Although insulin resistance is known to underlie type 2 diabetes, its role in the development of type 1 diabetes has been gaining increasing interest. In a model of type 1 diabetes, the nonobese diabetic (NOD) mouse, we found that insulin resistance driven by lipid- and glucose-independent mechanisms is already present in the liver of prediabetic mice. Hepatic insulin resistance is associated with a transient rise in mitochondrial respiration followed by increased production of lipid peroxides and c-Jun N-terminal kinase activity. At the onset of diabetes, increased adipose tissue lipolysis promotes myocellular diacylglycerol accumulation. This is paralleled by increased myocellular protein kinase C θ activity and serum fetuin A levels. Muscle mitochondrial oxidative capacity is unchanged at the onset but decreases at later stages of diabetes. In conclusion, hepatic and muscle insulin resistance manifest at different stages and involve distinct cellular mechanisms during the development of diabetes in the NOD mouse.

Insulin resistance not only is a feature of obesity and type 2 diabetes mellitus (T2DM) but also can be present in patients with type 1 diabetes mellitus (T1DM) (1,2), resulting in a condition termed “double diabetes.” T1DM is characterized by absolute insulin deficiency resulting from autoimmune-mediated β-cell destruction (3), leading to uncontrolled glucose homeostasis. Although insulin resistance in T1DM may occur independently, the accelerator hypothesis postulates that it can be triggered by autoimmune diabetes itself (4). The causes and mechanisms linking T1DM and insulin resistance remain unknown.

Prolonged hyperglycemia may induce insulin resistance by mechanisms summarized as glucotoxicity (5). On the other hand, insulin deficiency also decreases the suppression of lipolysis in adipose tissue and liver, thereby raising circulating triglyceride (TG) and fatty acid (FA) levels, which in turn impair insulin-stimulated glucose transport by various mechanisms summarized as lipotoxicity (6,7). Both the diacylglycerol (DAG)/protein kinase C (PKC) isoforms pathway (6) and toll-like receptor 4 (TLR4)/fetuin A-mediated ceramide synthesis (8) have been shown to inhibit insulin signaling.

Furthermore, glucotoxicity and lipotoxicity strongly associate with lower mitochondrial function. In muscle, lower mitochondrial activity is frequently associated with insulin resistance in humans with or at risk for T2DM (9,10). Insulin deprivation can also reduce muscle mitochondrial ATP production by altered expression of mitochondrial genes (11), and insulin-resistant patients with long-standing T1DM may have lower ATP synthase flux (1). Studies have shown that reduced hepatic energy metabolism also correlates with insulin resistance, hyperglycemia, and altered hepatic lipid content in T2DM (12,13). However, others questioned the role of mitochondrial function in insulin resistance (14). The contribution of mitochondrial function to insulin resistance has not yet been clarified in T1DM.

We hypothesized that the onset of T1DM is associated with impaired insulin sensitivity and lower mitochondrial function. Although glucolipotoxicity may cause insulin resistance in the liver, muscle insulin resistance may develop independent of ambient glycemia. To test this hypothesis, we examined diabetes-related effects in female nonobese diabetic (NOD) mice independently of other confounding factors, such as sex, obesity, aging, and inherited mitochondrial abnormalities.

Animals

NOD and wild-type (WT) C57BL/6 mice were maintained under specific pathogen-free conditions on a 12-h light-dark cycle and received a standard rodent diet (ssniff M-Z Extrudate, 4.5% fat; ssniff Spezialdiäten GmbH, Soest, Germany) and water ad libitum. We studied only female mice because 80% of female but only 30% of male NOD develop diabetes within 30 weeks of age (15). Experiments were performed 3 days (acute diabetic NOD [A-DM]) or 8 weeks after onset of diabetes (insulin-treated chronic diabetic NOD [C-DM]) in randomly assigned mice. Nondiabetic NOD (N-DM) mice were matched for age and body weight. Mice were studied in the fed state and after 6 h of fasting. Insulin-treated C-DM were examined only in the fed state to avoid hypoglycemia. After decapitation, trunk blood and tissues were collected, weighed, and used for high-resolution respirometry or snap frozen in liquid nitrogen. All experiments were performed according to the guidelines for the care and use of animals (GV-SOLAS [Society for Laboratory Animal Science]) and approved by the local council of animal care in line with the requirements of the German animal protection act.

Detection and Treatment of Hyperglycemic NOD Mice

Diabetes was detected by testing for glycosuria (Diabur-test 5000; Roche Diagnostics, Mannheim, Germany) and confirmed by tail blood glucose measurements >250 mg/dL on 2 consecutive days (Precision Xtra Plus; Abbott, Wiesbaden, Germany). Insulin was administered subcutaneously in LinBit pellets (LinShin Canada, Toronto, ON, Canada) for 8 weeks, aiming to maintain blood glucose between 250 and 500 mg/dL. At glucose levels >500 mg/dL for >1 week, an additional insulin pellet was administered.

Indirect Calorimetry

Mice were individually housed and placed in an eight-chamber indirect calorimetry system (PhenoMaster; TSE Systems, Bad Homburg, Germany). After 24 h of acclimatization, energy expenditure, respiratory quotient (RQ), physical activity, and food and water intake were simultaneously analyzed for 48 h.

Hyperinsulinemic-Euglycemic Clamp Test

A silicon catheter (Silastic Laboratory Tubing, Dow Corning, Midland, MI) was placed into the right-side jugular vein under Isofluran (CP Pharma, Burgdorf, Germany) anesthesia. Mice were allowed to recover for 4–5 days and fasted for 6 h on the day of the experiment (3:00–9:00 a.m.). To assess basal whole-body glucose disposal, d-[6,6-2H2]glucose (98% enriched) (Cambridge Isotope Laboratories, Andover, MA) was infused at a rate of 4 μmol/kg/min for 120 min. The hyperinsulinemic-euglycemic clamp was performed with a primed (40 mU/kg), continuous infusion (4 mU/kg/min) (Huminsulin; Lilly, Giessen, Germany) for 180 min. Euglycemia was maintained by periodically adjusting a variable 20% glucose infusion. d-[6,6-2H2]glucose was coinfused together with insulin solution (0.4 μmol/kg/min) and variable glucose infusion to obtain stable tracer concentrations during varying glucose infusion rates. Blood samples were taken at 10-min intervals during the last 30 min of basal and hyperinsulinemic-euglycemic clamps. Additional clamps were performed under identical conditions except for measuring FA levels at time points −15, 0, 5, 15, 30, 60, 90, 120, and 150 min to assess FA suppression and injecting 10 μCi of 2-deoxy-d-[1-14C]glucose at the end of the clamp to calculate rates of insulin-stimulated glucose uptake by gastrocnemius and soleus muscles (16). After the clamps, mice were exsanguinated through cervical incision and killed by cervical dislocation, and serum and organs were collected for analyses.

High-Resolution Respirometry

Ex vivo mitochondrial function was measured in fresh liver and gastrocnemius muscle using the Oxygraph-2k (Oroboros Instruments, Austria) as described (17). Defined respiratory states were obtained by the following protocol: 2 mmol/L malate, 10 mmol/L pyruvate, 10 mmol/L glutamate and 2.5 mmol/L ADP (state 3, complex I [CI]), 10 mmol/L succinate (state 3, CI + complex II [CII]), 10 μmol/L cytochrome c (mitochondrial membrane integrity check), carbonyl cyanide-p-trifluoromethoxyphenylhydrazone (stepwise increments of 0.25 μmol/L up to the final concentration of maximum 1.25 μmol/L, state u), and 2.5 μmol/L antimycin A. Addition of cytochrome c did not increase oxygen consumption, indicating integrity of the outer mitochondrial membrane after saponin permeabilization.

Mitochondrial Density

Tissue DNA was extracted with a Qiagen Tissue and Blood Kit (Qiagen, Hilden, Germany), and concentration was measured spectrophotometrically (GeneQuant; GE Healthcare, Munich, Germany). Mitochondrial DNA (mtDNA) copy number was quantified with RT-PCR (ABI Prism 7000; Applied Biosystems, Darmstadt, Germany) by using specific primers and 5′ FAM + 3′ TAMRA–labeled probes (Eurogentec, Liège, Belgium) for NADH dehydrogenase subunit 1 (forward primer: 5′-CTA-CAA-CCA-TTT-GCA-GAC-GC-3′, reverse primer 5′-GGA-ACT-CAT-AGA-CTT-AAT-GC-3′, probe 5′-CCA-ATA-CGC-CCT-TTA-ACA-ACC-TC-3′) and for lipoprotein lipase (forward primer: 5′-GGT-TTG-GAT-CCA-GCT-GGG-CC-3′, reverse primer 5′-GAT-TCC-AAT-ACT-TCG-ACC-AGG-3′, probe 5′-CTT-TGA-GTA-TGC-AGA-AGC-CC-3′). NADH dehydrogenase subunit 1 and lipoprotein lipase DNA copy numbers were determined by comparison with log-linear standard curves. The ratio of mtDNA to nuclear DNA is a measure of tissue concentration of mtDNA per cell.

Laboratory Analyses

Serum insulin (Mouse Insulin Elisa Kit; Mercodia, Uppsala, Sweden) and fetuin A (Mouse Fetuin A/AHSG DuoSet; R&D Systems, Abingdon, U.K.) were measured by ELISA. d-[6,6-2H2]glucose enrichment in deproteinized plasma was quantified with gas chromatography mass spectrometry (Agilent Technologies, Waldbronn, Germany) after derivatization of glucose to pentaacetate. Serum TG, cholesterol (Roche/Hitachi, Roche Diagnostics, Mannheim, Germany), FA, and β-hydroxybutyrate (Wako Chemicals GmbH, Neuss, Germany) levels were assessed photometrically.

Assessment of Lipid Peroxidation

Tissue concentrations of thiobarbituric acid reactive substances (TBARS) were measured fluorometrically (BioTek, Bad Friedrichshall, Germany) in 10 mg of frozen gastrocnemius muscle and liver (18). Protein content in homogenates was assessed with the bicinchoninic acid assay (BCA Protein Assay Kit; Thermo Fisher Scientific, Bonn, Germany).

Insulin Signaling Ex Vivo

Mice were injected intraperitoneally with insulin 1 unit/kg body weight or saline (control) and killed after 10 min, and liver and muscle tissues were snap frozen in liquid nitrogen.

RNA and Protein Analyses

Total RNA extraction, cDNA synthesis, and real-time quantitative RT-PCR expression analyses were performed as described (19). Data of gene-specific probes (Assays-on-Demand; Applied Biosystems, Darmstadt, Germany) were normalized to 18S rRNA content (19).

Proteins were extracted by homogenization and centrifugation (21,000g for 15 min at 4°C) and measured by the BCA Protein Assay Kit (Thermo Fisher Scientific). Membrane and cytosol fractions were prepared by using differential centrifugation (20).

Ten micrograms of protein were loaded onto SDS-PAGE gels and transferred to polyvinylidene difluoride membranes (Merck Millipore, Schwalbach, Germany). Membranes were blocked with 5% milk in PBS and incubated with antibodies recognizing total Akt isoforms 1 and 2, extracellular signal-regulated kinase phosphorylated at Thr202 and Tyr204 (pERK-Thr202/Tyr204), c-Jun N-terminal kinase phosphorylated at Thr183 and Tyr185 (pJNK-Thr183/Tyr185), nuclear factor κ-light-chain-enhancer of activated B cells phosphorylated at Ser536 (pNFκB-Ser536), p38 mitogen-activated protein kinase phosphorylated at Thr180 and Tyr182 (p-p38-Thr180/Tyr182), Ser1101 phosphorylation of insulin receptor substrate 1 (IRS1) (pIRS1-Ser1101) and Ser307 phosphorylation of IRS1 (pIRS1-Ser307) (Cell Signaling Technology, Danvers, MA), GLUT4 (Abcam, Cambridge, U.K.), PKCε and PKCθ (BD Biosciences, Franklin Lakes, NJ), and the insulin receptor β-subunit (Santa Cruz Biotechnology, Santa Cruz, CA). Horseradish peroxidase–conjugated secondary antibodies (Promega GmbH, Mannheim, Germany) and enhanced chemiluminescence (Merck Millipore) were used for detection. Immunoblots were quantified using VersaDoc 4000 MP (Bio-Rad) and Quantity One version 4.6.9 (Bio-Rad) software. Stripped membranes were reprobed with α-tubulin (Calbiochem, Darmstadt, Germany) and GAPDH (Abcam) antibodies to allow for loading corrections.

Quantification of Lipids

Oil red O staining was performed in fresh right-side liver lobes fixed in 4% paraformaldehyde (21). Stained sections were examined by light microscopy at ×40 magnification (Leica DMRB; Leica Microsystems, Wetzlar, Germany). Liver and muscle samples were solubilized in ethanolic KOH and glycerolipids measured as described (22) by using a TG standard (Lyonorm Calibrator; PLIVA-Lachema Diagnostika, Brno, Czech Republic).

Myocellular Lipid Metabolites

Membrane and cytosol fractions were extracted by homogenization and centrifugation (20) and DAG and ceramide levels measured by liquid chromatography-mass spectrometry (23).

Immunohistochemistry

Pancreatic tissue was embedded in paraffin, stained by hematoxylin-eosin, and examined with light microsopy (24). For β-cell and α-cell detection, double immunofluorescence staining was performed by using anti-insulin and antiglucagon antibodies (Dako, Hamburg, Germany), respectively. Images were acquired using inverted microscope (Leica Microsystems) and digital camera (Olympus Europa, Hamburg, Germany).

Calculations and Statistical Analyses

Glucose disposal (Rd) was calculated by Steele single-pool non–steady-state equations (25). Endogenous glucose production (EGP) is given as the difference between Rd and glucose infusion rate.

Surrogate indexes of insulin sensitivity and secretion were calculated from fasting blood glucose (G) and serum insulin (I) levels as follows: QUICKI = 1 / [log(I in μU/mL) + log(G in mg/dL)]) (26) and HOMA-B = [20 × (I in μU/mL)] / [(G in mmol/L) – 3.5] (27).

Data are presented as mean ± SD in the text and tables, and as mean ± SEM in figures. Groups were compared by Bonferroni test or nonparametric unpaired t test (Mann-Whitney) with Hochberg post hoc analysis. P < 0.05 is considered to indicate statistically significant differences.

Both Diabetic and Nondiabetic NOD Mice Are Insulin Resistant

Body weight was similar among A-DM, N-DM, and C-DM mice and slightly higher than that of WT mice (Table 1). In the fed state, blood glucose was fourfold higher in A-DM and C-DM versus N-DM and WT (Supplementary Table 1). Insulin levels were lower in A-DM than N-DM (Table 1). During fasting, circulating glucose, TG, and FA levels were increased in A-DM compared with N-DM and WT (Fig. 1A–C). QUICKI was 13% and 9% lower in A-DM and N-DM, respectively, than in WT, suggesting glucose-independent insulin resistance in the NOD model (Table 1). HOMA-B was 3.5- and 5.5-fold higher in N-DM than in WT and A-DM, respectively, fitting with the previous observation that prediabetic NOD mice have higher in vivo and ex vivo glucose-stimulated insulin secretion than C57BL/6 mice (28). A-DM had lower HOMA-B and insulin levels in the fed state than N-DM, illustrating the impaired β-cell function resulting from the severe insulitis (data not shown) typical for the NOD model (24).

Table 1

Characteristics of WT, N-DM, A-DM, and C-DM mice in the fasted and fed state

Fasted state
Fed state
WTN-DMA-DMWTN-DMA-DMC-DM
Females (n27 26 25 26 28 29 
Age (days) 152 ± 33 149 ± 25 156 ± 29 154 ± 36 165 ± 28 145 ± 40 180 ± 28 
Body weight (g) 22 ± 2 24 ± 2* 24 ± 2* 23 ± 2 26 ± 2# 25 ± 2* 27 ± 2* 
Insulin (pmol/L) 86 ± 45 267 ± 143 224 ± 404 99 ± 73 583 ± 623* 227 ± 390 554 ± 326 
QUICKI 0.32 ± 0.02 0.29 ± 0.03# 0.28 ± 0.03# NA NA NA NA 
HOMA-B 74 ± 39 260 ± 184# 47 ± 62§ NA NA NA NA 
Fasted state
Fed state
WTN-DMA-DMWTN-DMA-DMC-DM
Females (n27 26 25 26 28 29 
Age (days) 152 ± 33 149 ± 25 156 ± 29 154 ± 36 165 ± 28 145 ± 40 180 ± 28 
Body weight (g) 22 ± 2 24 ± 2* 24 ± 2* 23 ± 2 26 ± 2# 25 ± 2* 27 ± 2* 
Insulin (pmol/L) 86 ± 45 267 ± 143 224 ± 404 99 ± 73 583 ± 623* 227 ± 390 554 ± 326 
QUICKI 0.32 ± 0.02 0.29 ± 0.03# 0.28 ± 0.03# NA NA NA NA 
HOMA-B 74 ± 39 260 ± 184# 47 ± 62§ NA NA NA NA 

Data are mean ± SD unless otherwise indicated. P values by ANOVA with Bonferroni post hoc analysis or nonparametrical test (Mann-Whitney) with Hochberg post hoc analysis. NA, not assessed.

*P < 0.01 vs. WT in respective metabolic state.

#P < 0.001 vs. WT in respective metabolic state.

P < 0.01 vs. N-DM in respective metabolic state.

§P < 0.001 vs. N-DM in respective metabolic state.

Figure 1

Serum concentrations of glucose (A), TG (B), FA (C), and fetuin A (D) of WT, N-DM, and A-DM mice in the fasted state. Data are mean ± SEM (n = 8–26 per group). **P < 0.01, ***P < 0.001 by ANOVA with Bonferroni post hoc analysis.

Figure 1

Serum concentrations of glucose (A), TG (B), FA (C), and fetuin A (D) of WT, N-DM, and A-DM mice in the fasted state. Data are mean ± SEM (n = 8–26 per group). **P < 0.01, ***P < 0.001 by ANOVA with Bonferroni post hoc analysis.

Close modal

Diabetic NOD Mice Lose Fat Mass and Rely on Fat Oxidation

A-DM had substantially (60–75%) lower visceral and subcutaneous fat masses (Supplementary Fig. 1A and B). Muscle TG level was unchanged except for a small increase in ad libitum–fed N-DM (Supplementary Fig. 1C). Liver weight was comparable between groups, whereas hepatic TG level was lower in fasted A-DM (Supplementary Figs. 1A–D and 2).

Only WT and N-DM displayed normal diurnal energy expenditure patterns (Fig. 2A and B). A-DM showed a shift in RQ to 0.81 with preferential lipid oxidation (Fig. 2C and D), which was supported by a trend toward higher serum β-hydroxybutyrate levels (0.62 ± 0.22 mmol/L) versus N-DM (0.25 ± 0.07 mmol/L; P = 0.083). Finally, A-DM exhibited lower physical activity during the dark cycle (Fig. 2E) and increased overall food and water intake (Fig. 2F and G). Energy expenditure was similar among groups (Fig. 2H).

Figure 2

Energy balance of WT, N-DM, and A-DM mice. Time course of RQs of WT (A), N-DM (B), and A-DM (C) during light and dark cycles. Average RQ values (D), activity counts (E), food intake (F), water intake (G), and energy expenditure (H) during light and dark cycles. Data are mean ± SEM (n = 5 per group). *P < 0.05, **P < 0.01, ***P < 0.001 by ANOVA with Bonferroni post hoc analysis. D, dark; L, light.

Figure 2

Energy balance of WT, N-DM, and A-DM mice. Time course of RQs of WT (A), N-DM (B), and A-DM (C) during light and dark cycles. Average RQ values (D), activity counts (E), food intake (F), water intake (G), and energy expenditure (H) during light and dark cycles. Data are mean ± SEM (n = 5 per group). *P < 0.05, **P < 0.01, ***P < 0.001 by ANOVA with Bonferroni post hoc analysis. D, dark; L, light.

Close modal

Diabetic and Nondiabetic NOD Mice Show Tissue-Dependent Differences in Insulin Sensitivity

During the hyperinsulinemic-euglycemic clamps, steady-state blood glucose (Fig. 3A) and serum insulin (not shown) levels were comparable. A-DM had higher fasting EGP versus WT and N-DM (Fig. 3B). In A-DM, EGP was positively related to fasting glycemia (r = 0.95, P < 0.05, n = 9). Both A-DM and N-DM displayed lower insulin-mediated suppression of EGP than did WT (Fig. 3C), indicating hepatic insulin resistance. A-DM showed impaired insulin-stimulated Rd, which was 62% and 66% reduced compared with WT and N-DM (Fig. 3D). Uptake of 2-deoxyglucose by gastrocnemius and soleus muscles was also lower in A-DM (Fig. 3E and F). In A-DM, plasma FA level decreased similarly to WT and N-DM during the first 15 min but remained higher from 30 min to the end of the clamp (Fig. 2G). Overall percent suppression of FA in A-DM was comparable to N-DM (Fig. 2H), suggesting intact insulin sensitivity of adipose tissue.

Figure 3

Insulin sensitivity in WT, N-DM, and A-DM mice. A: Blood glucose levels before and during the hyperinsulinemic-euglycemic clamp. B: EGP under basal conditions. C: Insulin-mediated suppression of EGP. D: Insulin-mediated stimulation of Rd. Insulin-mediated 2-deoxyglucose (2-DG) uptake by gastrocnemius (gastroc) (E) and soleus (F) muscles. Plasma FA concentrations (G) and percent suppression during the clamp (H). Data are mean ± SEM (n = 6–10 per group [AD]; n = 3–4 per group [EH]). *P < 0.05, **P < 0.01, ***P < 0.001 (AF) by ANOVA with Bonferroni post hoc analysis. *P < 0.05: A-DM vs. WT and N-DM; #P < 0.05: A-DM vs. WT (GH).

Figure 3

Insulin sensitivity in WT, N-DM, and A-DM mice. A: Blood glucose levels before and during the hyperinsulinemic-euglycemic clamp. B: EGP under basal conditions. C: Insulin-mediated suppression of EGP. D: Insulin-mediated stimulation of Rd. Insulin-mediated 2-deoxyglucose (2-DG) uptake by gastrocnemius (gastroc) (E) and soleus (F) muscles. Plasma FA concentrations (G) and percent suppression during the clamp (H). Data are mean ± SEM (n = 6–10 per group [AD]; n = 3–4 per group [EH]). *P < 0.05, **P < 0.01, ***P < 0.001 (AF) by ANOVA with Bonferroni post hoc analysis. *P < 0.05: A-DM vs. WT and N-DM; #P < 0.05: A-DM vs. WT (GH).

Close modal

Diabetic and Nondiabetic NOD Mice Show Lower Fasting Muscle Mitochondrial Oxidation but Increased Lipid Peroxidation in the Fed State

In the fed state, CI respiration decreased by 38% in C-DM (Fig. 4A). During fasting, CI + CII and maximal electron transport system (ETS) capacities decreased by ∼20% in A-DM and N-DM (Fig. 4B). Results were normalized to mtDNA copy number, which did not differ between groups (not shown). Differences in mitochondrial respiration did not associate with changes in gene expression of peroxisome proliferator–activated receptor γ coactivator 1-α (PGC-1α), mitochondrial transcription factor A (TFAM), or nuclear respiratory factor-1 (NRF-1) (Fig. 4C and D).

Figure 4

Mitochondrial function and transcript levels of genes related to mitochondrial biogenesis in gastrocnemius muscle (AD) and liver (EH) of WT, N-DM, and A-DM mice. Mitochondrial respiration capacity through CI, CI + CII, and maximal respiratory capacities of ETS in gastrocnemius muscle (A and B) and liver (E and F) of fed and fasted mice (n = 5–14 per group). Respiratory data were individually corrected for mitochondrial content. Expression levels of PGC-1α, NRF-1, and TFAM in gastrocnemius muscle (C and D) and liver (G and H) of fed and fasted mice (n = 5 per group). Data are mean ± SEM. *P < 0.05, **P < 0.01, ***P < 0.001 by ANOVA with Bonferroni post hoc analysis. AU, arbitrary unit.

Figure 4

Mitochondrial function and transcript levels of genes related to mitochondrial biogenesis in gastrocnemius muscle (AD) and liver (EH) of WT, N-DM, and A-DM mice. Mitochondrial respiration capacity through CI, CI + CII, and maximal respiratory capacities of ETS in gastrocnemius muscle (A and B) and liver (E and F) of fed and fasted mice (n = 5–14 per group). Respiratory data were individually corrected for mitochondrial content. Expression levels of PGC-1α, NRF-1, and TFAM in gastrocnemius muscle (C and D) and liver (G and H) of fed and fasted mice (n = 5 per group). Data are mean ± SEM. *P < 0.05, **P < 0.01, ***P < 0.001 by ANOVA with Bonferroni post hoc analysis. AU, arbitrary unit.

Close modal

Measurement of TBARS in the muscle revealed that WT mice, unlike N-DM or A-DM, have lower lipid peroxide levels in the fed than in the fasting state (P < 0.01) (Supplementary Fig. 1E and F). Moreover, fed N-DM, A-DM, and C-DM mice had higher TBARS than WT mice (Supplementary Fig. 1E).

Diabetic NOD Mice Show Increased PKCθ-Mediated Serine Phosphorylation of IRS1 in Gastrocnemius Muscle

A-DM displayed reduced basal levels of insulin receptor (IR) (−32%) and IRS1 (−30%) in muscle compared with WT (Fig. 5A and B). Serine pIRS1-Ser1101 was increased by 149% and pIRS1-Ser307 by 246% upon intraperitoneal insulin injection (Fig. 5C and D). A-DM showed a profoundly decreased insulin-stimulated membrane-to-cytosol ratio of Akt (−61%) (Fig. 5E) and GLUT4 (−76%) (Fig. 5F). N-DM had reduced IR and intermediate decreases in Akt and GLUT4 membrane-to-cytosol ratio (Fig. 5A, E, and F).

Figure 5

Protein levels of cellular signaling pathway components in gastrocnemius muscle (AI) and liver (JR) of WT, N-DM, and A-DM mice. AF and JN: Basal and insulin-stimulated expression levels (n = 6 per group) of IR (A and J), IRS1 (B), IRS2 (K), pIRS1-Ser1101 (C and L), pIRS1-Ser307 (D and M), membrane-to-cytosol ratio of Akt (E and N), and GLUT4 (F). GI and OR: Fasted conditions: membrane-to-cytosol ratio of PKCθ (G) and PKCε (O) (n = 7–8 per group), cytosolic PKCε (P), pJNK (H and Q) (n = 6 per group), and pNFκB (I and R) (n = 6 per group). GAPDH was used as the loading control for all proteins. Data are mean ± SEM. *P < 0.05, **P < 0.01, ***P < 0.001 by ANOVA with Bonferroni post hoc analysis. AU, arbitrary unit.

Figure 5

Protein levels of cellular signaling pathway components in gastrocnemius muscle (AI) and liver (JR) of WT, N-DM, and A-DM mice. AF and JN: Basal and insulin-stimulated expression levels (n = 6 per group) of IR (A and J), IRS1 (B), IRS2 (K), pIRS1-Ser1101 (C and L), pIRS1-Ser307 (D and M), membrane-to-cytosol ratio of Akt (E and N), and GLUT4 (F). GI and OR: Fasted conditions: membrane-to-cytosol ratio of PKCθ (G) and PKCε (O) (n = 7–8 per group), cytosolic PKCε (P), pJNK (H and Q) (n = 6 per group), and pNFκB (I and R) (n = 6 per group). GAPDH was used as the loading control for all proteins. Data are mean ± SEM. *P < 0.05, **P < 0.01, ***P < 0.001 by ANOVA with Bonferroni post hoc analysis. AU, arbitrary unit.

Close modal

Levels of pERK-Thr202/Tyr204, p-p38-Thr180/Tyr182, phosphorylated inhibitor of κB (IκB) at Ser32 (pIκB-Ser32) (Supplementary Fig. 3), and pNFκB-Ser536 (Fig. 5I) were unchanged. pJNK-Thr183/Tyr186 was increased in A-DM and N-DM (Fig. 5H). Only the higher membrane-to-cytosol ratio of PKCθ was clearly associated with decreases in peripheral insulin sensitivity in A-DM (+148% vs. WT, +53% vs. N-DM) (Fig. 5G).

A-DM had higher membrane-to-cytosol DAGs (Fig. 6A) as a result of changes in several specific DAG species (Fig. 6B). Neither total nor individual species of ceramides were different between the groups (Fig. 6C and D).

Figure 6

Myocellular content of DAGs and ceramides in WT, N-DM, and A-DM mice. Membrane-to-cytosol total (A) or individual species (B) of DAG and whole-tissue content of total (C) or individual species (D) of ceramides in gastrocnemius muscle (n = 8 per group). Data are mean ± SEM. *P < 0.05, **P < 0.01, ***P < 0.001 by ANOVA with Bonferroni post hoc analysis.

Figure 6

Myocellular content of DAGs and ceramides in WT, N-DM, and A-DM mice. Membrane-to-cytosol total (A) or individual species (B) of DAG and whole-tissue content of total (C) or individual species (D) of ceramides in gastrocnemius muscle (n = 8 per group). Data are mean ± SEM. *P < 0.05, **P < 0.01, ***P < 0.001 by ANOVA with Bonferroni post hoc analysis.

Close modal

NOD Mice Have Increased Hepatic Lipid Peroxidation, but Only Diabetic NOD Mice Show Augmented Hepatic Mitochondrial Oxidation

In the fed state, hepatic O2 flux rates through CI, CI + CII, and maximal ETS capacity were increased in A-DM compared with N-DM and WT (Fig. 4E). In C-DM, CI and CI + CII respiration was increased versus WT but lower versus A-DM (Fig. 4E). No differences were observed in the fasted state (Fig. 4F). Hepatic mtDNA copy number was similar among all groups (data not shown). In contrast to muscle, increased respiration in the liver of A-DM was accompanied by higher transcription of PGC-1α TFAM and NRF-1 than in N-DM and WT (Fig. 4G).

Hepatic TBARS were unchanged in the fasted state but were higher in fed N-DM and A-DM (Supplementary Fig. 1E and F) and even more pronounced in C-DM. Similar to muscle, hepatic TBARS decreased during feeding in WT only (P < 0.001).

JNK Signaling Is Increased in the Liver of NOD Mice

Protein abundance of hepatic IR, IRS2 (Fig. 5J and K), and IRS1 (data not shown) were similar in all groups. However, A-DM had 125% and 74% higher levels of pIRS1-Ser1101 and pIRS1-Ser307, respectively, than WT at baseline (Fig. 5L and M). N-DM mice also showed 137% higher pIRS1-Ser1101 (Fig. 5L). Insulin-stimulated translocation of Akt to the membrane was suppressed by 76% in A-DM and to a lesser extent (by 40%) in N-DM (Fig. 5N).

Levels of pERK-Thr202/Tyr204, p-p38-Thr180/Tyr182, IκB, pIκB-Ser32 (Supplementary Fig. 4), and pNFκB-Ser536 (Fig. 5R) were unchanged. Of note, the membrane-to-cytosol ratio of PKCε was 36% and 27% lower in A-DM and N-DM, respectively (Fig. 5O). This was exclusively attributed to an increase in PKCε in the cytosolic fraction (Fig. 5P). Furthermore, pJNK-Thr183/Tyr186 was increased by 64% in A-DM and by 67% in N-DM (Fig. 5Q), rendering pJNK the only factor associated with changes in hepatic insulin sensitivity.

Circulating Fetuin A Levels Are Increased in NOD Mice

In A-DM and N-DM, fasting serum concentrations of fetuin A were 2.0- and 2.9-fold higher than in WT (Fig. 1D).

This study describes the early metabolic events occurring before and at the onset of insulin-dependent diabetes in NOD mice. The NOD model spontaneously develops autoimmune insulitis with insulin deficiency, resembling human T1DM. Because chemical (29,30) or surgical (31) induction of insulin deficiency may per se induce insulin resistance, NOD mice currently represent the most suitable model for studying metabolic changes associated with autoimmune diabetes. The study shows that the initiation of diabetes is associated with tissue-specific differences in metabolic flexibility and insulin sensitivity. Normoglycemic, nondiabetic NOD mice already exhibit hepatic insulin resistance associated with increased JNK phosphorylation and lipid peroxidation as well as lower muscle glucose transport. Acutely diabetic NOD mice also display muscle insulin resistance associated with increased intramyocellular DAG and PKCθ activation. Moreover, the liver transiently enhances its mitochondrial oxidative capacity at diabetes onset, possibly as an adaptation to increased lipolysis, whereas muscle oxidative capacity declines during later stages of diabetes.

A-DM mice show a reduction of visceral and subcutaneous fat, which reflects excessive fasting lipolysis and/or impaired stimulation of lipid synthesis by insulin in adipocytes. Blunted inhibition of lipolysis has been linked to the development of peripheral insulin resistance in adolescents with poorly controlled T1DM (32). In the current study, the intact reduction of FA during hyperinsulinemic clamps in NOD mice, unlike in humans, indicates that the absence of insulin-dependent control of lipid metabolism results from reduced insulin secretion rather than from insulin resistance of adipose tissue. The resulting rise in circulating FA and TG levels would favor redistribution of lipids toward ectopic fat storage in liver or muscle, which is associated with insulin resistance in these tissues (7,13). Of note, A-DM mice had unchanged muscle and even reduced liver TG content, indicating permanently ongoing lipid oxidation in the fed state. This was supported by a constant reduction and lack of the diurnal oscillation of RQ, reflecting restriction to fat oxidation, impaired switching to glucose utilization, and a trend toward ketonemia within 3 days after onset of hyperglycemia in A-DM. Likewise, T1DM patients with poor metabolic control have lower intrahepatic fat content along with moderate peripheral insulin resistance and increased whole-body lipid oxidation (2). On the other hand, N-DM mice had increased muscle fat content, likely reflecting higher lipid storage leading to unchanged circulating FA and TG levels. Both A-DM and N-DM mice also accumulated lipid peroxidation products in muscle and liver, indicating greater reactive oxygen species (ROS) production, which can further aggravate insulin resistance (33). Taken together, NOD mice are characterized by hyperlipidemia and oxidative stress in muscle and liver.

A-DM mice display markedly reduced muscle insulin sensitivity as assessed both in vivo and ex vivo. Impaired insulin-mediated glucose disposal and muscle glucose uptake, along with decreased membrane translocation of GLUT4 and Akt, could result from lipid-mediated intracellular alterations. In human T1DM, muscle insulin resistance is accompanied by decreased glucose transport into myocytes (34,35) and insulin-stimulated upregulation of GLUT4 mRNA (36) but increased muscle lipid content (37). FA-induced inhibition of insulin-stimulated glucose transport/phosphorylation (38) has been linked to intracellular accumulation of DAG, PKCθ activation, and serine phosphorylation of IRS1 (6). Supporting this contention, A-DM mice showed increased membrane-to-cytosol ratio of DAG and PKCθ, and Ser1101/Ser307 phosphorylation of IRS1 in skeletal muscle (Fig. 7). Moreover, the hyperglycemia-induced basal glucose uptake in A-DM mice could also contribute to PKC isoforms activation (40). On the other hand, plasma levels of fetuin A, an FA-induced hepatokine, were increased in A-DM and N-DM mice. Fetuin A positively correlates with hyperglycemia and insulin resistance during hyperlipidemia (41) and promotes lipid-induced insulin resistance by mediating the binding of FA to TLR4 (39). We found no changes in signaling downstream of TLR4, such as NFκB or ceramides, questioning the contribution of this pathway to muscle insulin resistance. Recent data also indicate that both high-saturated and high-unsaturated fat diets induce insulin resistance independently of TLR4 signaling (42).

Figure 7

Mechanisms of tissue-specific changes in glucose homeostasis occurring in diabetic and nondiabetic NOD mice. Effects occurring at the onset of T1DM (A-DM mice) are shown with red arrows and marks. Effects of the NOD genotype occurring independently of diabetes (both N-DM and A-DM mice) are shown with blue arrows and marks. The progression of T1DM is associated with increased circulating glucose and free FA levels resulting from impaired control of glucose and lipid metabolism by insulin. Intramyocellular lipotoxic intermediates DAGs activate PKCθ, which results in enhanced Ser1101 and Ser307 phosphorylation of IRS1 (pIRS1-Ser1101 and pIRS-Ser307). This inhibits IRS1 activity and the membrane translocation of Akt and GLUT4, thereby decreasing muscle glucose disposal. In the liver, increased production of ROS and lipid peroxidation is associated with increased pJNK and IRS1 (pIRS1-Ser307 and pIRS1-Ser1101). This leads to lower translocation of Akt and impaired insulin-mediated suppression of EGP. Furthermore, fetuin A (FetA) levels in serum are increased, which could contribute to the downregulation of the insulin signaling pathway by binding to insulin receptor β-subunit and shutting down the phosphorylation at tyrosine (pTyr) (50) or by increasing pJNK through TLR4 (39). G6P, glucose-6-phosphate; GS3K, glycogen synthase kinase 3; PI3K, phosphatidylinositol 3-kinase.

Figure 7

Mechanisms of tissue-specific changes in glucose homeostasis occurring in diabetic and nondiabetic NOD mice. Effects occurring at the onset of T1DM (A-DM mice) are shown with red arrows and marks. Effects of the NOD genotype occurring independently of diabetes (both N-DM and A-DM mice) are shown with blue arrows and marks. The progression of T1DM is associated with increased circulating glucose and free FA levels resulting from impaired control of glucose and lipid metabolism by insulin. Intramyocellular lipotoxic intermediates DAGs activate PKCθ, which results in enhanced Ser1101 and Ser307 phosphorylation of IRS1 (pIRS1-Ser1101 and pIRS-Ser307). This inhibits IRS1 activity and the membrane translocation of Akt and GLUT4, thereby decreasing muscle glucose disposal. In the liver, increased production of ROS and lipid peroxidation is associated with increased pJNK and IRS1 (pIRS1-Ser307 and pIRS1-Ser1101). This leads to lower translocation of Akt and impaired insulin-mediated suppression of EGP. Furthermore, fetuin A (FetA) levels in serum are increased, which could contribute to the downregulation of the insulin signaling pathway by binding to insulin receptor β-subunit and shutting down the phosphorylation at tyrosine (pTyr) (50) or by increasing pJNK through TLR4 (39). G6P, glucose-6-phosphate; GS3K, glycogen synthase kinase 3; PI3K, phosphatidylinositol 3-kinase.

Close modal

In vivo hepatic insulin sensitivity is impaired in both N-DM and A-DM mice, implying that hepatic insulin resistance can develop before the onset of hyperglycemia. In the absence of hyperglycemia and hyperlipidemia, the mechanism of hepatic insulin resistance in N-DM mice likely differ from that observed in muscle of A-DM. Indeed, inhibition of IRS1 in the liver of N-DM and A-DM was not associated with changes in PKCε but, rather, with an increase in pJNK. Although previous studies yielded conflicting data on the protective effects of liver-specific JNK deficiency against lipid-induced insulin resistance, it should be emphasized that the enhanced phosphorylation of JNK in the NOD model occurs independently of lipids. Alternatively, increased fetuin A (41) and hepatic oxidative stress (43), which have been linked to JNK activation, could account for increased pJNK, inhibition of IRS1, and hepatic insulin resistance.

We found no association of mitochondrial respiration with insulin sensitivity, excluding mitochondrial function as a potential mediator of insulin resistance in NOD mice. In muscle, mitochondrial oxidative capacity and transcript levels of mitochondrial biogenesis–related genes were comparable between A-DM and N-DM. However, C-DM showed lower oxidative capacity, in line with the observation that muscle ATP synthesis is decreased in vivo in human T1DM (1). This secondary decrease of mitochondrial function could result from mitochondrial damage caused by lipid peroxide accumulation (44), inhibitory effects of prolonged insulin treatment on mitochondrial biogenesis and function (45), or direct effects of the chronic diabetic state (1). In contrast, hepatic mitochondrial oxidative capacity as well as biogenesis were clearly higher in A-DM mice. Increased oxidative capacity may represent an early adaptation to increased lipid and glucose flux after diabetes onset. Expression of genes related to oxidative phosphorylation is upregulated in the liver of NOD mice 2 weeks after diabetes onset (46) as well as in the liver of obese humans with T2DM (47). Decreased Akt could mediate the increase in hepatic PGC-1α, FA oxidation (48), and overall mitochondrial respiration (49). Of note, upregulated hepatic mitochondrial oxidative capacity seems to be transient because it declines in C-DM. Similar to muscle, this could be a consequence of mitochondrial damage due to lipid peroxide accumulation.

In conclusion, insulin resistance in a murine nonobese model of T1DM develops in the liver before the onset of diabetes and is associated with increased oxidative stress and pJNK as possible cellular mediators. Muscle insulin sensitivity is impaired already 3 days after the onset of diabetes and accompanied by increased levels of DAG, PKCθ, and serum fetuin A. Furthermore, mitochondrial oxidative capacity is transiently enhanced in the liver and declines in muscle with longer disease duration. Knowledge of these mechanisms could help to develop new strategies to prevent and treat insulin resistance and subsequent complications in patients with T1DM.

Acknowledgments. The authors thank Mario Kahn of the Department of Internal Medicine, Yale University School of Medicine; Kay Jeruschke, Daniella Herzfeld de Wiza, Heidi Müller, and Conny Köllmer of the Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Düsseldorf; and Volker Burkart, Alexander Strom, Fariba Zivehe, Ilka Rokitta, and Olesja Ritter of the Institute for Clinical Diabetology, German Diabetes Center, Düsseldorf, for their expertise.

Funding. This study was supported in part by Schmutzler Stiftung, Skröder Stiftung, the German Research Foundation (SFB575, project 13, to M.R.), the German Federal Ministry of Education and Research to the German Center for Diabetes Research, the Ministry of Science and Research of the State of North Rhine-Westphalia, the German Federal Ministry of Health, and grants from the U.S. Public Health Service (R01-DK-40936, P30-DK-45735).

Duality of Interest. This study was supported in part by grants from the European Foundation for the Study of Diabetes (Novo Nordisk and GlaxoSmithKline grants). No other potential conflicts of interest relevant to this article were reported.

Author Contributions. T.J. and G.S. conceived of the experiments, researched data, contributed to the discussion, and wrote the manuscript. K.K., D.M.O., E.P., J.K., and B.K. researched data and edited and reviewed the manuscript. J.W., A.L.R., L.J., P.N., H.-J.P., and D.Z. researched data. G.I.S. contributed to the discussion and edited and reviewed the manuscript. J.S. conceived of the experiments, contributed to the discussion, and edited and reviewed the manuscript. M.R. conceived of the experiments, contributed to the discussion, and wrote the manuscript. M.R. is the guarantor of this work and, as such, had full access to all the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis.

Prior Presentation. Parts of this study were presented in abstract form at the 72nd Scientific Sessions of the American Diabetes Association, Philadelphia, PA, 8–12 June, 2012; and at the 48th Annual Meeting of the European Association for the Study of Diabetes, Berlin, Germany, 1–5 October 2012.

1.
Kacerovsky
M
,
Brehm
A
,
Chmelik
M
, et al
.
Impaired insulin stimulation of muscular ATP production in patients with type 1 diabetes
.
J Intern Med
2011
;
269
:
189
199
[PubMed]
2.
Perseghin
G
,
Lattuada
G
,
De Cobelli
F
, et al
.
Reduced intrahepatic fat content is associated with increased whole-body lipid oxidation in patients with type 1 diabetes
.
Diabetologia
2005
;
48
:
2615
2621
[PubMed]
3.
Gale
EA
.
The rise of childhood type 1 diabetes in the 20th century
.
Diabetes
2002
;
51
:
3353
3361
[PubMed]
4.
Wilkin
TJ
.
The accelerator hypothesis: a review of the evidence for insulin resistance as the basis for type I as well as type II diabetes
.
Int J Obes (Lond)
2009
;
33
:
716
726
[PubMed]
5.
Rossetti
L
.
Glucose toxicity: the implications of hyperglycemia in the pathophysiology of diabetes mellitus
.
Clin Invest Med
1995
;
18
:
255
260
[PubMed]
6.
Erion
DM
,
Shulman
GI
.
Diacylglycerol-mediated insulin resistance
.
Nat Med
2010
;
16
:
400
402
[PubMed]
7.
Roden
M
.
Muscle triglycerides and mitochondrial function: possible mechanisms for the development of type 2 diabetes
.
Int J Obes (Lond)
2005
;
29
(
Suppl. 2
):
S111
S115
[PubMed]
8.
Summers
SA
.
Ceramides in insulin resistance and lipotoxicity
.
Prog Lipid Res
2006
;
45
:
42
72
[PubMed]
9.
Kelley
DE
,
He
J
,
Menshikova
EV
,
Ritov
VB
.
Dysfunction of mitochondria in human skeletal muscle in type 2 diabetes
.
Diabetes
2002
;
51
:
2944
2950
[PubMed]
10.
Stump
CS
,
Short
KR
,
Bigelow
ML
,
Schimke
JM
,
Nair
KS
.
Effect of insulin on human skeletal muscle mitochondrial ATP production, protein synthesis, and mRNA transcripts
.
Proc Natl Acad Sci U S A
2003
;
100
:
7996
8001
[PubMed]
11.
Karakelides
H
,
Asmann
YW
,
Bigelow
ML
, et al
.
Effect of insulin deprivation on muscle mitochondrial ATP production and gene transcript levels in type 1 diabetic subjects
.
Diabetes
2007
;
56
:
2683
2689
[PubMed]
12.
Schmid
AI
,
Szendroedi
J
,
Chmelik
M
,
Krssák
M
,
Moser
E
,
Roden
M
.
Liver ATP synthesis is lower and relates to insulin sensitivity in patients with type 2 diabetes
.
Diabetes Care
2011
;
34
:
448
453
[PubMed]
13.
Szendroedi
J
,
Chmelik
M
,
Schmid
AI
, et al
.
Abnormal hepatic energy homeostasis in type 2 diabetes
.
Hepatology
2009
;
50
:
1079
1086
[PubMed]
14.
Pospisilik
JA
,
Knauf
C
,
Joza
N
, et al
.
Targeted deletion of AIF decreases mitochondrial oxidative phosphorylation and protects from obesity and diabetes
.
Cell
2007
;
131
:
476
491
[PubMed]
15.
Makino
S
,
Kunimoto
K
,
Muraoka
Y
,
Mizushima
Y
,
Katagiri
K
,
Tochino
Y
.
Breeding of a non-obese, diabetic strain of mice
.
Jikken Dobutsu
1980
;
29
:
1
13
[PubMed]
16.
Ayala
JE
,
Bracy
DP
,
Malabanan
C
, et al
.
Hyperinsulinemic-euglycemic clamps in conscious, unrestrained mice [article online]
.
J Vis Exp
2011
;(
57
):3188. Available from http://www.jove.com/video/3188/hyperinsulinemic-euglycemic-clamps-in-conscious-unrestrained-mice?id=3188. Accessed 12 April 2014
[PubMed]
17.
Phielix
E
,
Jelenik
T
,
Nowotny
P
,
Szendroedi
J
,
Roden
M
.
Reduction of non-esterified fatty acids improves insulin sensitivity and lowers oxidative stress, but fails to restore oxidative capacity in type 2 diabetes: a randomised clinical trial
.
Diabetologia
2014
;
57
:
572
581
[PubMed]
18.
Ohkawa
H
,
Ohishi
N
,
Yagi
K
.
Assay for lipid peroxides in animal tissues by thiobarbituric acid reaction
.
Anal Biochem
1979
;
95
:
351
358
[PubMed]
19.
Kotzka
J
,
Knebel
B
,
Avci
H
, et al
.
Phosphorylation of sterol regulatory element-binding protein (SREBP)-1a links growth hormone action to lipid metabolism in hepatocytes
.
Atherosclerosis
2010
;
213
:
156
165
[PubMed]
20.
Fu
M
,
Sun
T
,
Bookout
AL
, et al
.
A nuclear receptor atlas: 3T3-L1 adipogenesis
.
Mol Endocrinol
2005
;
19
:
2437
2450
[PubMed]
21.
Mukherjee
A
,
Sidis
Y
,
Mahan
A
, et al
.
FSTL3 deletion reveals roles for TGF-beta family ligands in glucose and fat homeostasis in adults
.
Proc Natl Acad Sci U S A
2007
;
104
:
1348
1353
[PubMed]
22.
Jelenik
T
,
Rossmeisl
M
,
Kuda
O
, et al
.
AMP-activated protein kinase α2 subunit is required for the preservation of hepatic insulin sensitivity by n-3 polyunsaturated fatty acids
.
Diabetes
2010
;
59
:
2737
2746
[PubMed]
23.
Neschen
S
,
Morino
K
,
Hammond
LE
, et al
.
Prevention of hepatic steatosis and hepatic insulin resistance in mitochondrial acyl-CoA:glycerol-sn-3-phosphate acyltransferase 1 knockout mice
.
Cell Metab
2005
;
2
:
55
65
[PubMed]
24.
Inoue
Y
,
Kaifu
T
,
Sugahara-Tobinai
A
,
Nakamura
A
,
Miyazaki
J
,
Takai
T
.
Activating Fc gamma receptors participate in the development of autoimmune diabetes in NOD mice
.
J Immunol
2007
;
179
:
764
774
[PubMed]
25.
Steele
R
.
Influences of glucose loading and of injected insulin on hepatic glucose output
.
Ann N Y Acad Sci
1959
;
82
:
420
430
[PubMed]
26.
Lee
S
,
Muniyappa
R
,
Yan
X
, et al
.
Comparison between surrogate indexes of insulin sensitivity and resistance and hyperinsulinemic euglycemic clamp estimates in mice
.
Am J Physiol Endocrinol Metab
2008
;
294
:
E261
E270
[PubMed]
27.
Uwaifo
GI
,
Fallon
EM
,
Chin
J
,
Elberg
J
,
Parikh
SJ
,
Yanovski
JA
.
Indices of insulin action, disposal, and secretion derived from fasting samples and clamps in normal glucose-tolerant black and white children
.
Diabetes Care
2002
;
25
:
2081
2087
[PubMed]
28.
Durant
S
,
Alves
V
,
Coulaud
J
,
Homo-Delarche
F
.
Nonobese diabetic (NOD) mouse dendritic cells stimulate insulin secretion by prediabetic islets
.
Autoimmunity
2002
;
35
:
449
455
[PubMed]
29.
Blondel
O
,
Bailbe
D
,
Portha
B
.
In vivo insulin resistance in streptozotocin-diabetic rats—evidence for reversal following oral vanadate treatment
.
Diabetologia
1989
;
32
:
185
190
[PubMed]
30.
Bevilacqua
S
,
Barrett
EJ
,
Smith
D
, et al
.
Hepatic and peripheral insulin resistance following streptozotocin-induced insulin deficiency in the dog
.
Metabolism
1985
;
34
:
817
825
[PubMed]
31.
Noguchi
Y
,
Younes
RN
,
Conlon
KC
,
Vydelingum
NA
,
Matsumoto
A
,
Brennan
MF
.
The effect of prolonged hyperglycemia on metabolic alterations in the subtotally pancreatectomized rat
.
Surg Today
1994
;
24
:
987
993
[PubMed]
32.
Bergman
BC
,
Howard
D
,
Schauer
IE
, et al
.
The importance of palmitoleic acid to adipocyte insulin resistance and whole-body insulin sensitivity in type 1 diabetes
.
J Clin Endocrinol Metab
2013
;
98
:
E40
E50
[PubMed]
33.
Anderson
EJ
,
Lustig
ME
,
Boyle
KE
, et al
.
Mitochondrial H2O2 emission and cellular redox state link excess fat intake to insulin resistance in both rodents and humans
.
J Clin Invest
2009
;
119
:
573
581
[PubMed]
34.
Cline
GW
,
Magnusson
I
,
Rothman
DL
,
Petersen
KF
,
Laurent
D
,
Shulman
GI
.
Mechanism of impaired insulin-stimulated muscle glucose metabolism in subjects with insulin-dependent diabetes mellitus
.
J Clin Invest
1997
;
99
:
2219
2224
[PubMed]
35.
Vuorinen-Markkola
H
,
Koivisto
VA
,
Yki-Jarvinen
H
.
Mechanisms of hyperglycemia-induced insulin resistance in whole body and skeletal muscle of type I diabetic patients
.
Diabetes
1992
;
41
:
571
580
[PubMed]
36.
Yki-Järvinen
H
,
Vuorinen-Markkola
H
,
Koranyi
L
, et al
.
Defect in insulin action on expression of the muscle/adipose tissue glucose transporter gene in skeletal muscle of type 1 diabetic patients
.
J Clin Endocrinol Metab
1992
;
75
:
795
799
[PubMed]
37.
Perseghin
G
,
Lattuada
G
,
Danna
M
, et al
.
Insulin resistance, intramyocellular lipid content, and plasma adiponectin in patients with type 1 diabetes
.
Am J Physiol Endocrinol Metab
2003
;
285
:
E1174
E1181
[PubMed]
38.
Roden
M
,
Price
TB
,
Perseghin
G
, et al
.
Mechanism of free fatty acid-induced insulin resistance in humans
.
J Clin Invest
1996
;
97
:
2859
2865
[PubMed]
39.
Pal
D
,
Dasgupta
S
,
Kundu
R
, et al
.
Fetuin-A acts as an endogenous ligand of TLR4 to promote lipid-induced insulin resistance
.
Nat Med
2012
;
18
:
1279
1285
[PubMed]
40.
Talior
I
,
Yarkoni
M
,
Bashan
N
,
Eldar-Finkelman
H
.
Increased glucose uptake promotes oxidative stress and PKC-delta activation in adipocytes of obese, insulin-resistant mice
.
Am J Physiol Endocrinol Metab
2003
;
285
:
E295
E302
[PubMed]
41.
Stefan
N
,
Häring
HU
.
Circulating fetuin-A and free fatty acids interact to predict insulin resistance in humans
.
Nat Med
2013
;
19
:
394
395
[PubMed]
42.
Galbo
T
,
Perry
RJ
,
Jurczak
MJ
, et al
.
Saturated and unsaturated fat induce hepatic insulin resistance independently of TLR-4 signaling and ceramide synthesis in vivo
.
Proc Natl Acad Sci U S A
2013
;
110
:
12780
12785
[PubMed]
43.
Lim
JH
,
Lee
HJ
,
Ho Jung
M
,
Song
J
.
Coupling mitochondrial dysfunction to endoplasmic reticulum stress response: a molecular mechanism leading to hepatic insulin resistance
.
Cell Signal
2009
;
21
:
169
177
[PubMed]
44.
Hruszkewycz
AM
.
Evidence for mitochondrial DNA damage by lipid peroxidation
.
Biochem Biophys Res Commun
1988
;
153
:
191
197
[PubMed]
45.
Liu
HY
,
Yehuda-Shnaidman
E
,
Hong
T
, et al
.
Prolonged exposure to insulin suppresses mitochondrial production in primary hepatocytes
.
J Biol Chem
2009
;
284
:
14087
14095
[PubMed]
46.
Liu
HY
,
Cao
SY
,
Hong
T
,
Han
J
,
Liu
Z
,
Cao
W
.
Insulin is a stronger inducer of insulin resistance than hyperglycemia in mice with type 1 diabetes mellitus (T1DM)
.
J Biol Chem
2009
;
284
:
27090
27100
[PubMed]
47.
Takamura
T
,
Misu
H
,
Matsuzawa-Nagata
N
, et al
.
Obesity upregulates genes involved in oxidative phosphorylation in livers of diabetic patients
.
Obesity (Silver Spring)
2008
;
16
:
2601
2609
[PubMed]
48.
Li
X
,
Monks
B
,
Ge
Q
,
Birnbaum
MJ
.
Akt/PKB regulates hepatic metabolism by directly inhibiting PGC-1alpha transcription coactivator
.
Nature
2007
;
447
:
1012
1016
[PubMed]
49.
Civitarese
AE
,
Carling
S
,
Heilbronn
LK
, et al
CALERIE Pennington Team
.
Calorie restriction increases muscle mitochondrial biogenesis in healthy humans
.
PLoS Med
2007
;
4
:
e76
[PubMed]
50.
Goustin
AS
,
Derar
N
,
Abou-Samra
AB
.
Ahsg-fetuin blocks the metabolic arm of insulin action through its interaction with the 95-kD β-subunit of the insulin receptor
.
Cell Signal
2013
;
25
:
981
988
[PubMed]

Supplementary data