Skip to main content

TWEAK promotes exercise intolerance by decreasing skeletal muscle oxidative phosphorylation capacity

Abstract

Background

Proinflammatory cytokine tumor necrosis factor (TNF)-like weak inducer of apoptosis (TWEAK) and its receptor Fn14 are the major regulators of skeletal muscle mass in many catabolic conditions. However, their role in muscle metabolism remains largely unknown. In the present study, we investigated the role of TWEAK on exercise capacity and skeletal muscle mitochondrial content and oxidative metabolism.

Methods

We employed wild-type and TWEAK-knockout (KO) mice and primary myotube cultures and performed biochemical, bioenergetics, and morphometric assays to evaluate the effects of TWEAK on exercise tolerance and muscle mitochondrial function and angiogenesis.

Results

TWEAK-KO mice showed improved exercise tolerance compared to wild-type mice. Electron microscopy analysis showed that the abundance of subsarcolemmal and intermyofibrillar mitochondria is significantly increased in skeletal muscle of TWEAK-KO mice compared to wild-type mice. Furthermore, age-related loss in skeletal muscle oxidative capacity was rescued in TWEAK-KO mice. Expression of a key transcriptional regulator peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) and several other molecules involved in oxidative metabolism were significantly higher in skeletal muscle of TWEAK-KO mice. Moreover, treatment of primary myotubes with soluble TWEAK inhibited the expression of PGC-1α and mitochondrial genes and decreased mitochondrial respiratory capacity. Deletion of TWEAK also improved angiogenesis and transcript levels of vascular endothelial growth factor in skeletal muscle of mice.

Conclusions

These results demonstrate that TWEAK decreases mitochondrial content and oxidative phosphorylation and inhibits angiogenesis in skeletal muscle. Neutralization of TWEAK is a potential approach for improving exercise capacity and oxidative metabolism in skeletal muscle.

Background

Skeletal muscle is the largest tissue of the human body which ensures basic functions such as locomotion, metabolism, and respiration. Skeletal muscle exhibits a high level of plasticity in response to physiological stressors. For example, in response to exercise training, the proportion of slow-type fibers and mitochondrial content within fibers is noticeably increased [1]. The biogenesis of new mitochondria and clearance of defunct mitochondria are essential to meet cellular energy demand especially during endurance exercise and to protect from many chronic conditions such as diabetes, heart failure, obesity, and aging [2, 3]. Peroxisome proliferator-activated receptor (PPAR)-γ coactivator 1α (PGC-1α) is a key player in regulating skeletal muscle fiber composition, mitochondrial content, and oxidative metabolism, and maintenance of glucose, lipid, and energy homeostasis in response to physiological demands [4–6]. Transgenic mice overexpressing physiological levels of PGC-1α in skeletal muscle have increased mitochondrial biogenesis and oxidative capacity, are more resistant to fatigue and have improved aerobic performance [7–10]. Forced expression of PGC-1α also preserves skeletal muscle mass in various catabolic states including denervation [11, 12]. PGC-1α improves mitochondrial biogenesis by coactivating nuclear respiratory factor (NRF)-1 [13], NRF-2 [14] and estrogen-related receptor α [15–17] which, in turn, augments the expression of hundreds of nuclear-coded mitochondrial genes [18]. Recent evidence also suggests a role for mitochondria in preventing muscle protein degradation in catabolic conditions [19, 20]. Thus, preserving mitochondria content and function is essential to countering muscle wasting and to improving muscle homeostasis.

Inflammatory cytokines such as tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) are some of the important mediators of loss of skeletal muscle mass and function in many catabolic conditions [21–24]. TNF-α has been reported to augment muscle protein degradation both in vitro and in vivo[25–27]. Moreover, TNF receptor I knockout (KO) mice are protected from diet-induced obesity due to increased thermogenesis [28]. Importantly, TNF-α inhibits the expression of PGC-1α levels in myotubes [29]. In animal models of cancer cachexia, mitochondrial content and biogenesis are reduced with increased levels of IL-6 during the progression of the condition whereas inhibition of IL-6 activity attenuates tumor-induced loss of skeletal muscle [30, 31]. It is also notable that while moderate exercise attenuates inflammation [32–34], acute exercise more robustly increases the levels of inflammatory cytokines in obese compared with lean subjects suggesting that rigorous exercise can further aggravate the condition of overweight individuals [35]. Despite these observations, cause-and-effect relationships between different inflammatory mediators, exercise tolerance, and mitochondrial function in skeletal muscle have not been clearly established using genetic mouse models.

TNF-like weak inducer of apoptosis (TWEAK) is a major proinflammatory cytokine of the TNF super family [36] that functions by binding to Fn14 receptors on target cells [37–39]. TWEAK-Fn14 signaling mediates unique and context-dependent pleiotropic effects [40]. TWEAK has been recently identified to be a key mediator that causes skeletal muscle loss that occurs in response to denervation, immobilization, and starvation [21, 41, 42]. Moreover, TWEAK-transgenic mice overexpressing physiological levels of TWEAK in skeletal muscle showed a higher proportion of fast-type fibers (glycolytic) in soleus and extensor digitorum longus (EDL) muscle [41] indicating that elevated levels of TWEAK cause a slow-to-fast type fiber switch. However, the role and mechanisms by which TWEAK affects exercise capacity and bioenergetic function have not been studied.

In the present study, we have used TWEAK-knockout (KO) mice to investigate the role of TWEAK in exercise tolerance and skeletal muscle bioenergetic function. Our results show that, compared with wild-type mice, TWEAK-KO mice have increased exercise tolerance, which is associated with higher levels of skeletal muscle subsarcolemmal and intermyofibrillar mitochondria and enhanced oxidative phosphorylation capacity. TWEAK represses the expression of PGC-1α and several other molecules involved in mitochondrial biogenesis and oxidative metabolism in vivo and in cultured myotubes. Furthermore, vascularization and expression of vascular endothelial growth factor (VEGF) are increased in skeletal muscle of TWEAK-KO mice compared with wild-type mice. These results support our hypothesis that TWEAK causes exercise intolerance by suppressing mitochondrial oxidative metabolism and angiogenesis.

Methods

Cell culture

Primary myoblasts were isolated from hind limb muscle of mice and cultured following the same protocol as previously described [43]. Briefly, hind limb skeletal muscles from mice were aseptically isolated, minced into a coarse slurry, and enzymatically digested at 37°C for one hour by adding 200 IU/ml collagenase I (cat # LS004196; Worthington, Lakewood, NJ, USA) and 0.1% pronase (EMD Chemicals, Billerica, MA, USA). The digested slurry was filtered through a 70 μm filter and spun, and isolated cells were resuspended and cultured initially in F-10 medium (containing 20% fetal bovine serum (FBS) and supplemented with 10 ng/ml basic fibroblast growth factor) and then in F-10 plus (Dulbecco’s) Modified Eagle’s Medium ((D)MEM) (1:1 ratio) based culture medium supplemented with 15% FBS on culture dishes coated with 10% matrigel (BD Biosciences, San Jose, CA, USA). Differentiation in primary myoblast cultures was induced by replacing the growth medium with differentiation medium (2% horse serum in (D)MEM).

Animals

TWEAK-KO mice were provided by Dr. Avi Ashkenazi (Genentech South, San Francisco, CA, USA) and have been previously described [40]. All the mice were in the C57BL/6 background, and their genotype was determined by PCR from tail DNA. All animal procedures were approved (protocol # 10129) by the Institutional Animal Care and Use Committee and conformed to the American Physiological Society’s Guiding Principles in the Care and Use of Animals.

Treadmill running protocol

Mice were subjected to treadmill running following the same previously described protocol [44]. In brief, 4.5-month-old wild-type and TWEAK-KO mice were matched for body weight and randomly assigned to either a sedentary or exercise group. Mice were subjected to an acute bout of treadmill (Eco3/6 treadmill; Columbus Instruments, Columbus, OH, USA) running at 15 m/minute for 90 minutes. All mice in the exercise group finished the 90-minute trial and were visibly exhausted. Mice were sacrificed within 30 minutes after exercise to study mitochondrial function.

Exercise tolerance test

The exercise tolerance test on mice was performed following a method as previously described [45]. Briefly, all animals were run on a treadmill (Columbus Instruments) at 10 m/minutes for five minutes at 0% degree incline for acclimation for three days. On the exercise testing day, animals ran on the treadmill with a fixed slope of 10%. Mice first ran at 10 m/minute for five minutes and the speed was increased by 2 m/minute every two minutes until they were exhausted or a maximal speed of 46 m/minute was achieved. The criterion of exhaustion was defined as the inability of the animal to run on the treadmill for 10 seconds despite mechanical prodding. Running time and maximum speed achieved was measured whereas running distance, work and power were calculated.

Transmission electron microscopy

Soleus muscles from wild-type and TWEAK-KO mice were fixed in 3% glutaraldehyde in 0.1 M cocodylate buffer overnight followed by fixing in 1% cocodylate-buffered osmium tetroxide. The tissue was dehydrated through a series of graded alcohols, and embedded in LX-112 plastic (Ladd Research Industries, Williston, VT, USA). Longitudinal sections (80 nm) were cut using an ultramicrotome (LKB, Rockville, MD, USA)) and stained with uranium acetate and lead citrate. Samples were analyzed using a transmission electron microscope (Philips CM 12; HZB, Berlin, Germany) operating at 60 kV. The pictures were captured at 8,800x magnification using a 3.2 megapixel digital camera (Sia-7C; Kodak, Rochester, NY, USA) at room temperature. No imaging medium was used to visualize the pictures, and images were stored as JPEG files. Image levels were equally adjusted using Photoshop CS2 software.

Succinate dehydrogenase (SDH) staining

SDH staining was performed as previously described [46]. Briefly, transverse sections (8 μm) were cut from the mid-belly of the TA muscles on a cryostat at −20°C and stored at −80°C until SDH staining was performed. The sections were dried at room temperature for 30 minutes before incubation in a solution made up of 0.2 M phosphate buffer (pH 7.4), 0.1 M MgCl2, 0.2 M succinic acid (Sigma Chemical Company, St. Louis, MO, USA) and 2.4 mM nitroblue tetrazolium (NBT, Sigma) at 37°C in a humidity chamber for 45 minutes. The sections were then washed in deionized water for three minutes, dehydrated in 50% ethanol for two minutes, and mounted for viewing with DPX mount medium (Electron Microscopy Sciences, Hatfield, PA, USA). Digital photographs were taken from each section at 10X magnification under a Nikon Eclipse TE 2000-U microscope (Nikon, Melville, NY, USA) with a Nikon digital camera (Digital Sight DS-Fi1), and fibers were quantified with imaging software (Image J, NIH). At least 700 fibers were counted to determine SDH-positive fibers in each section in a blinded fashion. The percentage of SDH stained fibers was then determined based on a criteria using integrated optical density.

Measurement of mitochondrial bioenergetics

Mitochondrial oxidative capacity was measured in isolated mitochondria and in cultured myotubes using a Seahorse Bioscience XF24 extracellular flux analyzer (Billerica, MA, USA). For measurements in isolated mitochondria, tissue from the limb muscle (approximately 50 mg) was isolated within 30 minutes of exercise and homogenized in 1 ml of isolation buffer (220 mM mannitol, 70 mM sucrose, 5 mM 3-(N-morpholino)propanesulfonic acid (MOPS), 1 mM ethylene glycol tetraacetic acid (EGTA), 0.3% fatty acid-free BSA, pH 7.2) using a Potter Elvehjem tube and a Teflon pestle. The homogenate was centrifuged at 500×g for five minutes at 4°C. The supernatant containing mitochondria was centrifuged at 10,000 × g for five minutes. After two wash-centrifugation steps in BSA-free isolation buffer, the mitochondria were suspended in respiration buffer (120 mM KCl, 25 mM sucrose, 10 mM 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES), 1 mM MgCl2, 5 mM KH2PO4, pH to 7.2). Protein in the mitochondrial suspension was estimated using the Lowry DC assay (Biorad, Hercules, CA, USA) and 5 to 12.5 μg of mitochondrial protein was sedimented in XF culture plates as described previously [47]. Complex I-mediated, state 3 respiratory activity was determined by measuring the oxygen consumption rate (OCR) after injection of pyruvate (5 mM), malate (2.5 mM) and ADP (1 mM). The OCR of mitochondria after exposure to oligomycin (1 μg/ml) was used to estimate state 4 activity; exposure to carbonyl cyanide p-trifluoromethoxyphenylhydrazone (FCCP, 2 μM) was used to examine the uncoupled rate of respiration. Finally, succinate (10 mM) and rotenone (1 μM) were injected to assess maximal Complex II-mediated respiratory capacity. Data are expressed as pmol O2/min/μg protein. For measurements in cultured cells, differentiated myoblasts were exposed to soluble TWEAK (R&D Systems, Minneapolis, MN, USA) for 72 hours followed by the mitochondrial function assay outlined previously [48–51].

RNA isolation and quantitative real time-PCR

RNA isolation and quantitative real time-PCR (qRT-PCR) were performed as previously described [41]. Briefly, total RNA was isolated from homogenized mouse tissues using the TRIzol reagent (Invitrogen, Grand Island, NY, USA) and an RNeasy Mini kit (QIAGEN, Valencia, CA, USA) according to the manufacturers’ protocols. First strand cDNA for PCR analyses were made using a reverse transcription system with 1 μg of purified RNA using oligo (dT) primer (Applied Biosystems, Grand Island, NY, USA) and the Omniscript reverse transcription kit (QIAGEN). The quantification of mRNA expression was performed using the SYBR green dye method on a sequence detection system (Applied Biosystems, model 7300). Primers were designed using Vector NTI software (Invitrogen). Primer sequences are available on request. The thermal conditions consisted of an initial denaturation at 95°C for 10 minutes, followed by 40 cycles of denaturation at 95°C for 15 seconds, annealing and extension at 60°C for one minute, and, for a final step, a melting curve of 95°C for 15 seconds, 60°C for 15 seconds, and 95°C for 15 seconds. All reactions were performed in duplicate to reduce variation. Data normalization was accomplished using the endogenous control (β-actin), and the normalized values were subjected to a 2-ΔΔCt formula to calculate the fold change between control and experimental groups.

Immunostaining for CD31

After cutting 8 μm thickness frozen sections of TA muscles, the sections were fixed by cold acetone for 10 minutes and dried in air for 30 minutes. The tissues were rinsed with phosphate buffered saline (PBS) twice, blocked in 2% BSA solution for one hour at room temperature followed by incubation with primary antibodies in 2% BSA solution (laminin, anti-rabbit 1:300, Sigma and CD31, anti-mouse 1:30, BD Biosciences, San Jose, CA, USA) at 4°C in a humidity chamber overnight. The next day, the sections were rinsed with PBS for five minutes three times and incubated with secondary antibody (Alexa Fluor-conjugated 488 anti-rabbit, 1:2500 and 546 anti-mouse, 1:2500, Invitrogen) in 2% BSA solution for one hour at room temperature. After washing twice with PBS and once with deionized water, the sections were mounted with DPX mounting medium. Digital photographs were taken from each section under a Nikon Eclipse TE 2000-U microscope (Nikon) with a Nikon digital camera (Digital Sight DS-Fi1). Number of CD31-postive vessels per myofiber was quantified using NIS Elements BR 3.00 software (Nikon).

Statistical analysis

The results are presented as means ± standard deviation (SD). Student’s t-test was used to compare the difference between control and treatment groups. A value P <0.05 was considered statistically significant.

Results

Improvement in exercise tolerance in TWEAK-KO mice

We first studied the role of TWEAK in exercise tolerance in mice. Male 4.5-month old wild-type and TWEAK-KO mice were acclimated for three days for treadmill running prior to the exercise tolerance test. After acclimation, the mice were run on a 10% slope until the mice were unable to run on the treadmill for 10 seconds despite mechanical prodding. TWEAK-KO mice ran longer compared to wild-type mice (1,483 seconds for TWEAK-KOs versus 1,170 seconds for the controls, Figure 1A). Since TWEAK-KO mice could keep running on higher speed (30.6 m/minute for TWEAK-KOs versus 25.5 m/minute for the controls, Figure 1B), the difference in running distance became greater than 47% (463 meter for TWEAK-KOs versus 314 meter for the controls, Figure 1C). Work and power generated by TWEAK-KO mice were also higher compared to wild-type mice. TWEAK-KO mice generated 22.2 J of work while control mice generated 15.2 J of work (a difference of 46%) during the exercise test (Figure 1D). In turn, TWEAK-KO mice exerted 14% more power than the controls (14.9 mW for TWEAK-KO versus 13.0 mW for the wild-type, Figure 1E) on the treadmill. These data established that TWEAK-KO mice have significantly improved exercise tolerance.

Figure 1
figure 1

Treadmill exercise tolerance test. After acclimatization, mice were run on a treadmill with a 10% slope and increasing speed to exhaustion. Maximum speed and running time were monitored, and distance, work, and power were calculated based on the individual performance. (A) Running time; (B) Speed; (C) Distance; (D) Work; and (E) Power. Data are represented as mean ± SD. N = 6 in each group. *P <0.05; values vary significantly between wild-type and TWEAK-KO mice. KO, knockout.

Ablation of TWEAK improves mitochondrial content in skeletal muscle of mice

We have previously shown that deletion of TWEAK increases the proportion of type I fibers in soleus and EDL muscles of mice [41]. In this study, we investigated whether TWEAK affects mitochondrial content in skeletal muscle of mice. The soleus muscle of 4.5-month old TWEAK-KO and wild-type mice were isolated and used to measure mitochondrial content by performing transmission electron microscopy. As shown in Figure 2A, the abundance and size of mitochondria was found to be increased in TWEAK-KO compared to wild-type mice. Quantitative analysis of mitochondria in electron micrographs showed that the levels of subsarcolemmal and intermyofibrillar mitochondria were increased by 42% and 32%, respectively, in TWEAK-KO mice compared to wild-type mice (Figure 2B and Figure 2C). Because gastrocnemius (GA) muscle contains a mixture of slow and fast twitch fibers and greatly influences running capacity on the treadmill, we measured mRNA levels of fiber type-specific myosin heavy chain (MyHC) isoforms in GA muscle of wild-type and TWEAK-KO mice. As shown in Figure 2D, mRNA levels of MyHC I and MyHC IIA were increased by 2- and 2.7-fold, respectively in TWEAK-KO mice compared to wild-type mice. These results further suggest that genetic ablation of TWEAK increases the slow-type fiber phenotype in skeletal muscle of mice.

Figure 2
figure 2

Transmission electron microscopy (TEM) analysis for mitochondria morphology and content and expression of muscle fiber type-specific genes in TWEAK-KO mice. Soleus muscle of six-month-old wild-type and TWEAK-KO mice were isolated and longitudinal sections were processed for TEM analysis. (A) Representative images of longitudinal soleus muscle are shown. The abundance and the size of mitochondria were increased in TWEAK-KO mice compared to wild-type mice. Arrows point to mitochondria in muscle sections. Circles are used to show representative enlarged mitochondria. Stars (‘*’) point to subsarcolemmal space in longitudinal sections. Scale bar: 1 μm. (B) Quantification of the number of subsarcolemmal mitochondria in soleus muscle of wild-type and TWEAK-KO mice. (C) Quantification of the number of intermyofibrillar mitochondria in soleus muscle sections of wild-type and TWEAK-KO mice. (D) Relative mRNA levels of MyHC type I, IIA and IIB in gastrocnemius muscle of 4.5-month-old wild-type and TWEAK-KO mice. Data are represented as mean ± SD. N = 3 in each group. *P <0.05; values vary significantly from wild-type mice. KO, knockout; TWEAK, TNF-like weak inducer of apoptosis.

SDH, also known as complex II in the mitochondrial respiratory chain, is a marker of oxidative capacity of skeletal muscle at the fiber level. Therefore, we performed SDH staining on TA muscle of 4.5-month- and 12-month-old wild-type mice and TWEAK-KO mice and quantified the percentage of SDH-positive fibers. Compared to wild-type mice, fibers in TWEAK-KO mice stained more darkly for SDH (Figure 3A). Furthermore, there was a significant increase in the number of SDH-positive fibers in TWEAK-KO mice compared to wild-type mice at the age of 4.5 months (Figure 3B). Likewise, 12-month-old TWEAK-KO had more SDH-positive fibers compared to age-matched wild-type mice (Figure 3A and Figure 3C). Taken together, these results suggest that ablation of TWEAK in mice is sufficient to improve mitochondrial content and oxidative capacity of skeletal muscle.

Figure 3
figure 3

Succinate dehydrogenase (SDH) staining analysis for oxidative capacity in TA muscle of TWEAK-KO mice. Frozen transverse TA muscle sections from 4.5- or 12-month old wild-type and TWEAK-KO mice were used to stain for SDH. (A) Representative SDH-stained images are presented here. TA muscle of TWEAK-KO showed relatively dark staining for SDH compared to wild-type mice. Scale bars: 50 μm. Quantification of SDH-positive fibers in TA muscle of (B) 4.5-month, and (C) 12-month old wild-type and TWEAK-KO mice. Data are presented as mean ± SD. *P <0.05; values vary significantly from wild-type mice. KO, knockout; TA, tibial anterior; TWEAK, TNF-like weak inducer of apoptosis.

TWEAK-KO mice demonstrate increased expression of PGC-1α and metabolic genes in skeletal muscle

To understand the mechanisms by which TWEAK affects exercise capacity and mitochondrial content in skeletal muscle, we investigated whether TWEAK modulates the expression of PGC-1α, a critical regulator of the mitochondrial biogenetic program in skeletal muscle [9, 10]. Interestingly, the mRNA levels of PGC-1α were found to be significantly higher in 4.5-month-old TWEAK-KO mice compared to age-matched wild-type mice (Figure 4). Similarly, mRNA levels of PPARδ were significantly higher in skeletal muscle of TWEAK-KO mice compared to corresponding wild-type mice (Figure 4). In contrast, mRNA levels of glycolytic enzymes such as hexokinase-2 (HK II) and phosphoglycerate mutase 2 (PGAM2) were reduced in skeletal muscle of TWEAK-KO mice compared to wild-type mice. No significant change was observed in the expression of pyruvate dehydrogenase kinase-4 (PDK4). Furthermore, mRNA levels of mitochondrial carnitine palmitoyltransferase I (mCPT1), which is required for the transport of long-chain fatty acyl-CoAs from the cytoplasm into the mitochondrion, were significantly elevated in TWEAK-KO mice compared to wild-type mice (Figure 4). These results suggest that a deficiency of TWEAK reduces expression of glycolytic genes and augments the expression of genes involved in oxidative metabolism in skeletal muscle.

Figure 4
figure 4

qRT-PCR analysis of transcript levels of metabolic genes in skeletal muscle of wild-type and TWEAK-KO mice. TA muscle from 4.5-month-old wild-type and TWEAK-KO mice were isolated and processed to measure mRNA levels of PGC-1α, PPARδ, HK II, PGAM2, PDK4, and mCPT1. Data are represented as mean ± SD. N = 4 in each group. *P <0.05. values vary significantly from wild-type mice. HK II, hexokinase-2; KO, knockout; mCPT1, mitochondrial carnitine palmitoyltransferase 1; PDK4, pyruvate dehydrogenase kinase-4; PGAM2, phosphoglycerate mutase 2; PGC-1α, PPAR coactivator 1α; PPAR, peroxisome proliferator-activated receptor; TA, tibial anterior; TWEAK, TNF-like weak inducer of apoptosis.

Ablation of TWEAK increases state 3 respiration in skeletal muscle mitochondria

We next sought to determine whether TWEAK regulates mitochondrial function in skeletal muscle. After an acute bout of treadmill running for 90 minutes, mitochondria were isolated from hind limb muscles of wild type and TWEAK-KO mice and mitochondrial function was examined by extracellular flux (XF) analysis. The sequential addition of respiratory substrates and inhibitors of oxidative phosphorylation were used to determine changes in electron transport chain activity: state 3 respiration was induced by addition of pyruvate, malate and ADP; state 4 respiration was induced by addition of oligomycin; FCCP was added to determine maximal complex I-mediated respiratory activity; and, lastly, succinate and rotenone were added to determine maximal complex II-mediated respiratory activity. As shown in Figure 5A and 5B, mitochondria from TWEAK-KO mice showed significantly increased state 3 respiratory activities compared with wild type mice. Mitochondrial coupling (as determined by calculating respiratory control ratios) and other indices of mitochondrial function were not significantly different between wild type and KO mice. These data suggest that absence of TWEAK augments mitochondrial oxidative capacity in skeletal muscle in exercised mice.

Figure 5
figure 5

Mitochondrial respiration is enhanced in TWEAK-KO mice. Respiratory activity of mitochondria isolated from wild-type and TWEAK-KO mice subjected to an acute bout of treadmill running: (A) Extracellular flux assay of isolated mitochondria: mitochondrial activity was measured in the absence of substrate (state 1 respiration) or in the presence of pyruvate (5 mM), malate (2.5 mM) and ADP (1 mM) (state 3 respiration). State 4 respiration was estimated by measuring the oxygen consumption rate (OCR) after the addition of oligomycin (1 μg/ml). FCCP (4 μM) was added to assess maximal activity of mitochondria respiring on substrates providing electrons for complex I (that is, pyruvate and malate) or complex II (that is, succinate in the presence of rotenone; 10 mM and 1 μM, respectively). (B) Group data derived from panel A. N = 3 mice per group. *P <0.05 versus wild-type. FCCP, carbonyl cyanide p-trifluoromethoxyphenylhydrazone; KO, knockout; TWEAK, TNF-like weak inducer of apoptosis.

TWEAK represses the expression of PGC-1α and genes regulating oxidative metabolism in cultured primary myotubes

We next studied whether TWEAK affects the expression of PGC-1α and other genes in cultured primary myotubes. Primary myoblasts prepared from wild-type mice were differentiated into myotubes by incubation in differentiation medium for 96 hours followed by treatment with TWEAK for 72 hours. As shown in Figure 6, addition of soluble TWEAK dramatically reduced the expression of PGC-1α in myotubes. Treatment with TWEAK also significantly reduced the mRNA levels of several mitochondrial genes encoding proteins related to oxidative metabolism, such as ATP synthase subunit beta (ATP5b), cytochrome c oxidase subunit I (Cox I), Cox subunit IV (COX IV), COX -7b, cytochrome c (Cyt c), medium-chain acyl-coenzyme A dehydrogenase (MCAD), PPARδ, and mCPT1 (Figure 6). These results further suggest that TWEAK inhibits the mitochondrial biogenetic program in skeletal muscle.

Figure 6
figure 6

Effect of TWEAK on expression of PGC-1α and mitochondria oxidative metabolism genes in cultured primary myotubes. Primary myotubes prepared from wild-type mice were treated with soluble TWEAK protein (10 ng/ml or 100 ng/ml) for 72 hours followed by RNA isolation and performing qRT-PCR to study transcript levels of PGC-1α, Tfam, Tfam2m, Cyt-c, Cox I, Cox IV, Cox 7b, and MCAD. Data are presented as mean ± SD. *P <0.05; values vary significantly from untreated myotubes. Cox, cytochroms c oxidase; Cyt c, cytochrome c; MCAD, medium-chain acyl-coenzyme A dehydrogenase; PGC-1α, PPAR coactivator 1α; PPAR, peroxisome proliferator-activated receptor; Tfam, transcription factor A mitochondrial; TWEAK, TNF-like weak inducer of apoptosis.

TWEAK decreases maximal respiratory capacity and increases glycolysis in cultured primary myotubes

Our studies in isolated mitochondria derived from skeletal muscle after exercise suggest that the absence of TWEAK increases mitochondrial oxidative capacity. To assess the effects of TWEAK on mitochondrial function directly, we exposed differentiated myotubes to TWEAK and then assessed mitochondrial function as described previously [48–51]. As shown in Figure 7A and 7B, exposure of myotubes to TWEAK for 72 hours did not affect the basal mitochondrial OCR, nor did it affect ATP-linked OCR or proton leak. However, the FCCP-stimulated OCR, indicative of maximal respiratory capacity, was remarkably diminished by TWEAK treatment. In addition, a modest, yet significant, increase in the non-mitochondrial OCR occurred in myotubes exposed to100 ng/ml TWEAK, which may be due to increased production of reactive oxygen species. The diminishment in mitochondrial respiratory capacity caused by TWEAK was associated with an increase in the extracellular acidification rate (ECAR), a surrogate measure of glycolysis (Figure 7C). Collectively, these results suggest that TWEAK directly modulates the bioenergetic capacity of skeletal muscle.

Figure 7
figure 7

TWEAK regulates mitochondrial oxidative capacity and glycolytic flux. Extracellular flux analysis of primary differentiated myotubes treated with 0, 10, or 100 ng/ml TWEAK for 72 hours. (A) Mitochondrial function assay: after three baseline measurements, inhibitors or activators of electron transport were added sequentially to intact myotubes. Oxygen consumption rate (OCR) measurements were recorded after each exposure. O, oligomycin; F, FCCP; and A, antimycin A/rotenone. The OCR was normalized to total protein in each well. (B) Indices of mitochondrial function calculated from assay results in panel A. (C) Extracellular acidification rates (ECAR) of cells treated without or with TWEAK; ECAR is a surrogate measure of lactate and therefore is used as a measure of glycolytic flux. N = 3 per group; *P <0.05 versus control. FCCP, carbonyl cyanide p-trifluoromethoxyphenylhydrazone; TWEAK, TNF-like weak inducer of apoptosis.

Ablation of TWEAK improves angiogenesis in skeletal muscle of mice

There is a causal relationship between PGC-1α and angiogenesis. Skeletal muscle-specific overexpression of PGC-1α in mice results in increased angiogenesis, which may contribute to enhanced exercise performance [52]. Since TWEAK suppresses PGC-α in skeletal muscle, we next sought to determine whether TWEAK also regulates angiogenesis in skeletal muscle of mice. TA muscle of 4.5-month-old wild-type and TWEAK-KO mice was isolated and immunostained with antibodies against CD31 (also known as platelet endothelial cell adhesion molecule-1), an endothelial-specific marker for capillaries. Laminin staining was used to mark the periphery of fibers. As shown in Figure 8A, the number of CD31-positive capillaries was considerably increased in TWEAK-KO mice compared with wild-type mice. The capillary-to-fiber ratio was increased by 29% in TWEAK-KO mice (Figure 8B). Since VEGF positively regulates angiogenesis, we also measured mRNA levels of VEGF in TA muscle of wild-type and TWEAK-KO mice. As shown in Figure 8C, mRNA levels of VEGF were significantly higher in TA muscle of TWEAK-KO mice compared to wild-type mice. Furthermore, treatment with TWEAK also reduced the mRNA levels of VEGF in primary myotubes (Figure 8D). These results suggest that ablation of TWEAK stimulates angiogenesis in skeletal muscle of mice potentially through increasing the expression of VEGF.

Figure 8
figure 8

Analysis of capillary density in skeletal muscle of wild-type and TWEAK-KO mice. Transverse sections of TA muscle prepared from 4.5-month-old wild-type and TWEAK-KO mice were immunostained for CD31 (red) and counterstained for laminin (green). (A) Representative CD31- and laminin-immunostained and merged images are presented here. Scale bars: 50 μm. (B) Quantification of CD31-postive capillaries per myofiber in TA muscle. N = 8 in each group. (C) Relative mRNA levels of VEGF in TA muscle of wild-type and TWEAK-KO mice (N = 4 in each group) measured by qRT-PCR analysis. (D) Primary myotubes were treated with the indicated concentration of TWEAK for 24 hours followed by measurement of mRNA levels of VEGF by qRT-PCR. Relative mRNA levels of VEGF are shown here. Data are presented as mean ± SD. *P <0.05; values vary significantly from untreated myotubes. KO, knockout; TA, tibial anterior; TWEAK, TNF-like weak inducer of apoptosis; VEGF, vascular endothelial growth factor.

Discussion

Exercise capacity in mammals is determined by multiple factors including skeletal muscle oxidative metabolism and vascularization. Our previous microarray study suggested that TWEAK can modulate the expression of several genes whose products are involved in mitochondrial dysfunction and fatty acid metabolism [53]. Our present study demonstrates that compared to wild-type mice, TWEAK-KO mice run longer and with higher speed during an exercise tolerance test (Figure 1). TWEAK-KO mice show augmented levels of subsarcolemmal and intermyofibrillar mitochondria, increased SDH-positive myofibers, and elevated expression of metabolic genes such as PGC-1α, PPARδ, and mCPT-1 compared to wild-type mice (Figures 2, 3, and 4). Oxidative phosphorylation is also increased in exercised TWEAK-KO mice compared with wild-type mice (Figure 5), and treatment of myotubes with TWEAK directly decreases mitochondrial biogenetic capacity and maximal respiratory activity (Figures 6 and 7). Moreover, vascularization and expression of VEGF in skeletal muscle are enhanced in TWEAK-KO mice compared to their controls (Figure 8). These data suggest that TWEAK directly regulates the mitochondrial biogenetic program and that loss of TWEAK improves mitochondrial respiratory capacity and increases vascularization of skeletal muscle tissue, which collectively lead to improved exercise performance.

One of the potential mechanisms by which TWEAK might be attenuating exercise capacity is through diminishing the levels of PGC-1α in skeletal muscle. PGC-1α augments mitochondrial biogenesis, oxidative metabolism, and promotes fast-to-slow type fiber transition [9, 13, 17, 54–56]. Overexpression of PGC-1α in rat primary culture cells leads to increased abundance of slow oxidative-associated MyHC isoform [57]. Skeletal muscle-specific PGC-1α KO mice demonstrate reduced endurance capacity with a shift from oxidative fiber type to glycolytic fibers and increased levels of TNF-α in skeletal muscle [45, 58]. By contrast, muscle-specific overexpression of PGC-1α enhances exercise performance with increased fatty acid oxidation and decreased glycogen usage during exercise [8, 52]. These findings suggest that PGC-1α is not only essential but sufficient to determining skeletal muscle fiber composition. Our experiments demonstrate that mRNA levels of PGC-1α are increased in skeletal muscle of TWEAK-KO mice with a concomitant increase in mitochondrial content and expression of genes whose products are involved in oxidative capacity (Figures 2 and 4). Furthermore, treatment of primary myotubes with TWEAK drastically reduced levels of PGC-1α and other mitochondrial genes (Figure 6) further suggesting that TWEAK represses the expression of PGC-1α leading to reduced mitochondrial content. These findings are consistent with functional analyses of mitochondrial function: ablation of TWEAK resulted in increased oxidative phosphorylation capacity in isolated mitochondria (Figure 5) and treatment of myotubes with TWEAK decreased maximal respiratory capacity (Figure 7). Although not tested in this study, it is possible that TWEAK also reduces the levels of PGC-1α in other organs which ultimately results in reduced exercise capacity. Indeed, Shi et al. recently reported that TWEAK represses the expression of PGC-1α and other molecules involved in mitochondrial oxidative phosphorylation in cardiomyocytes and forced expression of PGC-1α attenuates TWEAK-induced cardiac dysfunction in mice [59].

The PPARs are ligand-modulated transcription factors in which three subtypes have been identified: α, β/δ, and γ [60]. Previous studies have shown that their endogenous ligands are composed of fatty acids and lipid metabolites and, therefore, certain PPARs mediate the expression of genes whose products are involved in the regulation of fatty acid metabolism in response to changes in systemic fuel availability [60–62]. In skeletal muscle, levels of PPARδ are relatively higher compared to PPARα or PPARγ [63]. Treatment with synthetic PPARδ agonist or overexpression of PPARδ by retroviral infection induces the levels of molecules which are involved in lipid metabolism and fatty acid oxidation, whereas overexpression of a dominant-negative PPARδ mutant exerts opposite effects in C2C12 myotubes [64]. Furthermore, it has been shown that muscle-specific overexpression of PPARδ in mice increases exercise tolerance with a switch to increased number of type I muscle fibers and up-regulation of molecules related to fatty acid metabolism. In contrast, PPARδ-null mice show decreased exercise performance compared to wild-type mice [65, 66]. Although muscle relies mainly on fat and carbohydrate as energy resources, enhanced fatty acid utilization during exercise with glycogen sparing results in improved exercise endurance capacity [67–69]. Haramizu et al. demonstrated that the expression levels of gene products related to lipid metabolism in the muscle is correlated with the levels of fatty acid β-oxidation activity as well as exercise strength [70]. Our results demonstrate that transcript levels of PPARδ and mCPT1 are significantly increased while the expression of molecules that are associated with glycolysis such as HK II and PGAM2 is suppressed in skeletal muscle of TWEAK-KO mice compared to wild-type mice (Figure 6). These results suggest that enhanced fatty acid oxidation might be responsible for improvement in exercise capacity in TWEAK-KO mice. Data from cell culture experiments further support this possibility, as treatment of myotubes with TWEAK resulted in diminished maximal respiratory capacity and increased glycolytic flux (Figure 7).

Although the mechanisms by which TWEAK reduces levels of PGC-1α in skeletal muscle remain unknown, a recent study has shown that TWEAK increases membrane translocation of adaptor protein TNF receptor associated factor 2 (TRAF2), in an Fn14 dependent manner, in cardiomyocytes [59]. Furthermore, TWEAK treatment increases the activation of canonical nuclear factor-kappaB (NF-κB) signaling in both skeletal muscle [41] and cardiac myocytes [59]. Knockdown of TRAF2 using small hairpin RNA (shRNA) or selective blockade of IκB kinase-β (IKKβ, an upstream activator of canonical NF-κB signaling) prevented the TWEAK-mediated suppression of PGC-1α in cardiomyocytes suggesting that TWEAK repression of PGC-1α requires Fn14-TRAF2-IKKβ-NF-κB signaling cascade [59]. Previously, the effects of TNF-α on the mRNA expression of PPARδ and its target genes have been investigated. Treatment with TNF-α reduces PPARδ-target genes, such as mCPT1 and PGC-1α, whereas addition of PPARδ agonist rescues this reduction in adipocytes [71, 72]. Moreover, TNF-α impairs mitochondrial biogenesis and function in skeletal muscle [73]. While TWEAK can induce the expression of molecules associated with the autophagy-lysosomal system indicating it causes mitochondria dysfunction [74], muscle-specific ablation of TRAF6, which is involved in TWEAK signaling, suppresses the activation of autophagy in response to denervation and cancer cachexia [75]. These results suggest that TWEAK is involved in reducing PPARδ and PGC-1α levels and their target genes leading to mitochondrial dysfunction and content. Since no change of PPARδ expression was observed in skeletal muscle of PGC-1α KO mice or the muscle-specific PGC-1α overexpressing transgenic mice [66], PGC-1α is not an upstream regulator for PPARδ. Indeed, PGC-1α has a synergistic effect with PPARδ agonist to induce the expression of oxidative metabolic genes such as mCPT1 and PDK4[76]. Furthermore, it has been shown that PGC-1α directly coactivates the mCPT1 and PDK4 promoter via PPARδ in a ligand-dependent manner [76, 77]. Therefore, it is likely that PGC-1α is a coactivator of PPARδ in terms of up-regulating these metabolic genes.

Another interesting observation of the present study is that depletion of TWEAK improves angiogenesis in skeletal muscle of mice (Figure 8). Previous studies have shown that transgenic mice overexpressing PGC-1α show increased vascularization in skeletal muscle [56]. PGC-1α is also a positive regulator for the expression of VEGF1 which is known to promote angiogenesis [56]. Moreover, human endothelial cells treated with the PPARδ agonist GW501516 demonstrated increased production of VEGF and expression of VEGF receptor [78, 79]. Therefore, increased angiogenesis in skeletal muscle of TWEAK-KO mice appears to be a result of increased amounts of PGC-1α and PPAR δ (Figure 4). The enhancement of vascularization in TWEAK-KO mice might further augment the capacity of skeletal muscle mitochondria to respire by increasing blood flow and delivery of O2 and nutrients, especially during exercise.

Conclusion

In summary, our study provides initial evidence that inhibition of TWEAK increases mitochondrial biogenesis, oxidative metabolism and angiogenesis in skeletal muscle which potentially contribute to improved exercise capacity. These findings also have high clinical significance as inhibition of TWEAK activity using neutralizing antibodies or pharmacological compounds could improve muscle function and exercise capacity in patients with metabolic disorders.

Abbreviations

ATP5b:

ATP synthase subunit beta

BSA:

Bovine serum albumin

Cox:

Cytochrome c oxidase

FCCP:

Carbonyl cyanide p-trifluoromethoxyphenylhydrazone

GA:

Gastrocnemius

HK II:

Hexokinase II

KO:

Knockout

MCAD:

Medium-chain acyl-coenzyme A dehydrogenase

mCPT1:

Mitochondrial carnitine palmitoyltransferase I

MyHC:

Myosin heavy chain

NF-κB:

Nuclear factor-kappaB

OCR:

Oxygen consumption rate

PCR:

Polymerase chain reaction

PDK4:

Pyruvate dehydrogenase kinase 4

PGAM2:

Phosphoglycerate mutase 2

PGC-1α:

PPAR coactivator 1α

PPAR:

Peroxisome proliferator-activated receptor

qRT-PCR:

Quantitative real-time PCR

SDH:

Succinate dehydrogenase

shRNA:

Small hairpin RNA

TA:

Tibial anterior

TNF:

Tumor necrosis factor

TRAF:

TNF receptor-associated factor

TWEAK:

TNF-like weak inducer of apoptosis

VEGF:

Vascular endothelial growth factor.

References

  1. Pette D: The adaptive potential of skeletal muscle fibers. Can J Appl Physiol. 2002, 27: 423-448. 10.1139/h02-023.

    Article  PubMed  Google Scholar 

  2. Holloszy JO: Biochemical adaptations in muscle. Effects of exercise on mitochondrial oxygen uptake and respiratory enzyme activity in skeletal muscle. J Biol Chem. 1967, 242: 2278-2282.

    CAS  PubMed  Google Scholar 

  3. Lanza IR, Sreekumaran Nair K: Regulation of skeletal muscle mitochondrial function: genes to proteins. Acta Physiol (Oxf). 2010, 199: 529-547. 10.1111/j.1748-1716.2010.02124.x.

    Article  CAS  Google Scholar 

  4. Arany Z: PGC-1 coactivators and skeletal muscle adaptations in health and disease. Curr Opin Genet Dev. 2008, 18: 426-434. 10.1016/j.gde.2008.07.018.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  5. Finck BN, Kelly DP: PGC-1 coactivators: inducible regulators of energy metabolism in health and disease. J Clin Invest. 2006, 116: 615-622. 10.1172/JCI27794.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  6. Uldry M, Yang W, St-Pierre J, Lin J, Seale P, Spiegelman BM: Complementary action of the PGC-1 coactivators in mitochondrial biogenesis and brown fat differentiation. Cell Metab. 2006, 3: 333-341. 10.1016/j.cmet.2006.04.002.

    Article  CAS  PubMed  Google Scholar 

  7. Arany Z, Lebrasseur N, Morris C, Smith E, Yang W, Ma Y, Chin S, Spiegelman BM: The transcriptional coactivator PGC-1beta drives the formation of oxidative type IIX fibers in skeletal muscle. Cell Metab. 2007, 5: 35-46. 10.1016/j.cmet.2006.12.003.

    Article  CAS  PubMed  Google Scholar 

  8. Calvo JA, Daniels TG, Wang X, Paul A, Lin J, Spiegelman BM, Stevenson SC, Rangwala SM: Muscle-specific expression of PPARgamma coactivator-1alpha improves exercise performance and increases peak oxygen uptake. J Appl Physiol. 2008, 104: 1304-1312. 10.1152/japplphysiol.01231.2007.

    Article  CAS  PubMed  Google Scholar 

  9. Lin J, Wu H, Tarr PT, Zhang CY, Wu Z, Boss O, Michael LF, Puigserver P, Isotani E, Olson EN, Lowell BB, Bassel-Duby R, Spiegelman BM: Transcriptional co-activator PGC-1 alpha drives the formation of slow-twitch muscle fibres. Nature. 2002, 418: 797-801. 10.1038/nature00904.

    Article  CAS  PubMed  Google Scholar 

  10. Wende AR, Schaeffer PJ, Parker GJ, Zechner C, Han DH, Chen MM, Hancock CR, Lehman JJ, Huss JM, McClain DA, Holloszy JO, Kelly DP: A role for the transcriptional coactivator PGC-1alpha in muscle refueling. J Biol Chem. 2007, 282: 36642-36651. 10.1074/jbc.M707006200.

    Article  CAS  PubMed  Google Scholar 

  11. Sandri M, Lin J, Handschin C, Yang W, Arany ZP, Lecker SH, Goldberg AL, Spiegelman BM: PGC-1alpha protects skeletal muscle from atrophy by suppressing FoxO3 action and atrophy-specific gene transcription. Proc Natl Acad Sci U S A. 2006, 103: 16260-16265. 10.1073/pnas.0607795103.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  12. Brault JJ, Jespersen JG, Goldberg AL: Peroxisome proliferator-activated receptor gamma coactivator 1alpha or 1beta overexpression inhibits muscle protein degradation, induction of ubiquitin ligases, and disuse atrophy. J Biol Chem. 2010, 285: 19460-19471. 10.1074/jbc.M110.113092.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  13. Wu Z, Puigserver P, Andersson U, Zhang C, Adelmant G, Mootha V, Troy A, Cinti S, Lowell B, Scarpulla RC, Spiegelman BM: Mechanisms controlling mitochondrial biogenesis and respiration through the thermogenic coactivator PGC-1. Cell. 1999, 98: 115-124. 10.1016/S0092-8674(00)80611-X.

    Article  CAS  PubMed  Google Scholar 

  14. Mootha VK, Handschin C, Arlow D, Xie X, St Pierre J, Sihag S, Yang W, Altshuler D, Puigserver P, Patterson N, Willy PJ, Schulman IG, Heyman RA, Lander ES, Spiegelman BM: Erralpha and Gabpa/b specify PGC-1alpha-dependent oxidative phosphorylation gene expression that is altered in diabetic muscle. Proc Natl Acad Sci USA. 2004, 101: 6570-6575. 10.1073/pnas.0401401101.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  15. Huss JM, Kopp RP, Kelly DP: Peroxisome proliferator-activated receptor coactivator-1alpha (PGC-1alpha) coactivates the cardiac-enriched nuclear receptors estrogen-related receptor-alpha and -gamma. Identification of novel leucine-rich interaction motif within PGC-1alpha. J Biol Chem. 2002, 277: 40265-40274. 10.1074/jbc.M206324200.

    Article  CAS  PubMed  Google Scholar 

  16. Huss JM, Torra IP, Staels B, Giguere V, Kelly DP: Estrogen-related receptor alpha directs peroxisome proliferator-activated receptor alpha signaling in the transcriptional control of energy metabolism in cardiac and skeletal muscle. Mol Cell Biol. 2004, 24: 9079-9091. 10.1128/MCB.24.20.9079-9091.2004.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  17. 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. 10.1016/S0092-8674(00)81410-5.

    Article  CAS  PubMed  Google Scholar 

  18. Scarpulla RC: Transcriptional paradigms in mammalian mitochondrial biogenesis and function. Physiol Rev. 2008, 88: 611-638. 10.1152/physrev.00025.2007.

    Article  CAS  PubMed  Google Scholar 

  19. Romanello V, Guadagnin E, Gomes L, Roder I, Sandri C, Petersen Y, Milan G, Masiero E, Del Piccolo P, Foretz M, Scorrano L, Rudolf R, Sandri M: Mitochondrial fission and remodelling contributes to muscle atrophy. EMBO J. 2010, 29: 1774-1785. 10.1038/emboj.2010.60.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  20. Lokireddy S, Wijesoma IW, Teng S, Bonala S, Gluckman PD, McFarlane C, Sharma M, Kambadur R: The ubiquitin ligase Mul1 induces mitophagy in skeletal muscle in response to muscle-wasting stimuli. Cell Metab. 2012, 16: 613-624. 10.1016/j.cmet.2012.10.005.

    Article  CAS  PubMed  Google Scholar 

  21. Argiles JM, Busquets S, Toledo M, Lopez-Soriano FJ: The role of cytokines in cancer cachexia. Curr Opin Support Palliat Care. 2009, 3: 263-268. 10.1097/SPC.0b013e3283311d09.

    Article  PubMed  Google Scholar 

  22. Carson JA, Baltgalvis KA: Interleukin 6 as a key regulator of muscle mass during cachexia. Exerc Sport Sci Rev. 2010, 38: 168-176. 10.1097/JES.0b013e3181f44f11.

    Article  PubMed Central  PubMed  Google Scholar 

  23. Hardin BJ, Campbell KS, Smith JD, Arbogast S, Smith J, Moylan JS, Reid MB: TNF-alpha acts via TNFR1 and muscle-derived oxidants to depress myofibrillar force in murine skeletal muscle. J Appl Physiol. 2008, 104: 694-699. 10.1152/japplphysiol.00898.2007.

    Article  CAS  PubMed  Google Scholar 

  24. Reid MB, Lannergren J, Westerblad H: Respiratory and limb muscle weakness induced by tumor necrosis factor-alpha: involvement of muscle myofilaments. Am J Respir Crit Care Med. 2002, 166: 479-484. 10.1164/rccm.2202005.

    Article  PubMed  Google Scholar 

  25. Li YP, Schwartz RJ, Waddell ID, Holloway BR, Reid MB: Skeletal muscle myocytes undergo protein loss and reactive oxygen-mediated NF-kappaB activation in response to tumor necrosis factor alpha. FASEB J. 1998, 12: 871-880.

    CAS  PubMed  Google Scholar 

  26. Li YP, Reid MB: NF-kappaB mediates the protein loss induced by TNF-alpha in differentiated skeletal muscle myotubes. Am J Physiol Regul Integr Comp Physiol. 2000, 279: R1165-R1170.

    CAS  PubMed  Google Scholar 

  27. Reid MB, Moylan JS: Beyond atrophy: redox mechanisms of muscle dysfunction in chronic inflammatory disease. J Physiol. 2011, 589: 2171-2179. 10.1113/jphysiol.2010.203356.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  28. Romanatto T, Roman EA, Arruda AP, Denis RG, Solon C, Milanski M, Moraes JC, Bonfleur ML, Degasperi GR, Picardi PK, Hirabara S, Boschero AC, Curi R, Velloso LA: Deletion of tumor necrosis factor-alpha receptor 1 (TNFR1) protects against diet-induced obesity by means of increased thermogenesis. J Biol Chem. 2009, 284: 36213-36222. 10.1074/jbc.M109.030874.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  29. Tang K, Wagner PD, Breen EC: TNF-alpha-mediated reduction in PGC-1alpha may impair skeletal muscle function after cigarette smoke exposure. J Cell Physiol. 2010, 222: 320-327. 10.1002/jcp.21955.

    Article  CAS  PubMed  Google Scholar 

  30. White JP, Puppa MJ, Sato S, Gao S, Price RL, Baynes JW, Kostek MC, Matesic LE, Carson JA: IL-6 regulation on skeletal muscle mitochondrial remodeling during cancer cachexia in the ApcMin/+ mouse. Skelet Muscle. 2012, 2: 14-10.1186/2044-5040-2-14.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  31. White JP, Baltgalvis KA, Puppa MJ, Sato S, Baynes JW, Carson JA: Muscle oxidative capacity during IL-6-dependent cancer cachexia. Am J Physiol Regul Integr Comp Physiol. 2011, 300: R201-R211. 10.1152/ajpregu.00300.2010.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  32. Mangner N, Linke A, Oberbach A, Kullnick Y, Gielen S, Sandri M, Hoellriegel R, Matsumoto Y, Schuler G, Adams V: Exercise training prevents TNF-alpha induced loss of force in the diaphragm of mice. PLoS One. 2013, 8: e52274-10.1371/journal.pone.0052274.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  33. Santos RV, Viana VA, Boscolo RA, Marques VG, Santana MG, Lira FS, Tufik S, de Mello MT: Moderate exercise training modulates cytokine profile and sleep in elderly people. Cytokine. 2012, 60: 731-735. 10.1016/j.cyto.2012.07.028.

    Article  CAS  PubMed  Google Scholar 

  34. Rubin DA, Hackney AC: Inflammatory cytokines and metabolic risk factors during growth and maturation: influence of physical activity. Med Sport Sci. 2010, 55: 43-55.

    Article  CAS  PubMed  Google Scholar 

  35. Christiansen T, Bruun JM, Paulsen SK, Olholm J, Overgaard K, Pedersen SB, Richelsen B: Acute exercise increases circulating inflammatory markers in overweight and obese compared with lean subjects. Eur J Appl Physiol. 2013, 113: 1635-1642. 10.1007/s00421-013-2592-0.

    Article  CAS  PubMed  Google Scholar 

  36. Chicheportiche Y, Bourdon PR, Xu H, Hsu YM, Scott H, Hession C, Garcia I, Browning JL: TWEAK, a new secreted ligand in the tumor necrosis factor family that weakly induces apoptosis. J Biol Chem. 1997, 272: 32401-32410. 10.1074/jbc.272.51.32401.

    Article  CAS  PubMed  Google Scholar 

  37. Meighan-Mantha RL, Hsu DK, Guo Y, Brown SA, Feng SL, Peifley KA, Alberts GF, Copeland NG, Gilbert DJ, Jenkins NA, Richards CM, Winkles JA: The mitogen-inducible Fn14 gene encodes a type I transmembrane protein that modulates fibroblast adhesion and migration. J Biol Chem. 1999, 274: 33166-33176. 10.1074/jbc.274.46.33166.

    Article  CAS  PubMed  Google Scholar 

  38. Winkles JA, Tran NL, Brown SA, Stains N, Cunliffe HE, Berens ME: Role of TWEAK and Fn14 in tumor biology. Front Biosci. 2007, 12: 2761-2771. 10.2741/2270.

    Article  CAS  PubMed  Google Scholar 

  39. Wiley SR, Cassiano L, Lofton T, Davis-Smith T, Winkles JA, Lindner V, Liu H, Daniel TO, Smith CA, Fanslow WC: A novel TNF receptor family member binds TWEAK and is implicated in angiogenesis. Immunity. 2001, 15: 837-846. 10.1016/S1074-7613(01)00232-1.

    Article  CAS  PubMed  Google Scholar 

  40. Maecker H, Varfolomeev E, Kischkel F, Lawrence D, LeBlanc H, Lee W, Hurst S, Danilenko D, Li J, Filvaroff E, Yang B, Daniel D, Ashkenazi A: TWEAK attenuates the transition from innate to adaptive immunity. Cell. 2005, 123: 931-944. 10.1016/j.cell.2005.09.022.

    Article  CAS  PubMed  Google Scholar 

  41. Mittal A, Bhatnagar S, Kumar A, Lach-Trifilieff E, Wauters S, Li H, Makonchuk DY, Glass DJ, Kumar A: The TWEAK-Fn14 system is a critical regulator of denervation-induced skeletal muscle atrophy in mice. J Cell Biol. 2010, 188: 833-849. 10.1083/jcb.200909117.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  42. Kumar A, Bhatnagar S, Paul PK: TWEAK and TRAF6 regulate skeletal muscle atrophy. Curr Opin Clin Nutr Metab Care. 2012, 15: 233-239. 10.1097/MCO.0b013e328351c3fc.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  43. Dahiya S, Bhatnagar S, Hindi SM, Jiang C, Paul PK, Kuang S, Kumar A: Elevated levels of active matrix metalloproteinase-9 cause hypertrophy in skeletal muscle of normal and dystrophin-deficient mdx mice. Hum Mol Genet. 2011, 20: 4345-4359. 10.1093/hmg/ddr362.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  44. Safdar A, Little JP, Stokl AJ, Hettinga BP, Akhtar M, Tarnopolsky MA: Exercise increases mitochondrial PGC-1alpha content and promotes nuclear-mitochondrial cross-talk to coordinate mitochondrial biogenesis. J Biol Chem. 2011, 286: 10605-10617. 10.1074/jbc.M110.211466.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  45. Handschin C, Chin S, Li P, Liu F, Maratos-Flier E, Lebrasseur NK, Yan Z, Spiegelman BM: Skeletal muscle fiber-type switching, exercise intolerance, and myopathy in PGC-1alpha muscle-specific knock-out animals. J Biol Chem. 2007, 282: 30014-30021. 10.1074/jbc.M704817200.

    Article  CAS  PubMed  Google Scholar 

  46. Nachlas MM, Tsou KC, De Souza E, Cheng CS, Seligman AM: Cytochemical demonstration of succinic dehydrogenase by the use of a new p-nitrophenyl substituted ditetrazole. J Histochem Cytochem. 1957, 5: 420-436. 10.1177/5.4.420.

    Article  CAS  PubMed  Google Scholar 

  47. Sauerbeck A, Pandya J, Singh I, Bittman K, Readnower R, Bing G, Sullivan P: Analysis of regional brain mitochondrial bioenergetics and susceptibility to mitochondrial inhibition utilizing a microplate based system. J Neurosci Methods. 2011, 198: 36-43. 10.1016/j.jneumeth.2011.03.007.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  48. Hill BG, Dranka BP, Zou L, Chatham JC, Darley-Usmar VM: Importance of the bioenergetic reserve capacity in response to cardiomyocyte stress induced by 4-hydroxynonenal. Biochem J. 2009, 424: 99-107. 10.1042/BJ20090934.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  49. Dranka BP, Hill BG, Darley-Usmar VM: Mitochondrial reserve capacity in endothelial cells: the impact of nitric oxide and reactive oxygen species. Free Radic Biol Med. 2010, 48: 905-914. 10.1016/j.freeradbiomed.2010.01.015.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  50. Perez J, Hill BG, Benavides GA, Dranka BP, Darley-Usmar VM: Role of cellular bioenergetics in smooth muscle cell proliferation induced by platelet-derived growth factor. Biochem J. 2010, 428: 255-267. 10.1042/BJ20100090.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  51. Hill BG, Benavides GA, Lancaster JR, Ballinger S, Dell’italia L, Zhang J, Darley-Usmar VM: Integration of cellular bioenergetics with mitochondrial quality control and autophagy. Biol Chem. 2012, 393: 1485-1512.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  52. Tadaishi M, Miura S, Kai Y, Kano Y, Oishi Y, Ezaki O: Skeletal muscle-specific expression of PGC-1alpha-b, an exercise-responsive isoform, increases exercise capacity and peak oxygen uptake. PLoS One. 2011, 6: e28290-10.1371/journal.pone.0028290.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  53. Panguluri SK, Bhatnagar S, Kumar A, McCarthy JJ, Srivastava AK, Cooper NG, Lundy RF: Genomic profiling of messenger RNAs and microRNAs reveals potential mechanisms of TWEAK-induced skeletal muscle wasting in mice. PLoS One. 2010, 5: e8760-10.1371/journal.pone.0008760.

    Article  PubMed Central  PubMed  Google Scholar 

  54. Vega RB, Huss JM, Kelly DP: The coactivator PGC-1 cooperates with peroxisome proliferator-activated receptor alpha in transcriptional control of nuclear genes encoding mitochondrial fatty acid oxidation enzymes. Mol Cell Biol. 2000, 20: 1868-1876. 10.1128/MCB.20.5.1868-1876.2000.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  55. Liang H, Ward WF: PGC-1alpha: a key regulator of energy metabolism. Adv Physiol Educ. 2006, 30: 145-151. 10.1152/advan.00052.2006.

    Article  PubMed  Google Scholar 

  56. Chinsomboon J, Ruas J, Gupta RK, Thom R, Shoag J, Rowe GC, Sawada N, Raghuram S, Arany Z: The transcriptional coactivator PGC-1alpha mediates exercise-induced angiogenesis in skeletal muscle. Proc Natl Acad Sci U S A. 2009, 106: 21401-21406. 10.1073/pnas.0909131106.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  57. Mortensen OH, Frandsen L, Schjerling P, Nishimura E, Grunnet N: PGC-1alpha and PGC-1beta have both similar and distinct effects on myofiber switching toward an oxidative phenotype. Am J Physiol Endocrinol Metab. 2006, 291: E807-E816. 10.1152/ajpendo.00591.2005.

    Article  CAS  PubMed  Google Scholar 

  58. Leone TC, Lehman JJ, Finck BN, Schaeffer PJ, Wende AR, Boudina S, Courtois M, Wozniak DF, Sambandam N, Bernal-Mizrachi C, Chen Z, Holloszy JO, Medeiros DM, Schmidt RE, Saffitz JE, Abel ED, Semenkovich CF, Kelly DP: PGC-1alpha deficiency causes multi-system energy metabolic derangements: muscle dysfunction, abnormal weight control and hepatic steatosis. PLoS Biol. 2005, 3: e101-10.1371/journal.pbio.0030101.

    Article  PubMed Central  PubMed  Google Scholar 

  59. Shi J, Jiang B, Qiu Y, Guan J, Jain M, Cao X, Bauer M, Su L, Burkly LC, Leone TC, Kelly DP, Liao R: PGC1alpha plays a critical role in TWEAK-induced cardiac dysfunction. PLoS One. 2013, 8: e54054-10.1371/journal.pone.0054054.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  60. Kliewer SA, Xu HE, Lambert MH, Willson TM: Peroxisome proliferator-activated receptors: from genes to physiology. Recent Prog Horm Res. 2001, 56: 239-263. 10.1210/rp.56.1.239.

    Article  CAS  PubMed  Google Scholar 

  61. Takahashi S, Tanaka T, Sakai J: New therapeutic target for metabolic syndrome: PPARdelta. Endocr J. 2007, 54: 347-357. 10.1507/endocrj.KR-99.

    Article  CAS  PubMed  Google Scholar 

  62. Fredenrich A, Grimaldi PA: PPAR delta: an uncompletely known nuclear receptor. Diabetes Metab. 2005, 31: 23-27. 10.1016/S1262-3636(07)70162-3.

    Article  CAS  PubMed  Google Scholar 

  63. Muoio DM, MacLean PS, Lang DB, Li S, Houmard JA, Way JM, Winegar DA, Corton JC, Dohm GL, Kraus WE: Fatty acid homeostasis and induction of lipid regulatory genes in skeletal muscles of peroxisome proliferator-activated receptor (PPAR) alpha knock-out mice. Evidence for compensatory regulation by PPAR delta. J Biol Chem. 2002, 277: 26089-26097. 10.1074/jbc.M203997200.

    Article  CAS  PubMed  Google Scholar 

  64. Holst D, Luquet S, Nogueira V, Kristiansen K, Leverve X, Grimaldi PA: Nutritional regulation and role of peroxisome proliferator-activated receptor delta in fatty acid catabolism in skeletal muscle. Biochim Biophys Acta. 2003, 1633: 43-50. 10.1016/S1388-1981(03)00071-4.

    Article  CAS  PubMed  Google Scholar 

  65. Luquet S, Lopez-Soriano J, Holst D, Fredenrich A, Melki J, Rassoulzadegan M, Grimaldi PA: Peroxisome proliferator-activated receptor delta controls muscle development and oxidative capability. FASEB J. 2003, 17: 2299-2301.

    CAS  PubMed  Google Scholar 

  66. Wang YX, Zhang CL, Yu RT, Cho HK, Nelson MC, Bayuga-Ocampo CR, Ham J, Kang H, Evans RM: Regulation of muscle fiber type and running endurance by PPARdelta. PLoS Biol. 2004, 2: e294-10.1371/journal.pbio.0020294.

    Article  PubMed Central  PubMed  Google Scholar 

  67. Hawley JA, Brouns F, Jeukendrup A: Strategies to enhance fat utilisation during exercise. Sports Med. 1998, 25: 241-257. 10.2165/00007256-199825040-00003.

    Article  CAS  PubMed  Google Scholar 

  68. Helge JW: Long-term fat diet adaptation effects on performance, training capacity, and fat utilization. Med Sci Sports Exerc. 2002, 34: 1499-1504. 10.1097/00005768-200209000-00016.

    Article  CAS  PubMed  Google Scholar 

  69. Miller WC, Bryce GR, Conlee RK: Adaptations to a high-fat diet that increase exercise endurance in male rats. J Appl Physiol. 1984, 56: 78-83.

    CAS  PubMed  Google Scholar 

  70. Haramizu S, Nagasawa A, Ota N, Hase T, Tokimitsu I, Murase T: Different contribution of muscle and liver lipid metabolism to endurance capacity and obesity susceptibility of mice. J Appl Physiol. 2009, 106: 871-879. 10.1152/japplphysiol.90804.2008.

    Article  CAS  PubMed  Google Scholar 

  71. Rodriguez-Calvo R, Serrano L, Coll T, Moullan N, Sanchez RM, Merlos M, Palomer X, Laguna JC, Michalik L, Wahli W, Vazquez-Carrera M: Activation of peroxisome proliferator-activated receptor beta/delta inhibits lipopolysaccharide-induced cytokine production in adipocytes by lowering nuclear factor-kappaB activity via extracellular signal-related kinase 1/2. Diabetes. 2008, 57: 2149-2157. 10.2337/db08-0176.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  72. Serrano-Marco L, Chacon MR, Maymo-Masip E, Barroso E, Salvado L, Wabitsch M, Garrido-Sanchez L, Tinahones FJ, Palomer X, Vendrell J, Vazquez-Carrera M: TNF-alpha inhibits PPARbeta/delta activity and SIRT1 expression through NF-kappaB in human adipocytes. Biochim Biophys Acta. 2012, 1821: 1177-1185. 10.1016/j.bbalip.2012.05.006.

    Article  CAS  PubMed  Google Scholar 

  73. Valerio A, Cardile A, Cozzi V, Bracale R, Tedesco L, Pisconti A, Palomba L, Cantoni O, Clementi E, Moncada S, Carruba MO, Nisoli E: TNF-alpha downregulates eNOS expression and mitochondrial biogenesis in fat and muscle of obese rodents. J Clin Invest. 2006, 116: 2791-2798.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  74. Bhatnagar S, Mittal A, Gupta SK, Kumar A: TWEAK causes myotube atrophy through coordinated activation of ubiquitin-proteasome system, autophagy, and caspases. J Cell Physiol. 2012, 227: 1042-1051. 10.1002/jcp.22821.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  75. Paul PK, Gupta SK, Bhatnagar S, Panguluri SK, Darnay BG, Choi Y, Kumar A: Targeted ablation of TRAF6 inhibits skeletal muscle wasting in mice. J Cell Biol. 2010, 191: 1395-1411. 10.1083/jcb.201006098.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  76. Kleiner S, Nguyen-Tran V, Bare O, Huang X, Spiegelman B, Wu Z: PPAR{delta} agonism activates fatty acid oxidation via PGC-1{alpha} but does not increase mitochondrial gene expression and function. J Biol Chem. 2009, 284: 18624-18633. 10.1074/jbc.M109.008797.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  77. Dressel U, Allen TL, Pippal JB, Rohde PR, Lau P, Muscat GE: The peroxisome proliferator-activated receptor beta/delta agonist, GW501516, regulates the expression of genes involved in lipid catabolism and energy uncoupling in skeletal muscle cells. Mol Endocrinol. 2003, 17: 2477-2493. 10.1210/me.2003-0151.

    Article  CAS  PubMed  Google Scholar 

  78. Piqueras L, Reynolds AR, Hodivala-Dilke KM, Alfranca A, Redondo JM, Hatae T, Tanabe T, Warner TD, Bishop-Bailey D: Activation of PPARbeta/delta induces endothelial cell proliferation and angiogenesis. Arterioscler Thromb Vasc Biol. 2007, 27: 63-69. 10.1161/01.ATV.0000250972.83623.61.

    Article  CAS  PubMed  Google Scholar 

  79. Stephen RL, Gustafsson MC, Jarvis M, Tatoud R, Marshall BR, Knight D, Ehrenborg E, Harris AL, Wolf CR, Palmer CN: Activation of peroxisome proliferator-activated receptor delta stimulates the proliferation of human breast and prostate cancer cell lines. Cancer Res. 2004, 64: 3162-3170. 10.1158/0008-5472.CAN-03-2760.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We are grateful to Dr. Avi Ashkenazi (Genentech South San Francisco, CA) for providing TWEAK-KO mice. This work was supported by NIH grants R01AR059810 and RO1AG029623 to AK.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Ashok Kumar.

Additional information

Competing interests

The authors declare they have no competing interests.

Authors’ contributions

AK and BGH conceived and designed the study. SS, YO, VM, JS, BGH and SB performed experiments and analyzed the data. SS, BGH and AK wrote the manuscript. All authors read and approved the final manuscript.

Authors’ original submitted files for images

Rights and permissions

Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://creativecommons.org/licenses/by/2.0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Sato, S., Ogura, Y., Mishra, V. et al. TWEAK promotes exercise intolerance by decreasing skeletal muscle oxidative phosphorylation capacity. Skeletal Muscle 3, 18 (2013). https://doi.org/10.1186/2044-5040-3-18

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/2044-5040-3-18

Keywords