Murine protein serine-threonine kinase 38 (MPK38)/maternal embryonic leucine zipper kinase (MELK) is implicated in diverse biological processes, including the cell cycle, apoptosis, and tumorigenesis; however, its physiological role is unknown. Using mice lacking MPK38 (MPK38−/−), we found that MPK38−/− male, but not female, mice (7 months of age) became obese while consuming a standard diet, displayed impairments in metabolism and inflammation, became more obese than wild-type mice while consuming a high-fat diet, and exhibited no castration/testosterone replacement–induced metabolic changes. The adenoviral restoration of MPK38 ameliorated the obesity-induced adverse metabolic profile of the obese male, but not female, mice. Seven-month-old MPK38−/− males displayed typical postcastration concentrations of serum testosterone with an accompanying decrease in serum luteinizing hormone (LH) levels, suggesting a role for MPK38 in the age-related changes in serum testosterone in aged mature adult male mice. The stability and activity of MPK38 were increased by dihydrotestosterone but reduced by estradiol (E2). These findings suggest MPK38 as a therapeutic target for obesity-related metabolic disorders in males.

MPK38/MELK is an AMPK-related kinase that is highly phylogenetically conserved and has been implicated in many biological functions (13). Many studies are being conducted focusing on two functions of MPK38; one is a tumor-promoting function by its antiapoptotic action (1,47), and the other is a metabolic function by its proapoptotic action (818). In terms of the role of MPK38 in metabolism, recent studies have shown that MPK38 coordinately activates ASK1/TGF-β/p53 signaling, which is involved in metabolic homeostasis (811). MPK38 phosphorylates PDK1 at Thr354, thereby inhibiting its activity (12), and stimulates the activity of p53 by phosphorylating it at Ser15 (13). MPK38 also activates the apoptosis signal-regulating kinase 1 (ASK1) and transforming growth factor-β (TGF-β) signaling by directly phosphorylating ASK1 at Thr838 and SMADS (Ser245 of SMAD2, Ser204 of SMAD3, Ser343 of SMAD4, and Thr96 of SMAD7), respectively (14,15). ASK1 ablation has been shown to be involved in metabolic disorders, such as obesity, nonalcoholic fatty liver disease, and fibrosis, in adipose and liver tissues (1618). These findings suggest that MPK38 functions as a key regulator of the metabolism associated with ASK1/TGF-β/p53 signaling pathways. Indeed, obese mice display lower levels of activation of ASK1/TGF-β/p53 signaling and lower expression of MPK38 and its interacting partners (SMAD3, ASK1, and ZPR9) than control mice (19). The amelioration of the obesity-associated metabolic disturbance achieved by SMAD3 or ZPR9 replacement is mediated by an increase in MPK38 activity (11,20). These findings make MPK38 an attractive therapeutic target for obesity-associated metabolic disorders.

The current study shows that MPK38 plays a crucial role in the protection against late-onset obesity-induced metabolic disturbances and the age-related decline in testosterone concentrations in aged mature adult male, but not female, mice. We have also confirmed this sex-specific antiobesity effect of MPK38 by restoring its expression in diet-induced obese mice. These findings suggest that a reduction in MPK38 expression may be an important mediator of the obesity phenotype in aged mature adult male mice.

Antibodies, Oligonucleotides, Animals, and MPK38 Knockout Mice

Detailed information for antibodies, oligonucleotides (Supplementary Table 1), and animals can be found in the Supplementary Material. MPK38 knockout mice (MPK38−/−) were generated with C57BL/6N mouse embryonic stem cells harboring gene trap insertions (AR081; Mutant Mouse Regional Resource Center at UC Davis).

Glucose and Insulin Tolerance Tests

The tail blood glucose levels were measured at 0, 10, 20, 30, 45, 60, 90, and 120 min postinjection with an Accu-Check glucometer (6870228; Roche), according to the Mouse Metabolic Phenotyping Center recommendations for data presentation (21). The insulin level was measured by an ELISA kit (90080; Chrystal Chem), and the glucose level was quantified with an Accu-Check glucometer.

Lipolysis and Lipogenesis Assays

For the lipolysis assay, the glycerol content was determined by absorbance at a wavelength of 540 nm using the Free Glycerol Determination Kit (FG0100; Sigma-Aldrich). Isoproterenol-stimulated lipolysis was corrected for basal lipolysis by subtracting the amount of glycerol release in the absence of isoproterenol (19). The lipogenesis was assayed as previously described (19). The incorporated radiolabeled glucose was measured using liquid scintillation counting and normalized by total lipid content.

Assessment of Metabolic Parameters

Serum levels of insulin and free fatty acids (FFA) were measured by ELISA kits (for insulin, from Millipore [EZRMI-13K], and for FFA, from Merck [MAK044]). Serum levels of triglyceride, total cholesterol, HDL (HDL-C) and LDL (LDL-C) cholesterol, and glucose were determined with a Hitachi 7080 Chemistry analyzer (Hitachi High-Tech Corporation, Tokyo, Japan). The serum triglyceride determination kit (TR0100; Sigma-Aldrich) was used for measurement of liver triglyceride levels. Total ketone levels were quantified with an enzyme colorimetric assay kit (415-73301 and 411-73401; FUJIFILM Wako Chemicals, Richmond, VA). Fatty acid oxidation was measured in fresh liver and white adipose tissue (WAT) homogenates using the previously established methods (19).

Castration, Ovariectomy, Testosterone Replacement, and Testosterone/Estrogen/Luteinizing Hormone Measurements

Detailed methods used in castration and ovariectomy (OVX) are described in the Supplementary Material. For the testosterone replacement, testosterone pellets (25 µg/day, SA-151; Innovative Research of America) or placebo pellets (25 µg/day, SC-111; Innovative Research of America) were subcutaneously implanted at the time of castration as previously described in detail (22). The serum levels of testosterone and estrogen were determined by ultra-high-performance liquid chromatography–tandem mass spectrometry coupled to an Agilent 6490 triple quadrupole with an Agilent 6490 triple quadrupole (QqQ) mass spectrometer using the detailed protocol described by McLeod et al. (23). The luteinizing hormone (LH) levels were detected with ELISA kits (E-EL-M3053; Elabscience). The quantification of LH levels was obtained by a microplate reader at 550 nm, with the wavelength correction set at 450 nm.

Flow Cytometry Analysis of Macrophages in Adipose Stromal Vascular Fraction and Recombinant Adenoviruses

The stromal vascular fraction (SVF) cells were suspended in FACS buffer with propidium iodide (P4864; Sigma-Aldrich) and analyzed with BD LSRFortessa Cell Analyzer (BD Biosciences). Data analysis was carried out with FlowJo software version X.0.7 (Tree Star, Ashland, OR).

For generation of recombinant adenoviruses (11) expressing MPK38 (wild-type [WT] and kinase dead [K40R]), pEBG-MPK38 plasmids (WT and K40R) were used as templates for PCR with primers (Supplementary Table 1) with use of Advantage HD polymerase mix (cat. no. 639241; Takara Bio Inc.).

Statistical Analysis

Values are shown as mean ± SEM unless otherwise indicated, and results are representative of at least three independent experiments. The statistical analyses were performed by one- or two-way ANOVA, followed by Tukey multiple comparison test, with use of GraphPad Prism 7.0 software (GraphPad Software).

Data and Resource Availability

The data sets generated or analyzed during the current study are available from the corresponding author upon reasonable request.

MPK38 Deficiency Causes Obesity; Impairments in Glucose, Lipid, and Energy Metabolism; and Inflammation in Aged Mature Adult Male, but Not Female, Mice

To determine the functions of MPK38 in vivo, we generated MPK38−/− mice by gene trap insertion (Supplementary Fig. 1A). When fed a standard diet, 6- to 7-month-old MPK38−/− male mice were visibly larger than their WT counterparts (Fig. 1A); however, these differences were not observed in MPK38−/− female mice. MPK38−/− male mice had higher liver, spleen, and epididymal fat masses than their age- and sex-matched WT counterparts (Fig. 1A and B). When fed a standard diet, MPK38−/− and WT male mice had similar body masses for ∼4 months, but obesity became apparent in the MPK38−/− male mice at ∼6 months of age, despite there being no statistically significant difference in food intake (Fig. 1C). Heterozygous MPK38+/− male mice demonstrated intermediate levels of body mass gain (Fig. 1C, upper). These results suggest that the obesity induced by MPK38 deficiency is age, sex, and gene dose dependent.

Figure 1

MPK38-null male mice display late-onset obesity, impaired glucose and lipid metabolism, and inflammation. A: Appearance (top left), ventral views (middle left), cross-sectional views using MRI (bottom left), and morphology of the fat pads and organs (right) of 7-month-old MPK38+/+, MPK38+/−, and MPK38−/− male mice fed a standard diet (n = 13–15 mice per group). B and C: Volumes of adipose tissue depots, determined using computed tomography (B), and body masses and food intake (C). Epi., epididymal; Ret., retroperitoneal; Sub., subcutaneous. D and E: GTT/ITT results (D), glucose area under the curve (AUC) during GTT (D, upper right), glucose area above the curve (AAC) during ITT (D, lower right), and blood glucose and insulin concentrations in the fed and fasting states (24 h) (E). n = 6 mice per group, *P < 0.05, **P < 0.01, ***P < 0.001 vs. fasted MPK38+/+ mice; #P < 0.05, ##P < 0.01 vs. fed MPK38+/+ mice. The statistical analyses were performed with two-way ANOVA. F: In vitro 14C-2-deoxy-glucose uptake (19) into epididymal WAT and muscle was measured in the presence or absence of 100 nmol/L human insulin (left). n = 6 mice per group. *P < 0.05, **P < 0.01, ***P < 0.001 vs. MPK38+/+ mice treated with insulin. #P < 0.05, ##P < 0.01 vs. untreated MPK38+/+ mice. The statistical analyses were performed with two-way ANOVA. IRS-PI3K signaling after in vivo insulin stimulation by injection into the caudal vena cava was assessed by immunoblotting (right). G: mRNA expression of gluconeogenic genes in the liver (G, upper) and the concentrations of circulating glucose (G, lower). H: The concentrations of circulating FFA, triglyceride, total cholesterol, HDL-C, and LDL-C. I: The lipogenic capacity of adipocytes (left) and the mRNA expression of lipogenic genes in epididymal WAT and liver (right). J: The size distribution of adipocytes (upper) and paraffin-embedded epididymal WAT sections stained with hematoxylin-eosin (lower). Scale bar, 100 μm. K: The circulating concentrations of proinflammatory cytokines. L: Flow cytometry analysis of macrophages in the adipose SVFs from 7-month-old MPK38+/+ and MPK38−/− male mice (left). The percentages of M1 and M2 cells are also shown in the graph (right). M1 surface markers, F4/80+CD11c+CD206; M2 surface markers, F4/80+CD11cCD206+. n = 6 mice per group, *P < 0.05, **P < 0.01, ***P < 0.001 vs. MPK38+/+ mice (B and GL). Data were analyzed with one-way ANOVA unless otherwise indicated. Quantitative real-time PCR was performed in duplicate and repeated at least two to three times with similar results (G and I). Detailed quantitative real-time PCR methods and primers (Supplementary Table 1) used in this study are described in the Supplementary Material.

Figure 1

MPK38-null male mice display late-onset obesity, impaired glucose and lipid metabolism, and inflammation. A: Appearance (top left), ventral views (middle left), cross-sectional views using MRI (bottom left), and morphology of the fat pads and organs (right) of 7-month-old MPK38+/+, MPK38+/−, and MPK38−/− male mice fed a standard diet (n = 13–15 mice per group). B and C: Volumes of adipose tissue depots, determined using computed tomography (B), and body masses and food intake (C). Epi., epididymal; Ret., retroperitoneal; Sub., subcutaneous. D and E: GTT/ITT results (D), glucose area under the curve (AUC) during GTT (D, upper right), glucose area above the curve (AAC) during ITT (D, lower right), and blood glucose and insulin concentrations in the fed and fasting states (24 h) (E). n = 6 mice per group, *P < 0.05, **P < 0.01, ***P < 0.001 vs. fasted MPK38+/+ mice; #P < 0.05, ##P < 0.01 vs. fed MPK38+/+ mice. The statistical analyses were performed with two-way ANOVA. F: In vitro 14C-2-deoxy-glucose uptake (19) into epididymal WAT and muscle was measured in the presence or absence of 100 nmol/L human insulin (left). n = 6 mice per group. *P < 0.05, **P < 0.01, ***P < 0.001 vs. MPK38+/+ mice treated with insulin. #P < 0.05, ##P < 0.01 vs. untreated MPK38+/+ mice. The statistical analyses were performed with two-way ANOVA. IRS-PI3K signaling after in vivo insulin stimulation by injection into the caudal vena cava was assessed by immunoblotting (right). G: mRNA expression of gluconeogenic genes in the liver (G, upper) and the concentrations of circulating glucose (G, lower). H: The concentrations of circulating FFA, triglyceride, total cholesterol, HDL-C, and LDL-C. I: The lipogenic capacity of adipocytes (left) and the mRNA expression of lipogenic genes in epididymal WAT and liver (right). J: The size distribution of adipocytes (upper) and paraffin-embedded epididymal WAT sections stained with hematoxylin-eosin (lower). Scale bar, 100 μm. K: The circulating concentrations of proinflammatory cytokines. L: Flow cytometry analysis of macrophages in the adipose SVFs from 7-month-old MPK38+/+ and MPK38−/− male mice (left). The percentages of M1 and M2 cells are also shown in the graph (right). M1 surface markers, F4/80+CD11c+CD206; M2 surface markers, F4/80+CD11cCD206+. n = 6 mice per group, *P < 0.05, **P < 0.01, ***P < 0.001 vs. MPK38+/+ mice (B and GL). Data were analyzed with one-way ANOVA unless otherwise indicated. Quantitative real-time PCR was performed in duplicate and repeated at least two to three times with similar results (G and I). Detailed quantitative real-time PCR methods and primers (Supplementary Table 1) used in this study are described in the Supplementary Material.

Close modal

Glucose (GTT) and insulin (ITT) tolerance tests revealed that MPK38−/− male mice were glucose intolerant and less sensitive to insulin (Fig. 1D). In addition, they displayed significantly higher ratios of circulating glucose and insulin in fasting than in fed conditions (Fig. 1E). Consistent with this, insulin-stimulated in vitro 2-deoxy-glucose uptake was significantly lower in the WAT and muscle of MPK38−/− male mice than in WT mice (Fig. 1F, left). Furthermore, MPK38 deficiency reduced the activation of the insulin receptor substrate (IRS)–phosphatidylinositol 3-kinase (PI3K) pathway and the expression of GLUT type 4 (GLUT4) and type 1 (GLUT1) in WAT and soleus muscle (Fig. 1F, right). In addition, the expression of mRNAs encoding gluconeogenic proteins, including the catalytic subunit of glucose 6-phosphatase (G6PC), PEPCK-1 (PCK1), and peroxisome proliferator–activated receptor γ coactivator 1-α (PGC1α), were much higher in the livers of MPK38−/− mice than in WT mice (Fig. 1G, upper). When combined with the high blood glucose concentration in MPK38−/− male mice (Fig. 1G, lower), these findings implicate MPK38 in the inhibition of hepatic gluconeogenesis.

Abnormal lipid accumulation in the liver (Supplementary Fig. 1B) and substantial increases in the concentrations of circulating FFA, triglycerides, total cholesterol, HDL-C, and LDL-C were identified in the MPK38−/− mice (Fig. 1H). MPK38−/− mice showed a higher rate of lipogenesis (Fig. 1I, left) and higher expression of mRNAs encoding lipogenic proteins, including fatty acid synthase (FAS), sterol CoA desaturase 1 (SCD1), and sterol regulatory element–binding transcription factor 1c (SREBP1c), than WT mice (Fig. 1I, right). Furthermore, there were high serum activities of AST and ALT, which reflect the fatty liver phenotype (Supplementary Fig. 1C). Consistent with these findings, the mean adipocyte size was higher in the MPK38−/− mice than in their heterozygous (MPK38+/−) or WT counterparts (Fig. 1J). In addition, the expression of mRNAs encoding adipogenic regulators, including C/EBPα, peroxisome proliferator–activated receptor γ (PPARγ), and fatty acid binding protein 4 (FABP4), was much higher in MPK38−/− mice (Supplementary Fig. 1D). MPK38−/− mice also showed higher levels of inflammation, evidenced by high circulating concentrations of proinflammatory cytokines, including tumor necrosis factor-α (TNF-α), interleukin (IL)-6, IL-1β, and MCP1, and larger numbers of M1 macrophages, demonstrated with flow cytometry (Fig. 1K and L).

With regard to energy metabolism (Supplementary Fig. 2), MPK38−/− mice showed lower energy expenditure, 24-h O2 consumption, CO2 production, and respiratory exchange ratio. Consistent with this, the expression of mRNAs encoding uncoupling protein 1 (UCP1), PGC1α, and muscle-type carnitine palmitoyltransferase 1 (mCPT1), which are important for thermogenesis (24,25), was considerably lower in MPK38−/− than in WT mice. Overall, these results suggest that MPK38 has favorable effects on metabolism that prevent obesity in aged mature adult male mice.

MPK38 Deficiency Exacerbates Diet-Induced Obesity and Metabolic Disorders in Aged Mature Adult Male, but Not Female, Mice

We next determined whether MPK38 protects against high-fat diet (HFD)-induced metabolic disturbances specifically in male mice using 7-month-old WT, MPK38+/−, and MPK38−/− mice fed chow or an HFD. Compared with WT mice, MPK38−/− male, but not female, mice became more obese and developed dysregulated glucose and lipid metabolism, as well as developing higher levels of inflammation, under HFD-fed conditions (Fig. 2).

Figure 2

MPK38 deficiency exacerbates diet-induced metabolic disturbances in aged mature adult male mice. A: Appearance (top), body masses (middle), and morphology of the fat depots and organs (bottom) of 7-month-old MPK38+/+, MPK38+/−, and MPK38−/− male mice fed an HFD (n = 10–13 mice per group). B: Serum concentrations of FFA, triglyceride, AST, ALT, glucose, insulin, leptin, and adiponectin. C: Representative images of paraffin-embedded liver and epididymal WAT sections stained with hematoxylin-eosin (left), and liver triglyceride content (right). Scale bar, 100 μm. D: mRNA expression of adipogenic regulators in epididymal WAT. E and F: Blood glucose and insulin concentrations in the fed and fasted (24 h) states (E), the results of GTTs/ITTs (F), glucose area under the curve (AUC) during GTT (F, second), and glucose area above the curve (AAC) during ITT (F, fourth). *P < 0.05, **P < 0.01, ***P < 0.001 vs. fasted controls. #P < 0.05 vs. fed controls. The statistical analyses were performed with two-way ANOVA. G: In vitro 14C-2-deoxyglucose uptake into epididymal WAT and muscle (left). **P < 0.01, ***P < 0.001 vs. controls treated with insulin. ##P < 0.01 vs. untreated controls. The statistical analyses were performed using two-way ANOVA. IRS-PI3K signaling after in vivo insulin stimulation by injection into the caudal vena cava was assessed by immunoblotting (right, n = 2 mice per group). H: mRNA expression of gluconeogenic genes in the liver. I: mRNA expression of lipogenic genes in epididymal WAT (upper) and the lipogenic capacity of the adipocytes (lower). J: mRNA expression of fatty acid oxidative genes in epididymal WAT (upper) and the isoproterenol-stimulated lipolytic response of isolated adipocytes (lower). K: Measurement of hepatic β-oxidation with use of 14C-labeled palmitate. **P < 0.01, ***P < 0.001 vs. HFD-fed MPK38+/+ male mice, #P < 0.05 vs. MPK38+/+ male mice fed chow, determined by two-way ANOVA. L: The circulating concentrations of total cholesterol, HDL-C, and LDL-C. MO: The total ketone body concentration in fed and fasted (24 h) blood (M). Phosphorylation levels of S6 (Ser240/244) in liver lysates from ad libitum–fed, fasted (24 h), and refed (2 h) MPK38+/+, MPK38+/−, and MPK38−/− male mice fed an HFD (N, left); mTORC1 signaling pathway activation in liver lysates (N, right); and mRNA expression of ketogenic genes (Pparα, Cpt1, and 3-hydroxy-3-methylglutaryl-CoA synthase 2 [Hmgcs2]) in the liver (O). PEPCK 1 (Pck1), which is not a target of PPARα, was used as a reference gene. n = 6 mice per group. *P < 0.05, **P < 0.01, ***P < 0.001 vs. fasted control, and #P < 0.05, ##P < 0.01 vs. fed control, determined by two-way ANOVA. P: The circulating concentrations of proinflammatory cytokines. Q: Flow cytometry analysis of macrophages in adipose SVFs from 7-month-old MPK38+/+ and MPK38−/− male mice fed an HFD (upper). The percentages of M1 and M2 cells are also shown in the graph (lower). M1 surface markers, F4/80+CD11c+CD206; M2 surface markers, F4/80+CD11cCD206+. n = 6 mice per group. *P < 0.05, **P < 0.01, ***P < 0.001 vs. control (AD, HJ, L, P, and Q). The controls were HFD-fed MPK38+/+ male mice (7 months of age). Data were analyzed with one-way ANOVA unless otherwise indicated. Quantitative real-time PCR was performed in duplicate and repeated at least two to three times with similar results (D, HJ, and O).

Figure 2

MPK38 deficiency exacerbates diet-induced metabolic disturbances in aged mature adult male mice. A: Appearance (top), body masses (middle), and morphology of the fat depots and organs (bottom) of 7-month-old MPK38+/+, MPK38+/−, and MPK38−/− male mice fed an HFD (n = 10–13 mice per group). B: Serum concentrations of FFA, triglyceride, AST, ALT, glucose, insulin, leptin, and adiponectin. C: Representative images of paraffin-embedded liver and epididymal WAT sections stained with hematoxylin-eosin (left), and liver triglyceride content (right). Scale bar, 100 μm. D: mRNA expression of adipogenic regulators in epididymal WAT. E and F: Blood glucose and insulin concentrations in the fed and fasted (24 h) states (E), the results of GTTs/ITTs (F), glucose area under the curve (AUC) during GTT (F, second), and glucose area above the curve (AAC) during ITT (F, fourth). *P < 0.05, **P < 0.01, ***P < 0.001 vs. fasted controls. #P < 0.05 vs. fed controls. The statistical analyses were performed with two-way ANOVA. G: In vitro 14C-2-deoxyglucose uptake into epididymal WAT and muscle (left). **P < 0.01, ***P < 0.001 vs. controls treated with insulin. ##P < 0.01 vs. untreated controls. The statistical analyses were performed using two-way ANOVA. IRS-PI3K signaling after in vivo insulin stimulation by injection into the caudal vena cava was assessed by immunoblotting (right, n = 2 mice per group). H: mRNA expression of gluconeogenic genes in the liver. I: mRNA expression of lipogenic genes in epididymal WAT (upper) and the lipogenic capacity of the adipocytes (lower). J: mRNA expression of fatty acid oxidative genes in epididymal WAT (upper) and the isoproterenol-stimulated lipolytic response of isolated adipocytes (lower). K: Measurement of hepatic β-oxidation with use of 14C-labeled palmitate. **P < 0.01, ***P < 0.001 vs. HFD-fed MPK38+/+ male mice, #P < 0.05 vs. MPK38+/+ male mice fed chow, determined by two-way ANOVA. L: The circulating concentrations of total cholesterol, HDL-C, and LDL-C. MO: The total ketone body concentration in fed and fasted (24 h) blood (M). Phosphorylation levels of S6 (Ser240/244) in liver lysates from ad libitum–fed, fasted (24 h), and refed (2 h) MPK38+/+, MPK38+/−, and MPK38−/− male mice fed an HFD (N, left); mTORC1 signaling pathway activation in liver lysates (N, right); and mRNA expression of ketogenic genes (Pparα, Cpt1, and 3-hydroxy-3-methylglutaryl-CoA synthase 2 [Hmgcs2]) in the liver (O). PEPCK 1 (Pck1), which is not a target of PPARα, was used as a reference gene. n = 6 mice per group. *P < 0.05, **P < 0.01, ***P < 0.001 vs. fasted control, and #P < 0.05, ##P < 0.01 vs. fed control, determined by two-way ANOVA. P: The circulating concentrations of proinflammatory cytokines. Q: Flow cytometry analysis of macrophages in adipose SVFs from 7-month-old MPK38+/+ and MPK38−/− male mice fed an HFD (upper). The percentages of M1 and M2 cells are also shown in the graph (lower). M1 surface markers, F4/80+CD11c+CD206; M2 surface markers, F4/80+CD11cCD206+. n = 6 mice per group. *P < 0.05, **P < 0.01, ***P < 0.001 vs. control (AD, HJ, L, P, and Q). The controls were HFD-fed MPK38+/+ male mice (7 months of age). Data were analyzed with one-way ANOVA unless otherwise indicated. Quantitative real-time PCR was performed in duplicate and repeated at least two to three times with similar results (D, HJ, and O).

Close modal

HFD-fed MPK38−/− male mice displayed much higher ratios of circulating glucose and insulin in fasting than in fed conditions (Fig. 2E), along with glucose intolerance and insulin resistance (Fig. 2F). Consistent with these observations, insulin-stimulated in vitro 2-deoxy-glucose uptake was significantly lower in the WAT and muscle of HFD-fed MPK38−/− male mice (Fig. 2G). This contention was supported by the finding that HFD-fed MPK38−/− male mice had higher expression levels of mRNAs encoding gluconeogenic proteins, including G6PC, PCK1, and PGC1α, alongside their higher blood glucose concentrations, than HFD-fed WT male mice (Fig. 2B and H).

HFD-fed MPK38−/− males also demonstrated a higher rate of lipogenesis and higher expression levels of mRNAs encoding lipogenic proteins, including FAS, SCD1, and SREBP1c, in adipose tissue in comparison with HFD-fed WT males (Fig. 2I). In addition, HFD-fed MPK38−/− males demonstrated higher levels of expression of key adipogenic regulators, including C/EBPα, PPARγ, and FABP4 (Fig. 2D), and greater inflammation (Fig. 2P and Q). However, these adverse effects of MPK38 deficiency in HFD-fed mice were not observed in females (Supplementary Fig. 3). These results indicate that MPK38 protects against diet-induced metabolic abnormalities in aged mature adult male, but not female, mice.

Adenoviral Restoration of MPK38 Expression Ameliorates the Diet-Induced Metabolic Disorders in Aged Mature Adult Male, but Not Female, Mice

The activation of ASK1/TGF-β/p53 signaling by MPK38 was significantly less in the WAT of 7-month-old MPK38−/− male mice than in their age- and sex-matched WT counterparts (Fig. 3A). Similarly, lower levels of ASK1/TGF-β/p53 signaling, and MPK38 expression and kinase activity, were also identified in ob/ob and HFD-fed males (19,20) (Supplementary Fig. 4).

Figure 3

MPK38 ameliorates obesity-associated disturbances of glucose metabolism in male mice with diet-induced obesity. A: Downregulation of ASK1/TGF-β/p53 signaling in epididymal WAT from 7-month-old MPK38−/− male mice. P-, phosphorylated. B: Size distribution of adipocytes (upper) (19) and representative images of paraffin-embedded epididymal WAT sections stained with hematoxylin-eosin (lower) in HFD-fed C57BL/6N male mice (6–7 months of age; 8–12 days after adenoviral infection). Scale bar, 100 μm. C: mRNA expression of adipogenic genes in epididymal WAT. D and E: Blood glucose (D, left) and insulin (E, left) concentrations in the fed and fasted (16 h) states, the results of GTT and ITT (D and E; middle), glucose area under the curve (AUC) during GTT (D, right), and glucose area above the curve (AAC) during ITT (E, right). n = 6 mice per group. **P < 0.01, ***P < 0.001 vs. fasted controls; ##P < 0.01, ###P < 0.001 vs. fed controls. The statistical analyses were performed with two-way ANOVA. F: In vitro 14C-2-deoxyglucose uptake into epididymal WAT and muscle (left). n = 6 mice per group. ***P < 0.001 vs. controls treated with insulin. The statistical analyses were performed using two-way ANOVA. IRS-PI3K signaling after in vivo insulin stimulation by injection into the caudal vena cava was assessed by immunoblotting (right, n = 3 mice per group). G: The circulating concentration of glucose and mRNA expression of gluconeogenic genes in the liver. n = 6 mice per group, *P < 0.05, **P < 0.01, ***P < 0.001 vs. controls (B, C, and G). The controls were uninfected HFD-fed C57BL/6N male mice (6–7 months of age). Data were analyzed with one-way ANOVA unless otherwise indicated. Quantitative real-time PCR was performed in duplicate and repeated at least two to three times with similar results (C and G). All adenovirus-injected mice used in this study were analyzed 8–12 days after injection (Supplementary Fig. 5).

Figure 3

MPK38 ameliorates obesity-associated disturbances of glucose metabolism in male mice with diet-induced obesity. A: Downregulation of ASK1/TGF-β/p53 signaling in epididymal WAT from 7-month-old MPK38−/− male mice. P-, phosphorylated. B: Size distribution of adipocytes (upper) (19) and representative images of paraffin-embedded epididymal WAT sections stained with hematoxylin-eosin (lower) in HFD-fed C57BL/6N male mice (6–7 months of age; 8–12 days after adenoviral infection). Scale bar, 100 μm. C: mRNA expression of adipogenic genes in epididymal WAT. D and E: Blood glucose (D, left) and insulin (E, left) concentrations in the fed and fasted (16 h) states, the results of GTT and ITT (D and E; middle), glucose area under the curve (AUC) during GTT (D, right), and glucose area above the curve (AAC) during ITT (E, right). n = 6 mice per group. **P < 0.01, ***P < 0.001 vs. fasted controls; ##P < 0.01, ###P < 0.001 vs. fed controls. The statistical analyses were performed with two-way ANOVA. F: In vitro 14C-2-deoxyglucose uptake into epididymal WAT and muscle (left). n = 6 mice per group. ***P < 0.001 vs. controls treated with insulin. The statistical analyses were performed using two-way ANOVA. IRS-PI3K signaling after in vivo insulin stimulation by injection into the caudal vena cava was assessed by immunoblotting (right, n = 3 mice per group). G: The circulating concentration of glucose and mRNA expression of gluconeogenic genes in the liver. n = 6 mice per group, *P < 0.05, **P < 0.01, ***P < 0.001 vs. controls (B, C, and G). The controls were uninfected HFD-fed C57BL/6N male mice (6–7 months of age). Data were analyzed with one-way ANOVA unless otherwise indicated. Quantitative real-time PCR was performed in duplicate and repeated at least two to three times with similar results (C and G). All adenovirus-injected mice used in this study were analyzed 8–12 days after injection (Supplementary Fig. 5).

Close modal

To further investigate the sex-specific role of MPK38 in the amelioration of the metabolic disturbances associated with obesity, we restored WT MPK38 expression in 7-month-old HFD-fed male mice (Supplementary Fig. 5). This caused a significant reduction in adipocyte size (Fig. 3B) and in the expression of mRNAs encoding key adipogenic proteins (C/EBPα, PPARγ, and FABP4) (Fig. 3C). These findings indicate that MPK38 inhibits adipogenesis, leading to an increase in insulin sensitivity.

HFD-fed male mice infected with adenovirus expressing MPK38 (Ad-MPK38) displayed lower circulating glucose and insulin concentrations, and higher glucose tolerance and insulin sensitivity, than uninfected HFD-fed male mice (Fig. 3D and E). Ad-MPK38 infection increased 2-deoxyglucose uptake in both epididymal WAT and muscle (Fig. 3F, left), which may be explained by upregulation of the IRS-PI3K pathway and higher expression of GLUTs (Fig. 3F, right). In addition, the reduction in blood glucose may be explained by lower mRNA expression of hepatic gluconeogenic proteins (G6PC, PCK1, and PGC1α) (Fig. 3G).

Ad-MPK38 infection also reduced circulating FFA concentrations (Fig. 4A, left) and the expression of mRNAs encoding adipose lipogenic proteins (FAS, SCD1, and SREBP1c) (Fig. 4A, right) and increased isoproterenol-stimulated lipolysis (Fig. 4B, left) and the expression of mRNAs encoding adipose fatty acid oxidative proteins (PPARα, CPT1, and ACO) (Fig. 4B, right). Thus, Ad-MPK38 infection upregulated fatty acid utilization by increasing fatty acid oxidation via the mitochondrial and peroxisomal β-oxidation pathways (Fig. 4C). HFD-fed male mice infected with Ad-MPK38 also had lower circulating concentrations of triglycerides, total cholesterol, HDL-C, and LDL-C (Fig. 4D), as well as much lower hepatic lipid accumulation (Fig. 4E). These results suggest a positive role for MPK38 in lipid metabolism. Furthermore, Ad-MPK38 infection promoted ketone body production (Fig. 4F), increased the expression of mRNAs encoding ketogenic genes under fasting conditions (Fig. 4G), and reduced mTORC1 signaling (Fig. 4H). However, the beneficial effects of MPK38 restoration in 7-month-old HFD-fed male mice were not observed in age-matched female mice fed an HFD (Supplementary Fig. 6), indicating that the protective effect of MPK38 is sex specific.

Figure 4

MPK38 ameliorates obesity-associated disturbances of lipid metabolism in male mice with diet-induced obesity. A: The circulating concentration of FFA and mRNA expression of lipogenic genes in epididymal WAT. B: The isoproterenol-stimulated lipolytic response in isolated adipocytes (left) and mRNA expression of fatty acid oxidative genes in epididymal WAT (right). C: Measurement of hepatic β-oxidation with 14C-labeled palmitate. D: The circulating concentrations of triglyceride, total cholesterol, HDL-C, and LDL-C. E: Representative images of paraffin-embedded liver sections stained with hematoxylin-eosin (n = 6–8 mice per group). Scale bar, 100 μm. F and G: The total ketone body concentration in fed and fasted (24 h) blood (F) and mRNA expression of ketogenic genes in the liver (G). n = 6 mice per group. ***P < 0.001 vs. fasted controls. The statistical analyses were performed with two-way ANOVA. H: Phosphorylation levels of S6 (Ser240/244) in liver lysates from uninfected and infected HFD-fed C57BL/6N male mice fed ad libitum, which were fasted for 24 h or refed for 2 h (left). mTORC1 signaling in liver lysates (right). n = 6 mice per group. *P < 0.05, **P < 0.01, ***P < 0.001 vs. controls (AD). The controls were uninfected HFD-fed C57BL/6N male mice (6–7 months of age). Data were analyzed with one-way ANOVA unless otherwise indicated. Quantitative real-time PCR was performed in duplicate and repeated at least two to three times with similar results (A, B, and G). All adenovirus-injected mice used in this study were analyzed 8–12 days after injection (Supplementary Fig. 5). hr, hour; P-, phosphorylated.

Figure 4

MPK38 ameliorates obesity-associated disturbances of lipid metabolism in male mice with diet-induced obesity. A: The circulating concentration of FFA and mRNA expression of lipogenic genes in epididymal WAT. B: The isoproterenol-stimulated lipolytic response in isolated adipocytes (left) and mRNA expression of fatty acid oxidative genes in epididymal WAT (right). C: Measurement of hepatic β-oxidation with 14C-labeled palmitate. D: The circulating concentrations of triglyceride, total cholesterol, HDL-C, and LDL-C. E: Representative images of paraffin-embedded liver sections stained with hematoxylin-eosin (n = 6–8 mice per group). Scale bar, 100 μm. F and G: The total ketone body concentration in fed and fasted (24 h) blood (F) and mRNA expression of ketogenic genes in the liver (G). n = 6 mice per group. ***P < 0.001 vs. fasted controls. The statistical analyses were performed with two-way ANOVA. H: Phosphorylation levels of S6 (Ser240/244) in liver lysates from uninfected and infected HFD-fed C57BL/6N male mice fed ad libitum, which were fasted for 24 h or refed for 2 h (left). mTORC1 signaling in liver lysates (right). n = 6 mice per group. *P < 0.05, **P < 0.01, ***P < 0.001 vs. controls (AD). The controls were uninfected HFD-fed C57BL/6N male mice (6–7 months of age). Data were analyzed with one-way ANOVA unless otherwise indicated. Quantitative real-time PCR was performed in duplicate and repeated at least two to three times with similar results (A, B, and G). All adenovirus-injected mice used in this study were analyzed 8–12 days after injection (Supplementary Fig. 5). hr, hour; P-, phosphorylated.

Close modal

MPK38 Mediates Neutering-Induced Metabolic Alterations in Aged Mature Adult Male, but Not Female, Mice

To investigate the sexual dimorphism of late-onset obesity in MPK38−/− mice, we analyzed the glucose and lipid metabolism of age- and sex-matched WT (MPK38+/+) and MPK38−/− mice 6 weeks after neutering (castration/OVX) or sham surgery because the neutering of mice affects glucose and lipid metabolism (26). Castration of 7-month-old MPK38−/− male mice had little or no effect on the size distribution of adipocytes, according to histological analysis of WAT (Fig. 5A), or on the expression of mRNAs encoding key adipogenic regulators (C/EBPα, PPARγ, and FABP4) in WAT (Fig. 5B).

Figure 5

Lack of castration-induced changes in glucose metabolism in aged mature adult MPK38−/− males. A: Size distribution analysis of adipocytes (upper) and representative images of paraffin-embedded epididymal WAT sections stained with hematoxylin-eosin (lower) in castrated MPK38+/+ and MPK38−/− male mice and sham controls (4.5 and 7 months of age). n = 6 mice per group. Scale bar, 100 μm. B: mRNA expression of adipogenic genes. C and D: The results of GTTs/ITTs. n = 6 mice per group. *P < 0.05, **P < 0.01, ***P < 0.001 vs. MPK38+/+ sham controls; #P < 0.05, ##P < 0.01, ###P < 0.001 vs. MPK38−/− sham controls (C and D). E: In vitro 14C-2-deoxyglucose uptake into epididymal WAT and muscle (left). n = 6 mice per group. *P < 0.05, **P < 0.01 vs. sham controls treated with insulin. The statistical analyses were performed using two-way ANOVA. IRS-PI3K signaling was assessed with immunoblotting after in vivo insulin stimulation by injection into the caudal vena cava (right, n = 2 mice per group). F: mRNA expression of hepatic gluconeogenic genes. n = 6 mice per group. *P < 0.05, **P < 0.01, ***P < 0.001 vs. MPK38+/+ sham controls (AF); #P < 0.05, ##P < 0.01, ###P < 0.001 vs. MPK38−/− sham controls (C and D). The statistical analyses were performed using two-way ANOVA. Quantitative real-time PCR was performed in duplicate and repeated at least two to three times with similar results (B and F). N.S., no significant difference; P-, phosphorylated.

Figure 5

Lack of castration-induced changes in glucose metabolism in aged mature adult MPK38−/− males. A: Size distribution analysis of adipocytes (upper) and representative images of paraffin-embedded epididymal WAT sections stained with hematoxylin-eosin (lower) in castrated MPK38+/+ and MPK38−/− male mice and sham controls (4.5 and 7 months of age). n = 6 mice per group. Scale bar, 100 μm. B: mRNA expression of adipogenic genes. C and D: The results of GTTs/ITTs. n = 6 mice per group. *P < 0.05, **P < 0.01, ***P < 0.001 vs. MPK38+/+ sham controls; #P < 0.05, ##P < 0.01, ###P < 0.001 vs. MPK38−/− sham controls (C and D). E: In vitro 14C-2-deoxyglucose uptake into epididymal WAT and muscle (left). n = 6 mice per group. *P < 0.05, **P < 0.01 vs. sham controls treated with insulin. The statistical analyses were performed using two-way ANOVA. IRS-PI3K signaling was assessed with immunoblotting after in vivo insulin stimulation by injection into the caudal vena cava (right, n = 2 mice per group). F: mRNA expression of hepatic gluconeogenic genes. n = 6 mice per group. *P < 0.05, **P < 0.01, ***P < 0.001 vs. MPK38+/+ sham controls (AF); #P < 0.05, ##P < 0.01, ###P < 0.001 vs. MPK38−/− sham controls (C and D). The statistical analyses were performed using two-way ANOVA. Quantitative real-time PCR was performed in duplicate and repeated at least two to three times with similar results (B and F). N.S., no significant difference; P-, phosphorylated.

Close modal

The castration of WT male mice increased glucose tolerance and insulin sensitivity versus sham controls (Fig. 5C and D) and reduced their circulating insulin and glucose concentrations under fasting conditions (Supplementary Fig. 7A and B). However, these effects were not observed in 7-month-old MPK38−/− male mice when they were castrated. In addition, castration did not cause changes in insulin-stimulated in vitro 2-deoxyglucose uptake (Fig. 5E, left), IRS-PI3K pathway activation (Fig. 5E, right), or the expression of mRNAs encoding hepatic gluconeogenic genes (G6pc, Pck1, and Pgc1α) (Fig. 5F) in these mice.

The castrated WT male mice demonstrated more rapid lipogenesis than sham controls, which was accompanied by higher mRNA expression of hepatic and adipose lipogenic genes, including Fas, Scd1, and Srebp1c, but lower concentrations of circulating FFA (Fig. 6A and Supplementary Fig. 7C). However, castration of 7-month-old MPK38−/− male mice did not induce these effects; it had no effect on liver triglyceride storage or circulating total cholesterol, HDL-C, or LDL-C concentrations (Supplementary Fig. 7C and D).

Figure 6

Lack of castration-induced changes in lipid metabolism in aged mature adult MPK38−/− males. A: mRNA expression of lipogenic genes in liver (left) and the lipogenic capacity of hepatocytes (right). B: The mRNA expression of lipolytic genes in epididymal WAT. C: Measurement of hepatic β-oxidation with 14C-labeled palmitate. D: mRNA expression of fatty acid oxidative genes in epididymal WAT (left), circulating triglyceride concentration (middle), and the isoproterenol-stimulated lipolytic response in isolated adipocytes (right). E and F: The total ketone body concentrations in fed and fasted (24 h) blood (E) and mRNA expression of ketogenic genes in livers (F). n = 6 mice per group, *P < 0.05, **P < 0.01 vs. fasted sham controls (E). The statistical analyses were performed using two-way ANOVA. N.S., no significant difference. n = 6 mice per group. *P < 0.05, **P < 0.01, ***P < 0.001 vs. sham controls (AD). Quantitative real-time PCR was performed in duplicate and repeated at least two to three times with similar results (A, B, D, and F). hr, hours.

Figure 6

Lack of castration-induced changes in lipid metabolism in aged mature adult MPK38−/− males. A: mRNA expression of lipogenic genes in liver (left) and the lipogenic capacity of hepatocytes (right). B: The mRNA expression of lipolytic genes in epididymal WAT. C: Measurement of hepatic β-oxidation with 14C-labeled palmitate. D: mRNA expression of fatty acid oxidative genes in epididymal WAT (left), circulating triglyceride concentration (middle), and the isoproterenol-stimulated lipolytic response in isolated adipocytes (right). E and F: The total ketone body concentrations in fed and fasted (24 h) blood (E) and mRNA expression of ketogenic genes in livers (F). n = 6 mice per group, *P < 0.05, **P < 0.01 vs. fasted sham controls (E). The statistical analyses were performed using two-way ANOVA. N.S., no significant difference. n = 6 mice per group. *P < 0.05, **P < 0.01, ***P < 0.001 vs. sham controls (AD). Quantitative real-time PCR was performed in duplicate and repeated at least two to three times with similar results (A, B, D, and F). hr, hours.

Close modal

Furthermore, the effects of castration on the mRNA expression of lipolytic genes, such as hormone-sensitive lipase (HSL), adipose triglyceride lipase (ATGL), and β-3 adrenergic receptor (ADRB3) (Fig. 6B); fatty acid utilization (Fig. 6C); the mRNA expression of key genes involved in fatty acid oxidation, such as PPARα, CPT1, and ACO (Fig. 6D, left); isoproterenol-stimulated lipolysis (Fig. 6D, right); liver lipid and triglyceride content (Supplementary Fig. 7C and E); and circulating triglyceride concentrations (Fig. 6D, middle) were not found in 7-month-old MPK38−/− male mice. In addition, the castration of WT male mice markedly increased ketone body production and ketogenic gene expression under fasting conditions versus sham controls, but these effects were not induced by the castration of 7-month-old MPK38−/− male mice (Fig. 6E and F). As expected, castrated MPK38−/− male mice did not show changes in the phosphorylation of S6 at Ser240/244 in response to fasting or in mTORC1 signaling pathway activation (Supplementary Fig. 7F). All these effects were further confirmed by testosterone replacement exerting opposite effects (Supplementary Fig. 8). Moreover, the phenotype of castrated 7-month-old MPK38−/− male mice was not shared by age-matched OVX MPK38−/− female mice (Supplementary Fig. 9). Taken together, these findings suggest an age-specific role for MPK38 in castration, but not OVX-induced alterations, in the glucose and lipid metabolism of mice.

MPK38 Is Essential for Testosterone Production in Aged Mature Adult Male Mice

Because AMPK regulates steroid hormone synthesis (27,28), we performed liquid chromatography–tandem mass spectrometry to quantify serum testosterone and estrogen concentrations in sham-operated and neutered WT and MPK38−/− mice. Seven-month-old uncastrated MPK38−/− males exhibited serum testosterone concentrations similar to those of age-matched castrated WT males (Fig. 7A, left), but there was no difference between the serum estrogen concentrations of WT and MPK38−/− males of this age (Fig. 7A, right). By contrast, no equivalent effect was identified in non-OVX MPK38−/− females of the same age with regard to serum estrogen concentrations (Fig. 7B, left). As expected, OVX also had no effect on the serum testosterone concentrations of the female mice (Fig. 7B, right). All of these findings indicate that MPK38 may play a key role in testosterone production in 7-month-old male mice.

Figure 7

A role for MPK38 in testosterone production in aged mature adult male mice. A and B: The circulating concentrations of testosterone and estrogen in castrated or OVX MPK38+/+ and MPK38−/− mice (4.5 and 7 months of age) were quantified with liquid chromatography–tandem mass spectrometry. n = 6 mice per group. **P < 0.01, ***P < 0.001 vs. sham controls. CF: mRNA expression of genes involved in steroidogenesis in the testes (C and D) and ovaries (E and F) of MPK38+/+ and MPK38−/− mice fed a standard diet (4.5 and 7 months of age). n = 6 mice per group. *P < 0.05, **P < 0.01 vs. MPK38+/+ controls. N.S., no significant difference. Quantitative real-time PCR was performed in duplicate and repeated at least two to three times with similar results (CF).

Figure 7

A role for MPK38 in testosterone production in aged mature adult male mice. A and B: The circulating concentrations of testosterone and estrogen in castrated or OVX MPK38+/+ and MPK38−/− mice (4.5 and 7 months of age) were quantified with liquid chromatography–tandem mass spectrometry. n = 6 mice per group. **P < 0.01, ***P < 0.001 vs. sham controls. CF: mRNA expression of genes involved in steroidogenesis in the testes (C and D) and ovaries (E and F) of MPK38+/+ and MPK38−/− mice fed a standard diet (4.5 and 7 months of age). n = 6 mice per group. *P < 0.05, **P < 0.01 vs. MPK38+/+ controls. N.S., no significant difference. Quantitative real-time PCR was performed in duplicate and repeated at least two to three times with similar results (CF).

Close modal

We next determined whether alterations in steroidogenic gene expression are involved in the regulation of testosterone production by MPK38. The expression of 10 genes (29) was measured by quantitative PCR with use of RNA extracted from the testes of 7-month-old WT and MPK38−/− male mice that had been fed a standard diet: steroidogenic acute regulatory protein (Star) and scavenger receptor b1 (Scarb1), which are implicated in cholesterol transport, and transcriptional activators (Nr4a1, Nr4a3, Crebl2, Cited4, and cJun), a repressor of steroidogenesis (c-Fos), and protein phosphatase methylesterase 1 (Ppme1) and cyclin-dependent kinase 12 (Cdk12), which were used as reference genes. As expected, the expression of Star, Scarb1, and the five transcriptional activators was lower, but that of c-Fos was higher, in 7-month-old MPK38−/− male mice than in age-matched WT controls, whereas the expression of Ppme1 and Cdk12 was not affected by genotype (Fig. 7C). However, these differences in expression were not identified between 4.5-month-old MPK38−/− and WT males (Fig. 7D). Furthermore, equivalent differences were not found between the ovaries of WT and MPK38−/− females, regardless of age, suggesting that MPK38 plays no role in estrogen production in female mice (Fig. 7E and F). Similar results were also obtained in the analysis of other steroidogenic genes (30,31) including cytochrome P450 family 11 subfamily A member 1 (Cyp11a1), cytochrome P450 family 17 subfamily A member 1 (Cyp17a1), and 17β-hydroxysteroid dehydrogenase (17β-hsd), that were important for steroid hormone synthesis (Supplementary Fig. 10). When taken together, these findings suggest that MPK38 contributes to testosterone production in aged mature adult males but not estrogen production in females of the same age.

The Stability of MPK38 Is Increased by Testosterone but Reduced by Estrogen in Mice

To determine how sex hormones, such as testosterone and estrogen, regulate the kinase activity and expression of MPK38, we first performed immunoblot analysis of dihydrotestosterone (DHT)- and estradiol (E2)-treated adipocytes from 4.5-month-old WT male and female mice. DHT increased the stability of MPK38, whereas MPK38 stability was reduced by E2 treatment (Fig. 8A). However, exposure of the cells to MG132, a proteasomal inhibitor, increased the stability of MPK38 versus untreated controls (Fig. 8A), which implies that the proteasome pathway plays a role in the degradation of MPK38 (20). Consistent with this, DHT reduced, but E2 increased, the endogenous ubiquitination of MPK38, regardless of sex (Fig. 8B). We also determined whether DHT and E2 have effects on MPK38 degradation through MDM2 because MPK38 physically interacts with MDM2 (32). Compared with control cells that were not treated with DHT or E2, cells treated with DHT showed lower levels of endogenous MPK38-MDM2 complex formation, whereas those treated with E2 showed higher levels of complex formation, whether the cells were derived from male or female mice (Fig. 8C). To further dissect the mechanism whereby testosterone increases the stability of MPK38, we determined the effects of DHT on complex formation by MPK38 and ZPR9 or TRX because ZPR9 and Trx act as a stabilizer and destabilizer of MPK38, respectively (20,32). As expected, treatment with DHT significantly increased endogenous MPK38-ZPR9 complex formation but reduced MPK38-TRX complex formation in both male and female mice (Fig. 8D). However, E2 treatment had the opposite effect on complex formation. We also determined whether DHT and E2 influence the ASK1/TGF-β/p53 signaling induced by MPK38. Compared with untreated control cells, DHT treatment resulted in a significant increase in ASK1/TGF-β/p53 signaling, whereas E2 treatment had the opposite effect, in both male and female mice (Fig. 8E). Similar effects of DHT and E2 on lipid metabolism and MPK38 kinase activity were also observed in hepatocytes, suggesting that MPK38 deficiency equivalently affects WAT and liver in mice (Supplementary Fig. 11). These data indicate that testosterone positively regulates, and estrogen negatively regulates, MPK38-dependent ASK1, TGF-β, and p53 signaling by differentially regulating the stability of MPK38 in both sexes.

Figure 8

Testosterone increases and estrogen reduces the stability and activity of MPK38. A: MPK38 protein stability was assessed by immunoblot analysis with use of an anti‐MPK38 antibody. Primary adipocytes from male and female mice were treated with (+) or without (−) DHT or E2 (10 nmol/L each) for 24 h. The time intervals quoted are the number of minutes after treatment with 20 μg/mL cycloheximide (CHX) alone or with 10 μmol/L MG132. B: The ubiquitination of endogenous MPK38 was assessed in adipocytes from male and female mice treated with DHT, E2, or vehicle. C: Adipocyte cell lysates from male and female mice were treated with DHT, E2, or vehicle and then immunoprecipitated with an anti-MPK38 antibody (IP: α-MPK38), followed by immunoblotting with an anti‐MDM2 antibody for assessment of endogenous MPK38‐MDM2 complex formation. D: Adipocytes from male and female mice treated with DHT, E2, or vehicle were immunoprecipitated with an anti-MPK38 antibody, and the endogenous complex formation between MPK38 and ZPR9 (left) or TRX (right) was assessed by immunoblot analysis with use of anti-ZPR9 and anti-TRX antibodies, respectively. E: MPK38-dependent ASK1/TGF-β/p53 signaling in adipocytes from male and female mice treated with DHT, E2, or vehicle in the presence or absence of H2O2 (2 mmol/L, 30 min), TGF‐β1 (100 pmol/L, 20 h), and 5-fluorouracil (5FU) (0.38 mmol/L, 30 h) was determined by immunoblotting with the indicated antibodies. In all experiments that were performed, 4.5-month-old C57BL/6N male and female mice were used. WB, Western blot.

Figure 8

Testosterone increases and estrogen reduces the stability and activity of MPK38. A: MPK38 protein stability was assessed by immunoblot analysis with use of an anti‐MPK38 antibody. Primary adipocytes from male and female mice were treated with (+) or without (−) DHT or E2 (10 nmol/L each) for 24 h. The time intervals quoted are the number of minutes after treatment with 20 μg/mL cycloheximide (CHX) alone or with 10 μmol/L MG132. B: The ubiquitination of endogenous MPK38 was assessed in adipocytes from male and female mice treated with DHT, E2, or vehicle. C: Adipocyte cell lysates from male and female mice were treated with DHT, E2, or vehicle and then immunoprecipitated with an anti-MPK38 antibody (IP: α-MPK38), followed by immunoblotting with an anti‐MDM2 antibody for assessment of endogenous MPK38‐MDM2 complex formation. D: Adipocytes from male and female mice treated with DHT, E2, or vehicle were immunoprecipitated with an anti-MPK38 antibody, and the endogenous complex formation between MPK38 and ZPR9 (left) or TRX (right) was assessed by immunoblot analysis with use of anti-ZPR9 and anti-TRX antibodies, respectively. E: MPK38-dependent ASK1/TGF-β/p53 signaling in adipocytes from male and female mice treated with DHT, E2, or vehicle in the presence or absence of H2O2 (2 mmol/L, 30 min), TGF‐β1 (100 pmol/L, 20 h), and 5-fluorouracil (5FU) (0.38 mmol/L, 30 h) was determined by immunoblotting with the indicated antibodies. In all experiments that were performed, 4.5-month-old C57BL/6N male and female mice were used. WB, Western blot.

Close modal

In the current study, we found that functional inactivation of MPK38 led to late-onset obesity in males, even when fed a standard chow diet, which was associated with abnormal glucose, lipid, and energy metabolism, as well as inflammation, and that heterozygous MPK38+/− male mice displayed moderate levels of obesity-induced metabolic disturbances in comparison with MPK38−/− male mice (Fig. 1), proving haploinsufficiency of MPK38. In addition, Comparative Toxicogenomics Database (CTD) suggests a possibility that MPK38 may be associated with metabolic disorders even though genome-wide association studies are not available at the moment.

Testosterone is a male sex hormone that has important effects on carbohydrate, fat, and protein metabolism (33). In rodents, the insulin responsiveness of male WAT was increased by castration, whereas OVX reduced the insulin responsiveness of female WAT (26). However, in nonhuman primates, testosterone stimulates insulin-stimulated Akt phosphorylation and lipogenesis in the WAT of castrated males (34). In middle-aged men, the age-related decline in testosterone increases the obesity-associated risks of diabetes, cardiovascular diseases, and mortality by increasing body fat mass, in particular that of visceral fat (35,36). However, testosterone replacement therapy in hypogonadal males reverses these metabolic changes (37). The current study shows that the 7-month-old MPK38−/− male mice have circulating testosterone concentrations similar to those of castrated WT males (Fig. 7A), suggesting that MPK38 has a role in testosterone production in aged mature adult male mice. Furthermore, serum LH levels are significantly decreased in proportion to the testosterone levels in 7-month-old MPK38−/− males (Supplementary Fig. 12). These findings raise a strong possibility that MPK38 deficiency may affect the crucial components of the hypothalamic-pituitary-gonadal axis, leading to central hypogonadism associated with obesity. Nevertheless, the reverse effects of testosterone replacement are not found in 7-month-old MPK38−/− males in comparison with age-matched WT controls (Supplementary Fig. 8). Therefore, we cannot rule out the possibility that alterations in other potential targets including metabolic enzymes by MPK38 deficiency may contribute to the sexual dimorphism of late-onset obesity in MPK38−/− mice.

Estrogen, a female sex hormone, has also been shown to have a significant effect on metabolism. In rats, OVX leads to a marked increase in energy storage, and this is prevented by estrogen replacement (38). However, the results of the current study show that there is no difference in the effects of neutering on metabolism between WT and MPK38−/− female mice (Supplementary Fig. 9), suggesting that MPK38 does not have a role in the effects of estrogen on metabolism in female mice. Our data also show that E2, the principal circulating estrogen, inhibits the activity of MPK38, regardless of sex (Fig. 8E), but activates AMPK by increasing the phosphorylation of its α-catalytic subunit (39).

We also show that MPK38 stimulates steroidogenesis by increasing the expression of genes encoding proteins involved in cholesterol transport (Scarb1 and Star) (Fig. 7). In contrast to the effects of AMPK (29), MPK38 increases the expression of two activators (c-Jun and Nr4a1) of steroidogenesis. NR4A1 prevents AMPKα activation in hepatocytes by sequestering liver kinase B1 (LKB1), an AMPK upstream kinase, in the nucleus (40). The current study shows that NR4A1 is also involved in the inactivation of MPK38 because it stimulates the nuclear localization of MPK38 and its positive regulator ZPR9, and the cytoplasmic localization of its negative regulator TRX1, which leads to the inactivation of AMPK in the cytoplasm (Supplementary Fig. 13 and Supplementary Fig. 14A). However, the nuclear localization of LKB1 induced by NR4A1 does not affect the activity of MPK38 because LKB1 has no effect on MPK38 (Supplementary Fig. 13 and Supplementary Fig. 14B). These findings suggest that MPK38, unlike AMPK, modulates the expression of transcriptional regulators that play crucial roles in steroidogenesis in a sex- and age-dependent fashion (Fig. 7C–F).

In summary, the data presented here provide evidence that MPK38 acts as a metabolic regulator in aged mature adult male, but not female, mice, in which it prevents obesity-induced metabolic disturbances, and suggest that MPK38 may be an attractive therapeutic target for obesity in middle-aged men.

This article contains supplementary material online at https://doi.org/10.2337/figshare.13259669.

Acknowledgments. The authors thank E.-Y. Lee (National Institute of Horticultural and Herbal Science, Eumsung), J.-H. Mang (Clinical Research Associates Korea Incorporated, Cheongju), S.-H. Yeon (NEXEL Co., Ltd., Seoul), and J.-H. Kwak (Immunology Laboratory, Department of Biochemistry, Chungbuk National University) for excellent technical assistance during animal experiments. The authors also thank Dr. C.-S. Choi (Department of Internal Medicine, Gil Medical Center, Gachon University Graduate School of Medicine, Incheon, Korea) and Dr. K.-T. Kim (Department of Life Sciences, Pohang University of Science and Technology, Pohang, Korea) for warm and kind help in initial characterization of knockout mice and animal studies and Dr. K. Lee (Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, Korea) for excellent technical assistance in castration and OVX studies.

Funding. This work was supported by grants from the National Research Foundation of Korea (2018R1A2A2A05018692) and the Research Year of Chungbuk National University in 2019.

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

Author Contributions. H.-A.S. performed all experiments and data analysis and wrote all experimental protocols. H.H. designed, supervised, and interpreted all studies and wrote the manuscript. H.-A.S. and H.H. 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.

1.
Ganguly
R
,
Mohyeldin
A
,
Thiel
J
,
Kornblum
HI
,
Beullens
M
,
Nakano
I
.
MELK-a conserved kinase: functions, signaling, cancer, and controversy
.
Clin Transl Med
2015
;
4
:
11
2.
Gil
M
,
Yang
Y
,
Lee
Y
,
Choi
I
,
Ha
H
.
Cloning and expression of a cDNA encoding a novel protein serine/threonine kinase predominantly expressed in hematopoietic cells
.
Gene
1997
;
195
:
295
301
3.
Heyer
BS
,
Warsowe
J
,
Solter
D
,
Knowles
BB
,
Ackerman
SL
.
New member of the Snf1/AMPK kinase family, Melk, is expressed in the mouse egg and preimplantation embryo
.
Mol Reprod Dev
1997
;
47
:
148
156
4.
Jiang
P
,
Zhang
D
.
Maternal embryonic leucine zipper kinase (MELK): a novel regulator in cell cycle control, embryonic development, and cancer
.
Int J Mol Sci
2013
;
14
:
21551
21560
5.
Gray
D
,
Jubb
AM
,
Hogue
D
, et al
.
Maternal embryonic leucine zipper kinase/murine protein serine-threonine kinase 38 is a promising therapeutic target for multiple cancers
.
Cancer Res
2005
;
65
:
9751
9761
6.
Chung
S
,
Suzuki
H
,
Miyamoto
T
, et al
.
Development of an orally-administrative MELK-targeting inhibitor that suppresses the growth of various types of human cancer
.
Oncotarget
2012
;
3
:
1629
1640
7.
Nakano
I
,
Joshi
K
,
Visnyei
K
, et al
.
Siomycin A targets brain tumor stem cells partially through a MELK-mediated pathway
.
Neuro Oncol
2011
;
13
:
622
634
8.
Blüher
M
,
Bashan
N
,
Shai
I
, et al
.
Activated Ask1-MKK4-p38MAPK/JNK stress signaling pathway in human omental fat tissue may link macrophage infiltration to whole-body Insulin sensitivity
.
J Clin Endocrinol Metab
2009
;
94
:
2507
2515
9.
Yadav
H
,
Quijano
C
,
Kamaraju
AK
, et al
.
Protection from obesity and diabetes by blockade of TGF-β/Smad3 signaling
.
Cell Metab
2011
;
14
:
67
79
10.
Vousden
KH
,
Ryan
KM
.
p53 and metabolism
.
Nat Rev Cancer
2009
;
9
:
691
700
11.
Seong
HA
,
Manoharan
R
,
Ha
H
.
Smad proteins differentially regulate obesity-induced glucose and lipid abnormalities and inflammation via class-specific control of AMPK-related kinase MPK38/MELK activity
.
Cell Death Dis
2018
;
9
:
471
12.
Seong
HA
,
Jung
H
,
Manoharan
R
,
Ha
H
.
PDK1 protein phosphorylation at Thr354 by murine protein serine-threonine kinase 38 contributes to negative regulation of PDK1 protein activity
.
J Biol Chem
2012
;
287
:
20811
20822
13.
Seong
HA
,
Ha
H
.
Murine protein serine-threonine kinase 38 activates p53 function through Ser15 phosphorylation
.
J Biol Chem
2012
;
287
:
20797
20810
14.
Jung
H
,
Seong
HA
,
Ha
H
.
Murine protein serine/threonine kinase 38 activates apoptosis signal-regulating kinase 1 via Thr 838 phosphorylation
.
J Biol Chem
2008
;
283
:
34541
34553
15.
Seong
HA
,
Jung
H
,
Ha
H
.
Murine protein serine/threonine kinase 38 stimulates TGF-beta signaling in a kinase-dependent manner via direct phosphorylation of Smad proteins
.
J Biol Chem
2010
;
285
:
30959
30970
16.
Lucchini
FC
,
Wueest
S
,
Challa
TD
, et al
.
ASK1 inhibits browning of white adipose tissue in obesity
.
Nat Commun
2020
;
11
:
1642
17.
Challa
TD
,
Wueest
S
,
Lucchini
FC
, et al
.
Liver ASK1 protects from non-alcoholic fatty liver disease and fibrosis
.
EMBO Mol Med
2019
;
11
:
e10124
18.
Liles
JT
,
Corkey
BK
,
Notte
GT
, et al
.
ASK1 contributes to fibrosis and dysfunction in models of kidney disease
.
J Clin Invest
2018
;
128
:
4485
4500
19.
Seong
HA
,
Manoharan
R
,
Ha
H
.
Coordinate activation of redox-dependent ASK1/TGF-β signaling by a multiprotein complex (MPK38, ASK1, SMADs, ZPR9, and TRX) improves glucose and lipid metabolism in mice
.
Antioxid Redox Signal
2016
;
24
:
434
452
20.
Seong
HA
,
Manoharan
R
,
Ha
H
.
Zinc finger protein ZPR9 functions as an activator of AMPK-related serine/threonine kinase MPK38/MELK involved in ASK1/TGF-β/p53 signaling pathways
.
Sci Rep
2017
;
7
:
42502
21.
McGuinness
OP
,
Ayala
JE
,
Laughlin
MR
,
Wasserman
DH
.
NIH experiment in centralized mouse phenotyping: the Vanderbilt experience and recommendations for evaluating glucose homeostasis in the mouse
.
Am J Physiol Endocrinol Metab
2009
;
297
:
E849
E855
22.
Markman
JL
,
Porritt
RA
,
Wakita
D
, et al
.
Loss of testosterone impairs anti-tumor neutrophil function
.
Nat Commun
2020
;
11
:
1613
23.
McLeod
VM
,
Lau
CL
,
Chiam
MDF
, et al
.
Androgen receptor antagonism accelerates disease onset in the SOD1G93A mouse model of amyotrophic lateral sclerosis
.
Br J Pharmacol
2019
;
176
:
2111
2130
24.
Puigserver
P
,
Wu
Z
,
Park
CW
,
Graves
R
,
Wright
M
,
Spiegelman
BM
.
A cold-inducible coactivator of nuclear receptors linked to adaptive thermogenesis
.
Cell
1998
;
92
:
829
839
25.
Esser
V
,
Brown
NF
,
Cowan
AT
,
Foster
DW
,
McGarry
JD
.
Expression of a cDNA isolated from rat brown adipose tissue and heart identifies the product as the muscle isoform of carnitine palmitoyltransferase I (M-CPT I). M-CPT I is the predominant CPT I isoform expressed in both white (epididymal) and brown adipocytes
.
J Biol Chem
1996
;
271
:
6972
6977
26.
Macotela
Y
,
Boucher
J
,
Tran
TT
,
Kahn
CR
.
Sex and depot differences in adipocyte insulin sensitivity and glucose metabolism
.
Diabetes
2009
;
58
:
803
812
27.
Tosca
L
,
Froment
P
,
Solnais
P
,
Ferré
P
,
Foufelle
F
,
Dupont
J
.
Adenosine 5′-monophosphate-activated protein kinase regulates progesterone secretion in rat granulosa cells
.
Endocrinology
2005
;
146
:
4500
4513
28.
Tosca
L
,
Chabrolle
C
,
Uzbekova
S
,
Dupont
J
.
Effects of metformin on bovine granulosa cells steroidogenesis: possible involvement of adenosine 5′ monophosphate-activated protein kinase (AMPK)
.
Biol Reprod
2007
;
76
:
368
378
29.
Abdou
HS
,
Bergeron
F
,
Tremblay
JJ
.
A cell-autonomous molecular cascade initiated by AMP-activated protein kinase represses steroidogenesis
.
Mol Cell Biol
2014
;
34
:
4257
4271
30.
Ramayya
MS
,
Zhou
J
,
Kino
T
,
Segars
JH
,
Bondy
CA
,
Chrousos
GP
.
Steroidogenic factor 1 messenger ribonucleic acid expression in steroidogenic and nonsteroidogenic human tissues: Northern blot and in situ hybridization studies
.
J Clin Endocrinol Metab
1997
;
82
:
1799
1806
31.
Hanley
NA
,
Ball
SG
,
Clement-Jones
M
, et al
.
Expression of steroidogenic factor 1 and Wilms’ tumour 1 during early human gonadal development and sex determination
.
Mech Dev
1999
;
87
:
175
180
32.
Manoharan
R
,
Seong
HA
,
Ha
H
.
Thioredoxin inhibits MPK38-induced ASK1, TGF-β, and p53 function in a phosphorylation-dependent manner
.
Free Radic Biol Med
2013
;
63
:
313
324
33.
Kelly
DM
,
Jones
TH
.
Testosterone: a metabolic hormone in health and disease
.
J Endocrinol
2013
;
217
:
R25
R45
34.
Varlamov
O
,
White
AE
,
Carroll
JM
, et al
.
Androgen effects on adipose tissue architecture and function in nonhuman primates
.
Endocrinology
2012
;
153
:
3100
3110
35.
Navarro
G
,
Allard
C
,
Xu
W
,
Mauvais-Jarvis
F
.
The role of androgens in metabolism, obesity, and diabetes in males and females
.
Obesity (Silver Spring)
2015
;
23
:
713
719
36.
De Maddalena
C
,
Vodo
S
,
Petroni
A
,
Aloisi
AM
.
Impact of testosterone on body fat composition
.
J Cell Physiol
2012
;
227
:
3744
3748
37.
Brodsky
IG
,
Balagopal
P
,
Nair
KS
.
Effects of testosterone replacement on muscle mass and muscle protein synthesis in hypogonadal men--a clinical research center study
.
J Clin Endocrinol Metab
1996
;
81
:
3469
3475
38.
Richard
D
.
Effects of ovarian hormones on energy balance and brown adipose tissue thermogenesis
.
Am J Physiol
1986
;
250
:
R245
R249
39.
Lipovka
Y
,
Chen
H
,
Vagner
J
,
Price
TJ
,
Tsao
TS
,
Konhilas
JP
.
Oestrogen receptors interact with the α-catalytic subunit of AMP-activated protein kinase
.
Biosci Rep
2015
;
35
:
e00264
40.
Zhan
YY
,
Chen
Y
,
Zhang
Q
, et al
.
The orphan nuclear receptor Nur77 regulates LKB1 localization and activates AMPK
.
Nat Chem Biol
2012
;
8
:
897
904
Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. More information is available at https://www.diabetesjournals.org/content/license.