Exercise is a first-line treatment for type 2 diabetes and preserves β-cell function by hitherto unknown mechanisms. We postulated that proteins from contracting skeletal muscle may act as cellular signals to regulate pancreatic β-cell function. We used electric pulse stimulation (EPS) to induce contraction in C2C12 myotubes and found that treatment of β-cells with EPS-conditioned medium enhanced glucose-stimulated insulin secretion (GSIS). Transcriptomics and subsequent targeted validation revealed growth differentiation factor 15 (GDF15) as a central component of the skeletal muscle secretome. Exposure to recombinant GDF15 enhanced GSIS in cells, islets, and mice. GDF15 enhanced GSIS by upregulating the insulin secretion pathway in β-cells, which was abrogated in the presence of a GDF15 neutralizing antibody. The effect of GDF15 on GSIS was also observed in islets from GFRAL-deficient mice. Circulating GDF15 was incrementally elevated in patients with pre- and type 2 diabetes and positively associated with C-peptide in humans with overweight or obesity. Six weeks of high-intensity exercise training increased circulating GDF15 concentrations, which positively correlated with improvements in β-cell function in patients with type 2 diabetes. Taken together, GDF15 can function as a contraction-induced protein that enhances GSIS through activating the canonical signaling pathway in a GFRAL-independent manner.

Article Highlights

  • Exercise improves glucose-stimulated insulin secretion through direct interorgan communication.

  • Contracting skeletal muscle releases growth differentiation factor 15 (GDF15), which is required to synergistically enhance glucose-stimulated insulin secretion.

  • GDF15 enhances glucose-stimulated insulin secretion by activating the canonical insulin release pathway.

  • Increased levels of circulating GDF15 after exercise training are related to improvements in β-cell function in patients with type 2 diabetes.

Type 2 diabetes is characterized by hyperglycemia and loss of pancreatic insulin secretion. The penultimate step in disease onset involves compensatory insulin secretion from pancreatic β-cells to overcome peripheral insulin resistance. The ensuing hyperinsulinemia leads to progressive β-cell fatigue, dysfunction, apoptosis, dedifferentiation, and senescence, resulting in reduced insulin secretory capacity and dysregulated glucose homeostasis (1). Exercise is a first-line therapy in the prevention and management of type 2 diabetes (2,3). Importantly, exercise enhances pancreatic β-cell function independent of changes in body weight, circulating lipids, or peripheral insulin sensitivity (48). However, the mechanisms underlying exercise-induced improvements in insulin secretory function remain unclear.

It has been proposed that biologically active factors secreted in response to muscle contraction may contribute to improved β-cell function (9,10). This is exemplified by recent observations in which exposure to conditioned medium (CM) derived from contracted skeletal muscle cells prevented β-cell apoptosis (11) and potentiated insulin secretion (12,13). However, the specific molecular transducers mediating cross talk between contracting skeletal muscle and pancreatic β-cells remain largely unknown. Growth differentiation factor 15 (GDF15) is a secreted ligand and member of the transforming growth factor-β (TGF-β) superfamily (14,15). Circulating levels of GDF15 are elevated in marathon runners and cyclists after strenuous physical exercise (16,17). Pharmacologic treatment and overexpression of GDF15 in mice reduce food intake and body weight while improving insulin sensitivity (1827). Conversely, loss of GDF15 has been shown to increase body weight and impair glycemic control (25,28,29). It was recently demonstrated that clinical markers of insulin resistance and secretory function are associated with circulating GDF15 in patients with obesity and prediabetes or type 2 diabetes (30,31). More recently, GDF15 was also found to be expressed in mouse β-cells and human pancreatic islets and involved in glucose-stimulated insulin secretion (GSIS) (32,33). GDF15 expression was lower in islets of patients with type 1 diabetes, and treatment with exogenous GDF15 prevents cytokine-induced apoptosis (34). Furthermore, GDF15 was protective of β-cells, and administration of GDF15 prevented diabetes onset in mice (34). In addition, 12 weeks of GDF15 treatment improved exercise capacity in obese ZSF1 rats (35). Collectively, these reports indicate that skeletal muscle GDF15 may synergistically enhance β-cell function. Thus, in this study, we examined whether GDF15 secreted from skeletal muscle acts as a signaling peptide to regulate β-cell function.

In Vitro Studies

Muscle Contraction

Electrical pulse stimulation (EPS) was used to induce contraction in fully differentiated C2C12 myotubes as previously described (36).

RNA-seq

Following 16 h of stimulation, RNA was extracted from the cell pellet (RNAeasy; Qiagen) for untargeted RNA-seq. Libraries were prepared using the Illumina TruSeq Stranded Total RNA kit with 100-bp paired-end RNA-seq strategy. Illumina reads were mapped to mouse genome release mm10 using STAR Aligner, version 2.5.1. Differential expression was analyzed by Cuffdiff, version 2.2.1. Targets with a positive fold change were filtered through three secretome databases: SignalP (37), SPOCTUPUS (38), and Phobius (39).

CM Analysis

After 1, 2, 4, 8, 16, or 24 h of EPS, CM was collected for quantification of interleukin-6 by ELISA (Sigma). After 16 h of EPS in the culture medium from C2C12 or primary human skeletal muscle cells, GDF15 concentrations were determined by ELISA (R&D Systems).

GSIS

After treatment as described below, medium and cell lysates were harvested to determine insulin concentrations by ELISA (Mercodia). Total protein concentrations were determined by bicinchoninic acid assay (Thermo Fisher Scientific).

Glucose Titration.

β-TC-6 cells were stimulated with increasing concentrations of glucose (0.5, 1, 3, 5, and 10 mmol/L) in KRH buffer for 3 h. Culture medium and cell lysates were harvested for analysis.

EPS-CM Treatment.

β-TC-6 and MIN6 cells (AddexBio) were treated with CM from non-EPS or EPS-exposed C2C12 myotubes for 16 h. For GDF15-neutralizing experiment, 10 μg/mL anti-GDF15 monoclonal antibody (R&D Systems) was added to EPS-CM prior to a 16-h treatment in β-TC-6 cells. β-TC-6 cells were also treated with non-EPS CM supplemented with recombinant mouse GDF15 (rmGDF15) (R&D Systems) at 130 pg/mL or vehicle for 16 h. MIN6 cells were incubated with 0 or 10 mmol/L glucose in KRH buffer for 60 min. β-TC-6 cells were incubated with 0 or 1 mmol/L glucose in KRH buffer for 3 h.

GDF15 Treatment in β-Cells.

β-TC-6 cells were treated with 300 pg/mL and 10 ng/mL rmGDF15 protein or vehicle. rmGDF15 was supplemented in 0.1% BSA in KRH buffer and used for β-cell treatment in the presence or absence of 1 mmol/L glucose for 3 h.

GDF15 Treatment in Islets.

Human islets (Lonza) were obtained from a 33-year-old, Hispanic male donor, plated into cell culture inserts (MilliporeSigma), and equilibrated for 1 h in 2 mmol/L glucose–Krebs-Ringer bicarbonate buffer. Mouse islets were isolated from WT (Gfral+/+) and Gfral-knockout (Gfral−/−) mice as described previously (27,40). Islets were sequentially incubated for 1 h in Krebs-Ringer bicarbonate buffer plus 2 mmol/L glucose (basal) and Krebs-Ringer bicarbonate buffer plus 16 mmol/L glucose (stimulated), with or without 10 ng/mL recombinant human GDF15 (rhGDF15) (R&D Systems). Insulin was measured in supernatants and insulin content was determined in islet lysate.

Quantitative Real-Time PCR

Total RNA was extracted (RNAeasy; Qiagen) and converted to cDNA using an iScript cDNA synthesis kit (BIORAD). Quantitative real-time PCR was carried out with SYBR Green Master Mix (Thermo Fisher Scientific), as described previously (41). Gene expression was normalized to GAPDH and analyzed by the comparative cycle threshold method. Primer sequences are listed in Supplementary Table 1.

Glycolysis

Extracellular acidification rates were determined on a Seahorse XF24, as described previously (41). Maximal glycolytic flux was determined in the presence of glucose (1 mmol/L) and oligomycin (2 μmol/L).

ADP to ATP Ratio

Cellular ATP to ADP ratios were determined using a commercially available bioluminescent luciferase assay (Biovision) according to manufacturer instructions.

Intracellular Calcium

Intracellular calcium flux was determined by incubation of β-TC-6 cells with Fura-2 a.m. (Molecular Devices) in 0.1% DMSO for 50 min prior to harvest. Fluorescent intensity was determined on a Flexstation3 at baseline and after addition of KCL (27 mmol/L). Data are reported as the ratio of 340:380 (maximum to minimum).

Viability, Proliferation, and Cytotoxicity

Cell viability (Dojindo), proliferation (Abcam), and lactate dehydrogenase cytotoxicity (Thermo Fisher Scientific) were determined via commercial colorimetric assays per manufacturer instructions.

Flow Cytometry

β-TC-6 cells were expanded in a 10-cm dish, harvested by scraping, transferred to flow cytometry tubes, and incubated at 37°C for 1 h to recover cell surface receptors. Cells were then washed, resuspended in FACS buffer (BD Biosciences), and stained with anti-ErbB2/Her2-PE (Bio-Techne R&D Systems), anti–CD44-FITC (eBioscience Invitrogen) and anti–CD48-PerCP/Cy5.5 (Biolegend). Data were acquired on an Aurora flow cytometer (Cytek). UltraComp eBeads (Thermo Scientific) were used for compensation, and FlowJo, version 10.8.1 software (Treestar Inc) was used for data analysis.

Mouse Studies

Animal Husbandry

Male C57BL/6J (catalog 000664, Jackson) mice were housed under a standard 12-h light/dark cycle at a temperature of 21–22°C and fed 5053 PicoLab standard Rodent Diet 20 (catalog 0007688, LabDiet) with ad libitum access to food and water. GFRAL-deficient mice (Gfral−/−) and littermate controls (Gfral+/+) were housed under a standard 12-h light/dark cycle at a temperature of 25°C and fed standard Rodent Diet (PicoLab) with ad libitum access to food and water. Mice were euthanized at 12 weeks of age for isolation of primary mouse islets.

In vivo Insulin Secretion

Twelve-week-old mice were fasted overnight for 12 h and, at 0800 the next day, were moved to the operating room for adaptation prior to treatment. After 1 h, the mice received either intraperitoneal injections of 0.25 mg/kg rhGDF15 (R&D Systems) or vehicle. After 2 h, the mice were given an intraperitoneal injection of 1 g/kg d-glucose. Blood was collected prior to injection, at 2 h after rhGDF15 injection via tail snip, and 30 min after glucose challenge via terminal cardiac puncture. Blood was collected in EDTA-treated tubes and plasma was isolated by centrifugation at 2000g for 20 min at 4°C. Insulin and C-peptide concentrations were determined by a multiplex assay (Millipore), and GDF15 by ELISA (R&D). All experiments and procedures involving animals were approved by the Pennington Biomedical Research Center and University of Michigan Institutional Animal Care and Use Committee.

Human Studies

All human data and specimens (vastus lateralis muscle samples, plasma samples) were obtained from previously approved and reported studies (4247). In Human Study 1, skeletal muscle specimens were collected from older adults with obesity (mean ± SD: age 66.0 ± 4.4 years; BMI 33.7 ± 3.5) before and after 12 weeks of daily aerobic exercise training, as described previously (48). The supervised aerobic training program consisted of treadmill exercise set at 85% of heart rate maximum for 1 h/day, 5 days/week, over 12 weeks. Patient characteristics are provided in Table 1. Muscle cDNA samples were prepared as previously described (49). GAPDH was used as an internal standard. Data were calculated using the comparative cycle threshold method.

Table 1

Baseline participant characteristics for the clinical cohorts in Human Studies 1 and 3

CharacteristicClinical cohort in Fig. 1H (Human Study 1)Clinical cohort in Fig. 6E and F (Human Study 3)
Participants; male participants; female participants, n 19; 10; 9 12; 4; 8 
Age, years 66 ± 4.4 54 ± 6.9 
Weight, kg 98.5 ± 16.1 98.5 ± 12.5 
Height, cm 170.6 ± 10.0 169.1 ± 10.4 
BMI, kg/m2 33.7 ± 3.5 34.5 ± 3.8 
Fat, % 42.7 ± 6.5 44.0 ± 6.2 
Fasting plasma glucose, mg/dL 98.6 ± 13.1 179.1 ± 57.5 
Insulin, µU/mL 14 ± 4.8 23.1 ± 20.1 
HbA1c, % 5.6 ± 0.9 8.5 ± 2.1 
HbA1c, mmol/mol 38 ± 9.9 69 ± 23.1 
Vo2max, mL/kg/min 21.4 ± 3.9 23.5 ± 3.8 
Type 2 diabetes, n 12 
CharacteristicClinical cohort in Fig. 1H (Human Study 1)Clinical cohort in Fig. 6E and F (Human Study 3)
Participants; male participants; female participants, n 19; 10; 9 12; 4; 8 
Age, years 66 ± 4.4 54 ± 6.9 
Weight, kg 98.5 ± 16.1 98.5 ± 12.5 
Height, cm 170.6 ± 10.0 169.1 ± 10.4 
BMI, kg/m2 33.7 ± 3.5 34.5 ± 3.8 
Fat, % 42.7 ± 6.5 44.0 ± 6.2 
Fasting plasma glucose, mg/dL 98.6 ± 13.1 179.1 ± 57.5 
Insulin, µU/mL 14 ± 4.8 23.1 ± 20.1 
HbA1c, % 5.6 ± 0.9 8.5 ± 2.1 
HbA1c, mmol/mol 38 ± 9.9 69 ± 23.1 
Vo2max, mL/kg/min 21.4 ± 3.9 23.5 ± 3.8 
Type 2 diabetes, n 12 

Data are reported as mean ± SD, unless otherwise indicated.

In Human Study 2, in the cross-sectional analysis, 62 sedentary, weight-stable (± 2.5 kg for prior 6 months) adults were assigned to the following groups: 1) healthy controls: normal fasting glucose and glucose tolerance; 2) prediabetes: impaired fasting glucose or glucose tolerance; and 3) type 2 diabetes, based upon the American Diabetes Association guidelines for classification and diagnosis of diabetes (50). The participants’ characteristics are described in Table 2. Fasting plasma samples were collected as previously reported (46) and assayed for GDF15 concentration by ELISA (R&D Systems).

Table 2

Baseline characteristics of participants in the clinical cohort in Human Study 2 (Fig. 6A–D)

CharacteristicNFG/NGTIFG/IGTT2D
Participants; male participants; female participants, n 17; 12; 5 21; 11; 10 24; 19; 5 
Age, years 45 ± 16.5 61 ± 13.7 55 ± 9.8 
Weight, kg 82.2 ± 17.7 100.8 ± 15.1 93.9 ± 14.7 
BMI, kg/m2 29.1 ± 5.8 35.3 ± 3.7 34.3 ± 4.9 
Fat, % 37.8 ± 13.6 45.9 ± 6.4 45.7 ± 5.4 
Fasting plasma glucose, mg/dL 92.3 ± 7.0 101.4 ± 7.8 155 ± 49.0 
Fasting plasma insulin, µU/mL 13.9 ± 6.6 25.0 ± 18.3 29.1 ± 23.5 
HbA1c, % 5.4 ± 0.4 5.6 ± 0.5 7.5 ± 2.0 
HbA1c, mmol/mol 36 ± 4.4 38 ± 5.5 58 ± 22 
Vo2max, mL/kg/min 28.6 ± 8.2 21.9 ± 4.6 21.8 ± 3.9 
CharacteristicNFG/NGTIFG/IGTT2D
Participants; male participants; female participants, n 17; 12; 5 21; 11; 10 24; 19; 5 
Age, years 45 ± 16.5 61 ± 13.7 55 ± 9.8 
Weight, kg 82.2 ± 17.7 100.8 ± 15.1 93.9 ± 14.7 
BMI, kg/m2 29.1 ± 5.8 35.3 ± 3.7 34.3 ± 4.9 
Fat, % 37.8 ± 13.6 45.9 ± 6.4 45.7 ± 5.4 
Fasting plasma glucose, mg/dL 92.3 ± 7.0 101.4 ± 7.8 155 ± 49.0 
Fasting plasma insulin, µU/mL 13.9 ± 6.6 25.0 ± 18.3 29.1 ± 23.5 
HbA1c, % 5.4 ± 0.4 5.6 ± 0.5 7.5 ± 2.0 
HbA1c, mmol/mol 36 ± 4.4 38 ± 5.5 58 ± 22 
Vo2max, mL/kg/min 28.6 ± 8.2 21.9 ± 4.6 21.8 ± 3.9 

Data are reported as mean ± SD. IFG, impaired fasting glucose; IGT, impaired glucose tolerance; NFG, normal fasting glucose; NGT, normal glucose tolerance; T2D, type 2 diabetes.

In Human Study 3, plasma samples were collected from patients with type 2 diabetes before and after 6 weeks high-intensity exercise training intervention (46,47). The participants’ characteristics are described in Table 1. Fasting plasma samples were analyzed for GDF15 by ELISA (R&D Systems) per manufacturer’s instructions.

Quantification and Statistical Analysis

All data points are reported as biological replicates, not technical replicates. Technical replicates were averaged when appropriate. In figures, data are reported as mean ± SD, unless otherwise indicated in the figure legend. GraphPad Prism8 software was used for statistical analysis. Detailed descriptions of statistical procedures are provided in the respective figure legends. Normality was assessed by the Kolmogorov-Smirnov test. Pearson correlation was used to investigate the associations between the change in GDF15 and the disposition index. Significance was accepted at P < 0.05.

Data and Resource Availability

RNA-seq data sets were submitted to the National Center for Biotechnology Information’s Gene Expression Omnibus database under the accession number GSE139872. All other data sets generated and/or analyzed in this study are available from the corresponding author upon request.

The Skeletal Muscle Secretome Enhances Insulin Secretion in Pancreatic β-Cells

We first explored the relationship between skeletal muscle contraction and insulin secretion by using an in vitro model to induce muscle contraction (Supplementary Fig. 1). We and others have previously validated this EPS model and used it to demonstrate the effects of isolated cell contraction on glucose uptake and insulin signaling in C2C12 and primary human myotubes (36,51). In the present experiments, contraction was validated by accumulation of interleukin-6 in the CM from C2C12 myotubes (Supplementary Fig. 2A), a widely accepted, exercise-induced myokine (52). Cultured β-cells were then pretreated with EPS or non–EPS-CM for 16 h and evaluated for glucose-stimulated insulin secretion (GSIS) (Fig. 1A). EPS-CM increased GSIS by 30% in β-TC-6 cells and by 150% in MIN6 cells compared with non–EPS-CM–treated β-cells (Fig. 1B). To address anticipated concerns that EPS may produce non–cell-mediated effects (53), we assessed GSIS after EPS in cell-free conditions and confirmed that the phenotype observed was due to EPS-stimulated contraction of myotubes (Supplementary Fig. 2BD). In addition, EPS-CM from C2C12 cells did not affect β-cell viability (Supplementary Fig. 2E). Together, these data demonstrate that EPS-CM from C2C12 myotubes potentiates GSIS.

Figure 1

GDF15 secretion is increased in response to muscle contraction and exercise. A: Experimental flow to investigate cross talk between skeletal muscle and pancreatic β-cells. B: Insulin secretion in pancreatic β-cells after 16 h of treatment with CM collected from non-EPS or EPS-exposed C2C12 myotubes. Left: β-TC-6 cells treated with and without 1 mmol/L glucose for 3 h (n = 10). Right: MIN6 cells were treated with and without 10 mmol/L glucose for 1 h (n = 10). C: C2C12 myotubes were subjected to RNA-seq after 16 h of EPS or non-EPS, followed by filtration through a secretome database. A total of 71 secreted genes were significantly changed relative to the non-EPS condition (n = 4 repeated culture). D: Heat map of expression of 33 significantly upregulated genes. E: Scatterplot displaying the distribution of the significantly changed genes, which are presented as statistical significance (P value) vs. magnitude of change (fold change). F: Gdf15 gene expression in EPS-stimulated C2C12 myotubes. Left: Results from transcriptome analysis expressed in fragments per kilobase of sequence per million (FPKM) mapped reads (n = 4 repeated culture). Right: Results from qPCR expressed in A.U. (arbitrary units) (n = 6). G: GDF15 protein secretion in the CM of EPS-exposed C2C12 myotubes (n = 4 repeated culture). H: GDF15 gene expression in human skeletal muscle tissue. Gene expression was accessed in participants in Human Study 1 before and after 12-week aerobic exercise training (n = 19). Data are expressed as mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001 compared with non-EPS by two-way ANOVA with Bonferroni post hoc test for B and unpaired t test for F and G. **P < 0.01 compared with pretraining by paired t test for H.

Figure 1

GDF15 secretion is increased in response to muscle contraction and exercise. A: Experimental flow to investigate cross talk between skeletal muscle and pancreatic β-cells. B: Insulin secretion in pancreatic β-cells after 16 h of treatment with CM collected from non-EPS or EPS-exposed C2C12 myotubes. Left: β-TC-6 cells treated with and without 1 mmol/L glucose for 3 h (n = 10). Right: MIN6 cells were treated with and without 10 mmol/L glucose for 1 h (n = 10). C: C2C12 myotubes were subjected to RNA-seq after 16 h of EPS or non-EPS, followed by filtration through a secretome database. A total of 71 secreted genes were significantly changed relative to the non-EPS condition (n = 4 repeated culture). D: Heat map of expression of 33 significantly upregulated genes. E: Scatterplot displaying the distribution of the significantly changed genes, which are presented as statistical significance (P value) vs. magnitude of change (fold change). F: Gdf15 gene expression in EPS-stimulated C2C12 myotubes. Left: Results from transcriptome analysis expressed in fragments per kilobase of sequence per million (FPKM) mapped reads (n = 4 repeated culture). Right: Results from qPCR expressed in A.U. (arbitrary units) (n = 6). G: GDF15 protein secretion in the CM of EPS-exposed C2C12 myotubes (n = 4 repeated culture). H: GDF15 gene expression in human skeletal muscle tissue. Gene expression was accessed in participants in Human Study 1 before and after 12-week aerobic exercise training (n = 19). Data are expressed as mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001 compared with non-EPS by two-way ANOVA with Bonferroni post hoc test for B and unpaired t test for F and G. **P < 0.01 compared with pretraining by paired t test for H.

Close modal

GDF15 Is Increased in Response to Muscle Contraction and Exercise Training

To identify specific components of the secretome that may regulate β-cell function, we conducted untargeted RNA-seq on C2C12 myotubes after EPS (Fig. 1A). RNA-seq revealed 832 differentially regulated mRNA transcripts in response to EPS. We then refined the pool of putative transcripts to those with known secretory functions by filtering through three protein databases: SignalP, SPOCTUPUS, and Phobius (Fig. 1C). This approach yielded 71 transcripts that were differentially expressed after EPS, 33 of which were upregulated (Fig. 1D). Gdf15 was the transcript with the most significantly increased expression and was subsequently chosen for validation (Fig. 1E). Indeed, Gdf15 expression was significantly increased in C2C12 myotubes after EPS, compared with non-EPS control myotubes (Fig. 1F). To validate protein expression after muscle contraction, CM from EPS-exposed myotubes was collected for assessment (Fig. 1A). GDF15 was increased by 76.3% in the CM of C2C12 myotubes (Fig. 1G) and by 57.6% in primary human muscle cells (Supplementary Fig. 3). To support these findings, we assessed changes in muscle GDF15 expression after exercise training in humans. Characteristics of participants in Human Study 1 are listed in Table 1. In skeletal muscle tissue from this cohort of older adults with obesity, gene expression of GDF15 was increased by 3.7-fold after 12 weeks of daily aerobic exercise training compared with the pretraining expression level (Fig. 1H). Taken together, these data suggest that GDF15 gene expression is upregulated by muscle contraction and that exercise training promotes increased protein secretion.

GDF15 Potentiates GSIS

To determine whether GDF15 regulates β-cell function, isolated human islets and mouse β-TC-6 cells were exposed to rhGDF15. Under low-glucose (unstimulated) conditions, rhGDF15 treatment did not change insulin secretion in islets from a donor with prediabetes. Notably, in high-glucose (stimulated) conditions, insulin secretion was increased by 45% in rhGDF15-treated islets compared with control islets isolated from the same donor (Fig. 2A). This rhGDF15-induced increase in GSIS was independent of changes in total insulin content in human islets (Fig. 2B). To further explore the effect of GDF15 on β-cell function, pancreatic β-TC-6 cells were exposed to rmGDF15 for 3 h in the presence or absence of glucose. rmGDF15 alone did not promote insulin secretion but did potentiate GSIS (Fig. 2C). Like human islets, total insulin content was not changed in β-TC-6 cells with rmGDF15 treatment (Fig. 2D). In addition, Ins1 and Ins2 gene expression, as well as β-cell mass, were unaffected by rmGDF15 treatment under basal or glucose-stimulated conditions (Fig. 2E–H).

Figure 2

GDF15 increases GSIS in human islets and mouse pancreatic β-cells. A and B: Insulin secretion (A) and insulin content (B) in human islets treated with rhGDF15. Participants (n = 1); experiment replicates: n = 12/vehicle and n = 14/rhGDF15. C and D: Insulin secretion (C) and insulin content (D) in mouse pancreatic β-TC-6 cells after 3 h of rmGDF15 treatment in the presence or absence of 1 mmol/L glucose (n = 4 repeated culture, n = 4 replicates/culture). E and F: Gene expression of Ins1 (E) and Ins2 (F) in β-TC-6 cells treated with rmGDF15 in the presence or absence of 1 mmol/L glucose (n = 5–8). G: Effect of rmGDF15 on proliferation of β-TC-6 (n = 4 repeated culture, n = 4–10 replicates/culture). H: Toxicity of rmGDF15 in β-TC-6 cells (n = 4 repeated culture). Data are expressed as mean ± SD. *P < 0.05 compared with control by two-way ANOVA with Bonferroni post hoc test.

Figure 2

GDF15 increases GSIS in human islets and mouse pancreatic β-cells. A and B: Insulin secretion (A) and insulin content (B) in human islets treated with rhGDF15. Participants (n = 1); experiment replicates: n = 12/vehicle and n = 14/rhGDF15. C and D: Insulin secretion (C) and insulin content (D) in mouse pancreatic β-TC-6 cells after 3 h of rmGDF15 treatment in the presence or absence of 1 mmol/L glucose (n = 4 repeated culture, n = 4 replicates/culture). E and F: Gene expression of Ins1 (E) and Ins2 (F) in β-TC-6 cells treated with rmGDF15 in the presence or absence of 1 mmol/L glucose (n = 5–8). G: Effect of rmGDF15 on proliferation of β-TC-6 (n = 4 repeated culture, n = 4–10 replicates/culture). H: Toxicity of rmGDF15 in β-TC-6 cells (n = 4 repeated culture). Data are expressed as mean ± SD. *P < 0.05 compared with control by two-way ANOVA with Bonferroni post hoc test.

Close modal

To determine the effect of GDF15 on GSIS in vivo, chow-fed male C57BL/6J mice were injected with saline or rhGDF15 and evaluated for insulin and C-peptide secretion (Fig. 3A). Mice were weight matched and glucose levels in circulation were not different in vehicle- or rhGDF15-injected mice (Supplementary Fig. 4A and B). We observed that GDF15 alone did not increase plasma C-peptide (Fig. 3B) or insulin concentrations (Fig. 3C). However, GDF15 increased GSIS and glucose-stimulated C-peptide release in mice (Fig. 3B and C). Collectively, these data support that GDF15 increases insulin secretion in isolated human islets, mouse β-cells, and chow diet–fed mice in a glucose-dependent manner.

Figure 3

GDF15 increases GSIS in C57BL/6J mice. A: Fasting plasma rhGDF15 levels in C57BL/6J mice 2 h after intraperitoneal injection of vehicle or 0.25 mg/kg rhGDF15 (n = 6–7). B: Plasma C-peptide and (C) insulin levels in C57BL/6J mice pretreated with vehicle or rhGDF15 and after 1 g/kg d-glucose challenge (n = 6 in control group, n = 7 in the rhGDF15 treatment group). Data are expressed as mean ± SD. **P < 0.01 compared with control by unpaired t test for A, and two-way ANOVA with Bonferroni post hoc test for B and C.

Figure 3

GDF15 increases GSIS in C57BL/6J mice. A: Fasting plasma rhGDF15 levels in C57BL/6J mice 2 h after intraperitoneal injection of vehicle or 0.25 mg/kg rhGDF15 (n = 6–7). B: Plasma C-peptide and (C) insulin levels in C57BL/6J mice pretreated with vehicle or rhGDF15 and after 1 g/kg d-glucose challenge (n = 6 in control group, n = 7 in the rhGDF15 treatment group). Data are expressed as mean ± SD. **P < 0.01 compared with control by unpaired t test for A, and two-way ANOVA with Bonferroni post hoc test for B and C.

Close modal

GDF15 Is Required to Potentiate Contraction-Induced GSIS

To determine if GDF15 was required for EPS-CM to enhance GSIS, native protein was neutralized by incubation with an IgG anti-GDF15 antibody (5456). Consistent with our previous experiments, GSIS increased by ∼30% after exposure to EPS-CM compared with β-cells exposed to non–EPS-CM. Blocking GDF15 with neutralizing antibody abrogated the effects of EPS-CM, and the insulin secretion rate did not differ significantly from non-EPS controls (Fig. 4A). Although, GDF15-neutralizing antibody in non–EPS-CM did not affect GSIS, the insulin secretion rate was still significantly lower compared with the effects of EPS-CM with the neutralizing antibody. Interestingly, replacement of non–EPS-CM with rmGDF15 to the concentration detected in EPS-CM (120 pg/mL) (Fig. 1G and Supplementary Fig. 6A) increased the basal insulin secretion rate by 13% and GSIS by 20% in β-TC-6 cells (Fig. 4B). Furthermore, short-term exposure of β-TC-6 cells with rmGDF15 at a physiological concentration increased GSIS by 30% (Fig. 4C and Supplementary Fig. 5B).

Figure 4

GDF15 at physiological doses increases GSIS in pancreatic β-cells. A: GSIS (1 mmol/L) in β-TC-6 cells after 16 h of treatment with CM: non-EPS, EPS, and supplemented with 10 μg/mL neutralizing antibody (nAb) anti-GDF15 at growth day 4. GSIS levels in non-EPC control cells were set as 100% (n = 2 repeated cultures, n = 6–7 replicates/culture). B: Insulin secretion in TC-6 cells in the presence or absence of 1 mmol/L glucose after 16 h of treatment with non–EPS-CM supplemented with vehicle or 130 pg/mL rmGDF15 at growth day 4 (n = 3 repeated cultures). C: Insulin secretion in β-TC-6 cells after 3 h of vehicle or 300 pg/mL rmGDF15 treatment in the presence or absence of 1 mmol/L glucose at growth day 3 (n = 3 repeated cultures, n = 4–6 replicates/culture). D: Insulin secretion in islets from Gfral+/+ and Gfral−/− mice treated with rmGDF15 (n = 7–8 mice; n = 7–10 replicates/treatment). Data are expressed as mean ± SD. *P < 0.05 compared with vehicle-treated control by unpaired t test and two-way ANOVA with Bonferroni post hoc test.

Figure 4

GDF15 at physiological doses increases GSIS in pancreatic β-cells. A: GSIS (1 mmol/L) in β-TC-6 cells after 16 h of treatment with CM: non-EPS, EPS, and supplemented with 10 μg/mL neutralizing antibody (nAb) anti-GDF15 at growth day 4. GSIS levels in non-EPC control cells were set as 100% (n = 2 repeated cultures, n = 6–7 replicates/culture). B: Insulin secretion in TC-6 cells in the presence or absence of 1 mmol/L glucose after 16 h of treatment with non–EPS-CM supplemented with vehicle or 130 pg/mL rmGDF15 at growth day 4 (n = 3 repeated cultures). C: Insulin secretion in β-TC-6 cells after 3 h of vehicle or 300 pg/mL rmGDF15 treatment in the presence or absence of 1 mmol/L glucose at growth day 3 (n = 3 repeated cultures, n = 4–6 replicates/culture). D: Insulin secretion in islets from Gfral+/+ and Gfral−/− mice treated with rmGDF15 (n = 7–8 mice; n = 7–10 replicates/treatment). Data are expressed as mean ± SD. *P < 0.05 compared with vehicle-treated control by unpaired t test and two-way ANOVA with Bonferroni post hoc test.

Close modal

GDF15 has been shown to exert its peripheral metabolic actions via the GFRAL receptor (1820). As such, we opted to evaluate whether GFRAL was required to potentiate the effects of GDF15 on GSIS. To this end, GDF15 increased GSIS in isolated islets obtained from GFRAL-deficient and control mice to a similar extent with no difference in insulin content in the islets (Fig. 4D). Interestingly, Gfral gene expression was not detected in β-TC-6 cells (Supplementary Fig. 6). However, we did detect expression of other putative GDF15 receptors, such as ErbB2, CD44, and CD48, in β-TC-6 cells (Supplementary Figs. 6 and 8). Taken together, these data demonstrate that GDF15 is required for muscle contraction–induced potentiation of GSIS and that replacement with recombinant protein at a physiological dose was sufficient to rapidly increase GSIS independent of GFRAL.

GDF15 Increases Glycolysis, ATP to ADP Ratio, and Calcium Release in β-cells

Having demonstrated that GDF15 increases GSIS in β-cells in vitro and in vivo, individual components of the canonical insulin-release pathway were investigated to identify how GDF15 was enhancing GSIS. First, glycolytic flux was assessed in real time by measuring extracellular acidification in response to glucose, oligomycin, 2-deoxy-d-glucose, and a cocktail of rotenone and antimycin A in cultured β-cells (Fig. 5A). GDF15 increased glucose-stimulated extracellular acidification, a measure of net glycolytic activity (Fig. 5B). GDF15 treatment also increased the cytosolic ATP to ADP ratio (Fig. 5C) and the net intracellular calcium influx (Fig. 5D). Interestingly, the GDF15-stimulated increase in glycolysis was reversed with anti-GDF15 neutralizing antibody, and the glucose-stimulated extracellular acidification rate was not significantly different compared with the vehicle-treated rate (Fig. 5E). Additionally, GDF15 treatment did not significantly change expression of genes that maintain insulin secretion (e.g., PDX-1, FOXO1, MafA, MafB, NeuronD1, NKX6.1, KLF11, GDF15, GLUT-2, GIPR, GLPIR, Kir6.1, SUR1, and Rab27a) (Supplementary Fig. 7). Taken together, these data indicate that GDF15 enhances GSIS through activation of the canonical insulin secretion–signaling pathway (Fig. 5F).

Figure 5

GDF15 upregulates the insulin secretion pathway. A: Extracellular acidification rate (ECAR) in β-TC-6 cells treated with rmGDF15 under 0.5 mmol/L glucose condition was determined using Seahorse XF24 extracellular flux analysis (n = 6). B: Glucose-stimulated (1 mmol/L) ECAR in β-TC-6 cells treated with rmGDF15 (n = 6). C: The ratio of ATP to ADP (ATP/APD) in β-TC-6 cells treated with rmGDF15 in the presence or absence of 1 mmol/L glucose (n = 9–12). D: Representative image of intracellular calcium influx in response to K+ stimulation in β-TC-6 cells treated with vehicle or rmGDF15 (n = 4). Changes in intracellular calcium levels in β-TC-6 cells treated with rmGDF15 in response to K+ stimulation are shown in the inserted graph (n = 3 repeated cultures, n = 6 replicates/culture). E: Glucose-stimulated ECAR in β-TC-6 cells at passage 5 treated with 5 ng/mL rmGDF15 under 1 mmol/L glucose conditions was determined by Seahorse XF24 extracellular flux analysis (n = 4 repeated cultures, n = 7–8 replicates/culture). F: Illustration of GDF15 effect on the insulin secretion pathway. Data are expressed as mean ± SD. *P < 0.05, **P < 0.01 compared with control by paired t test for (A and B), two-way ANOVA with Bonferroni post hoc test for C, and one-way ANOVA followed by unpaired t test for D. Ab, antibody; max, maximum; min, minimum; neut, neutralizing; Veh, vehicle. Rot, rotenone; AmA, antimycin A; Oligo, oligomycin.

Figure 5

GDF15 upregulates the insulin secretion pathway. A: Extracellular acidification rate (ECAR) in β-TC-6 cells treated with rmGDF15 under 0.5 mmol/L glucose condition was determined using Seahorse XF24 extracellular flux analysis (n = 6). B: Glucose-stimulated (1 mmol/L) ECAR in β-TC-6 cells treated with rmGDF15 (n = 6). C: The ratio of ATP to ADP (ATP/APD) in β-TC-6 cells treated with rmGDF15 in the presence or absence of 1 mmol/L glucose (n = 9–12). D: Representative image of intracellular calcium influx in response to K+ stimulation in β-TC-6 cells treated with vehicle or rmGDF15 (n = 4). Changes in intracellular calcium levels in β-TC-6 cells treated with rmGDF15 in response to K+ stimulation are shown in the inserted graph (n = 3 repeated cultures, n = 6 replicates/culture). E: Glucose-stimulated ECAR in β-TC-6 cells at passage 5 treated with 5 ng/mL rmGDF15 under 1 mmol/L glucose conditions was determined by Seahorse XF24 extracellular flux analysis (n = 4 repeated cultures, n = 7–8 replicates/culture). F: Illustration of GDF15 effect on the insulin secretion pathway. Data are expressed as mean ± SD. *P < 0.05, **P < 0.01 compared with control by paired t test for (A and B), two-way ANOVA with Bonferroni post hoc test for C, and one-way ANOVA followed by unpaired t test for D. Ab, antibody; max, maximum; min, minimum; neut, neutralizing; Veh, vehicle. Rot, rotenone; AmA, antimycin A; Oligo, oligomycin.

Close modal

GDF15 Is Associated With Enhanced β-Cell Function in Humans

To contextualize these findings, we then examined the relationship between GDF15 and β-cell function in humans. First, we conducted a cross-sectional analysis of circulating levels of GDF15 in sedentary adults across the spectrum of glucose tolerance (Human Study 2). We observed that fasting plasma GDF15 was increased in individuals with prediabetes and type 2 diabetes compared with healthy control participants (Fig. 6A). Of note, the relative increase in C-peptide (Fig. 6B) across the metabolic spectrum was accompanied by an incremental increase in GDF15. Fasting blood glucose was significantly greater in individuals with prediabetes compared with healthy control participants, although glucose levels in these individuals remained below 126 mg/dL (Fig. 6C). However, fasting glucose was further elevated in patients with type 2 diabetes compared with healthy control participants and individuals with prediabetes (Fig. 6C). Notably, C-peptide concentrations were positively correlated with circulating GDF15 in this cohort (Fig. 6D).

Next, we evaluated the effects of 6 weeks of high-intensity exercise training on circulating GDF15 and β-cell function in patients with type 2 diabetes (Human Study 3). Fasting plasma GDF15 was increased after training (from 768.5 ± 227.4 to 1,015.7 ± 397.6 pg/mL) (Fig. 6E), while fasting blood glucose (from 179.1 ± 57.5 to 157.0 ± 61.3, mg/dL; P = 0.2) and insulin (from 23.1 ± 20.2 to 25.8 ± 16.8 µU/mL; P = 0.5) remained relatively unchanged. Importantly, the change in circulating GDF15 levels between pre- and postexercise positively correlated with the insulin disposition index, a validated assessment of β-cell function (Fig. 6F). Together, these data provide clinical evidence to support a role for GDF15 in regulating glucose homeostasis and β-cell function.

Figure 6

GDF15 is associated with improved insulin secretion in humans. A: Fasting plasma GDF15, (B) C-peptide, and (C) blood glucose levels in healthy control participants (n = 19), patients with prediabetes (n = 24), and those with type 2 diabetes (n = 27) from Human Study 2. D: Correlation between C-peptide and plasma GDF15 levels in participants in Human Study 2 (n = 62). E: Fasting plasma GDF15 levels in patients with type 2 diabetes in Human Study 3 before and after 6-weeks of high-intensity exercise training (n = 12). F: Association between exercise-induced circulating GDF15 levels and an improvement in the disposition index (DI) (β-cell function) in patients with type 2 diabetes (r = 0.69; P = 0.01; n = 12). Data are expressed as mean ± SD. *P < 0.05, **P ≤ 0.01, ***P < 0.001 by one-way ANOVA followed by unpaired t test for AD and by paired t test for E.

Figure 6

GDF15 is associated with improved insulin secretion in humans. A: Fasting plasma GDF15, (B) C-peptide, and (C) blood glucose levels in healthy control participants (n = 19), patients with prediabetes (n = 24), and those with type 2 diabetes (n = 27) from Human Study 2. D: Correlation between C-peptide and plasma GDF15 levels in participants in Human Study 2 (n = 62). E: Fasting plasma GDF15 levels in patients with type 2 diabetes in Human Study 3 before and after 6-weeks of high-intensity exercise training (n = 12). F: Association between exercise-induced circulating GDF15 levels and an improvement in the disposition index (DI) (β-cell function) in patients with type 2 diabetes (r = 0.69; P = 0.01; n = 12). Data are expressed as mean ± SD. *P < 0.05, **P ≤ 0.01, ***P < 0.001 by one-way ANOVA followed by unpaired t test for AD and by paired t test for E.

Close modal

GDF15 is a stress-sensitive cytokine (57) that regulates food intake, body weight, and glucose metabolism via direct action on the central nervous system (24,26,28,29). It has also been noted that GDF15 is highly enriched in hepatic tissue, suggesting that peripheral mechanisms may also contribute to its metabolic effects (58). Here, we provide comprehensive evidence that GDF15 is increased in response to skeletal muscle contraction and acts on pancreatic β-cells to improve GSIS. This observation is further supported by clinical studies demonstrating elevated GDF15 after exercise training and associations with β-cell function and glycemic control in patients with type 2 diabetes.

Using an in vitro contraction model, we found that GDF15 was secreted from C2C12 myotubes in response to EPS. These data are consistent with those in recent reports showing increasing GDF15 secretion after both acute and chronic exposure to EPS (56). Furthermore, others have now observed that circulating and skeletal muscle GDF15 is increased after exercise training in humans with obesity (45,56,5961). In agreement with these observations, we demonstrated that GDF15 gene expression was increased in skeletal muscle tissue of adults with obesity and circulating GDF15 concentrations elevated in patients with type 2 diabetes after exercise training. In contrast, circulating GDF15 is unchanged in mice after acute treadmill running (62). Interestingly, acute cycling increased arterial and venous GDF15, whereas the arterial-venous difference was unaffected, indicating that exercise stimulates GDF15 secretion but that under these conditions, skeletal muscle may not substantially contribute to the circulating pool (59). It is likely that differences in exercise protocols, study populations, exercise duration, and intensity or timing of sample collections may influence GDF15 release from skeletal muscle and contribute to these disparate findings.

Exercise volume and mechanical loading are particularly significant (63); other studies have shown that C/EBP-homologous protein (CHOP), which is upstream in the unfolded protein response pathway, directly binds the GDF15 promoter to activate its transcription under endoplasmic reticulum stress conditions in the liver (64). Additionally, GDF15-deficient mice exposed to acute exercise exhibit hyperactivation of skeletal muscle Atf3 and Atf6, and Xbp1 (62), all of which induce unfolded protein response target gene expression (62). It is also known that muscle contraction upregulates synthesis, posttranslational modification, and signaling of membrane trafficking proteins (65). However, it was recently demonstrated that CHOP was not required for contraction-induced GDF15 release (66), whereas it was shown that AMPK is a central mediator when energy deficiency is present in the liver (67). Given that exercise activates AMPK signaling in skeletal muscle, there is the possibility that contraction-stimulated AMPK activation is a GDF15-driven mechanism in muscle. Whether GDF15 secretion from contracting muscle is mediated by other forms of stress, by changes in stress machinery or vesicle trafficking, or both, are areas ripe for further investigation.

We have previously shown that an acute bout of intense exercise increases GSIS in healthy young adults (68). After intense exercise, plasma insulin concentrations can increase two- to threefold to offset increased glucose production in response to prolonged glycogenolysis (69). In this situation, insulin secretion may be triggered by glucose-sensing mechanisms but may also involve direct communication between contracting muscle and pancreatic β-cells (13). There is also strong evidence that exercise training enhances β-cell function in humans with type 2 diabetes (4,6,46). Herein, we show that GDF15 is increased after exercise training in patients with type 2 diabetes and the increase correlates with improved β-cell function. Furthermore, our cell culture and animal data demonstrate that GDF15 stimulates insulin secretion in β-cells, human and mouse islets, and C57BL/6J mice. However, we recognize that GDF15’s in vivo regulation of insulin secretion in humans was not directly confirmed in the present set of studies. This will require the development and validation of endogenous labeling techniques to trace GDF15 from source to site of action.

Presumably, GDF15 synergistically mediates insulin secretion by receptor-mediated signal transduction. However, GFRAL, the widely acknowledged receptor for GDF15, is not present in the human pancreas or β-TC-6 cells (1821), though it appears to be present in pancreatic ductal adenocarcinoma (70). Nonetheless, our data rule out a direct GDF15/GFRAL signaling pathway in β-cells in normal conditions. In support of this, GDF15 is present in immortalized β-cell lineages, murine islets, and pancreatic sections from humans with type 1 diabetes and nondiabetic and type 2 diabetic human islets despite no detection of GFRAL (33,34).

Compellingly, recent work in rodents demonstrated that GDF15 activates the hypothalamic-pituitary-adrenal axis through hindbrain receptor, GFRAL (71). Endocrine responses including by the hypothalamic-pituitary-adrenal axis to exercise in humans vary depending upon the mode, frequency, and duration of exercise (72). Cortisol released in intensely exercising men suppressed the subsequent cortisol response to acute stressors (stress-buffering effect of exercise) (73,74). Thus, regardless of the absence of expression of GFRAL in the pancreas, involvement of GFRAL signaling in insulin secretion at the whole-body level as it relates to exercise-induced GDF15 and hypothalamic-pituitary-adrenal axis activation needs further investigation.

On the other hand, ErbB2 in cervical cancer cells and CD44 and CD48 in immune cells such as dendritic and regulatory T cells in a human cancer environment have been revealed recently as potential GDF15 receptors (7577). CD44 also can function as a coreceptor to the ERBB family of receptor tyrosine kinases in cancer cells (7880). Interestingly, we detected all three receptors in β-TC-6 cells expressed at the gene and surface protein levels. Thus, GDF15 may bind to an individual receptor or a complex of the coreceptors to synergistically enhance GSIS.

Conversely, GDF15 may regulate β-cell function through a glucose-dependent but nonreceptor-mediated mechanism, because GDF15 alone does not affect β-cell function. Previous studies have also reported that endogenous GDF15 is triggered in a glucose-dependent manner in HepG2 cells (81), which would be consistent with a glucose-coupled mechanism. Our data suggest that another potential mechanism may act via regulation of the insulin-release pathway. β-cells treated with GDF15 displayed enhanced glycolytic function, an ATP to ADP ratio, and intracellular calcium flux independent of altering insulin gene expression, insulin content, β-cell toxicity, or growth. These observations are consistent with studies showing that GDF15 amplifies intracellular calcium flux by increasing CaV1.3 expression (82) and promoting CaV3.1 and CaV3.3 α-subunit membrane trafficking in neurons (83).

Increased insulin secretion may also have consequences for how the β-cell itself functions, because increased glycolytic flux, decreased ADP concentration, and increased intracellular calcium concentrations can all contribute to mitochondrial oxidative stress, in turn leading to a progressive decline in insulin secretion from β-cells (84). The clinical term for this phenomenon is secondary failure, and it manifests as short-term glycemic control but worse long-term type 2 diabetes disease progression (85). However, in our study, GDF15 treatment did not induce β-cell toxicity. This may be due to increased antioxidant activity or reduced oxidative stress resulting from β-cell detoxification (86). In fact, chronic GDF15 treatment of diet-induced obese mice for 1 year reduced islet hypertrophy and improved glucose homoeostasis (26), although it is not clear whether β-cell preservation was directly mediated by GDF15 or secondary to GDF15-induced weight loss. A similar phenotype was observed in ob/ob and diet-induced obese mice (1826), making it more striking as a potential therapeutic target for type 2 diabetes.

We conclude that GDF15 secretion is increased in response to skeletal muscle contraction and exercise, and that the synergistic action of GDF15 and glucose enhances β-cell function. The mechanistic underpinnings are driven largely by acute potentiation of the insulin secretion pathway, independent of β-cell mass and GFRAL. Collectively, these findings advance our understanding of how GDF15 may contribute to regulation of glycemic control and provide important evidence for a role for GDF15 in the treatment of type 2 diabetes and related diseases.

This article contains supplementary material online at https://doi.org/10.2337/figshare.23059439.

Acknowledgments. We thank all the volunteers for participating in the human studies and the IRB staff and students who collected samples and data. We thank the Pennington Biomedical Research Center Cell Biology Core Facility for support with flow cytometry. We thank the genomics core at the Cleveland Clinic and Pennington Biomedical Research Center for support with RNA-sequencing and bioinformatic analyses. We thank the Comparative Biology Core staff at Pennington Biomedical and University of Michigan for support with animal husbandry. We thank previous and current members of the Integrated Physiology and Molecular Medicine Laboratory for their contributions to this work.

Funding. This research was supported in part by the National Institutes of Health (NIH) National Institute of Arthritis and Musculoskeletal and Skin Diseases (grant R21 AR067477), National Institute on Aging (grant R01 AG12834 to J.P.K.), National Institute of Diabetes and Digestive and Kidney Diseases (grants R01 DK108089 [to J.P.K.] and R01 DK119188 [to R.J.S.]), National Institute of General Medical Sciences (grant U54 GM104940 to J.P.K.), and UL1 RR024989 (Cleveland, OH). This work was also supported in part by an investigator-initiated award from Crossfit, Inc. (J.P.K.) and a Department of Veterans Affairs grant IK2BX005715 (to N.B.K.). This project used core facilities at Pennington Biomedical Research Center that are supported, in part, by NIH center awards P30GM118430, P20GM135002, and P30DK072476.

Duality of Interest. R.J.S. has received research support from Ethicon Endo-Surgery, Zafgen, Novo Nordisk, Kallyope, and MedImmune; has served on scientific advisory boards for Ethicon Endo-Surgery, Novo Nordisk, Sanofi, Janssen, Kallyope, Scohia, and Ironwood Pharma; and is a stakeholder in Zafgen. No other potential conflicts of interest relevant to this article were reported.

Author Contributions. H.Z. contributed to the study conceptualization, methodology, validation, formal analysis, investigation, data curation, visualization, and writing of the original draft. A.M. contributed to the study conceptualization, methodology, validation, formal analysis, investigation, review and editing of the manuscript, and project administration. S.N. contributed to the study conceptualization, methodology, validation, formal analysis, investigation, data curation, review and editing of the manuscript. B.V. contributed to the study conceptualization, methodology, formal analysis, investigation, data curation, and writing of the original draft. R.M. contributed to the study conceptualization, investigation, and review and editing of the manuscript. E.C.H. contributed to the formal analysis, investigation, and review and editing of the manuscript. E.R.M.Z. contributed to study methodology, investigation, and review and editing of the manuscript. J.J.C. contributed to the study conceptualization, methodology, resources, and review and editing of the manuscript. N.B.-K. and R.J.S contributed to the investigation, resources, and review and editing of the manuscript. C.L.A. contributed to the study conceptualization, formal analysis, data curation, writing of the original draft, visualization, supervision, and project administration. J.P.K. contributed to the study conceptualization, resources, investigation, review and editing of the manuscript, supervision, project administration, and funding acquisition, and is the guarantor and takes full responsibility for the work as a whole, including the study design, access to data, and the decision to submit and publish the manuscript.

Prior Presentation. The data in this study were presented, in part, at the American Diabetes Association’s 78th Scientific Sessions, Orlando, FL, 22–26 2018, and 79th Scientific Sessions, San Francisco, CA, 7–11 June 2019.

1.
Cernea
S
,
Dobreanu
M
.
Diabetes and beta cell function: from mechanisms to evaluation and clinical implications
.
Biochem Med (Zagreb)
2013
;
23
:
266
280
2.
Kirwan
JP
,
Heintz
EC
,
Rebello
CJ
,
Axelrod
CL
.
Exercise in the prevention and treatment of type 2 diabetes
.
Compr Physiol
2023
;
13
:
4559
4585
3.
Kirwan
JP
,
Sacks
J
,
Nieuwoudt
S
.
The essential role of exercise in the management of type 2 diabetes
.
Cleve Clin J Med
2017
;
84
(
Suppl. 1
):
S15
S21
4.
Solomon
TP
,
Haus
JM
,
Kelly
KR
,
Rocco
M
,
Kashyap
SR
,
Kirwan
JP
.
Improved pancreatic beta-cell function in type 2 diabetic patients after lifestyle-induced weight loss is related to glucose-dependent insulinotropic polypeptide
.
Diabetes Care
2010
;
33
:
1561
1566
5.
Heiskanen
MA
,
Motiani
KK
,
Mari
A
, et al
.
Exercise training decreases pancreatic fat content and improves beta cell function regardless of baseline glucose tolerance: a randomised controlled trial
.
Diabetologia
2018
;
61
:
1817
1828
6.
Dela
F
,
von Linstow
ME
,
Mikines
KJ
,
Galbo
H
.
Physical training may enhance beta-cell function in type 2 diabetes
.
Am J Physiol Endocrinol Metab
2004
;
287
:
E1024
E1031
7.
Dela
F
.
Functional adaptation of the human β-cells after frequent exposure to noradrenaline
.
J Physiol
2015
;
593
:
3199
3206
8.
Bloem
CJ
,
Chang
AM
.
Short-term exercise improves beta-cell function and insulin resistance in older people with impaired glucose tolerance
.
J Clin Endocrinol Metab
2008
;
93
:
387
392
9.
Barlow
JP
,
Solomon
TP
.
Do skeletal muscle-secreted factors influence the function of pancreatic β-cells?
Am J Physiol Endocrinol Metab
2018
;
314
:
E297
E307
10.
Pedersen
BK
,
Febbraio
MA
.
Muscle as an endocrine organ: focus on muscle-derived interleukin-6
.
Physiol Rev
2008
;
88
:
1379
1406
11.
Christensen
CS
,
Christensen
DP
,
Lundh
M
, et al
.
Skeletal muscle to pancreatic β-cell cross-talk: the effect of humoral mediators liberated by muscle contraction and acute exercise on β-cell apoptosis
.
J Clin Endocrinol Metab
2015
;
100
:
E1289
E1298
12.
Barlow
JP
,
Karstoft
K
,
Vigelsø
A
, et al
.
Beta-aminoisobutyric acid is released by contracting human skeletal muscle and lowers insulin release from INS-1 832/3 cells by mediating mitochondrial energy metabolism
.
Metabol Open
2020
;
7
:
100053
13.
Barlow
J
,
Solomon
TPJ
.
Conditioned media from contracting skeletal muscle potentiates insulin secretion and enhances mitochondrial energy metabolism of pancreatic beta-cells
.
Metabolism
2019
;
91
:
1
9
14.
Böttner
M
,
Suter-Crazzolara
C
,
Schober
A
,
Unsicker
K
.
Expression of a novel member of the TGF-beta superfamily, growth/differentiation factor-15/macrophage-inhibiting cytokine-1 (GDF-15/MIC-1) in adult rat tissues
.
Cell Tissue Res
1999
;
297
:
103
110
15.
Strelau
J
,
Böttner
M
,
Lingor
P
, et al
.
GDF-15/MIC-1 a novel member of the TGF-beta superfamily
.
J Neural Transm Suppl
2000
;(
60
):
273
276
16.
Campderrós
L
,
Sánchez-Infantes
D
,
Villarroya
J
, et al
.
Altered GDF15 and FGF21 levels in response to strenuous exercise: a study in marathon runners
.
Front Physiol
2020
;
11
:
550102
17.
Conte
M
,
Martucci
M
,
Mosconi
G
, et al
.
GDF15 plasma level is inversely associated with level of physical activity and correlates with markers of inflammation and muscle weakness
.
Front Immunol
2020
;
11
:
915
18.
Emmerson
PJ
,
Wang
F
,
Du
Y
, et al
.
The metabolic effects of GDF15 are mediated by the orphan receptor GFRAL
.
Nat Med
2017
;
23
:
1215
1219
19.
Mullican
SE
,
Lin-Schmidt
X
,
Chin
CN
, et al
.
GFRAL is the receptor for GDF15 and the ligand promotes weight loss in mice and nonhuman primates
.
Nat Med
2017
;
23
:
1150
1157
20.
Yang
L
,
Chang
CC
,
Sun
Z
, et al
.
GFRAL is the receptor for GDF15 and is required for the anti-obesity effects of the ligand
.
Nat Med
2017
;
23
:
1158
1166
21.
Hsu
JY
,
Crawley
S
,
Chen
M
, et al
.
Non-homeostatic body weight regulation through a brainstem-restricted receptor for GDF15
.
Nature
2017
;
550
:
255
259
22.
Tsai
VWW
,
Husaini
Y
,
Sainsbury
A
,
Brown
DA
,
Breit
SN
.
The MIC-1/GDF15-GFRAL pathway in energy homeostasis: implications for obesity, cachexia, and other associated diseases
.
Cell Metab
2018
;
28
:
353
368
23.
Johnen
H
,
Lin
S
,
Kuffner
T
, et al
.
Tumor-induced anorexia and weight loss are mediated by the TGF-beta superfamily cytokine MIC-1
.
Nat Med
2007
;
13
:
1333
1340
24.
Macia
L
,
Tsai
VW
,
Nguyen
AD
, et al
.
Macrophage inhibitory cytokine 1 (MIC-1/GDF15) decreases food intake, body weight and improves glucose tolerance in mice on normal & obesogenic diets
.
PLoS One
2012
;
7
:
e34868
25.
Chung
HK
,
Ryu
D
,
Kim
KS
, et al
.
Growth differentiation factor 15 is a myomitokine governing systemic energy homeostasis
.
J Cell Biol
2017
;
216
:
149
165
26.
Xiong
Y
,
Walker
K
,
Min
X
, et al
.
Long-acting MIC-1/GDF15 molecules to treat obesity: evidence from mice to monkeys
.
Sci Transl Med
2017
;
9
:
eaan8732
27.
Frikke-Schmidt
H
,
Hultman
K
,
Galaske
JW
,
Jørgensen
SB
,
Myers
MG
Jr
,
Seeley
RJ
.
GDF15 acts synergistically with liraglutide but is not necessary for the weight loss induced by bariatric surgery in mice
.
Mol Metab
2019
;
21
:
13
21
28.
Tsai
VW
,
Macia
L
,
Johnen
H
, et al
.
TGF-b superfamily cytokine MIC-1/GDF15 is a physiological appetite and body weight regulator
.
PLoS One
2013
;
8
:
e55174
29.
Tran
T
,
Yang
J
,
Gardner
J
,
Xiong
Y
.
GDF15 deficiency promotes high fat diet-induced obesity in mice
.
PLoS One
2018
;
13
:
e0201584
30.
Schernthaner-Reiter
MH
,
Itariu
BK
,
Krebs
M
, et al
.
GDF15 reflects beta cell function in obese patients independently of the grade of impairment of glucose metabolism
.
Nutr Metab Cardiovasc Dis
2019
;
29
:
334
342
31.
Vila
G
,
Riedl
M
,
Anderwald
C
, et al
.
The relationship between insulin resistance and the cardiovascular biomarker growth differentiation factor-15 in obese patients
.
Clin Chem
2011
;
57
:
309
316
32.
Asrih
M
,
Dusaulcy
R
,
Gosmain
Y
, et al
.
Growth differentiation factor-15 prevents glucotoxicity and connexin-36 downregulation in pancreatic beta-cells
.
Mol Cell Endocrinol
2022
;
541
:
111503
33.
Mohammad
MG
,
Saeed
R
,
Mohammed
AK
, et al
.
GDF15 plays a critical role in insulin secretion in INS-1 cells and human pancreatic islets
.
Exp Biol Med (Maywood)
2023
;
248
:
339
349
34.
Nakayasu
ES
,
Syed
F
,
Tersey
SA
, et al
.
Comprehensive proteomics analysis of stressed human islets identifies GDF15 as a target for type 1 diabetes intervention
.
Cell Metab
2020
;
31
:
363
374.e6
35.
Stolina
M
,
Luo
X
,
Dwyer
D
, et al
.
The evolving systemic biomarker milieu in obese ZSF1 rat model of human cardiometabolic syndrome: characterization of the model and cardioprotective effect of GDF15
.
PLoS One
2020
;
15
:
e0231234
36.
Nieuwoudt
S
,
Mulya
A
,
Fealy
CE
, et al
.
In vitro contraction protects against palmitate-induced insulin resistance in C2C12 myotubes
.
Am J Physiol Cell Physiol
2017
;
313
:
C575
C583
37.
Petersen
TN
,
Brunak
S
,
von Heijne
G
,
Nielsen
H
.
SignalP 4.0: discriminating signal peptides from transmembrane regions
.
Nat Methods
2011
;
8
:
785
786
38.
Viklund
H
,
Bernsel
A
,
Skwark
M
,
Elofsson
A
.
SPOCTOPUS: a combined predictor of signal peptides and membrane protein topology
.
Bioinformatics
2008
;
24
:
2928
2929
39.
Käll
L
,
Krogh
A
,
Sonnhammer
EL
.
A combined transmembrane topology and signal peptide prediction method
.
J Mol Biol
2004
;
338
:
1027
1036
40.
Patel
AR
,
Frikke-Schmidt
H
,
Bezy
O
, et al
.
LPS induces rapid increase in GDF15 levels in mice, rats, and humans but is not required for anorexia in mice
.
Am J Physiol Gastrointest Liver Physiol
2022
;
322
:
G247
G255
41.
Axelrod
CL
,
King
WT
,
Davuluri
G
, et al
.
BAM15-mediated mitochondrial uncoupling protects against obesity and improves glycemic control
.
EMBO Mol Med
2020
;
12
:
e12088
42.
Kelly
KR
,
Haus
JM
,
Solomon
TP
, et al
.
A low-glycemic index diet and exercise intervention reduces TNF(alpha) in isolated mononuclear cells of older, obese adults
.
J Nutr
2011
;
141
:
1089
1094
43.
Solomon
TP
,
Sistrun
SN
,
Krishnan
RK
, et al
.
Exercise and diet enhance fat oxidation and reduce insulin resistance in older obese adults
.
J Appl Physiol (1985)
2008
;
104
:
1313
1319
44.
Haus
JM
,
Solomon
TP
,
Kelly
KR
, et al
.
Improved hepatic lipid composition following short-term exercise in nonalcoholic fatty liver disease
.
J Clin Endocrinol Metab
2013
;
98
:
E1181
E1188
45.
Zhang
H
,
Fealy
CE
,
Kirwan
JP
.
Exercise training promotes a GDF15-associated reduction in fat mass in older adults with obesity
.
Am J Physiol Endocrinol Metab
2019
;
316
:
E829
E836
46.
Nieuwoudt
S
,
Fealy
CE
,
Foucher
JA
, et al
.
Functional high-intensity training improves pancreatic β-cell function in adults with type 2 diabetes
.
Am J Physiol Endocrinol Metab
2017
;
313
:
E314
E320
47.
Fealy
CE
,
Nieuwoudt
S
,
Foucher
JA
, et al
.
Functional high-intensity exercise training ameliorates insulin resistance and cardiometabolic risk factors in type 2 diabetes
.
Exp Physiol
2018
;
103
:
985
994
48.
Axelrod
CL
,
Fealy
CE
,
Mulya
A
,
Kirwan
JP
.
Exercise training remodels human skeletal muscle mitochondrial fission and fusion machinery towards a pro-elongation phenotype
.
Acta Physiol (Oxf)
2019
;
225
:
e13216
49.
Mulya
A
,
Haus
JM
,
Solomon
TP
, et al
.
Exercise training-induced improvement in skeletal muscle PGC-1α-mediated fat metabolism is independent of dietary glycemic index
.
Obesity (Silver Spring)
2017
;
25
:
721
729
50.
American Diabetes Association
.
2. Classification and diagnosis of diabetes: Standards of Medical Care in Diabetes-2020
.
Diabetes Care
2020
;
43
(
Suppl. 1
):
S14
S31
51.
Park
S
,
Turner
KD
,
Zheng
D
, et al
.
Electrical pulse stimulation induces differential responses in insulin action in myotubes from severely obese individuals
.
J Physiol
2019
;
597
:
449
466
52.
Fischer
CP
.
Interleukin-6 in acute exercise and training: what is the biological relevance?
Exerc Immunol Rev
2006
;
12
:
6
33
53.
Evers-van Gogh
IJ
,
Alex
S
,
Stienstra
R
,
Brenkman
AB
,
Kersten
S
,
Kalkhoven
E
.
Electric pulse stimulation of myotubes as an in vitro exercise model: cell-mediated and non-cell-mediated effects
.
Sci Rep
2015
;
5
:
10944
54.
Olsen
OE
,
Skjærvik
A
,
Størdal
BF
,
Sundan
A
,
Holien
T
.
TGF-β contamination of purified recombinant GDF15
.
PLoS One
2017
;
12
:
e0187349
55.
Radwanska
A
,
Cottage
CT
,
Piras
A
, et al
.
Increased expression and accumulation of GDF15 in IPF extracellular matrix contribute to fibrosis
.
JCI Insight
2022
;
7
:
e153058
56.
Laurens
C
,
Parmar
A
,
Murphy
E
, et al
.
Growth and differentiation factor 15 is secreted by skeletal muscle during exercise and promotes lipolysis in humans
.
JCI Insight
2020
;
5
:
131870
57.
Patel
S
,
Alvarez-Guaita
A
,
Melvin
A
, et al
.
GDF15 provides an endocrine signal of nutritional stress in mice and humans
.
Cell Metab
2019
;
29
:
707
718.e8
58.
Chung
HK
,
Kim
JT
,
Kim
HW
, et al
.
GDF15 deficiency exacerbates chronic alcohol- and carbon tetrachloride-induced liver injury
.
Sci Rep
2017
;
7
:
17238
59.
Kleinert
M
,
Clemmensen
C
,
Sjøberg
KA
, et al
.
Exercise increases circulating GDF15 in humans
.
Mol Metab
2018
;
9
:
187
191
60.
Tchou
I
,
Margeli
A
,
Tsironi
M
, et al
.
Growth-differentiation factor-15, endoglin and N-terminal pro-brain natriuretic peptide induction in athletes participating in an ultramarathon foot race
.
Biomarkers
2009
;
14
:
418
422
61.
Galliera
E
,
Lombardi
G
,
Marazzi
MG
, et al
.
Acute exercise in elite rugby players increases the circulating level of the cardiovascular biomarker GDF-15
.
Scand J Clin Lab Invest
2014
;
74
:
492
499
62.
Gil
CI
,
Ost
M
,
Kasch
J
,
Schumann
S
,
Heider
S
,
Klaus
S
.
Role of GDF15 in active lifestyle induced metabolic adaptations and acute exercise response in mice
.
Sci Rep
2019
;
9
:
20120
63.
Klein
AB
,
Nicolaisen
TS
,
Ørtenblad
N
, et al
.
Pharmacological but not physiological GDF15 suppresses feeding and the motivation to exercise
.
Nat Commun
2021
;
12
:
1041
64.
Li
D
,
Zhang
H
,
Zhong
Y
.
Hepatic GDF15 is regulated by CHOP of the unfolded protein response and alleviates NAFLD progression in obese mice
.
Biochem Biophys Res Commun
2018
;
498
:
388
394
65.
Towler
MC
,
Kaufman
SJ
,
Brodsky
FM
.
Membrane traffic in skeletal muscle
.
Traffic
2004
;
5
:
129
139
66.
Townsend
LK
,
Medak
KD
,
Weber
AJ
,
Dibe
H
,
Shamshoum
H
,
Wright
DC
.
CHOP is dispensable for exercise-induced increases in GDF15
.
J Appl Physiol
1985
;
132
:
413
422
67.
Townsend
LK
,
Weber
AJ
,
Day
EA
, et al
.
AMPK mediates energetic stress-induced liver GDF15
.
FASEB J
2021
;
35
:
e21218
68.
Kirwan
JP
,
Bourey
RE
,
Kohrt
WM
,
Staten
MA
,
Holloszy
JO
.
Effects of treadmill exercise to exhaustion on the insulin response to hyperglycemia in untrained men
.
J Appl Physiol (1985)
1991
;
70
:
246
250
69.
Marliss
EB
,
Vranic
M
.
Intense exercise has unique effects on both insulin release and its roles in glucoregulation: implications for diabetes
.
Diabetes
2002
;
51
(
Suppl. 1
):
S271
S283
70.
Zhao
Z
,
Zhang
J
,
Yin
L
, et al
.
Upregulated GDF-15 expression facilitates pancreatic ductal adenocarcinoma progression through orphan receptor GFRAL
.
Aging (Albany NY)
2020
;
12
:
22564
22581
71.
Cimino
I
,
Kim
H
,
Tung
YCL
, et al
.
Activation of the hypothalamic-pituitary-adrenal axis by exogenous and endogenous GDF15
.
Proc Natl Acad Sci USA
2021
;
118
:
e2106868118
72.
Athanasiou
N
,
Bogdanis
GC
,
Mastorakos
G
.
Endocrine responses of the stress system to different types of exercise
.
Rev Endocr Metab Disord
2023
;
24
:
251
266
73.
Zschucke
E
,
Renneberg
B
,
Dimeo
F
,
Wüstenberg
T
,
Ströhle
A
.
The stress-buffering effect of acute exercise: evidence for HPA axis negative feedback
.
Psychoneuroendocrinology
2015
;
51
:
414
425
74.
Caplin
A
,
Chen
FS
,
Beauchamp
MR
,
Puterman
E
.
The effects of exercise intensity on the cortisol response to a subsequent acute psychosocial stressor
.
Psychoneuroendocrinology
2021
;
131
:
105336
75.
Li
S
,
Ma
YM
,
Zheng
PS
,
Zhang
P
.
GDF15 promotes the proliferation of cervical cancer cells by phosphorylating AKT1 and Erk1/2 through the receptor ErbB2
.
J Exp Clin Cancer Res
2018
;
37
:
80
76.
Gao
Y
,
Xu
Y
,
Zhao
S
, et al
.
Growth differentiation factor-15 promotes immune escape of ovarian cancer via targeting CD44 in dendritic cells
.
Exp Cell Res
2021
;
402
:
112522
77.
Wang
Z
,
He
L
,
Li
W
, et al
.
GDF15 induces immunosuppression via CD48 on regulatory T cells in hepatocellular carcinoma
.
J Immunother Cancer
2021
;
9
:
e002787
78.
Wobus
M
,
Kuns
R
,
Sheyn
I
, et al
.
Endometrial carcinoma cells are nonpermissive for CD44-erbB2 interactions
.
Appl Immunohistochem Mol Morphol
2002
;
10
:
242
246
79.
Ponta
H
,
Sherman
L
,
Herrlich
PA
.
CD44: from adhesion molecules to signalling regulators
.
Nat Rev Mol Cell Biol
2003
;
4
:
33
45
80.
Pereira
PMR
,
Ragupathi
A
,
Shmuel
S
,
Mandleywala
K
,
Viola
NT
,
Lewis
JS
.
HER2-targeted PET imaging and therapy of hyaluronan-masked HER2-overexpressing breast cancer
.
Mol Pharm
2020
;
17
:
327
337
81.
Schernthaner-Reiter
MH
,
Kasses
D
,
Tugendsam
C
, et al
.
Growth differentiation factor 15 increases following oral glucose ingestion: effect of meal composition and obesity
.
Eur J Endocrinol
2016
;
175
:
623
631
82.
Lu
JM
,
Wang
CY
,
Hu
C
,
Fang
YJ
,
Mei
YA
.
GDF-15 enhances intracellular Ca2+ by increasing Cav1.3 expression in rat cerebellar granule neurons
.
Biochem J
2016
;
473
:
1895
1904
83.
Liu
DD
,
Lu
JM
,
Zhao
QR
,
Hu
C
,
Mei
YA
.
Growth differentiation factor-15 promotes glutamate release in medial prefrontal cortex of mice through upregulation of T-type calcium channels
.
Sci Rep
2016
;
6
:
28653
84.
Fridlyand
LE
,
Philipson
LH
.
Does the glucose-dependent insulin secretion mechanism itself cause oxidative stress in pancreatic beta-cells?
Diabetes
2004
;
53
:
1942
1948
85.
Sola
D
,
Rossi
L
,
Schianca
GP
, et al
.
Sulfonylureas and their use in clinical practice
.
Arch Med Sci
2015
;
11
:
840
848
86.
Stancill
JS
,
Broniowska
KA
,
Oleson
BJ
,
Naatz
A
,
Corbett
JA
.
Pancreatic β-cells detoxify H2O2 through the peroxiredoxin/thioredoxin antioxidant system
.
J Biol Chem
2019
;
294
:
4843
4853
Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. More information is available at https://www.diabetesjournals.org/journals/pages/license.