Journal List > Korean J Physiol Pharmacol > v.21(6) > 1026208

Heo, No, Park, Kang, Seo, Han, Neufer, and Kwak: Effects of exercise on obesity-induced mitochondrial dysfunction in skeletal muscle

Abstract

Obesity is known to induce inhibition of glucose uptake, reduction of lipid metabolism, and progressive loss of skeletal muscle function, which are all associated with mitochondrial dysfunction in skeletal muscle. Mitochondria are dynamic organelles that regulate cellular metabolism and bioenergetics, including ATP production via oxidative phosphorylation. Due to these critical roles of mitochondria, mitochondrial dysfunction results in various diseases such as obesity and type 2 diabetes. Obesity is associated with impairment of mitochondrial function (e.g., decrease in O2 respiration and increase in oxidative stress) in skeletal muscle. The balance between mitochondrial fusion and fission is critical to maintain mitochondrial homeostasis in skeletal muscle. Obesity impairs mitochondrial dynamics, leading to an unbalance between fusion and fission by favorably shifting fission or reducing fusion proteins. Mitophagy is the catabolic process of damaged or unnecessary mitochondria. Obesity reduces mitochondrial biogenesis in skeletal muscle and increases accumulation of dysfunctional cellular organelles, suggesting that mitophagy does not work properly in obesity. Mitochondrial dysfunction and oxidative stress are reported to trigger apoptosis, and mitochondrial apoptosis is induced by obesity in skeletal muscle. It is well known that exercise is the most effective intervention to protect against obesity. Although the cellular and molecular mechanisms by which exercise protects against obesity-induced mitochondrial dysfunction in skeletal muscle are not clearly elucidated, exercise training attenuates mitochondrial dysfunction, allows mitochondria to maintain the balance between mitochondrial dynamics and mitophagy, and reduces apoptotic signaling in obese skeletal muscle.

INTRODUCTION

Obesity, which has emerged as the largest health problem in modern society, induces numerous diseases such as cardiovascular disease (CVD), type 2 diabetes (insulin resistance), neurodegenerative diseases, and cancer [1]. Skeletal muscle is the largest organ of the human body, accounting for 40~50% of human body mass. In skeletal muscle, obesity is related to decreased glucose uptake, abnormal protein turnover, dysregulation of lipid metabolism, and mitochondrial dysfunction [234]. Specifically, over the last decade, many studies have demonstrated that mitochondrial contents and oxidative capacity are reduced while oxidative stress and intramyocellular triglyceride levels are increased in metabolic diseases such as obesity-induced models in skeletal muscle [56789].
Mitochondria are dynamic organelles that regulate cellular functions such as cellular respiration, calcium homeostasis, and reactive oxygen species (ROS) production. The most important role of mitochondria is energy metabolism, which involves production of adenosine triphosphate (ATP) through oxidative phosphorylation in skeletal muscle mitochondria [10]. In addition, mitochondria have a morphological and structural cycle referred to as mitochondrial dynamics (fusion, fission, and mitophagy) [11]. Due to these critical roles of mitochondria, mitochondrial dysfunction is directly or indirectly associated with various diseases, ranging from mild to severe diseases such as obesity and type 2 diabetes. In support of this notion, mitochondrial O2 respiration was shown to be reduced and mitochondrial ROS emission increased in high fat diet-induced obesity models [12]. Under obese conditions, the mitochondrial fusion and fission balance is altered, favorably shifting to fission [8], whereas mitophagy-related protein levels are decreased [13]. Furthermore, obesity induces apoptosis (programmed cell death), resulting in an increase in pro-apoptotic proteins and decrease in anti-apoptotic proteins in skeletal muscle mitochondria [14].
Although mitochondrial function, mitochondrial dynamics (fusion and fission), mitophagy, and apoptosis are exacerbated by obesity, the underlying mechanisms by which obesity induces mitochondrial dysfunction, imbalance of mitochondrial dynamics, mitophagy, and mitochondrial-mediated apoptosis in skeletal muscle have not been clearly elucidated. Therefore, this review paper is mainly focused on the potential mechanisms by which obesity affects mitochondrial function, dynamics, mitophagy, and apoptosis, including the role of exercise in obesity-related mitochondrial impairment in skeletal muscle.

OBESITY AND MITOCHONDRIAL FUNCTION IN SKELETAL MUSCLE

Obesity and mitochondrial function

Mitochondrial function includes various metabolic factors, including control of oxidative stress, cellular respiration, calcium homeostasis, as well as the production of ATP. This review focuses on mitochondrial O2 respiration and mitochondrial ROS production in skeletal muscle.
Mitochondria play a key role in the production of ATP, phosphorylating ADP in complex V (ATP synthase) of the electron transport chain (ETC). ATP plays an essential role in various organs such as skeletal muscle. To generate ATP, mitochondrial respiration operates via complex IV of the ETC through oxidative phosphorylation. Mitochondrial O2 respiration is directly associated with mitochondrial function. Thus, decreased mitochondrial respiration may entail mitochondrial dysfunction, including reduced ATP production. Paradoxically, during production of ATP, ROS (i.e., superoxide) can be generated by complexes I and III of the ETC, and mitochondrial superoxide (O2) can be converted into hydrogen peroxide (H2O2) by Mn superoxide dismutase (MnSOD). Finally, superoxide and hydrogen peroxide can damage various macromolecules such as DNA, lipids, and proteins [15]. However, several studies reported that the mitochondrial H2O2 emission level is physiologically considered to be an essential marker of mitochondrial function and the cellular redox environment [161718]. Recently, Shadel and Horvath [19] demonstrated that appropriate emission of ROS positively plays vital role in cell proliferation, differentiation, and adaptive responses. In contrast, excessive oxidative stress may induce cellular damage as well as mitochondrial damage, resulting in release of cytochrome c as an electron transporter of mitochondrial complexes III and IV. Eventually, this released cytochrome c induces apoptosis, facilitating expression of pro-apoptotic proteins such as Bax and inhibition of anti-apoptotic proteins such as Bcl-2 [20]. Furthermore, mitochondrial DNA (mtDNA) is more susceptible to oxidative stress due to its localization near to the ETC than nuclear DNA and possesses a less efficient defense system [21]. In particular, mtDNA mutations synthesize defective ETC components, resulting in impaired oxidative phosphorylation, reduced ATP production, and ROS production. Given these conflicting views, ROS can play a negative or positive role in cells depending on their abundance.
Obesity is associated with impairment of mitochondrial function, including reduced mitochondrial O2 respiration and ATP production as well as increased mitochondrial ROS emission (Fig. 1). Table 1 summarizes the effects of obesity on mitochondrial function in skeletal muscle. For example, Bonnard et al. [12] reported that mitochondrial respiration was reduced upon consumption of a high fat and high sucrose diet for 16 weeks, demonstrating that mitochondrial state 3 respiration is reduced in permeabilized muscle. Recently, Konopka et al. [22] reported that the respiratory control ratio (RCR) was reduced in obese women compared with lean women. Additionally, in obese skeletal muscle, excessive oxidative stress and ROS production were found to be a major risk factors of skeletal muscle atrophy and mitochondrial dysfunction [23]. Anderson et al. [3] reported that lipid accumulation played a major role in the elevation of mitochondrial H2O2 emission in permeabilized skeletal muscle with high fat diet-induced rodent model for 3 days and 3 weeks compared with the standard chow group. Further, almost 2-fold higher H2O2 emission was detected in obese individuals versus lean individuals, which suggests that excessive ROS can negatively alter mitochondrial function. In rodents fed a high fat diet for 16 weeks, the ratio of mtDNA to nuclear DNA in skeletal muscle was significantly reduced compared with their standard chow counterparts [12].

Exercise and obesity-induced mitochondrial dysfunction

It is well known that obesity is a strongly associated with impairment of mitochondrial function in skeletal muscle. However, the relationship between exercise training as an effective remedy and obesity-induced mitochondrial dysfunction in skeletal muscle has not been well studied. Fig. 1 shows the potential role of exercise in obesity-induced mitochondrial dysfunction in skeletal muscle. In addition, Table 2 summarizes the effects of exercise training on mitochondrial function, dynamics, and apoptosis in obese skeletal muscle. Konopka et al. [22] reported that reduction of mitochondrial RCR and O2 respiration by obesity was attenuated by aerobic exercise training for 10 weeks, and conversely, increased H2O2 emission induced by obesity was reduced by aerobic exercise training via increased catalase activity and myocellular antioxidant production. In addition, 8-oxo-2′deoxyguanosine, a marker of DNA oxidative damage, was shown to be reduced by exercise training [22], whereas uncoupling protein isotype 3 (UCP3) protein, which plays a protective role against ROS emission [24], was up-regulated by 72% in the aerobic exercise training group compared with non-exercise training group in obese individuals [25]. Li et al. [26] also reported an increased superoxide anion level in obese rats fed a high fat diet for 12 weeks. However, exercise training for 8 weeks attenuated superoxide anion levels in obese skeletal muscle.

OBESITY AND MITOCHONDRIAL DYNAMICS IN SKELETAL MUSCLE

Obesity and mitochondrial dynamics (fusion and fission)

Skeletal muscle mitochondria, regarded as dynamic organelles, undergo a constant structural and morphological cycle involving fusion and fission, which are essential for cell survival as well as cell growth and division during cell differentiation [27]. Mitochondrial fusion can compensate for damaged mitochondria by binding damaged mitochondria to healthy mitochondria, whereas mitochondria fission can maintain mitochondrial function by separating damaged mitochondrial sites from healthy mitochondria [28].
Mitochondrial fusion plays essential role in the regulating the fusion proteins Mitofusins 1 and 2 (Mfn1 and Mfn2) as well as Optic atrophy 1 (Opa1). Mfn1 and Mfn2, which are dynaminrelated GTPases, are responsible for the fusion of mitochondrial outer membranes while Opa1, also a dynamin-related GTPase, is recruited for the fusion of mitochondrial inner membranes and regulates cristae remodeling [28]. Mitochondrial fission is largely mediated by dynamin-related protein 1 (Drp-1), which is mostly localized in the cytoplasm [29] and interacts with several mitochondrial outer membrane receptors such as mitochondrial fission factor (MFF), fission protein 1 (Fis1), and mitochondrial dynamics proteins (Mid49/51) when mitochondria are damaged by loss of membrane potential or oxidative stress [3031]. To generate the fission process, Drp1 is recruited from the cytosol to the dysfunctional site to cleave the damaged mitochondrial site through the receptors Fis1, Mff, and Mid49/51 [3132].
The balance between mitochondrial fusion and fission is important for maintaining mitochondrial health in skeletal muscle (Fig. 2). However, obesity impairs mitochondrial dynamics and alters the balance between mitochondrial fusion and fission, thereby reducing mitochondrial contents and inducing mitochondrial dysfunction in skeletal muscle [333435]. Table 3 summarizes the effects of obesity on mitochondrial dynamics (fusion, fission, and mitophagy) in skeletal muscle. Particularly, a recent study reported that inhibition of Mfn2 is related to diminished substrate oxidation, cellular metabolism, and reduction of membrane potential in ETC complexes under obese conditions [34]. In addition, Liu et al. [8] reported that high fat diet consumption for 40 weeks reduced both Mfn1 and Mfn2 protein levels in skeletal muscle by 20%, whereas protein levels of Fis1 and Drp1 were elevated by 50%. Furthermore, Jheng et al. [33] reported that mitochondrial fusion protein (Mfn1, Mfn2, and Opa1) levels were unaltered while mitochondrial fission protein (Drp1 and Fis1) levels were significantly increased in genetically induced obese mice (ob/ob) and high fat diet-induced obese mice compared with lean mice, demonstrating the unbalance between fusion and fission in obesity.

Exercise and obesity-induced mitochondrial dynamics

The balance between mitochondrial fusion and fission can be broken through lipid accumulation, which induces mitochondrial dysfunction such as loss of mitochondrial membrane potential, reduction of oxygen consumption, and elevation of ROS production. Although several studies have reported that exercise training maintains the balance between mitochondrial fusion and fission under normal conditions [3637], few studies have assessed the impact of exercise training on obesity-induced dysfunction of mitochondrial dynamics in skeletal muscles.
Recently, one study reported that exercise training attenuates obesity-induced imbalance between mitochondrial fusion and fission through increasing mitochondrial fusion proteins and decreasing or maintaining mitochondrial fission protein levels [13]. In particular, Mfn2 and Opa1 expression levels were shown to be elevated in the physical activity group for promotion of mitochondrial fusion, whereas mitochondrial fission protein expression levels were unaltered in the physical activity group, suggesting a shift to mitochondrial fusion [13]. This study demonstrated that physical activity maintains healthy mitochondria under obese conditions. Although only one study has demonstrated the relationship between mitochondrial dynamics altered by obese conditions and physical activity, more studies are needed on whether exercise training has positive or negative effects on cellular and molecular levels.

OBESITY AND MITOPHAGY IN SKELETAL MUSCLE

Obesity and mitophagy

Autophagy is the process of catabolism and the removal of damaged or unnecessary cellular proteins or organelles. Excessive autophagy or defective autophagy is associated with skeletal muscle atrophy [38]. In other words, excessive autophagy induces cellular stress and causes skeletal muscle loss through increased protein degradation, whereas deficiency of intracellular autophagy leads to accumulation of abnormal proteins.
Mitochondria in skeletal muscle damaged by obesity lose their membrane potential and are self-eliminated by an autophagy process known as mitophagy (mitochondria+autophagy). Specifically, mitophagy is the process of catabolism and the removal of damaged or unnecessary mitochondria. Mitophagy also plays an important role in mitochondrial quality control for protection against mitochondrial dysfunction that has undergone inefficient oxidative phosphorylation and emit more oxidative byproducts [3940]. After damaged mitochondria lose their membrane potential, autophagocytosis selectively mediates a catabolic process.
According to a recent study, mitophagy consists of two signaling pathways: Parkin-dependent mitophagy, which is centered on Parkin protein, and Parkin-independent mitophagy, which occurs regardless of Parkin protein expression [28] (Fig. 3). Parkin, which is an E3 ubiquitin ligase, and Pink1, which is a mitochondrial serine/threonine kinase, are cleaved by PARL (Presenilinsassociated-rhomboid-like) [41] and play central roles in the mitochondrial Parkin-dependent pathway [42]. Upon loss of mitochondrial membrane potential, Pink1 is no longer cleaved by PARL and becomes stabilized in the mitochondrial outer membrane [41]. Stabilized Pink1 recruits Parkin from the cytoplasm to the mitochondrial outer membrane. Recruitment of Parkin mediates ubiquitination of mitochondrial outer membrane proteins such as Mfn1, Mfn2, Mitochondrial Rho GTPase 1 (Miro-1), Translocase of Outer Mitochondrial Membrane (TOMM7), and Voltage-Dependent Anion Channel (VDAC) to promote mitophagy [4344]. Parkin-mediated ubiquitination results in recruitment of p62 and optineurin, an autophagy adapter protein, whereupon p62 interacts with LC3 [4546]. LC3 participates in autophagosome formation, which results in lysosomal clearance of damaged mitochondria in the cytoplasm [47].
Recent studies have reported that mitophagy can be mediated even without Parkin. Some autophagy receptor proteins such as Bcl-2/adenovirus E1B Interacting Protein 3 (BNIP3), Nip3-like Protein X (NIX), and Fun14 Domain-Containing Protein 1 (FUNDC1) participate in the regulation of mitophagy. BNIP3, NIX, and FUNDC1 directly interact with LC3, promoting mitophagy through autophagosome formation for destruction of damaged mitochondria [48]. Once mitochondria are damaged or lose membrane potential, cardiolipin synthesized in the mitochondrial inner membrane shifts to outer membrane to induce mitophagy through LC3 [4950]. In addition, SMURF1, another E3 ubiquitin ligase, is involved in the removal of damaged mitochondria by autophagosomes during mitophagy [51].
Although many studies have reported that mitophagy is associated with various diseases such as aging [5253], mitophagy in obese skeletal muscle has been rarely studied. However, as obesity reduces mitochondrial biogenesis in skeletal muscle and is associated with accumulation of dysfunctional cellular organelles, it can be assumed that mitophagy does not function properly during obesity (Fig. 4). In support of this assumption, Greene et al. [13] recently demonstrated that the level of p62, an autophagy adapter protein, was lower in those who consumed a Western diet. However, to elucidate the mechanism underlying the relationship between mitophagy and obesity, additional research is needed.

Exercise and obesity-induced mitophagy

Exercise training is effective for maintaining emission of autophagy proteins and may even facilitate expression of skeletal muscle autophagy proteins, demonstrating the beneficial role of exercise training [36]. In support of this notion, it was recently demonstrated that exercise training can induce autophagy in normal skeletal muscle [3637]. Similarly, many studies have already assessed the relationship between mitophagy and exercise training. However, few studies have examined the role of exercise in obesity-related mitophagy. Recently, one study investigated mitochondrial quality control associated with obesity, suggesting that obesity-induced impairment of mitophagy was protected by physical activity [13].

OBESITY AND MITOCHONDRIAL APOPTOSIS IN SKELETAL MUSCLE

Obesity and mitochondrial apoptosis

Mitochondrial dysfunction and oxidative stress have been reported to induce apoptosis [54]. Specifically, mitochondria are the major sites that induce apoptosis [55]. Cytochrome c is released from mitochondria into the cytoplasm through mitochondrial permeability transition pore (mPTP) openings induced by mitochondrial imbalance between pro-apoptotic proteins (Bax, Bid) and anti-apoptotic proteins (Bcl-2, Bcl-XL). The released cytochrome c binds to apoptotic protease-activating factor-1 (Apaf-1), dATP, and pro-caspase-9, activating caspase-3 and eventually causing DNA fragmentation, a hallmark of apoptosis [56]. In addition, as a caspase-independent pathway, apoptosis-inducing factor (AIF) and endonuclease G (Endo G) in mitochondria cause direct DNA fragmentation without caspase-mediated apoptosis of mitochondria [5758].
Previous studies have demonstrated that obesity induces mitochondrial apoptosis in skeletal muscle (Fig. 5). Table 4 summarizes the effects of obesity on mitochondrial apoptosis in skeletal muscle. For example, in rats fed a high fat diet, levels of cleaved caspase-3, a pro-apoptotic protein, were shown to be significantly elevated compared with rats fed a low-fat diet [59]. In addition, the high fat diet rats showed high levels of cytochrome c and cleaved caspase-3 compared with the normal chow diet rats [6061], and the Bax/Bcl-2 ratio and apoptotic nuclei were elevated in high fat diet-induced obese mice compared with control mice [14]. Moreover, mitochondrial DNA (mtDNA) to nuclear DNA ratio in skeletal muscle was significantly reduced in mice fed a high fat diet for 16 weeks [12]. However, there are contradictory studies showing that mitochondrial apoptotic signaling molecules, including Bax, Bcl-2, cytochrome c, caspase-9, caspase-3, and DNA fragmentation factors, were unaltered in an obese Zucker rat model [6263]. In addition, Peterson et al. [63] demonstrated that the AIF level is unaltered by obesity. Therefore, due to these contradictory studies, further research is needed to establish the precise mechanism between obesity and apoptotic signaling.

Exercise and obesity-induced mitochondrial apoptosis

Although the topic remains controversial, many studies have reported that apoptotic signaling is induced by obesity based on up-regulation of pro-apoptotic proteins and down-regulation of anti-apoptotic proteins in the context of high fat diet-induced obesity. Indeed, there have been many studies showing the effects of exercise training on mitochondrial apoptosis in aging and various disease states [646566]. However, only one study has assessed the relationship between exercise training and apoptosis induced by obesity and demonstrated that apoptotic signaling induced by obesity is not altered by exercise training in skeletal muscle [62]. Therefore, the beneficial or detrimental role of exercise training on apoptotic signaling in obese skeletal muscle should be further studied.

CONCLUSION

High fat diet-induced obesity is a potential cause of mitochondrial dysfunction, increased oxidative stress, impaired mitochondrial dynamics (fusion and fission), mitophagy dysfunction, and increased mitochondrial apoptosis. Although more research is needed, exercise training might improve mitochondrial function, mitochondrial dynamics, mitophagy, and anti-apoptotic signaling in obese skeletal muscle. In order to elucidate the cellular and molecular mechanisms by which exercise training improves mitochondrial function, including dynamics, mitophagy, and apoptosis in obese skeletal muscle, more studies should be performed.

Figures and Tables

Fig. 1

Effects of obesity and exercise training on mitochondrial dysfunction.

Exercise training protects against obesity-induced mitochondrial dysfunction (e.g., O2 respiration, ATP production, ROS emission, β-oxidation, markers of TCA cycle, mtDNA mutation) in skeletal muscle. TCA cycle, tricarboxylic acid cycle; ROS, reactive oxygen species; ADP, adenosine diphosphate; ATP, adenosine triphosphate.
kjpp-21-567-g001
Fig. 2

Schematic overview of mitochondrial dynamics impaired by obesity and adaptation to exercise training.

Obesity impairs mitochondrial membrane potential and triggers oxidative stress, resulting in imbalance of mitochondrial fusion and fission and elevation of fission proteins. However, exercise training allows mitochondria to maintain the balance between fusion and fission by up-regulating fusion proteins and down-regulating fission proteins. ↓, decrease; ↑, increase; =, no change.
kjpp-21-567-g002
Fig. 3

Mitophagy pathways, including (i) Parkin-dependent pathway and (ii) Parkin-independent pathway.

In the Parkin-dependent pathway, Pink1 recruits Parkin from the cytoplasm to mitochondrial outer membrane. After Parkin has been recruited to mitochondria, activated Parkin ubiquitinates proteins in the mitochondrial outer membrane such as MFN and VDAC (Voltage-Dependent Anion Channel) to facilitate mitophagy. Parkinmediated ubiquitination recruits autophagy adapter proteins such as p62 and optineurin, and these proteins interact with LC3. LC3 participates in formation of an autophagosome, which is fated for lysosomal destruction to clear out damaged mitochondrion. The second pathway called the Parkin-independent pathway is generated without Parkin protein. When mitochondria are damaged, several autophagy receptor proteins such as BNIP3, NIX, and FUNDC1 are recruited to regulate mitophagy. These autophagy receptors directly interact with LC3 to form an autophagosome and degrade damaged mitochondria in the lysosome. ↓, decrease; ↑, increase; PARL, presenilins-associated-rhomboid-like; VDAC, Voltage-Dependent Anion Channel; MFN, mitofusin; UB, ubiquitination; BNIP3, bcl-2/adenovirus E1B interacting protein 3; NIX, nip3-like protein x; FUNDC1, fun14 domain-containing protein 1.
kjpp-21-567-g003
Fig. 4

Potential mechanisms of obesity-induced impairment in mitophagy.

Obesity may induce defective or excessive mitophagy. Specifically, excessive mitophagy induces cellular/mitochondrial stress and causes skeletal muscle loss through increased protein degradation, whereas deficiency of mitophagy leads to accumulation of dysfunctional mitochondria in skeletal muscle.
kjpp-21-567-g004
Fig. 5

Schematic overview of two obesity-induced apoptotic signaling pathways, including caspase-dependent pathway and caspase-independent pathway.

As a caspase-dependent pathway, obesity increases the Bax/Bcl-2 ratio and facilitates mPTP opening. Upon mPTP opening, cytochrome c is released from mitochondria to the cytosol. Released cytochrome c activates caspase-9 and cleaves caspase-3. Cleaved caspase-3 induces DNA fragmentation, leading to apoptosis. The relationship between obesity and the caspase-independent pathway has been rarely studied. ↓, decrease; ↑, increase; mPTP, mitochondrial permeability transition pore; AIF, apoptosis-inducing factor; Endo G, endonuclease G.
kjpp-21-567-g005
Table 1

Effects of obesity on mitochondrial function in skeletal muscle

kjpp-21-567-i001

RCR, respiratory control ratio; CS, citrate synthase; Mfn2, mitofusins 2; H2O2, hydrogen peroxide; ↓, decrease; ↑, increase.

Table 2

Effects of exercise training on mitochondrial function, dynamics, and apoptosis in obese skeletal muscle

kjpp-21-567-i002

CS, citrate synthase; RCR, respiratory control ratio; H2O2, hydrogen peroxide; UCP3, uncoupling protein 3; ↓, decrease; ↑, increase; =, no change.

Table 3

Effects of obesity on mitochondrial dynamics (fusion, fission, and mitophagy) in skeletal muscle

kjpp-21-567-i003

Mfn 1,2, mitofusins 1,2; OPA 1, optic atrophy 1; Drp 1, Dynamin-1-like protein; Fis 1, fission protein 1; ↓, decrease; ↑, increase; =, no change.

Table 4

Effects of obesity on mitochondrial apoptosis in skeletal muscle

kjpp-21-567-i004

AIF, apoptosis-inducing factor; Apaf-1, apoptotic protease-activating factor-1; ↓, decrease; ↑, increase; =, no change.

ACKNOWLEGEMENTS

This work was supported by the Ministry of Education of the Republic of Korea and the National Research Foundation of Korea (NRF-2015S1A2A1A01026121) and WCSL at Inha University.

Notes

CONFLICTS OF INTEREST The authors declare no conflicts of interest.

References

1. Ng M, Fleming T, Robinson M, Thomson B, Graetz N, Margono C, Mullany EC, Biryukov S, Abbafati C, Abera SF, Abraham JP, Abu-Rmeileh NM, Achoki T, AlBuhairan FS, Alemu ZA, Alfonso R, Ali MK, Ali R, Guzman NA, Ammar W, Anwari P, Banerjee A, Barquera S, Basu S, Bennett DA, Bhutta Z, Blore J, Cabral N, Nonato IC, Chang JC, Chowdhury R, Courville KJ, Criqui MH, Cundiff DK, Dabhadkar KC, Dandona L, Davis A, Dayama A, Dharmaratne SD, Ding EL, Durrani AM, Esteghamati A, Farzadfar F, Fay DF, Feigin VL, Flaxman A, Forouzanfar MH, Goto A, Green MA, Gupta R, Hafezi-Nejad N, Hankey GJ, Harewood HC, Havmoeller R, Hay S, Hernandez L, Husseini A, Idrisov BT, Ikeda N, Islami F, Jahangir E, Jassal SK, Jee SH, Jeffreys M, Jonas JB, Kabagambe EK, Khalifa SE, Kengne AP, Khader YS, Khang YH, Kim D, Kimokoti RW, Kinge JM, Kokubo Y, Kosen S, Kwan G, Lai T, Leinsalu M, Li Y, Liang X, Liu S, Logroscino G, Lotufo PA, Lu Y, Ma J, Mainoo NK, Mensah GA, Merriman TR, Mokdad AH, Moschandreas J, Naghavi M, Naheed A, Nand D, Narayan KM, Nelson EL, Neuhouser ML, Nisar MI, Ohkubo T, Oti SO, Pedroza A, Prabhakaran D, Roy N, Sampson U, Seo H, Sepanlou SG, Shibuya K, Shiri R, Shiue I, Singh GM, Singh JA, Skirbekk V, Stapelberg NJ, Sturua L, Sykes BL, Tobias M, Tran BX, Trasande L, Toyoshima H, van de Vijver S, Vasankari TJ, Veerman JL, Velasquez-Melendez G, Vlassov VV, Vollset SE, Vos T, Wang C, Wang X, Weiderpass E, Werdecker A, Wright JL, Yang YC, Yatsuya H, Yoon J, Yoon SJ, Zhao Y, Zhou M, Zhu S, Lopez AD, Murray CJ, Gakidou E. Global, regional, and national prevalence of overweight and obesity in children and adults during 1980-2013: a systematic analysis for the Global Burden of Disease Study 2013. Lancet. 2014; 384:766–781.
2. Koves TR, Ussher JR, Noland RC, Slentz D, Mosedale M, Ilkayeva O, Bain J, Stevens R, Dyck JR, Newgard CB, Lopaschuk GD, Muoio DM. Mitochondrial overload and incomplete fatty acid oxidation contribute to skeletal muscle insulin resistance. Cell Metab. 2008; 7:45–56.
3. Anderson EJ, Lustig ME, Boyle KE, Woodlief TL, Kane DA, Lin CT, Price JW 3rd, Kang L, Rabinovitch PS, Szeto HH, Houmard JA, Cortright RN, Wasserman DH, Neufer PD. Mitochondrial H2O2 emission and cellular redox state link excess fat intake to insulin resistance in both rodents and humans. J Clin Invest. 2009; 119:573–581.
4. Greene NP, Nilsson MI, Washington TA, Lee DE, Brown LA, Papineau AM, Shimkus KL, Greene ES, Crouse SF, Fluckey JD. Impaired exercise-induced mitochondrial biogenesis in the obese Zucker rat, despite PGC-1α induction, is due to compromised mitochondrial translation elongation. Am J Physiol Endocrinol Metab. 2014; 306:E503–E511.
5. Pich S, Bach D, Briones P, Liesa M, Camps M, Testar X, Palacín M, Zorzano A. The Charcot-Marie-Tooth type 2A gene product, Mfn2, up-regulates fuel oxidation through expression of OXPHOS system. Hum Mol Genet. 2005; 14:1405–1415.
6. Dahlmans D, Houzelle A, Schrauwen P, Hoeks J. Mitochondrial dynamics, quality control and miRNA regulation in skeletal muscle: implications for obesity and related metabolic disease. Clin Sci (Lond). 2016; 130:843–852.
7. Chen H, Chomyn A, Chan DC. Disruption of fusion results in mitochondrial heterogeneity and dysfunction. J Biol Chem. 2005; 280:26185–26192.
8. Liu R, Jin P, Yu L, Wang Y, Han L, Shi T, Li X. Impaired mitochondrial dynamics and bioenergetics in diabetic skeletal muscle. PLoS One. 2014; 9:e92810.
9. Erlich AT, Tryon LD, Crilly MJ, Memme JM, Moosavi ZSM, Oliveira AN, Beyfuss K, Hood DA. Function of specialized regulatory proteins and signaling pathways in exercise-induced muscle mitochondrial biogenesis. Integr Med Res. 2016; 5:187–197.
10. Seo DY, Lee SR, Kim N, Ko KS, Rhee BD, Han J. Age-related changes in skeletal muscle mitochondria: the role of exercise. Integr Med Res. 2016; 5:182–186.
11. Kirkwood SP, Munn EA, Brooks GA. Mitochondrial reticulum in limb skeletal muscle. Am J Physiol. 1986; 251:C395–C402.
12. Bonnard C, Durand A, Peyrol S, Chanseaume E, Chauvin MA, Morio B, Vidal H, Rieusset J. Mitochondrial dysfunction results from oxidative stress in the skeletal muscle of diet-induced insulin-resistant mice. J Clin Invest. 2008; 118:789–800.
13. Greene NP, Lee DE, Brown JL, Rosa ME, Brown LA, Perry RA, Henry JN, Washington TA. Mitochondrial quality control, promoted by PGC-1α, is dysregulated by Western diet-induced obesity and partially restored by moderate physical activity in mice. Physiol Rep. 2015; 3:e12470.
14. Abrigo J, Rivera JC, Aravena J, Cabrera D, Simon F, Ezquer F, Ezquer M, Cabello-Verrugio C. High fat diet-induced skeletal muscle wasting is decreased by mesenchymal stem cells administration: implications on oxidative stress, ubiquitin proteasome pathway activation, and myonuclear apoptosis. Oxid Med Cell Longev. 2016; 2016:9047821.
15. Bisbal C, Lambert K, Avignon A. Antioxidants and glucose metabolism disorders. Curr Opin Clin Nutr Metab Care. 2010; 13:439–446.
16. Schafer FQ, Buettner GR. Redox environment of the cell as viewed through the redox state of the glutathione disulfide/glutathione couple. Free Radic Biol Med. 2001; 30:1191–1212.
17. Jones DP. Radical-free biology of oxidative stress. Am J Physiol Cell Physiol. 2008; 295:C849–C868.
18. Jones DP. Disruption of mitochondrial redox circuitry in oxidative stress. Chem Biol Interact. 2006; 163:38–53.
19. Shadel GS, Horvath TL. Mitochondrial ROS signaling in organismal homeostasis. Cell. 2015; 163:560–569.
20. Martindale JL, Holbrook NJ. Cellular response to oxidative stress: signaling for suicide and survival. J Cell Physiol. 2002; 192:1–15.
21. Yakes FM, Van Houten B. Mitochondrial DNA damage is more extensive and persists longer than nuclear DNA damage in human cells following oxidative stress. Proc Natl Acad Sci U S A. 1997; 94:514–519.
22. Konopka AR, Asante A, Lanza IR, Robinson MM, Johnson ML, Dalla Man C, Cobelli C, Amols MH, Irving BA, Nair KS. Defects in mitochondrial efficiency and H2O2 emissions in obese women are restored to a lean phenotype with aerobic exercise training. Diabetes. 2015; 64:2104–2115.
23. Mootha VK, Lindgren CM, Eriksson KF, Subramanian A, Sihag S, Lehar J, Puigserver P, Carlsson E, Ridderstråle M, Laurila E, Houstis N, Daly MJ, Patterson N, Mesirov JP, Golub TR, Tamayo P, Spiegelman B, Lander ES, Hirschhorn JN, Altshuler D, Groop LC. PGC-1alpha-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat Genet. 2003; 34:267–273.
24. Nabben M, Hoeks J, Briedé JJ, Glatz JF, Moonen-Kornips E, Hesselink MK, Schrauwen P. The effect of UCP3 overexpression on mitochondrial ROS production in skeletal muscle of young versus aged mice. FEBS Lett. 2008; 582:4147–4152.
25. Hey-Mogensen M, Højlund K, Vind BF, Wang L, Dela F, Beck-Nielsen H, Fernström M, Sahlin K. Effect of physical training on mitochondrial respiration and reactive oxygen species release in skeletal muscle in patients with obesity and type 2 diabetes. Diabetologia. 2010; 53:1976–1985.
26. Li G, Liu JY, Zhang HX, Li Q, Zhang SW. Exercise training attenuates sympathetic activation and oxidative stress in diet-induced obesity. Physiol Res. 2015; 64:355–367.
27. van der Bliek AM, Shen Q, Kawajiri S. Mechanisms of mitochondrial fission and fusion. Cold Spring Harb Perspect Biol. 2013; 5:a011072.
28. Ni HM, Williams JA, Ding WX. Mitochondrial dynamics and mitochondrial quality control. Redox Biol. 2015; 4:6–13.
29. Picard M, Shirihai OS, Gentil BJ, Burelle Y. Mitochondrial morphology transitions and functions: implications for retrograde signaling? Am J Physiol Regul Integr Comp Physiol. 2013; 304:R393–R406.
30. Westermann B. Mitochondrial fusion and fission in cell life and death. Nat Rev Mol Cell Biol. 2010; 11:872–884.
31. Palmer CS, Osellame LD, Laine D, Koutsopoulos OS, Frazier AE, Ryan MT. MiD49 and MiD51, new components of the mitochondrial fission machinery. EMBO Rep. 2011; 12:565–573.
32. Santel A, Frank S. Shaping mitochondria: The complex posttranslational regulation of the mitochondrial fission protein DRP1. IUBMB Life. 2008; 60:448–455.
33. Jheng HF, Tsai PJ, Guo SM, Kuo LH, Chang CS, Su IJ, Chang CR, Tsai YS. Mitochondrial fission contributes to mitochondrial dysfunction and insulin resistance in skeletal muscle. Mol Cell Biol. 2012; 32:309–319.
34. Bach D, Pich S, Soriano FX, Vega N, Baumgartner B, Oriola J, Daugaard JR, Lloberas J, Camps M, Zierath JR, Rabasa-Lhoret R, Wallberg-Henriksson H, Laville M, Palacín M, Vidal H, Rivera F, Brand M, Zorzano A. Mitofusin-2 determines mitochondrial network architecture and mitochondrial metabolism. A novel regulatory mechanism altered in obesity. J Biol Chem. 2003; 278:17190–17197.
35. Sebastián D, Hernández-Alvarez MI, Segalés J, Sorianello E, Muñoz JP, Sala D, Waget A, Liesa M, Paz JC, Gopalacharyulu P, Orešič M, Pich S, Burcelin R, Palacín M, Zorzano A. Mitofusin 2 (Mfn2) links mitochondrial and endoplasmic reticulum function with insulin signaling and is essential for normal glucose homeostasis. Proc Natl Acad Sci U S A. 2012; 109:5523–5528.
36. Yan Z, Lira VA, Greene NP. Exercise training-induced regulation of mitochondrial quality. Exerc Sport Sci Rev. 2012; 40:159–164.
37. Ding H, Jiang N, Liu H, Liu X, Liu D, Zhao F, Wen L, Liu S, Ji LL, Zhang Y. Response of mitochondrial fusion and fission protein gene expression to exercise in rat skeletal muscle. Biochim Biophys Acta. 2010; 1800:250–256.
38. Petrovski G, Das DK. Does autophagy take a front seat in lifespan extension? J Cell Mol Med. 2010; 14:2543–2551.
39. Twig G, Elorza A, Molina AJ, Mohamed H, Wikstrom JD, Walzer G, Stiles L, Haigh SE, Katz S, Las G, Alroy J, Wu M, Py BF, Yuan J, Deeney JT, Corkey BE, Shirihai OS. Fission and selective fusion govern mitochondrial segregation and elimination by autophagy. EMBO J. 2008; 27:433–446.
40. Campello S, Strappazzon F, Cecconi F. Mitochondrial dismissal in mammals, from protein degradation to mitophagy. Biochim Biophys Acta. 2014; 1837:451–460.
41. Jin SM, Lazarou M, Wang C, Kane LA, Narendra DP, Youle RJ. Mitochondrial membrane potential regulates PINK1 import and proteolytic destabilization by PARL. J Cell Biol. 2010; 191:933–942.
42. Youle RJ, Narendra DP. Mechanisms of mitophagy. Nat Rev Mol Cell Biol. 2011; 12:9–14.
43. Chan NC, Salazar AM, Pham AH, Sweredoski MJ, Kolawa NJ, Graham RL, Hess S, Chan DC. Broad activation of the ubiquitin-proteasome system by Parkin is critical for mitophagy. Hum Mol Genet. 2011; 20:1726–1737.
44. Gegg ME, Cooper JM, Chau KY, Rojo M, Schapira AH, Taanman JW. Mitofusin 1 and mitofusin 2 are ubiquitinated in a PINK1/parkin-dependent manner upon induction of mitophagy. Hum Mol Genet. 2010; 19:4861–4870.
45. Huang C, Andres AM, Ratliff EP, Hernandez G, Lee P, Gottlieb RA. Preconditioning involves selective mitophagy mediated by Parkin and p62/SQSTM1. PLoS One. 2011; 6:e20975.
46. Wong YC, Holzbaur EL. Optineurin is an autophagy receptor for damaged mitochondria in parkin-mediated mitophagy that is disrupted by an ALS-linked mutation. Proc Natl Acad Sci U S A. 2014; 111:E4439–E4448.
47. 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.
48. Kumar AR, Snyder JM. Differential regulation of SP-A1 and SP-A2 genes by cAMP, glucocorticoids, and insulin. Am J Physiol. 1998; 274:L177–L185.
49. Ren M, Phoon CK, Schlame M. Metabolism and function of mitochondrial cardiolipin. Prog Lipid Res. 2014; 55:1–16.
50. Chu CT, Ji J, Dagda RK, Jiang JF, Tyurina YY, Kapralov AA, Tyurin VA, Yanamala N, Shrivastava IH, Mohammadyani D, Wang KZQ, Zhu J, Klein-Seetharaman J, Balasubramanian K, Amoscato AA, Borisenko G, Huang Z, Gusdon AM, Cheikhi A, Steer EK, Wang R, Baty C, Watkins S, Bahar I, Bayir H, Kagan VE. Cardiolipin externalization to the outer mitochondrial membrane acts as an elimination signal for mitophagy in neuronal cells. Nat Cell Biol. 2013; 15:1197–1205.
51. Orvedahl A, Sumpter R Jr, Xiao G, Ng A, Zou Z, Tang Y, Narimatsu M, Gilpin C, Sun Q, Roth M, Forst CV, Wrana JL, Zhang YE, Luby-Phelps K, Xavier RJ, Xie Y, Levine B. Image-based genome-wide siRNA screen identifies selective autophagy factors. Nature. 2011; 480:113–117.
52. Joseph AM, Adhihetty PJ, Wawrzyniak NR, Wohlgemuth SE, Picca A, Kujoth GC, Prolla TA, Leeuwenburgh C. Dysregulation of mitochondrial quality control processes contribute to sarcopenia in a mouse model of premature aging. PLoS One. 2013; 8:e69327.
53. Carnio S, LoVerso F, Baraibar MA, Longa E, Khan MM, Maffei M, Reischl M, Canepari M, Loefler S, Kern H, Blaauw B, Friguet B, Bottinelli R, Rudolf R, Sandri M. Autophagy impairment in muscle induces neuromuscular junction degeneration and precocious aging. Cell Rep. 2014; 8:1509–1521.
54. Dirks AJ, Hofer T, Marzetti E, Pahor M, Leeuwenburgh C. Mitochondrial DNA mutations, energy metabolism and apoptosis in aging muscle. Ageing Res Rev. 2006; 5:179–195.
55. Quadrilatero J, Alway SE, Dupont-Versteegden EE. Skeletal muscle apoptotic response to physical activity: potential mechanisms for protection. Appl Physiol Nutr Metab. 2011; 36:608–617.
56. Li P, Nijhawan D, Budihardjo I, Srinivasula SM, Ahmad M, Alnemri ES, Wang X. Cytochrome c and dATP-dependent formation of Apaf-1/caspase-9 complex initiates an apoptotic protease cascade. Cell. 1997; 91:479–489.
57. Li LY, Luo X, Wang X. Endonuclease G is an apoptotic DNase when released from mitochondria. Nature. 2001; 412:95–99.
58. Susin SA, Lorenzo HK, Zamzami N, Marzo I, Snow BE, Brothers GM, Mangion J, Jacotot E, Costantini P, Loeffler M, Larochette N, Goodlett DR, Aebersold R, Siderovski DP, Penninger JM, Kroemer G. Molecular characterization of mitochondrial apoptosis-inducing factor. Nature. 1999; 397:441–446.
59. Dungan CM, Li J, Williamson DL. Caloric restriction normalizes obesity-induced alterations on regulators of skeletal muscle growth signaling. Lipids. 2016; 51:905–912.
60. Yuzefovych LV, Musiyenko SI, Wilson GL, Rachek LI. Mitochondrial DNA damage and dysfunction, and oxidative stress are associated with endoplasmic reticulum stress, protein degradation and apoptosis in high fat diet-induced insulin resistance mice. PLoS One. 2013; 8:e54059.
61. Sishi B, Loos B, Ellis B, Smith W, du Toit EF, Engelbrecht AM. Diet-induced obesity alters signalling pathways and induces atrophy and apoptosis in skeletal muscle in a prediabetic rat model. Exp Physiol. 2011; 96:179–193.
62. Peterson JM, Bryner RW, Sindler A, Frisbee JC, Alway SE. Mitochondrial apoptotic signaling is elevated in cardiac but not skeletal muscle in the obese Zucker rat and is reduced with aerobic exercise. J Appl Physiol (1985). 2008; 105:1934–1943.
63. Peterson JM, Bryner RW, Alway SE. Satellite cell proliferation is reduced in muscles of obese Zucker rats but restored with loading. Am J Physiol Cell Physiol. 2008; 295:C521–C528.
64. Ljubicic V, Joseph AM, Adhihetty PJ, Huang JH, Saleem A, Uguccioni G, Hood DA. Molecular basis for an attenuated mitochondrial adaptive plasticity in aged skeletal muscle. Aging (Albany NY). 2009; 1:818–830.
65. Song W, Kwak HB, Lawler JM. Exercise training attenuates age-induced changes in apoptotic signaling in rat skeletal muscle. Antioxid Redox Signal. 2006; 8:517–528.
66. Chae CH, Jung SL, An SH, Jung CK, Nam SN, Kim HT. Treadmill exercise suppresses muscle cell apoptosis by increasing nerve growth factor levels and stimulating p-phosphatidylinositol 3-kinase activation in the soleus of diabetic rats. J Physiol Biochem. 2011; 67:235–241.
TOOLS
Similar articles