieve lasting preservation of β-cell function, which indicates the need to combine other strategies with β-cell therapy [32]. However, in this study, we demonstrated that transplantation of adipose SVF cells could also significantly reduce liver inflammation-related expression of TNF-α, IL-33, iNOS, and DPP4, as well as increase systemic anti-inflammatory factor IL-10 and liver Treg expression in diabetic Akita mice. As previously mentioned, certain studies demonstrated that mesenchymal stem cells (MSCs) could improve diabetic hyperglycemia and increase the number of cells of pancreatic islet β cells in mice and rats [48, 49]. Ezquer et al. demonstrated that injecting murine bone marrow multipotent mesenchymal stromal cells into STZ-treated mice increases insulin levels and significantly reduces hyperglycemia, with these effects lasting for at least 2 months [50]. Fluorescence tracing confirmed that MSCs do not directly into pancreatic progenitors but resulted in secondary lymphoid organs after their graft in vivo [50, 51]. However, it restored both systemic and local regulatory T-cell balance, increased the production of the anti-inflammatory factor IL-13, and reduced the levels of the proinflammatory factors IL-1β, IL-18, TNF-α, and MCP-1 [51]. Mice also showed an increase in the levels of epidermal growth factor (EGF), a pancreatic trophic factor involved in cell survival, and total insulin. These data imply that the reduction of inflammation is responsible for at least some of the beneficial effects of MSC treatment. Although MSCs are emerging as the most promising source for allogeneic cell therapy [52], their therapeutic use in T1D clinical trials remains highly controversial [53]. Intriguingly, we demonstrated that transplantation of adipose tissue-derived SVF cells into Akita mice not only showed a high anti-inflammatory property but also increased insulin sensitivity. It also induced the expression of glucose-response c-peptides (Fig. 6). In this context, this evidence suggests that stem cells in adipose tissue-derived SVF cells may promote the repair ability or regulate immune mediators in damaged, injured, or dysfunctional cells to modulate T1D. This may be due to the multifaceted nature of adipose tissue-derived SVF cells, which contain pluripotency stem cells that differentiate into insulin-positive cells and maintain survival of β cells in the pancreas via modulation of immunogenicity. Further study is needed to clarify their roles more elaborately.

Previous studies have also reported that fibroblast growth factor 21 (FGF21) plays a crucial role in regulating liver gluconeogenesis, and the liver is the only major organ that produces circulating FGF21 in T1D [54, 55]. Glucose transport activity in the target tissue stimulated by FGF21 is accomplished mainly by the upregulation of GLUT1 transcription [56]. The basal level of FGF21 was significantly lower in T1D patients than that in healthy controls [57]. Wente et al. demonstrated that continuous administration of FGF21 increased the activity of pancreatic β cells by activating extracellular signal-regulated kinase 1/2 (ERK1/2) and protein kinase B in animal models with diabetes [58]. As demonstrated in this study (Fig. 4), transplantation of adipose tissue-derived SVF cells into Akita mice could significantly increase phosphorylated ERK expression compared with control Akita mice. Although the role of FGF21 in this study was not explored, it may be another reason why adipose SVF cells transplanted could reverse the biological effects in the liver.

However, transplantation of adipose tissue-derived SVF cells from wild-type mice does not completely reverse the effects of hyperglycemia-induced liver dysfunction in this study. Therefore, other mechanisms were involved in our experiment. Similarly, the number of adipose tissue-derived SVF cells was transplanted in a dose-dependent manner in this mouse model. Kim et al. indicated that cell number density is a crucial factor in inducing intracellular reactive oxygen species (ROS), leading to senescence of stem cells derived from human bone marrow [59]. Other studies also demonstrated that low Treg cell number density had high growth rates of viability and FOXP3 expression associated with expression of inhibitory molecules, lower intracellular oxygen, extracellular nutrient concentrations, and extracellular lactate accumulation on human Treg production in vitro [60]. Environmental factors such as ROS effects should be considered in cell transplantation medical efficacy of T1D treatment in the future.

Furthermore, another limitation of this study is that we focused on the biological effect within 2 weeks by transplanting adipose tissue-derived SVF cells into Akita mice for early intervention in T1D, and we did not extend the observation period beyond this time frame. Because SVF treatments often mediate short term effects, the longevity of the observed benefits remains an important question. In future studies, we intend to extend the observation period to determine whether the beneficial effects persist beyond 14 days. This may provide a clearer understanding of the long-term efficacy of SVF treatment. Additionally, we used cell number as a method of standardization, but we did not examine the frequency of different cell types in the adipose-derived SVFs in our model.

Conclusion

Transplantation of adipose tissue-derived SVF cells from wild-type mice into peritoneal cavity of diabetic Akita mice decreased the mRNA expression of proinflammatory-related genes; reduced the protein expression of TNF-α, DPP4, and iNOS; and increased the expression of IL-10 and Foxp3 in the liver. This transplantation also reduced the expression of the liver gluconeogenesis genes G6pc and Pck1 and increased C-peptide levels. This study presents that such transplantation inhibited hepatic gluconeogenesis, improved insulin sensitivity, maintained blood glucose stability, and regulated the immune response in a T1D mouse model. Furthermore, adipose tissue-derived SVF cells are easy to harvest and purify. This research provided a novel clinical approach to cell therapy that is simpler, more cost-effective, safer, and scalable than insulin replacement therapy. This novel approach would reduce the occurrence of severe complications associated with T1D, such as acute DKA.

Data availability

All relevant data and material to reproduce the findings are available within the paper.

Abbreviations

DM:

Diabetes mellitus

DKA:

Diabetic ketoacidosis

SVFs:

Stromal vascular fractions

HGP:

Hepatic glucose production

PC:

Pyruvate carboxylase

PEPCK:

Phosphoenolpyruvate carboxykinase

G6Pase:

Glucose 6-phosphatase

TNF:

Tumor Necrosis Factor-α

IL-1β:

Interleukin-1β

IL-33:

Interleukin-33

CCL2:

Monocyte chemoattractant protein-1

iNOS:

Inducible nitric oxide synthase

DPP-4:

Dipeptidyl peptidase-4

IL-10:

Interleukin-10

Foxp3:

Forkhead box protein P3

MSC:

Mesenchymal stem cells

FGF21:

Fibroblast growth factor 21

References

  1. Sun H, Saeedi P, Karuranga S, Pinkepank M, Ogurtsova K, Duncan BB, et al. Idf Diabetes atlas: global, regional and country-level diabetes prevalence estimates for 2021 and projections for 2045. Diabetes Res Clin Pract. 2022;183:109119.

    Article  PubMed  Google Scholar 

  2. Kampoli AM, Tousoulis D, Briasoulis A, Latsios G, Papageorgiou N, Stefanadis C. Potential pathogenic inflammatory mechanisms of endothelial dysfunction induced by type 2 diabetes mellitus. Curr Pharm Des. 2011;17(37):4147–58.

    Article  PubMed  CAS  Google Scholar 

  3. Foster NC, Beck RW, Miller KM, Clements MA, Rickels MR, DiMeglio LA, et al. State of type 1 diabetes management and outcomes from the t1d exchange in 2016–2018. Diabetes Technol Ther. 2019;21(2):66–72.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Calimag APP, Chlebek S, Lerma EV, Chaiban JT. Diabetic ketoacidosis. Dis Mon. 2023;69(3):101418.

    Article  PubMed  Google Scholar 

  5. Demirci H, Cosar R, Ciftci O, Sari IK. Atypical diabetic ketoacidosis: Case report. Balkan Med J. 2015;32(1):124–6.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Calimag APP, Chlebek S, Lerma EV, Chaiban JT. Diabetic ketoacidosis. Dis Mon 2022:101418.

  7. Tao Y, Jiang Q, Wang Q. Adipose tissue macrophages in remote modulation of hepatic glucose production. Front Immunol. 2022;13:998947.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Nuttall FQ, Ngo A, Gannon MC. Regulation of hepatic glucose production and the role of gluconeogenesis in humans: is the rate of gluconeogenesis constant? Diabetes Metab Res Rev. 2008;24(6):438–58.

    Article  PubMed  CAS  Google Scholar 

  9. McCommis KS, Finck BN. Mitochondrial pyruvate transport: a historical perspective and future research directions. Biochem J. 2015;466(3):443–54.

    Article  PubMed  CAS  Google Scholar 

  10. Hatting M, Tavares CDJ, Sharabi K, Rines AK, Puigserver P. Insulin regulation of gluconeogenesis. Ann N Y Acad Sci. 2018;1411(1):21–35.

    Article  PubMed  CAS  Google Scholar 

  11. Sharabi K, Tavares CD, Rines AK, Puigserver P. Molecular pathophysiology of hepatic glucose production. Mol Aspects Med. 2015;46:21–33.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Hoang DM, Pham PT, Bach TQ, Ngo ATL, Nguyen QT, Phan TTK, et al. Stem cell-based therapy for human diseases. Signal Transduct Target Ther. 2022;7(1):272.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Sterner RC, Sterner RM. Car-t cell therapy: current limitations and potential strategies. Blood Cancer J. 2021;11(4):69.

    Article  PubMed  PubMed Central  Google Scholar 

  14. McCall AL, Farhy LS. Treating type 1 diabetes: from strategies for insulin delivery to dual hormonal control. Minerva Endocrinol. 2013;38(2):145–63.

    PubMed  PubMed Central  CAS  Google Scholar 

  15. Singh A, Afshan N, Singh A, Singh SK, Yadav S, Kumar M, Sarma DK, Verma V. Recent trends and advances in type 1 diabetes therapeutics: a comprehensive review. Eur J Cell Biol. 2023;102(2):151329.

    Article  PubMed  CAS  Google Scholar 

  16. Higashiyama R, Inagaki Y, Hong YY, Kushida M, Nakao S, Niioka M, et al. Bone marrow-derived cells express matrix metalloproteinases and contribute to regression of liver fibrosis in mice. Hepatology. 2007;45(1):213–22.

    Article  PubMed  CAS  Google Scholar 

  17. Kuo TK, Hung SP, Chuang CH, Chen CT, Shih YR, Fang SC, Yang VW, Lee OK. Stem cell therapy for liver disease: parameters governing the success of using bone marrow mesenchymal stem cells. Gastroenterology. 2008;134(7):2111–21. 2121 e2111-2113.

    Article  PubMed  Google Scholar 

  18. Choi JS, Chae DS, Ryu HA, Kim SW. Transplantation of human adipose tissue derived-svf enhance liver function through high anti-inflammatory property. Biochim Biophys Acta Mol Cell Biol Lipids. 2019;1864(12):158526.

    Article  PubMed  CAS  Google Scholar 

  19. Brusko TM, Russ HA, Stabler CL. Strategies for durable beta cell replacement in type 1 diabetes. Science. 2021;373(6554):516–22.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Han S, Sun HM, Hwang KC, Kim SW. Adipose-derived stromal vascular fraction cells: update on clinical utility and efficacy. Crit Rev Eukaryot Gene Expr. 2015;25(2):145–52.

    Article  PubMed  Google Scholar 

  21. Yano M, Nasti A, Seki A, Ishida K, Yamato M, Inui H, et al. Characterization of adipose tissue-derived stromal cells of mice with nonalcoholic fatty liver disease and their use for liver repair. Regen Ther. 2021;18:497–507.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Chen LW, Chen PH, Tang CH, Yen JH. Adipose-derived stromal cells reverse insulin resistance through inhibition of m1 expression in a type 2 diabetes mellitus mouse model. Stem Cell Res Ther. 2022;13(1):357.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Gearty SV, Dundar F, Zumbo P, Espinosa-Carrasco G, Shakiba M, Sanchez-Rivera FJ, et al. An autoimmune stem-like cd8 t cell population drives type 1 diabetes. Nature. 2022;602(7895):156–61.

    Article  PubMed  CAS  Google Scholar 

  24. Barber AJ, Antonetti DA, Kern TS, Reiter CE, Soans RS, Krady JK, Levison SW, Gardner TW, Bronson SK. The ins2akita mouse as a model of early retinal complications in diabetes. Invest Ophthalmol Vis Sci. 2005;46(6):2210–8.

    Article  PubMed  Google Scholar 

  25. Bae CS, Lee Y, Ahn T. Therapeutic treatments for diabetes mellitus-induced liver injury by regulating oxidative stress and inflammation. Appl Microsc. 2023;53(1):4.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Lai HC, Chen PH, Tang CH, Chen LW. Il-10 enhances the inhibitory effect of adipose-derived stromal cells on insulin resistance/liver gluconeogenesis by treg cell induction. Int J Mol Sci 2024;25(15).

  27. Ouyang W, O’Garra A. Il-10 family cytokines il-10 and il-22: from basic science to clinical translation. Immunity. 2019;50(4):871–91.

    Article  PubMed  CAS  Google Scholar 

  28. Pierson W, Liston A. A new role for interleukin-10 in immune regulation. Immunol Cell Biol. 2010;88(8):769–70.

    Article  PubMed  Google Scholar 

  29. Hsu P, Santner-Nanan B, Hu M, Skarratt K, Lee CH, Stormon M, Wong M, Fuller SJ, Nanan R. Il-10 potentiates differentiation of human induced regulatory t cells via stat3 and foxo1. J Immunol. 2015;195(8):3665–74.

    Article  PubMed  CAS  Google Scholar 

  30. Toda G, Soeda K, Okazaki Y, Kobayashi N, Masuda Y, Arakawa N, et al. Insulin- and lipopolysaccharide-mediated signaling in adipose tissue macrophages regulates postprandial glycemia through akt-mtor activation. Mol Cell. 2020;79(1):43–e5344.

    Article  PubMed  CAS  Google Scholar 

  31. Sulen A, Aouadi M. Fed macrophages hit the liver’s sweet spot with il-10. Mol Cell. 2020;79(1):1–3.

    Article  PubMed  CAS  Google Scholar 

  32. Roep BO, Thomaidou S, van Tienhoven R, Zaldumbide A. Type 1 diabetes mellitus as a disease of the beta-cell (do not blame the immune system?). Nat Rev Endocrinol. 2021;17(3):150–61.

    Article  PubMed  CAS  Google Scholar 

  33. Chiasson JL, Aris-Jilwan N, Belanger R, Bertrand S, Beauregard H, Ekoe JM, Fournier H, Havrankova J. Diagnosis and treatment of diabetic ketoacidosis and the hyperglycemic hyperosmolar state. CMAJ. 2003;168(7):859–66.

    PubMed  PubMed Central  Google Scholar 

  34. Norlin S, Axelsson J, Ericsson M, Edlund H. O304 ameliorates hyperglycemia in mice by dually promoting muscle glucose effectiveness and preserving beta-cell function. Commun Biol. 2023;6(1):877.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Cintra DE, Pauli JR, Araujo EP, Moraes JC, de Souza CT, Milanski M, et al. Interleukin-10 is a protective factor against diet-induced insulin resistance in liver. J Hepatol. 2008;48(4):628–37.

    Article  PubMed  CAS  Google Scholar 

  36. Uccelli A, Moretta L, Pistoia V. Mesenchymal stem cells in health and disease. Nat Rev Immunol. 2008;8(9):726–36.

    Article  PubMed  CAS  Google Scholar 

  37. Yu Y, Mu J, Fan Z, Lei G, Yan M, Wang S, et al. Insulin-like growth factor 1 enhances the proliferation and osteogenic differentiation of human periodontal ligament stem cells via erk and jnk mapk pathways. Histochem Cell Biol. 2012;137(4):513–25.

    Article  PubMed  CAS  Google Scholar 

  38. Sordi V, Malosio ML, Marchesi F, Mercalli A, Melzi R, Giordano T, et al. Bone marrow mesenchymal stem cells express a restricted set of functionally active chemokine receptors capable of promoting migration to pancreatic islets. Blood. 2005;106(2):419–27.

    Article  PubMed  CAS  Google Scholar 

  39. Miklosz A, Chabowski A. Adipose-derived mesenchymal stem cells therapy as a new treatment option for diabetes mellitus. J Clin Endocrinol Metab. 2023;108(8):1889–97.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Gepts W. Islet changes suggesting a possible immune aetiology of human diabetes mellitus. Acta Endocrinol Suppl (Copenh). 1976;205:95–106.

    PubMed  CAS  Google Scholar 

  41. Bottazzo GF, Dean BM, McNally JM, MacKay EH, Swift PG, Gamble DR. In situ characterization of autoimmune phenomena and expression of hla molecules in the pancreas in diabetic insulitis. N Engl J Med. 1985;313(6):353–60.

    Article  PubMed  CAS  Google Scholar 

  42. Roep BO. The role of t-cells in the pathogenesis of type 1 diabetes: from cause to cure. Diabetologia. 2003;46(3):305–21.

    Article  PubMed  CAS  Google Scholar 

  43. El-Sheikh A, Suarez-Pinzon WL, Power RF, Rabinovitch A. Both cd4(+)and cd8(+)t cells are required for ifn-gamma gene expression in pancreatic islets and autoimmune diabetes development in biobreeding rats. J Autoimmun. 1999;12(2):109–19.

    Article  PubMed  CAS  Google Scholar 

  44. Phillips JM, Parish NM, Raine T, Bland C, Sawyer Y, De La Pena H, Cooke A. Type 1 diabetes development requires both cd4 + and cd8 + t cells and can be reversed by non-depleting antibodies targeting both t cell populations. Rev Diabet Stud. 2009;6(2):97–103.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Richardson SJ, Morgan NG, Foulis AK. Pancreatic pathology in type 1 diabetes mellitus. Endocr Pathol. 2014;25(1):80–92.

    Article  PubMed  CAS  Google Scholar 

  46. Tsalamandris S, Antonopoulos AS, Oikonomou E, Papamikroulis GA, Vogiatzi G, Papaioannou S, Deftereos S, Tousoulis D. The role of inflammation in diabetes: current concepts and future perspectives. Eur Cardiol. 2019;14(1):50–9.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Kirino Y, Sato Y, Kamimoto T, Kawazoe K, Minakuchi K, Nakahori Y. Interrelationship of dipeptidyl peptidase iv (dpp4) with the development of diabetes, dyslipidaemia and nephropathy: a streptozotocin-induced model using wild-type and dpp4-deficient rats. J Endocrinol. 2009;200(1):53–61.

    Article  PubMed  CAS  Google Scholar 

  48. Hu J, Fu Z, Chen Y, Tang N, Wang L, Wang F, Sun R, Yan S. Effects of autologous adipose-derived stem cell infusion on type 2 diabetic rats. Endocr J. 2015;62(4):339–52.

    Article  PubMed  CAS  Google Scholar 

  49. Hu J, Wang Y, Wang F, Wang L, Yu X, Sun R, et al. Effect and mechanisms of human wharton’s jelly-derived mesenchymal stem cells on type 1 diabetes in nod model. Endocrine. 2015;48(1):124–34.

    Article  PubMed  CAS  Google Scholar 

  50. Ezquer FE, Ezquer ME, Parrau DB, Carpio D, Yanez AJ, Conget PA. Systemic administration of multipotent mesenchymal stromal cells reverts hyperglycemia and prevents nephropathy in type 1 diabetic mice. Biol Blood Marrow Transpl. 2008;14(6):631–40.

    Article  CAS  Google Scholar 

  51. Ezquer F, Ezquer M, Contador D, Ricca M, Simon V, Conget P. The antidiabetic effect of mesenchymal stem cells is unrelated to their transdifferentiation potential but to their capability to restore th1/th2 balance and to modify the pancreatic microenvironment. Stem Cells. 2012;30(8):1664–74.

    Article  PubMed  CAS  Google Scholar 

  52. Song N, Scholtemeijer M, Shah K. Mesenchymal stem cell immunomodulation: mechanisms and therapeutic potential. Trends Pharmacol Sci. 2020;41(9):653–64.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. de Klerk E, Hebrok M. Stem cell-based clinical trials for diabetes mellitus. Front Endocrinol (Lausanne). 2021;12:631463.

    Article  PubMed  Google Scholar 

  54. Kim JH, Bae KH, Choi YK, Go Y, Choe M, Jeon YH, et al. Fibroblast growth factor 21 analogue ly2405319 lowers blood glucose in streptozotocin-induced insulin-deficient diabetic mice by restoring brown adipose tissue function. Diabetes Obes Metab. 2015;17(2):161–9.

    Article  PubMed  CAS  Google Scholar 

  55. Zhang J, Weng W, Wang K, Lu X, Cai L, Sun J. The role of fgf21 in type 1 diabetes and its complications. Int J Biol Sci. 2018;14(9):1000–11.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Kharitonenkov A, Wroblewski VJ, Koester A, Chen YF, Clutinger CK, Tigno XT, Hansen BC, Shanafelt AB, Etgen GJ. The metabolic state of diabetic monkeys is regulated by fibroblast growth factor-21. Endocrinology. 2007;148(2):774–81.

    Article  PubMed  CAS  Google Scholar 

  57. Zibar K, Blaslov K, Bulum T, Cuca JK, Smircic-Duvnjak L. Basal and postprandial change in serum fibroblast growth factor-21 concentration in type 1 diabetic mellitus and in healthy controls. Endocrine. 2015;48(3):848–55.

    Article  PubMed  CAS  Google Scholar 

  58. Wente W, Efanov AM, Brenner M, Kharitonenkov A, Koster A, Sandusky GE, et al. Fibroblast growth factor-21 improves pancreatic beta-cell function and survival by activation of extracellular signal-regulated kinase 1/2 and akt signaling pathways. Diabetes. 2006;55(9):2470–8.

    Article  PubMed  CAS  Google Scholar 

  59. Kim SN, Choi B, Lee CJ, Moon JH, Kim MK, Chung E, Song SU. Culturing at low cell density delays cellular senescence of human bone marrow-derived mesenchymal stem cells in long-term cultures. Int J Stem Cells. 2021;14(1):103–11.

    Article  PubMed  CAS  Google Scholar 

  60. MacDonald KN, Hall MG, Ivison S, Gandhi S, Klein Geltink RI, Piret JM, Levings MK. Consequences of adjusting cell density and feed frequency on serum-free expansion of thymic regulatory t cells. Cytotherapy. 2022;24(11):1121–35.

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by KAFGH-ZY-A-112017, MOST 110-2314-B-A49A-540-MY3 and KSVGH112-082.

Author information

Authors and Affiliations

Authors

Contributions

Yen-Ju Lee & Hsiao-Chi Lai: Investigation, Validation, Data curation, Writing-Original draft preparation. Pei-Hsuan Chen & Chia-Hua Tang: Methodology. Lee-Wei Chen: Project administration, Conceptualization, Visualization, Supervision, Writing-Reviewing and Editing.

Corresponding author

Correspondence to Lee-Wei Chen.

Ethics declarations

Ethics approval and consent to participate

(1) Title of the approved project: The effect of adipose stromal vascular fractions on type 1 diabetes- induced complications by regulating PPAR-γ; (2) Name of the institutional approval committee or unit: Institutional Animal Care and Use Committee of Kaohsiung Veterans General Hospital; (3) Approval number: IACUC-2408-2607-23112-NSTC (4) Date of approval: 15th Jan 2024. The authors declare that they have not use AI-generated work in this manuscript.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Supplementary Material 2

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lai, HC., Lee, YJ., Chen, PH. et al. Adipose stromal cells increase insulin sensitivity and decrease liver gluconeogenesis in a mouse model of type 1 diabetes mellitus. Stem Cell Res Ther 16, 133 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13287-025-04225-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13287-025-04225-5

Keywords