Uncoupling of mitochondrial respiration by chemical uncouplers has proven effective in ameliorating obesity, insulin resistance, and hyperglycemia. However, development of uncoupler-based therapy remains challenging due to its potentially lethal adverse effects. Here, we identify pyruvate dehydrogenase (PDH) as a key modifier of the toxicity profile of 2, 4-dinitrophenol (DNP), a prototypical mitochondrial uncoupler. PDH activation by dichloroacetic acid (DCA) protects mice from DNP-induced hyperlactacidemia, hyperthermia, and death while preserving the ability of DNP to promote fuel oxidation and improve insulin sensitivity in mice. Mechanistically, PDH activation switches on mitochondrial glucose oxidation to accommodate increased glycolytic flux, leading to reduced lactate secretion during uncoupler treatments. We devised a chemical screening strategy and discovered compound 6j as a dual-action compound that simultaneously activates PDH and uncouples mitochondrial respiration. Compound 6j exhibits an excellent efficacy and safety profile in restoring glucose homeostasis in diabetic mice. This work establishes a new principle to safely harness the power of chemical uncouplers for the treatment of metabolic disease.

Type 2 diabetes (T2D), which is characterized by hyperglycemia and insulin resistance, has become a global epidemic. Although several antidiabetic drugs have been applied in the clinic, few of them are effective in correcting the underlying pathology of insulin resistance. Therefore, the identification of novel drugs with new mechanisms to improve insulin resistance is urgently needed.

Obesity is a highly intricate disease and is one of the strongest risk factors for T2D through obesity-driven insulin resistance in liver, adipose tissue, and skeletal muscle, combined with insufficient insulin secretion to overcome this resistance (1). It has been reported that excessive ectopic lipid accumulation is an important contributor to insulin resistance through the development of lipid-derived lipotoxicity in insulin-sensitive tissue (2). How lipid accumulates in peripheral tissue has not been precisely clarified, but several mechanisms involved in the development of insulin resistance have been well documented, including increased circulating cytokines, inflammatory cell infiltration, and intracellular lipid metabolites (37). Therefore, reducing ectopic lipid accumulation in peripheral tissue might be an attractive strategy for treating T2D. Mitochondrial uncoupling, which promotes the proton gradient across the mitochondrial inner member through non-ATP synthase and creates a futile cycle of glucose and lipid oxidation (810), is an energy-wasting strategy to combat ectopic lipid accumulation.

There is well-known evidence for the effectiveness of the mitochondrial uncoupler 2,4-dinitrophenol (DNP), which was approved for weight loss in the clinic. However, DNP therapy ceased in the 1930s due to its severe adverse effects, such as hyperlactacidemia, hyperthermia, and even death (1115), which limited the further development of uncoupling agents. The efficacy of DNP administered in vivo was largely attributable to the expense of fat as fuel, and fat combustion resulted in weight loss but also heat overproduction by uncoupling actions. Therefore, stimulation of lipid oxidation by uncoupling activity seems to be a double-edged sword (16). In addition, ATP production was dramatically decreased through the mitochondrial electronic chain by uncoupling actions, and then glucose was forced to undergo anaerobic glycolysis for ATP production, leading to lactate overproduction (17,18). The toxicities of DNP were mostly on-target effects related to uncoupling actions (19). Thus, it is crucial to dissociate the therapeutic benefits of uncouplers from their adverse effects.

To relieve the main adverse effects derived from fat combustion and anaerobic glycolysis, we hypothesized that switching anaerobic glycolysis to glucose oxidation would remit fat burning and lactate overproduction and maintain hypoglycemic actions. Dichloroacetic acid (DCA), a pyruvate dehydrogenase (PDH) activator via the inhibition of PDH kinases (PDKs) (2022), was identified as meeting our expectations. We verified that DCA combined with DNP in vivo ameliorated hyperglycemia by preferentially inducing the oxidization of glucose over fatty acids and protected mice from hyperlactacidemia, hyperthermia, and death. Encouraged by the excellent profile of DCA combined with DNP, a novel compound possessing both mitochondrial uncoupling and PDH activation effects was identified and confirmed to improve hyperglycemia without associated systemic toxicities in vivo. Therefore, our work opens the possibility of exploring a new class of mitochondrial uncouplers for treating T2D.

Materials

DNP was obtained from Sinopharm Chemical Reagent. Sodium dichloroacetate and streptozocin (STZ) were obtain from Sigma-Aldrich. 9,10-[3H]-palmitic acid, 1-[14C]-glucose, and 1,2-[3H]-deoxy-d-glucose were obtained from PerkinElmer. The primary antibodies of PDH, protein kinase C (PKC)-θ, PKC-ε, Na+-K+-ATPase, pSer473-Akt, Akt, and GAPDH were from Cell Signaling Technology. The pSer473-PDH antibody was from Novus Biologicals.

Cell Culture

L6 myoblasts were cultured in DMEM supplemented with 10% FBS (Invitrogen). For the differentiation of L6 myotubes, the concentration of FBS was reduced to 2% for 6 days. Mouse primary hepatocytes were isolated using Selgen’s two-step perfusion method (23) and were maintained in low-glucose DMEM supplemented with 10% FBS. The 3T3-L1 cells were cultured in DMEM supplemented with 10% CS. The differentiation of 3T3-L1 cells was conducted as described previously (24).

Measurement of Cellular Oxygen Consumption Rate and Extracellular Acidification Rate

L6 myotubes, hepatocytes, and 3T3-L1 cells were plated in an XF96 plate (Seahorse Biosciences). Cells were equilibrated in DMEM containing 25 mmol/L glucose and 2 mmol/L sodium pyruvate. The baseline oxygen consumption rate (OCR) was recorded, and then oligomycin, test compounds, and carbonyl cyanide 4-(trifluoromethoxy)phenylhydrazone were injected, in turn, during continuous oxygen measurements. For combination OCR and extracellular acidification rate (ECAR) assay with DNP, the baseline OCR and ECAR values were recorded, and then test compounds and DNP were injected, in turn, during continuous oxygen and pH measurements.

Measurement of Adenine Nucleotide Levels

L6 myotubes, hepatocytes, and 3T3-L1 cells in six-well cell culture plates were scraped into 100 μL of 4% (v/v) ice-cold HClO4. The acidic supernatants were neutralized and used for high-performance liquid chromatography determination as described previously (25).

Determination of Lactate Concentration

Cells were cultured in a 96-well plate and treated with compounds in serum-free cell culture medium containing 0.5% BSA for the indicated time. Lactate in the medium was measured by the lactate assay kit.

Determination of Cellular Glucose Uptake

The 2-deoxyglucose uptake was measured as described previously (26). Briefly, L6 myotubes were incubated in serum-free medium containing the respective treatments in 24-well plate for 2.5 h, followed by the addition of 100 μmol/L 2-deoxyglucose and 0.5 μCi per well of 2-deoxy-[3H]-d-glucose in HBS buffer. After 10-min incubation, cells were washed with ice-cold PBS and lysed using 0.1% Triton X-100. Radioactivity was quantitated by a scintillation counter.

Determination of Cellular Glucose Oxidation

L6 myotubes were incubated in serum-free medium containing 1,2-[14C]-glucose with the appropriate treatments in a 24-well plate for 4 h. Each well was then covered with a piece of Whatman paper. Reactions were stopped following the injection of 70% perchloric acid into the medium. In the NaOH filter paper, 14C-labeled CO2 formed from glucose oxidation was then collected. The filter paper traps were counted in a scintillation counter.

Determination of Cellular Palmitate Oxidation

Fatty acid oxidation was measured as reported previously (25), with some modifications. Cells were exposed to DMEM containing 0.25 mmol/L palmitate and 1.5 μCi of 9,10-[3H]-palmitic acid for 4 h. The nonmetabolized palmitate in the medium was absorbed with charcoal slurry (0.1 g/mL charcoal in 20 mmol/L Tris-HCl pH 7.5) for 30 min and then removed by centrifugation. The radioactivity of the medium was measured by a scintillation counter.

Animal Studies

All animal experiments were approved by the Animal Ethics Committee of the Shanghai Institute of Materia Medica. All animals were housed in a temperature-controlled room (22°C ± 2°C) with a 12-h light/dark cycle. The hyperglycemic mouse model was implemented with C57BL/6J mice injected intraperitoneally daily with 60 mg/kg STZ for 6 days. For high-fat diet (HFD) mice, 6-week-old male C57BL/6J mice were fed with an HFD (D12492; Research Diets). The 14-week-old mice were randomly assigned and gavaged with 300 mg/kg DCA and 10 mg/kg DNP individually or together for 4 weeks. For db/db mice, 7-week-old mice were assigned randomly and gavaged with either vehicle or 6j for 7 weeks. Body weight and food intake were recorded daily. Blood glucose levels were measured following 6-h starvation. Glucose tolerance test and insulin tolerance test were performed in 6-h or 4-h starvation mice. Finally, HFD mice and db/db mice were killed after injection with insulin for 15 min, and tissues were freeze clamped for further experiments.

Measurement of Metabolic Cage and Core Temperature

The energy expenditure (EE), OCR, respiratory exchange rate (RER), and activity of the mice were determined by the Oxymax system (TSE PhenoMaster). Rectal core temperature was detected by BAT-12 microprobe digital thermometer and RET-3 mouse rectal probe (Physitemp Instruments, Clifton, NJ).

Metabolic Profile Measurements in Tissues

For the quantification of intrahepatic and intramuscular triglyceride (TG), tissue was homogenized with 0.5 mL PBS, and then total TG was extracted with 1.5 mL chloroform and methanol (2:1 v/v) overnight. The samples were centrifuged, and the bottom liquid phase was transferred to a clean tube and air dried. TG was then dissolved in 1 mL of ethanol containing 1% Triton X-100. TG contents were determined using the TG determination kit and normalized to the tissue weight. Tissue diacylglycerol (DAG) was detected by a DAG assay kit (EIAab, Wuhan, China).

Pharmacokinetic Studies

Pharmacokinetic analyses of oral DCA and DNP were performed in C57BL/6J mice. Briefly, 1 g/kg DCA and 30 mg/kg DNP individually or together were dissolved in 0.5% methyl cellulose and given to male C57BL/6J mice, and blood samples and tissue were collected at the indicated time. Tissue distribution of oral 6j was determined in db/db mice. Briefly, 40 mg/kg 6j was dissolved in 0.5% methyl cellulose and given to db/db mice. Tissue was collected 2 h after administration. All samples were subjected to liquid chromatography–tandem mass spectrometry analyses.

Mouse Liver Histological Analysis

For liver histological studies, mice were euthanized by decapitation, and liver tissues were fixed with neutral buffered formalin 10% and embedded in paraffin. Tissue sections were prepared and stained with hematoxylin-eosin.

RNA Interference

L6 myotubes and hepatocytes were seeded in a 24-well plate and transfected with synthesized siRNA by Lipofectamine RNAiMAX Reagent (Thermo Fisher) for 48 h for follow-up experiments. The sequences of oligonucleotide used are described in Supplementary Table 1.

RNA Isolation and Real-time PCR

Total RNA was extracted from mouse muscle, liver, or inguinal white adipose tissue (iWAT) lysates using RNAiso Plus reagent (Takara, Kusatsu, Japan) and standard protocol. Total RNA from each sample was then used to generate high-fidelity cDNA by PrimeScript RT reagent kit (Takara) for quantitative PCR analyses. The relative expression levels of each gene were normalized against the expression of 18s. The primers used are described in Supplementary Table 2.

Immunoblotting

Cell and tissue lysates were subjected to electrophoresis through SDS-PAGE and blotted with antibodies. For the translocation of PKC-θ and PKC-ε in muscle and liver, protein was isolated from the membrane and cytosol by a membrane and cytosol protein extraction kit (Yeasen, Shanghai, China). Then protein was electrophoresed through SDS-PAGE and blotted with antibodies. The immunoblots were visualized by chemiluminescence using the enhanced chemiluminescence Western Blotting System. Quantification was determined by measuring band intensities using ImageJ software.

Statistical Analysis

Results represented means ± SEM. Differences between two groups were examined using the unpaired two-tailed Student’s t test. Differences between four groups were assessed by one-way ANOVA. P < 0.05 was regarded as significant.

Data and Resource Availability

All data needed to evaluate the conclusions in the study are present in the article or Supplementary Data. Additional data related to this article may be requested from the authors.

DNP Induces Lipid Oxidation and Anaerobic Glycolysis In Vitro

The adverse effects of DNP are mostly on-target effects related to uncoupling actions (19); thus, we examined the uncoupling effects on energy metabolism in vitro. In myotubes, hepatocytes, and 3T3-L1 cells, DNP stimulated the OCR in the presence of oligomycin and increased the ADP/ATP ratio, meeting the criteria for uncoupling agents (Supplementary Fig. 1A and B). As expected, lactate production and fatty acid oxidation were robustly stimulated by DNP in cells (Supplementary Fig. 1C and D). However, the increase in glucose oxidation induced by DNP in myotubes was weaker than its effect on fatty acid oxidation (Supplementary Fig. 1E). Increased glucose uptake by DNP was also observed in myotubes, but it was induced at lower concentrations of DNP than those required for glucose oxidation, suggesting that part of the glucose uptake was contributed by lactate production (Supplementary Fig. 1F).

Identification of PDH Activator DCA From the Combination Assays Based on the OCR and ECAR Upon DNP Treatment

On the basis of the striking adverse effects of DNP derived from fat combustion and anaerobic glycolysis, we assumed that inhibition of lipid oxidation or lactate production would achieve our toxicity-reducing goal. We tried to combine DNP with etomoxir (carnitine palmitoyl-transferase I inhibitor), and we found that the combination treatment inhibited the increased OCR induced by DNP without affecting the ECAR in myotubes (Supplementary Fig. 2A and B). Otherwise, DNP combined with galloflavin (27) (lactate dehydrogenase inhibitor) decreased the ECAR and increased the OCR without affecting DNP-induced lipid oxidation upon etomoxir challenge (Supplementary Fig. 2C and D). Thus, inhibition of lipid oxidation or lactate production derived from DNP might not achieve the toxicity-reducing goal.

Next, we hypothesized that switching anaerobic glycolysis to glucose oxidation might remit fat burning and lactate overproduction. We designed a combination assay with DNP based on the OCR and ECAR to test diverse compounds and determine the different characteristics of metabolic profiles. On the basis of the ratio of combination treatment to DNP treatment alone in myotubes, DCA and amiloride were shown to meet our expectations, each decreasing the ECAR without affecting the OCR upon DNP uncoupling (Fig. 1A and B and Supplementary Table 3). However, amiloride treatment was reported to cause hyperkalemia in the clinic (28,29), which is similar to DNP. Thus, DCA, a classical PDH activator, acts as a switch from anaerobic glycolysis and lipid oxidation to glucose oxidation for cellular energy homeostasis (20,22,30) and was considered further as a candidate. In myotubes, hepatocytes, and 3T3-L1 cells, DCA dose-dependently inhibited the ECAR without affecting the OCR (Fig. 1C). Moreover, these phenomena also occurred in cells treated with different uncouplers and PDH activators (Supplementary Fig. 2E).

Figure 1

DNP combined with DCA preferentially stimulates glucose oxidation over fatty acid oxidation. A: The species of the compounds selected for the assay. B: Dot distribution of ECAR and OCR for DNP combined with compounds relative to DNP alone. C: Maximal respiration and glycolysis in myotubes, hepatocytes, and 3T3-L1 cells incubated with DCA and DNP or DNP alone. D: Immunoblot analyses of phosphorylated Ser293-PDH in cells incubated with DNP, DCA, or both for 3 h. E: Detection of lactate content released from cells incubated with DNP, DCA, or both for 12 h. F: Fatty acid oxidation was detected with 3H-labeled palmitate in cells treated with DNP for 3 h and then treated with DCA combined with DNP for another 1 h. G: Glucose oxidation was detected with 14C-labeled glucose in myotubes incubated with DNP, DCA, or both for 4 h. n = 3–4 for all groups. *P < 0.05, **P < 0.01, and ***P < 0.001 compared with the DMSO group. #P < 0.05, ##P < 0.01, and ###P < 0.001 compared with the combination treatment group. $P < 0.05, $$P < 0.01, and $$$P < 0.001 compared with the DNP group. H: C57BL/6J mice were orally treated with 10 mg/kg DNP, 1 g/kg DCA, or both at 8:00 in the morning (time 0 in the curve graph). EE, OCR, RER, and physical activity were calculated at 5 h after administration. n = 6–8 for all groups. *P < 0.05 and **P < 0.001 compared with vehicle group. #P < 0.05 and ##P < 0.01 compared with the combination treatment group. Statistical significance (P) was calculated by one-way ANOVA. Error bars represent SEM. MPTP, mitochondrial permeability transition pore; veh, vehicle.

Figure 1

DNP combined with DCA preferentially stimulates glucose oxidation over fatty acid oxidation. A: The species of the compounds selected for the assay. B: Dot distribution of ECAR and OCR for DNP combined with compounds relative to DNP alone. C: Maximal respiration and glycolysis in myotubes, hepatocytes, and 3T3-L1 cells incubated with DCA and DNP or DNP alone. D: Immunoblot analyses of phosphorylated Ser293-PDH in cells incubated with DNP, DCA, or both for 3 h. E: Detection of lactate content released from cells incubated with DNP, DCA, or both for 12 h. F: Fatty acid oxidation was detected with 3H-labeled palmitate in cells treated with DNP for 3 h and then treated with DCA combined with DNP for another 1 h. G: Glucose oxidation was detected with 14C-labeled glucose in myotubes incubated with DNP, DCA, or both for 4 h. n = 3–4 for all groups. *P < 0.05, **P < 0.01, and ***P < 0.001 compared with the DMSO group. #P < 0.05, ##P < 0.01, and ###P < 0.001 compared with the combination treatment group. $P < 0.05, $$P < 0.01, and $$$P < 0.001 compared with the DNP group. H: C57BL/6J mice were orally treated with 10 mg/kg DNP, 1 g/kg DCA, or both at 8:00 in the morning (time 0 in the curve graph). EE, OCR, RER, and physical activity were calculated at 5 h after administration. n = 6–8 for all groups. *P < 0.05 and **P < 0.001 compared with vehicle group. #P < 0.05 and ##P < 0.01 compared with the combination treatment group. Statistical significance (P) was calculated by one-way ANOVA. Error bars represent SEM. MPTP, mitochondrial permeability transition pore; veh, vehicle.

Close modal

DCA Coordinated With DNP Is an Effect-Enhancing Way to Improve Hyperglycemia via Preferential Glucose Oxidation

Next, we explored the effects of DNP combined with DCA (combination treatment) on metabolic profiles in vitro. PDH is regulated by four PDKs, which phosphorylate and inhibit PDH activity (31). Reduced phosphorylated Ser293-PDH was observed with DCA treatment and combination treatment in vitro, while DNP alone had no effects (Fig. 1D and Supplementary Fig. 2F). On the basis of the inhibition of anaerobic glycolysis and fatty acid oxidation by DCA, the stimulation of lactate production and fatty acid oxidation by DNP decreased after combination treatment in cells. Furthermore, fatty acid oxidation was adjusted to control levels by combination treatment (Fig. 1E and F). Because of the distinct mechanisms exerted by DCA/DNP alone on glucose oxidation, combination treatment further promoted glucose oxidation in myotubes (Fig. 1G). These data implied that combination treatment resulted in a preference for glucose as an oxidative substrate in vitro.

Then, we examined the metabolic effects of DNP/DCA treatment and combination treatment in mice through indirect calorimetry (IDC). IDC showed that DNP or combination treatment, but not DCA, increased the EE and the OCR without affecting activity for 2 h after oral administration. Meanwhile, DCA and combination treatment exhibited a higher RER than that of the DNP group, which indicated that a larger proportion of RER occurring with combination treatment was derived from glucose oxidation by DCA. In the next 3–5 h, the effect on RER induced by DCA disappeared, while the OCR and EE were still elevated in the DNP and combination treatment groups (Fig. 1H). Consistently, IDC performance after combination treatment was evidenced by lower phosphorylated Ser293-PDH levels in skeletal muscle and liver but not in iWAT (Supplementary Fig. 3). Previous research reported that glucose-derived oxidation was not significantly affected by DCA-induced PDH activation in adipose tissue (32), suggesting that PDH activation in adipose tissue might contribute little to EE. Our data implied that combination treatment drives metabolic cells to prefer glucose over fatty acids as an oxidative substrate in vivo.

The effects of combination treatment on the preferential promotion of glucose oxidation prompted us to investigate the effect on glycemic control in vivo. DNP and DCA individually enhanced glucose clearance in normal and hyperglycemic mice based on glucose challenge. Moreover, combination treatment further promoted glucose clearance in both mouse models compared with DNP alone (Fig. 2A and B). Thus, these data suggested that DNP combined with DCA enhanced the hypoglycemic effect in vivo.

Figure 2

DCA enhances the efficacy of DNP in glucose clearance with improved toxicity profile. A and B: STZ-induced (STZ-treated) hyperglycemic mice or normal (untreated) C57BL/6J mice were fasted for 6 h and then gavaged with 10 mg/kg DNP, 1 g/kg DCA, or both. Glucose (2 g/kg, intraperitoneally) tolerance was assessed at 1 h after administration of compounds. Blood glucose concentration and the area under the glucose curve were detected at 2 h after glucose administration. n = 7–8 for all groups. C and D: C57BL/6J mice were orally treated with 1 g/kg DCA for 2 weeks (multidose DCA), single-dose DCA, or 30 mg/kg DNP. C: The relative expression of PDK1, 2, 3, and 4 normalized to 18s expression in skeletal muscle, liver, and iWAT. n = 5–7 for all groups. D: Immunoblot and quantification analyses of the phosphorylated Ser293-PDH in skeletal muscle, liver, and iWAT and the density of P-PDH/PDH calculated and normalized to the NC (no treatment) group. n = 2 for all groups. For EH, C57BL/6J mice were orally treated with 1 g/kg DCA or vehicle for 2 weeks. Then, mice were orally treated with single-dose 30 or 60 mg/kg DNP at the last day. E: The percentage of surviving mice was detected at 24 h after administration of 60 mg/kg DNP. n = 10 for all groups. F: Plasma LDH was detected at 1 h after administration of 60 mg/kg DNP. n = 6 for all groups. G: Rectal temperature was detected at 2 h after administration of 30 mg/kg DNP. H: Plasma lactate concentration was detected at 3 h after administration of 30 mg/kg DNP. n = 9–10 for all groups. *P < 0.05, **P < 0.01 and ***P < 0.001 compared with the vehicle group. #P < 0.05, ##P < 0.01, and ###P < 0.001 compared with the combination treatment group. Statistical significance (P) was calculated by one-way ANOVA. Error bars reprsent SEM. AUC units are mg/dL × min × 103. AUC, area under the curve; NC, mice without any treatment; Veh, vehicle.

Figure 2

DCA enhances the efficacy of DNP in glucose clearance with improved toxicity profile. A and B: STZ-induced (STZ-treated) hyperglycemic mice or normal (untreated) C57BL/6J mice were fasted for 6 h and then gavaged with 10 mg/kg DNP, 1 g/kg DCA, or both. Glucose (2 g/kg, intraperitoneally) tolerance was assessed at 1 h after administration of compounds. Blood glucose concentration and the area under the glucose curve were detected at 2 h after glucose administration. n = 7–8 for all groups. C and D: C57BL/6J mice were orally treated with 1 g/kg DCA for 2 weeks (multidose DCA), single-dose DCA, or 30 mg/kg DNP. C: The relative expression of PDK1, 2, 3, and 4 normalized to 18s expression in skeletal muscle, liver, and iWAT. n = 5–7 for all groups. D: Immunoblot and quantification analyses of the phosphorylated Ser293-PDH in skeletal muscle, liver, and iWAT and the density of P-PDH/PDH calculated and normalized to the NC (no treatment) group. n = 2 for all groups. For EH, C57BL/6J mice were orally treated with 1 g/kg DCA or vehicle for 2 weeks. Then, mice were orally treated with single-dose 30 or 60 mg/kg DNP at the last day. E: The percentage of surviving mice was detected at 24 h after administration of 60 mg/kg DNP. n = 10 for all groups. F: Plasma LDH was detected at 1 h after administration of 60 mg/kg DNP. n = 6 for all groups. G: Rectal temperature was detected at 2 h after administration of 30 mg/kg DNP. H: Plasma lactate concentration was detected at 3 h after administration of 30 mg/kg DNP. n = 9–10 for all groups. *P < 0.05, **P < 0.01 and ***P < 0.001 compared with the vehicle group. #P < 0.05, ##P < 0.01, and ###P < 0.001 compared with the combination treatment group. Statistical significance (P) was calculated by one-way ANOVA. Error bars reprsent SEM. AUC units are mg/dL × min × 103. AUC, area under the curve; NC, mice without any treatment; Veh, vehicle.

Close modal

DCA Coordinated With DNP Partially Reduces Adverse Effects From Uncoupling Effects

Then, we further examined whether combination treatment achieved the toxicity-reducing goal. The metabolic effects of PDH activation in mice were demonstrated with a single dose of DCA applied for a short time (Supplementary Fig. 3). With 2 weeks of daily DCA pretreatment (multidose DCA), continuous activation of PDH was observed in skeletal muscle, liver, and iWAT, accompanied by downregulated PDK1, 2, 3, and 4 (Fig. 2C and D). Following gavage with multidose DCA, the death caused by 60 mg/kg DNP was avoided (Fig. 2E). In addition, lactate dehydrogenase release was significantly reduced by DCA protection, suggesting that tissue injury might be relieved (Fig. 2F). Mice gavaged with 30 mg/kg DNP did not die, but hyperthermia emerged. Inhibition of lipid oxidation by PDH activation in PDK2/PDK4 double-deficiency mice was reported to induce hypothermia (33). Consistently, hypothermia occurred following DCA treatment. However, this aberrant rectal temperature was adjusted by combination treatment (Fig. 2G). Also, the increase in plasma lactate by DNP was reduced by combination treatment (Fig. 2H). Additionally, we observed that the pharmacokinetic profiles and tissue distribution of DNP and DCA were unchanged with combination treatment compared with DCA/DNP–treated mice (Supplementary Fig. 4). These data implied that combination treatment protected against adverse effects due to uncoupling by DNP.

Uncoupling of Mitochondria Restores the Sensitivity to Insulin Impaired by PDH Activation in HFD Mice

We identified an effect-enhancing and toxicity-reducing method to harness PDH activation with DNP to improve hyperglycemia. However, from the phenotype of muscle-specific PDK2/PDK4 double-knockout mice, the hypoglycemic actions of PDH activation driving HFD mice to switch their oxidative substrates from fatty acids to glucose led to increased re-esterification of fatty acids into DAG and TG (34). DAG has been confirmed to be closely related to insulin resistance through activation of PKC isoform and decreased insulin signaling in vivo (3538). Thus, we tested whether combination treatment could ameliorate insulin resistance in HFD mice relative to DCA treatment. Although DCA treatment decreased blood glucose, insulin resistance was aggravated (Fig. 3A and B). Additionally, DCA treatment increased the organ index of the liver over body weight (Fig. 3C). Furthermore, histological analyses showed that mice treated with DCA showed exaggerative lipid accumulation in the liver, which was confirmed by quantification of the TG and DAG contents (Fig. 3D, E, and H). Additionally, we assessed intramuscular lipid contents and found that DCA treatment induced intramuscular TG and DAG accumulation (Fig. 3F and G). Next, increase in the membrane translocation of PKC-θ and PKC-ε and reduction in insulin-stimulated phosphorylated Ser473-Akt occurred in skeletal muscle and liver (Fig. 3I–K). With combination treatment, the exaggerated hepatic steatosis induced by DCA was relieved according to histological analyses. Combination treatment reduced intramuscular and intrahepatic lipid accumulation and membrane translocation of PKC-θ in skeletal muscle and PKC-ε in liver, leading to the restoration of insulin sensitivity in skeletal muscle and liver (Fig. 3). Taken together, these data suggested that DNP restored the sensitivity of metabolic tissues to insulin impaired by DCA, which might be partially attributed to the rectification of DAG accumulation and activation of PKC-ε and PKC-θ.

Figure 3

DNP improves DCA-induced insulin resistance by normalizing the status of the DAG/PKC axis in HFD mice. For AK, HFD mice were orally treated with 300 mg/kg DCA, 10 mg/kg DNP, or both for 4 weeks. A: Insulin (0.75 units/kg, intraperitoneally) tolerance was assessed in mice fasted for 4 h at the 4th week. B: Fasting blood glucose concentration was assessed in mice fasted for 6 h. C: Liver index. D and E: TG and DAG contents in liver. F and G: TG and DAG contents in skeletal muscle. n = 8–10 for all groups. H: Representative liver sections stained with H-E (scale bars, 100 μm; upper H-E images). I: Immunoblot and quantification analyses of PKC-θ translocation in skeletal muscle. J: Immunoblot and quantification analyses of PKC-ε translocation in liver. K: Immunoblot and quantification analyses of phosphorylated Ser473-Akt in skeletal muscle and liver from mice intraperitoneally injected with insulin (0.75 units/kg) for 15 min or not. n = 3–4 for all groups. *P < 0.05, **P < 0.01, and ***P < 0.001 compared with the vehicle group, &P < 0.05 compared among mice treated with insulin or not. #P < 0.05, ##P < 0.01, and ###P < 0.001 compared with the combination treatment group. Statistical significance (P) was determined by one-way ANOVA. Error bars represent SEM. H-E, hematoxylin-eosin; Veh, vehicle.

Figure 3

DNP improves DCA-induced insulin resistance by normalizing the status of the DAG/PKC axis in HFD mice. For AK, HFD mice were orally treated with 300 mg/kg DCA, 10 mg/kg DNP, or both for 4 weeks. A: Insulin (0.75 units/kg, intraperitoneally) tolerance was assessed in mice fasted for 4 h at the 4th week. B: Fasting blood glucose concentration was assessed in mice fasted for 6 h. C: Liver index. D and E: TG and DAG contents in liver. F and G: TG and DAG contents in skeletal muscle. n = 8–10 for all groups. H: Representative liver sections stained with H-E (scale bars, 100 μm; upper H-E images). I: Immunoblot and quantification analyses of PKC-θ translocation in skeletal muscle. J: Immunoblot and quantification analyses of PKC-ε translocation in liver. K: Immunoblot and quantification analyses of phosphorylated Ser473-Akt in skeletal muscle and liver from mice intraperitoneally injected with insulin (0.75 units/kg) for 15 min or not. n = 3–4 for all groups. *P < 0.05, **P < 0.01, and ***P < 0.001 compared with the vehicle group, &P < 0.05 compared among mice treated with insulin or not. #P < 0.05, ##P < 0.01, and ###P < 0.001 compared with the combination treatment group. Statistical significance (P) was determined by one-way ANOVA. Error bars represent SEM. H-E, hematoxylin-eosin; Veh, vehicle.

Close modal

Identification of 6j as a Novel Compound Possessing Uncoupling and PDH Activation Activity In Vitro and In Vivo

Through the aforementioned evidence, we demonstrated that mitochondrial uncoupling combined with PDH activation might be a potentially safe and effective strategy for hyperglycemia therapy (Fig. 4A). Then, we assumed that a small molecule possessing both the above dual actions might safely ameliorate hyperglycemia. On the basis of our previous research on high-throughput screening for modulators of mitochondrial membrane potential in myotubes (39), LGH00277 was selected as an active hit, and its weak uncoupling activity was also confirmed (Supplementary Fig. 5A–C). Then, a series of derivatives were synthesized (Medicinal Chemistry Section in the Supplementary Data), the OCR was determined to reflect uncoupling activity, and ECAR was determined to indirectly reflect lactate production upon uncoupling challenge (Fig. 4B). Then, the phosphorylated Ser293-PDH was confirmed. With the increase in OCR, a decrease in phosphorylated Ser293-PDH, and no increase in ECAR, lactate content was examined following 12-h treatment in myotubes. By analyzing uncoupling activity, Ser293-PDH phosphorylation, and lactate production, 6j was identified, in myotubes, as a potential compound for validation of our novel concept (Fig. 4B and Supplementary Fig. 5D–F).

Figure 4

6j selectively stimulates glucose oxidative metabolism in vitro. A: Summary of the effects of DCA combined with DNP on hyperglycemia therapy. B: The route of identification of the candidate compound (6j) possessing uncoupling action and PDH activity in myotubes. C: OCR detected by Seahorse Analyzer in myotubes, 3T3-L1 cells, and hepatocytes. D: The ADP/ATP ratio detected in myotubes and hepatocytes incubated with 6j for 3 h. E: Immunoblot analyses of phosphorylated Ser293-PDH in myotubes and hepatocytes incubated with 6j for 3 h. F: Glucose uptake was detected through 3H-labeled 2-deoxyglucose in myotubes incubated with 6j for 3 h. G: Glucose oxidation detected through 14C-labeled glucose in myotubes treated with 6j for 4 h. H: Quantification of lactate release from myotubes and hepatocytes treated with 6j for 12 h. I: Fatty acid oxidation detected through 3H-labeled palmitate in myotubes and hepatocytes treated with 6j for 4 h. n = 3–4 for all groups. *P < 0.05, **P < 0.01, and ***P < 0.001 compared with DMSO group. Statistical significance (P) was determined by one-way ANOVA. Error bars represent SEM.

Figure 4

6j selectively stimulates glucose oxidative metabolism in vitro. A: Summary of the effects of DCA combined with DNP on hyperglycemia therapy. B: The route of identification of the candidate compound (6j) possessing uncoupling action and PDH activity in myotubes. C: OCR detected by Seahorse Analyzer in myotubes, 3T3-L1 cells, and hepatocytes. D: The ADP/ATP ratio detected in myotubes and hepatocytes incubated with 6j for 3 h. E: Immunoblot analyses of phosphorylated Ser293-PDH in myotubes and hepatocytes incubated with 6j for 3 h. F: Glucose uptake was detected through 3H-labeled 2-deoxyglucose in myotubes incubated with 6j for 3 h. G: Glucose oxidation detected through 14C-labeled glucose in myotubes treated with 6j for 4 h. H: Quantification of lactate release from myotubes and hepatocytes treated with 6j for 12 h. I: Fatty acid oxidation detected through 3H-labeled palmitate in myotubes and hepatocytes treated with 6j for 4 h. n = 3–4 for all groups. *P < 0.05, **P < 0.01, and ***P < 0.001 compared with DMSO group. Statistical significance (P) was determined by one-way ANOVA. Error bars represent SEM.

Close modal

We then examined the uncoupling activity and PDH activation effects of 6j in hepatocytes and 3T3-L1 cells and found that 6j stimulated the OCR in hepatocytes but not in 3T3-L1 cells. Increased ADP/ATP ratios and reduced Ser293-PDH phosphorylation by 6j were also observed in myotubes and hepatocytes (Fig. 4C–E). These results suggested that 6j was a myotube- and hepatocyte-specific mitochondrial uncoupler and PDH activator. To address the uncoupling action of 6j, glucose uptake and glucose oxidation was stimulated by 6j in myotubes (Fig. 4F and G). However, fatty acid oxidation and lactate production were not induced by 6j in myotubes and hepatocytes (Fig. 4H and I). Then, we examined the metabolic effects of orally administered 6j in vivo. A tissue distribution analysis showed that the 6j concentration in liver and skeletal muscle might reach the working concentration for myotubes or hepatocytes for PDH activation and uncoupling in db/db mice (Supplementary Fig. 6A). The reduction in phosphorylated Ser293-PDH was observed in skeletal muscle and liver but not in iWAT (Supplementary Fig. 6B). Meanwhile, IDC studies showed that mice gavaged with 6j had increased EE, OCR, and RER without effects on activity, indicating a preferential proportion of EE was obtained from glucose oxidation (Supplementary Fig. 6C). These data implied that 6j, with the ability to induce uncoupling and PDH activation effects, preferentially promoted glucose oxidation in vitro and in vivo.

Use of 6j Improves Hyperglycemia and Insulin Resistance in HFD Mice and db/db Mice

We further examined whether 6j was effective in improving glycemic control and insulin resistance in HFD mice. Daily treatment with 6j for 4 weeks led to a reduction in blood glucose. Also, the capacities of glucose tolerance and sensitivity to insulin were improved in mice that received 6j chronically (Supplementary Fig. 7). Then, we further examined the similar effects in db/db mice. The reduced blood glucose and enhanced glucose tolerance were observed during 6j treatment chronically (Fig. 5A–C). Furthermore, improved sensitivity to insulin and increased insulin-stimulated phosphorylation of Ser473-Akt in skeletal muscle and liver were observed in the 6j-treated mice (Fig. 5D–F). Then, we determined whether 6j improved insulin sensitivity via reducing ectopic lipid accumulation. Histological analysis of livers showed that 6j-treated mice had little intracellular lipid accumulation, which was evidenced by the quantification of TG and DAG contents (Fig. 5G and H). Also, a decrease in intramuscular TG and DAG was also observed in the 6j-treated group (Fig. 5I). On the basis of the reduction of intrahepatic and intramuscular DAG, membrane translocation of PKC-θ in skeletal muscle and PKC-ε in liver was significantly decreased (Fig. 5J and K). We also analyzed the uncoupling effects of 6j on body temperature and lactate production during chronic treatment and did not observe any differences between vehicle- and 6j-treated mice (Fig. 5L and M). Collectively, the in vivo results supported that 6j could improve hyperglycemia and insulin resistance without lactate and heat overproduction.

Figure 5

Oral 6j treatment improves glycemic control and insulin sensitivity in db/db mice. For AM, db/db mice were gavaged with 40 mg/kg 6j for 7 weeks. A: Fasting blood glucose concentrations were assessed in mice fasted for 6 h. B and C: Glucose (1.5 g/kg, intraperitoneally) tolerance was assessed on mice fasted for 6 h at the 5th week. *P < 0.05, *P < 0.01, and ***P < 0.001 compared with the vehicle group. Statistical significance (P) was determined by Student’s t test. D: Insulin (0.75 units/kg, intraperitoneally) tolerance was assessed in mice fasted for 4 h at the sixth week. n = 7–8 for all groups. E and F: Immunoblot and quantification analyses of phosphorylated Ser473-Akt in liver (E) and skeletal muscle (F) from mice intraperitoneally injected with insulin (0.75 units/kg) for 15 min or not. n = 3–4 for all groups. *P < 0.05, *P < 0.01, and ***P < 0.001 compared with the vehicle group; a <0.05 compared with the vehicle group treated with insulin. Statistical significance (P) was determined by one-way ANOVA. G: Representative liver sections stained with H-E (scale bars, 100 μm; upper H-E images). H and I: TG and DAG contents in liver and skeletal muscle. n = 7–8 for all groups. J and K: Immunoblot and quantification analyses of PKC-ε translocation in liver (J) and PKC-θ translocation in skeletal muscle (K). n = 3–4 for all groups. L: Detection of plasma lactate. M: Detection of rectal temperature. n = 7–8 for all groups. *P < 0.05, *P < 0.01, and ***P < 0.001 compared with the vehicle group. Statistical significance (P) was determined by Student’s t test. Error bars represent SEM. AUC units are mg/dL × min × 103. AUC, area under the curve; H-E, hematoxylin-eosin.

Figure 5

Oral 6j treatment improves glycemic control and insulin sensitivity in db/db mice. For AM, db/db mice were gavaged with 40 mg/kg 6j for 7 weeks. A: Fasting blood glucose concentrations were assessed in mice fasted for 6 h. B and C: Glucose (1.5 g/kg, intraperitoneally) tolerance was assessed on mice fasted for 6 h at the 5th week. *P < 0.05, *P < 0.01, and ***P < 0.001 compared with the vehicle group. Statistical significance (P) was determined by Student’s t test. D: Insulin (0.75 units/kg, intraperitoneally) tolerance was assessed in mice fasted for 4 h at the sixth week. n = 7–8 for all groups. E and F: Immunoblot and quantification analyses of phosphorylated Ser473-Akt in liver (E) and skeletal muscle (F) from mice intraperitoneally injected with insulin (0.75 units/kg) for 15 min or not. n = 3–4 for all groups. *P < 0.05, *P < 0.01, and ***P < 0.001 compared with the vehicle group; a <0.05 compared with the vehicle group treated with insulin. Statistical significance (P) was determined by one-way ANOVA. G: Representative liver sections stained with H-E (scale bars, 100 μm; upper H-E images). H and I: TG and DAG contents in liver and skeletal muscle. n = 7–8 for all groups. J and K: Immunoblot and quantification analyses of PKC-ε translocation in liver (J) and PKC-θ translocation in skeletal muscle (K). n = 3–4 for all groups. L: Detection of plasma lactate. M: Detection of rectal temperature. n = 7–8 for all groups. *P < 0.05, *P < 0.01, and ***P < 0.001 compared with the vehicle group. Statistical significance (P) was determined by Student’s t test. Error bars represent SEM. AUC units are mg/dL × min × 103. AUC, area under the curve; H-E, hematoxylin-eosin.

Close modal

Requirements of PDH Activation for Preferential Glucose Oxidation Induced by 6j

The in vitro and in vivo results support the hypothesis that 6j improves glycemia and insulin resistance through mitochondrial uncoupling combined with PDH activation and consequently preferentially promotes glucose oxidation. To investigate the dependency of preferential glucose oxidation on PDH activation, we examined the effect of 6j on PDH in vitro. The decrease in phosphorylated Ser293-PDH by 6j in myotubes and hepatocytes disappeared upon PDH knockdown by the application of siRNA (Fig. 6A). As expected, glucose oxidation induced by 6j declined with deficient PDH activation in myotubes, but 6j still had a slight effect on promoting glucose oxidation because of its uncoupling action (Fig. 6B). Meanwhile, fatty acid oxidation and lactate production were dramatically enhanced by 6j under deficient PDH activation in myotubes and hepatocytes (Fig. 6C–F). These results indicate that the effects of 6j on preferential glucose oxidation depend on PDH activation.

Figure 6

Preferential glucose oxidation induced by 6j is dependent on PDH activation. A: Immunoblot analyses of phosphorylated Ser293-PDH in myotubes and hepatocytes incubated with 6j for 3 h. B: Glucose oxidation through 14C-labeled glucose in myotubes incubated with 6j for 3 h. C and D: Fatty acid oxidation detected through 3H-labeled palmitate in myotubes (C) and hepatocytes (D) incubated with 6j for 4 h. E: Lactate release from myotubes incubated with 6j for 12 h. F: Lactate release from hepatocytes incubated with 6j for 12 h. For AF, myotubes and hepatocytes were incubated with PDH siRNA or not for 48 h before the assay. n = 3–4 for all groups. *P < 0.05, **P < 0.01, and ***P < 0.001 compared with the siCtrl-DMSO group. #P < 0.05 and ##P < 0.01 compared with the siPDH-DMSO group. $$$P < 0.001 compared with the siPDH-6j group. Statistical significance (P) was calculated by one-way ANOVA. Error bars represent SEM. Ctrl, control; siPDH, small interfering RNA targeting PDH.

Figure 6

Preferential glucose oxidation induced by 6j is dependent on PDH activation. A: Immunoblot analyses of phosphorylated Ser293-PDH in myotubes and hepatocytes incubated with 6j for 3 h. B: Glucose oxidation through 14C-labeled glucose in myotubes incubated with 6j for 3 h. C and D: Fatty acid oxidation detected through 3H-labeled palmitate in myotubes (C) and hepatocytes (D) incubated with 6j for 4 h. E: Lactate release from myotubes incubated with 6j for 12 h. F: Lactate release from hepatocytes incubated with 6j for 12 h. For AF, myotubes and hepatocytes were incubated with PDH siRNA or not for 48 h before the assay. n = 3–4 for all groups. *P < 0.05, **P < 0.01, and ***P < 0.001 compared with the siCtrl-DMSO group. #P < 0.05 and ##P < 0.01 compared with the siPDH-DMSO group. $$$P < 0.001 compared with the siPDH-6j group. Statistical significance (P) was calculated by one-way ANOVA. Error bars represent SEM. Ctrl, control; siPDH, small interfering RNA targeting PDH.

Close modal

The promotion of mitochondrial oxidative metabolism by uncoupling actions represents a conceptual approach for the treatment of diabetes, nonalcoholic fatty liver disease, or other metabolic diseases. In view of this, a pharmacological uncoupling agent, DNP, was applied clinically in the 1930s. However, DNP was taken off the market because of its narrow therapeutic index, evidenced by the emergence of severe adverse effects, including hyperlactacidemia, hyperthermia, and even death (1115). Thus, it is a challenge to discover safe mitochondrial uncouplers for practical use. In our study, we demonstrated that PDH activation combined with mitochondrial uncoupling might be an alternative safe and effective approach for treating T2D.

It is critical and challenging to separate adverse effects from uncoupling activity. Several studies focused on the development of new classes of chemical mitochondrial uncouplers to safely treat metabolic diseases. On the basis of the excellent effects and large therapeutic indexes of liver-specific mitochondrial uncouplers, including liver-targeted derivatives of DNP, controlled-release mitochondrial protonophore, and niclosamide ethanolamine, on hyperglycemia and hyperlipidemia (4043), it might be possible that tissue-specific chemical mitochondrial uncouplers have potential safety and efficacy. In our study, we focused on exploring a novel strategy of developing a new class of mitochondrial uncouplers. Previous research showed that the adverse effects of DNP mainly resulted from uncoupling-induced excessive lipid oxidation and anaerobic glycolysis. However, we found that inhibition of lipid oxidation alone by etomoxir or lactate production alone by galloflavin did not affect lactate production or DNP-derived lipid oxidation, suggesting that DNP-induced adverse effects could not be relieved by combination with etomoxir or galloflavin. Thus, we explored whether other targets combined with DNP could differentiate its hypoglycemic actions from its adverse effects by switching anaerobic glycolysis to oxidation to decrease fat burning and lactate overproduction. DCA, as a fat-to-glucose switcher, was selected to prove our hypothesis. DNP combined with amiloride had an effect similar to its effects with DCA in myotubes, but hyperkalemia was observed with amiloride treatment in the clinic (28,29,4446), similar to the adverse effects of DNP. However, linking other safe potassium-retaining diuretic drugs to DNP might provide another effect-enhancing and toxicity-reducing option for T2D therapy.

Heat production from fat combustion is far higher than that produced from glucose (47). Inhibition of lipolysis abolished the mobilization of lipids and cold-induced brown adipose tissue thermogenesis, suggesting that fat is a standby source of heat for adaption to environmental changes (48). Once excessive fat mobilization by uncoupling actions occurs, hyperthermia develops. Conversely, inhibition of fatty acid oxidation by PDH activation in a double-knockout PDK2/PDK4 mouse model resulted in hypothermia (33). In our study with mice treated with DNP and DCA together, normal rectal temperature was maintained, suggesting that the abnormal status of fatty acid oxidation was corrected. Furthermore, a normal rectal temperature might partially contribute to protecting mice from death. In addition, increased plasma LDH, which is released from tissue injured by the toxic effects of DNP, declined with DCA treatment, suggesting that decreased LDH might be an indicator of survival against DNP toxicity. However, neither normal rectal temperature nor LDH release solely accounts for the survival and its underlying mechanism against DNP-induced mortality, and the mechanism associated with combination treatment required further investigation.

The nutrient storage pathways evolved to maximize efficient energy utilization that exposed to chronic energy surplus causes obesity and obesity-derived insulin resistance. Several previous studies clarified that obesity-derived ectopic lipid accumulation or obesity-induced tissue inflammation was closely associated with the progression of insulin resistance. Samuel and Shulman (49) proved that DAG directly activated PKC-θ in skeletal muscle and PKC-ε in liver, which decreased downstream insulin signaling. Meanwhile, it was demonstrated that an increase in glucose utilization and a decrease in fatty acid oxidation by PDH activation led to increased intramuscular DAG and an impaired insulin pathway (34). In our study, the lowered blood glucose and DAG accumulation–induced exaggerative insulin resistance were observed by DCA treatment. Interestingly, these phenomena were corrected by combination treatment. These results might be explained by the fact that the DNP contributed to relieve the inhibition of fatty acid oxidation by PDH activation. According to histological analyses of mouse livers under combination treatment, hepatic steatosis was markedly alleviated compared with that in the DCA group, but it was still severe compared with that in the DNP group. However, sensitivity to insulin was similar between the DNP and combination treatment group. Apart from the DAG/PKC pathway, hepatic lipid overload might increase the expression of proinflammatory cytokines, such as tumor necrosis factor-α (TNF-α) and interleukin (IL)–6, or the accumulation of specific lipid metabolites, such as ceramides, which decrease insulin signaling (6,50). Thus, it is possible that the decreased expression of TNF-α and IL-6 or ceramide accumulation in liver in the combination treatment group partially contributed to insulin sensitivity, which still requires further investigation.

On the basis of the metabolic benefits of DNP combined with DCA, the compound 6j was identified and confirmed to possess mitochondrial uncoupling and PDH activation in vitro and in vivo. Chronic administration of 6j significantly improved glycemic control in db/db mice. Correlated with the reduction of DAG and TG accumulation in liver and skeletal muscle, insulin resistance was restored by chronic 6j treatment. Apart from DAG accumulation, the decrease in the expression of proinflammatory cytokines was observed (Supplementary Fig. 8), which also might have contributed to the improvement of insulin sensitivity. In this study, 6j was a classical chemical mitochondrial uncoupler, promoting the proton across mitochondrial inner membrane through non-ATP synthase. However, the way that 6j affected the proton leakage through mitochondrial transporters such as uncoupling proteins, adenine nucleotide translocators, or the other proton transporters still needs to be further explored. Unlike classical mitochondrial uncouplers, the effects of 6j on PDH activation altered the uncoupling actions and stimulated oxidative substrate flux from fatty acids to glucose, which might have partially contributed to the reduction in adverse effects. Also, PDH activity increased by 6j still requires further investigation.

In summary, our research demonstrated that PDH activation combined with mitochondrial uncoupling promoted hypoglycemic effects and partially reduced adverse effects through preferential glucose oxidative flux. A novel compound, 6j, was identified and exhibited hypoglycemic effects in vivo. Thus, our research provides the possibility of further exploring a new class of mitochondrial uncouplers for treating T2D.

Funding. This work was supported by National Natural Science Foundation of China grants 81125023 and 81673493, Strategic Priority Research Program of Chinese Academy of Sciences grant XDA12040301, Shanghai Science and Technology Commission grant 16JC1405000, and K.C. Wong Education Foundation.

Duality of Interest. No potential conflicts of interest relevant to this article were reported.

Author Contributions. H.J. contributed to the design and conducted the experiments, collected the research data, provided analyses and discussion, and wrote the manuscript. J.J. and H.L. contributed to the synthesis compounds. Y.D., Z.X., Y.L., X.Z., C.P., C.X., and T.D. assisted with the animal experiments. Y.D. and A.G. assisted with the cellular experiments. M.G. contributed to the measurement of adenine nucleotide levels. L.Y., J.T., and F.Y. contributed to design, analysis, and manuscript drafting for the chemistry. Jin. Li and Jia Li contributed to the research design, analysis, discussion, and drafting of the manuscript. F.Y., Jin. Li, and Jia Li are the guarantors of this work and, as such, had full access to all the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis.

Prior Presentation. Parts of this study were presented at the 79th Scientific Sessions of the American Diabetes Association, San Francisco, CA, 7–11 June 2019.

See accompanying article, p. 2195.

1.
Kusminski
CM
,
Bickel
PE
,
Scherer
PE
.
Targeting adipose tissue in the treatment of obesity-associated diabetes
.
Nat Rev Drug Discov
2016
;
15
:
639
660
2.
Holland
WL
,
Knotts
TA
,
Chavez
JA
,
Wang
LP
,
Hoehn
KL
,
Summers
SA
.
Lipid mediators of insulin resistance
.
Nutr Rev
2007
;
65
:
S39
S46
3.
Parker
R
,
Weston
CJ
,
Miao
Z
, et al
.
CC chemokine receptor 2 promotes recruitment of myeloid cells associated with insulin resistance in nonalcoholic fatty liver disease
.
Am J Physiol Gastrointest Liver Physiol
2018
;
314
:
G483
G493
4.
Yamauchi
T
,
Kamon
J
,
Waki
H
, et al
.
The fat-derived hormone adiponectin reverses insulin resistance associated with both lipoatrophy and obesity
.
Nat Med
2001
;
7
:
941
946
5.
Steppan
CM
,
Bailey
ST
,
Bhat
S
, et al
.
The hormone resistin links obesity to diabetes
.
Nature
2001
;
409
:
307
312
6.
Uysal
KT
,
Wiesbrock
SM
,
Marino
MW
,
Hotamisligil
GS
.
Protection from obesity-induced insulin resistance in mice lacking TNF-alpha function
.
Nature
1997
;
389
:
610
614
7.
Apostolopoulou
M
,
Gordillo
R
,
Koliaki
C
, et al
.
Specific hepatic sphingolipids relate to insulin resistance, oxidative stress, and inflammation in nonalcoholic steatohepatitis
.
Diabetes Care
2018
;
41
:
1235
1243
8.
Terada
H
.
Uncouplers of oxidative phosphorylation
.
Environ Health Perspect
1990
;
87
:
213
218
9.
Nedergaard
J
,
Ricquier
D
,
Kozak
LP
.
Uncoupling proteins: current status and therapeutic prospects
.
EMBO Rep
2005
;
6
:
917
921
10.
Mookerjee
SA
,
Divakaruni
AS
,
Jastroch
M
,
Brand
MD
.
Mitochondrial uncoupling and lifespan
.
Mech Ageing Dev
2010
;
131
:
463
472
11.
Hoch
FL
,
Hogan
FP
.
Hyperthermia, muscle rigidity, and uncoupling in skeletal muscle mitochondria in rats treated with halothane and 2,4-dinitrophenol
.
Anesthesiology
1973
;
38
:
237
243
12.
McFee
RB
,
Caraccio
TR
,
McGuigan
MA
,
Reynolds
SA
,
Bellanger
P
.
Dying to be thin: a dinitrophenol related fatality
.
Vet Hum Toxicol
2004
;
46
:
251
254
13.
Wallace
KB
,
Starkov
AA
.
Mitochondrial targets of drug toxicity
.
Annu Rev Pharmacol Toxicol
2000
;
40
:
353
388
14.
Parascandola
J
.
Dinitrophenol and bioenergetics: an historical perspective
.
Mol Cell Biochem
1974
;
5
:
69
77
15.
Tewari
A
,
Ali
T
,
O’Donnell
J
,
Butt
MS
.
Weight loss and 2,4-dinitrophenol poisoning [published correction appears in Br J Anaesth 2009;102:728]
.
Br J Anaesth
2009
;
102
:
566
567
16.
Bryan
AH
,
Ricketts
HT
,
Dine
WC
.
Effect of dinitrophenol on experimental diabetes
.
Proc Soc Exp Biol Med
1937
;
37
:
4
8
17.
Rognstad
R
,
Katz
J
.
The effect of 2,4-dinitrophenol on adipose-tissue metabolism
.
Biochem J
1969
;
111
:
431
444
18.
Simon
EW
.
Mechanisms of dinitrophenol toxicity
.
Biol Rev Camb Philos Soc
1953
;
28
:
453
479
19.
Grundlingh
J
,
Dargan
PI
,
El-Zanfaly
M
,
Wood
DM
.
2,4-dinitrophenol (DNP): a weight loss agent with significant acute toxicity and risk of death
.
J Med Toxicol
2011
;
7
:
205
212
20.
Lee
IK
.
The role of pyruvate dehydrogenase kinase in diabetes and obesity
.
Diabetes Metab J
2014
;
38
:
181
186
21.
Constantin-Teodosiu
D
.
Regulation of muscle pyruvate dehydrogenase complex in insulin resistance: effects of exercise and dichloroacetate
.
Diabetes Metab J
2013
;
37
:
301
314
22.
Evans
OB
,
Stacpoole
PW
.
Prolonged hypolactatemia and increased total pyruvate dehydrogenase activity by dichloroacetate
.
Biochem Pharmacol
1982
;
31
:
1295
1300
23.
Hommes
FA
,
Draisma
MI
,
Molenaar
I
.
Preparation and some properties of isolated rat liver cells
.
Biochim Biophys Acta
1970
;
222
:
361
371
24.
Kim
JB
,
Spiegelman
BM
.
ADD1/SREBP1 promotes adipocyte differentiation and gene expression linked to fatty acid metabolism
.
Genes Dev
1996
;
10
:
1096
1107
25.
Fu
YY
,
Zhang
M
,
Turner
N
, et al
.
A novel chemical uncoupler ameliorates obesity and related phenotypes in mice with diet-induced obesity by modulating energy expenditure and food intake
.
Diabetologia
2013
;
56
:
2297
2307
26.
Cheng
Z
,
Pang
T
,
Gu
M
, et al
.
Berberine-stimulated glucose uptake in L6 myotubes involves both AMPK and p38 MAPK
.
Biochim Biophys Acta
2006
;
1760
:
1682
1689
27.
Farabegoli
F
,
Vettraino
M
,
Manerba
M
,
Fiume
L
,
Roberti
M
,
Di Stefano
G
.
Galloflavin, a new lactate dehydrogenase inhibitor, induces the death of human breast cancer cells with different glycolytic attitude by affecting distinct signaling pathways
.
Eur J Pharm Sci
2012
;
47
:
729
738
28.
Cremades
A
,
Vicente-Ortega
V
,
Peñafiel
R
.
Influence of murine renal sexual dimorphism on amiloride-induced hyperkalemia
.
Nephron, Physiol
2003
;
95
:
57
66
29.
McNay
JL
,
Oran
E
.
Possible predisposition of diabetic patients to hyperkalemia following administration of potassium-retaining diuretic, amiloride (MK 870)
.
Metabolism
1970
;
19
:
58
70
30.
Patel
MS
,
Korotchkina
LG
.
Regulation of the pyruvate dehydrogenase complex
.
Biochem Soc Trans
2006
;
34
:
217
222
31.
Holness
MJ
,
Sugden
MC
.
Regulation of pyruvate dehydrogenase complex activity by reversible phosphorylation
.
Biochem Soc Trans
2003
;
31
:
1143
1151
32.
Whitehouse
S
,
Cooper
RH
,
Randle
PJ
.
Mechanism of activation of pyruvate dehydrogenase by dichloroacetate and other halogenated carboxylic acids
.
Biochem J
1974
;
141
:
761
774
33.
Jeoung
NH
,
Rahimi
Y
,
Wu
P
,
Lee
WN
,
Harris
RA
.
Fasting induces ketoacidosis and hypothermia in PDHK2/PDHK4-double-knockout mice
.
Biochem J
2012
;
443
:
829
839
34.
Rahimi
Y
,
Camporez
JP
,
Petersen
MC
, et al
.
Genetic activation of pyruvate dehydrogenase alters oxidative substrate selection to induce skeletal muscle insulin resistance
.
Proc Natl Acad Sci U S A
2014
;
111
:
16508
16513
35.
Ter Horst
KW
,
Gilijamse
PW
,
Versteeg
RI
, et al
.
Hepatic diacylglycerol-associated protein kinase Cε translocation links hepatic steatosis to hepatic insulin resistance in humans
.
Cell Rep
2017
;
19
:
1997
2004
36.
Petersen
MC
,
Madiraju
AK
,
Gassaway
BM
, et al
.
Insulin receptor Thr1160 phosphorylation mediates lipid-induced hepatic insulin resistance
.
J Clin Invest
2016
;
126
:
4361
4371
37.
Kim
JK
,
Fillmore
JJ
,
Sunshine
MJ
, et al
.
PKC-theta knockout mice are protected from fat-induced insulin resistance
.
J Clin Invest
2004
;
114
:
823
827
38.
Petersen
MC
,
Shulman
GI
.
Roles of diacylglycerols and ceramides in hepatic insulin resistance
.
Trends Pharmacol Sci
2017
;
38
:
649
665
39.
Qiu
BY
,
Turner
N
,
Li
YY
, et al
.
High-throughput assay for modulators of mitochondrial membrane potential identifies a novel compound with beneficial effects on db/db mice
.
Diabetes
2010
;
59
:
256
265
40.
Perry
RJ
,
Zhang
D
,
Zhang
XM
,
Boyer
JL
,
Shulman
GI
.
Controlled-release mitochondrial protonophore reverses diabetes and steatohepatitis in rats
.
Science
2015
;
347
:
1253
1256
41.
Perry
RJ
,
Kim
T
,
Zhang
XM
, et al
.
Reversal of hypertriglyceridemia, fatty liver disease, and insulin resistance by a liver-targeted mitochondrial uncoupler
.
Cell Metab
2013
;
18
:
740
748
42.
Abulizi
A
,
Perry
RJ
,
Camporez
JPG
, et al
.
A controlled-release mitochondrial protonophore reverses hypertriglyceridemia, nonalcoholic steatohepatitis, and diabetes in lipodystrophic mice
.
FASEB J
2017
;
31
:
2916
2924
43.
Tao
H
,
Zhang
Y
,
Zeng
X
,
Shulman
GI
,
Jin
S
.
Niclosamide ethanolamine-induced mild mitochondrial uncoupling improves diabetic symptoms in mice
.
Nat Med
2014
;
20
:
1263
1269
44.
Loffing
J
,
Kaissling
B
.
Sodium and calcium transport pathways along the mammalian distal nephron: from rabbit to human
.
Am J Physiol Renal Physiol
2003
;
284
:
F628
F643
45.
Benos
DJ
.
Amiloride: a molecular probe of sodium transport in tissues and cells
.
Am J Physiol
1982
;
242
:
C131
C145
46.
Canessa
CM
,
Schild
L
,
Buell
G
, et al
.
Amiloride-sensitive epithelial Na+ channel is made of three homologous subunits
.
Nature
1994
;
367
:
463
467
47.
Sessler
DI
.
Perioperative thermoregulation and heat balance
.
Ann N Y Acad Sci
1997
;
813
:
757
777
48.
Blondin
DP
,
Frisch
F
,
Phoenix
S
, et al
.
Inhibition of intracellular triglyceride lipolysis suppresses cold-induced brown adipose tissue metabolism and increases shivering in humans
.
Cell Metab
2017
;
25
:
438
447
49.
Samuel
VT
,
Shulman
GI
.
Mechanisms for insulin resistance: common threads and missing links
.
Cell
2012
;
148
:
852
871
50.
Holland
WL
,
Brozinick
JT
,
Wang
LP
, et al
.
Inhibition of ceramide synthesis ameliorates glucocorticoid-, saturated-fat-, and obesity-induced insulin resistance
.
Cell Metab
2007
;
5
:
167
179
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 http://www.diabetesjournals.org/content/license.

Supplementary data