Skip to content
Publicly Available Published by De Gruyter December 19, 2015

Intramyocellular fat storage in metabolic diseases

  • Claire Laurens and Cedric Moro EMAIL logo

Abstract

Over the past decades, obesity and its metabolic co-morbidities such as type 2 diabetes (T2D) developed to reach an endemic scale. However, the mechanisms leading to the development of T2D are still poorly understood. One main predictor for T2D seems to be lipid accumulation in “non-adipose” tissues, best known as ectopic lipid storage. A growing body of data suggests that these lipids may play a role in impairing insulin action in metabolic tissues, such as liver and skeletal muscle. This review aims to discuss recent literature linking ectopic lipid storage and insulin resistance, with emphasis on lipid deposition in skeletal muscle. The link between skeletal muscle lipid content and insulin sensitivity, as well as the mechanisms of lipid-induced insulin resistance and potential therapeutic strategies to alleviate lipotoxic lipid pressure in skeletal muscle will be discussed.

Introduction

Nowadays, obesity has become one of the most prevalent disease worldwide, leading to the development of metabolic and cardiovascular pathologies [1]. Obesity is currently the strongest risk factor known for type 2 diabetes (T2D). T2D is characterized by a fasting hyperglycemia that is due to an impaired action of insulin on insulin sensitive tissues (i.e. adipose tissue, liver and skeletal muscle), the so-called insulin resistance [2, 3]. In the last decades, at least two mutually not exclusive hypotheses have emerged linking obesity to insulin resistance as recently reviewed by Samuel and Shulman [4]. One prevailing hypothesis is lipotoxicity caused by lipid overflow out of adipose tissue and ectopic lipid storage in lean tissues. This situation typically occurs when excess dietary lipids cannot anymore be appropriately stored into adipose tissue. Thus an enhanced expansion of fat mass is thought to protect against metabolic disturbances and to convey a healthy obese profile [5]. As a consequence, excess lipid accumulation in pancreas causes beta cell dysfunction. Some data suggest that, in a context of hyperglycemia, fatty acids can induce beta-cell death by apoptosis, a process called glucolipotoxicity [6].

In liver, ectopic lipid accumulation is referred as nonalcoholic fatty liver disease (NAFLD). Increased hepatocellular lipid content correlates negatively with both whole body and hepatic insulin sensitivity, leading to impaired suppression of endogenous glucose production and decreased hepatic glycogen synthesis during euglycemic hyperinsulinemic clamp [7]. When T2D patients are subjected to a hypocaloric diet, a strong decrease of 85% of hepatic lipid content is observed, associated with a normalization of hepatic insulin sensitivity as well as decreased hyperglycemia and hepatic glucose production. Importantly, these changes occur without any modifications of skeletal muscle lipid content, suggesting that the reduction of hepatic lipids per se could improve insulin sensitivity [8].

Numerous studies have linked ectopic lipid accumulation in skeletal muscle and insulin resistance [913]. Skeletal muscle plays a major role in whole body glucose homeostasis, as it is responsible for 80% of glucose disposal in response to insulin in the postprandial phase [14, 15]. Thus, as this organ acts as a metabolic sink for glucose, it is easy to figure out that impaired insulin action in muscle will lead to whole body insulin resistance and further development of T2D.

The purpose of this review was to summarize current understanding of how lipids emerge in skeletal muscle and to what extent they can contribute to muscle and whole body insulin resistance.

Skeletal muscle ectopic lipids and insulin resistance

Lipids can be found under two different forms in skeletal muscle: adipose cells, located between muscle fibers (i.e. intramuscular adipose tissue), and lipid droplets inside muscle fibers (i.e. intramyocellular triglycerides) [16, 17] (Figure 1).

Figure 1: Model linking obesity, muscle lipids and insulin resistance.This cartoon illustrates that in young and lean subjects, a physiological amount of lipids is present in skeletal muscle. These lipids are stored in intramuscular adipocytes located between muscle fibers and in intramyocellular lipid droplets (imLD) within muscle fibers. When obesity develops, and this is also observed with aging, these lipids are stored in excess in skeletal muscle, leading to lipotoxicity responsible for insulin and anabolic resistance.
Figure 1:

Model linking obesity, muscle lipids and insulin resistance.

This cartoon illustrates that in young and lean subjects, a physiological amount of lipids is present in skeletal muscle. These lipids are stored in intramuscular adipocytes located between muscle fibers and in intramyocellular lipid droplets (imLD) within muscle fibers. When obesity develops, and this is also observed with aging, these lipids are stored in excess in skeletal muscle, leading to lipotoxicity responsible for insulin and anabolic resistance.

Intramuscular adipose tissue

Intramuscular fat is made of adipose cells (i.e. adipocytes) located between muscle fibers and muscle groups. Different studies in rodents and humans report a positive correlation between intramuscular adipose tissue (IMAT) and insulin resistance [1820], and even when body mass index (BMI) is accounted for, it remains a strong predictor of fasting glucose and insulin levels [21].

However, it is still unclear if these adipocytes emerge from resident adipogenic progenitors or from satellite cells in skeletal muscle [16, 22]. Recent data from our group suggest that resident adipogenic progenitors, with potential to differentiate into functional adipocytes in vitro, are present in human skeletal muscle. We suggest that the proximity of these adipocytes to muscle fibers may impair muscle insulin action in a paracrine manner, and thus impact on whole body insulin sensitivity. On the other hand, one could speculate that these adipogenic progenitors differentiate into mature adipocytes as an adaptive mechanism during periods of lipid overload, as a buffer system to prevent lipid accumulation within myofibers. Future studies should investigate if IMAT can cause muscle dysfunction.

Intramyocellular triglycerides

In muscle fibers, lipids are stored within lipid droplets (LD) as triacylglycerols (TAG). Intramyocellular triglycerides (IMTG) constitute an important energy fuel source during muscle contraction [23, 24]. Indeed, elegant studies from Jensen’s group demonstrated that plasma fatty acids are first esterified into IMTG pools before being oxidized, both at rest [25, 26] and during exercise [27]. They further demonstrated that during submaximal exercise IMTG turnover is high, without changes in the IMTG pool size. However, other studies report that 60%–100% of radiolabeled oleate or palmitate taken up in skeletal muscle was directly oxidized [24, 28]. Such discrepancies may be due to differences in experimental protocol and/or training status of the subjects, which really differs from one study to another [23].

Importantly, maximal fatty acid oxidation in skeletal muscle is observed at moderate exercise intensities between 45 and 65% of maximal oxygen uptake depending on the type of subjects [24].

However, increased IMTG content is also found in obese and T2D people [10], and studies in lean healthy individuals report IMTG content as a stronger predictor of muscle insulin resistance than circulating fatty acids [29]. However, these studies did not account for the sedentary or active status of the subjects. In sedentary individuals, a strong inverse relationship has been reported between IMTG content and insulin sensitivity, independently of body mass index [30]. However, this relationship has proven much more complex than initially thought. Indeed, Goodpaster and colleagues have demonstrated that the skeletal muscle of endurance-trained athletes is highly insulin sensitive despite having an elevated IMTG content [31]. This could be explained by a more efficient lipid handling in skeletal muscle of athletes, allowing an optimal coupling between lipid storage and utilization to avoid lipotoxic insults. Interestingly, a close physical association between LD and mitochondria has been reported in active versus sedentary subjects, as well as a higher turnover of the IMTG pool [3236].

Lipotoxic lipids

In the last few years, several studies aimed to identify the molecular actors responsible for the impairment of insulin signaling in skeletal muscle. It is now well established that IMTG per se do not cause insulin resistance, and several evidence dissociate TAG content from insulin resistance in humans [37, 38]. These observations raised the question of which molecular triggers could link increased IMTG content and insulin resistance in T2D subjects. Several lipid species emerged as potential candidates and in particular diacylglycerols (DAG) and ceramides (CER) [39, 40]. Indeed, increased DAG and CER content in skeletal muscle has been associated with insulin resistance in rodents [41, 42] and humans [43, 44]. A study from Goodpaster and colleagues demonstrated that diet-induced weight loss and exercise improved insulin resistance while reducing muscle DAG and CER content [45]. However, controversies exist related to the role of these lipid metabolites in impairing muscle insulin signalling, as some studies describe no change in muscle DAG and CER content associated with insulin resistance [17, 4648], although data are not consistent [17, 49]. Taken together, these studies reveal that it seems that subcellular localization and composition of DAG [50] and CER, as well as their stereospecificity play an important role in their ability to impede insulin action, and needs to be further investigated.

Diacylglycerols pools

In rats, intralipid/heparin induces a strong increase of plasma fatty acids, and starts to reduce insulin sensitivity 3h after the beginning of infusion concomitant with the accumulation of DAG, impaired insulin signalling and glucose uptake in skeletal muscle. Of importance, insulin resistance occurred without any changes in IMTG content [40, 51]. DAGs act as signalling molecules, and are allosteric activators of protein kinase C (PKC). It has been shown that DAG-induced activation of novel PKC isoforms, and particularly PKCθ and PKCε in skeletal muscle, induces serine phosphorylation of insulin receptor substrate 1 (IRS1), thereby inhibiting insulin signalling [40, 52, 53].

Ceramides pools

It is interesting to note that mice fed for 12 weeks with a high-fat diet are protected from glucose intolerance if they are treated with myriocin, an inhibitor of serine palmitoyl tranferase 1 which decreases skeletal muscle CER content [54]. However, the role of CER in insulin resistance appears restricted to saturated fat, as myriocin prevents skeletal muscle insulin resistance after infusion of palmitate but not oleate [55].

CER seems to activate atypical PKC isoforms such as PKCξ, and the protein phosphatase 2A (PP2A), leading, respectively, to the phosphorylation of Akt on a Thr34 residue decreasing its ability to bind to membrane PI3P and to the dephosphorylation of Ser473 activating residue, thus impairing insulin-induced activation of Akt, and subsequent translocation of GLUT4 vesicles to the plasma membrane [56, 57].

Molecular mechanisms leading to intramyocellular triglycerides accumulation

Defects in muscle fatty acids storage

IMTG emerge from circulating fatty acids, and their uptake inside muscle fibers is partly regulated by cluster of differenciation 36 (cd36) and fatty acid transport protein 1 (FATP1) transporters. Knock-out studies in mice have shown that CD36 [58, 59] and FATP1 [60] deletion protects mice from muscle lipid accumulation and muscle insulin resistance under high-fat diet. The first step of TAG synthesis is controlled by glycerol-3-phosphate acyltransferase (GPAT), which catalyzes the formation of lysophosphatidic acid from fatty acyl CoA and glycerol-3-phosphate. The final step of TAG synthesis is catalyzed by diacylglycerol acyltransferases 1 and 2 (DGAT1 and DGAT2), which acylate DAG into TAG. Muscle-specific DGAT1 [61] or DGAT2 [62] overexpression leads to an accumulation of TAG in skeletal muscle, concomitant with a decrease in DAG content. In addition, unilateral overexpression of DGAT1 in rat skeletal muscle increased TAG content and rescued muscle insulin sensitivity when animals were fed high-fat diet [63]. Recently, Sparks and coworkers showed that muscle homogenates and human primary myotubes from obese individuals with T2D exhibit a reduced ability to incorporate FA into TAG pools [64]. Altogether, these studies indicate that increased rates of esterification of FA into TAG pools could be beneficial for insulin sensitivity by lowering lipotoxic lipid pressure in skeletal muscle.

Dysregulation of lipid droplets homeostasis

Perilipins

LDs are dynamic organelles, composed of a neutral lipid core surrounded by a phospholipid monolayer [6567]. The surface of LDs is coated by different membrane-associated proteins, and the major structural components of LDs belong to the family of perilipins. Five members of the perilipin family have been identified so far, with various tissue expression patterns. In skeletal muscle, the most represented isoforms are PLIN2, PLIN3, PLIN4 and PLIN5 [68]. By forming a “lipolytic barrier”, they also protect LDs against their hydrolysis by lipases. PLIN5 is highly expressed in oxidative tissues such as skeletal muscle, and PLIN5 knock-out mice display an accumulation of CER in muscle fibers and skeletal muscle insulin-resistance [69]. However, upon specific conditions, perilipins could also facilitate lipolysis. For instance, in adipose tissue, PLIN1 facilitates lipases access to LDs upon adrenergic stimulation of lipolysis [7072]. In skeletal muscle, PLIN5 may play a role in fatty acids channeling to mitochondria, promoting by this way their oxidation upon stimulated conditions [73]. Indeed, independent studies from two different groups have demonstrated that PLIN5 also localizes to mitochondria, providing a physical linkage between LDs and mitochrondria [74, 75]. Further work is needed to clarify the physiological role of perilipins in skeletal muscle, with emphasis on the apparent discrepancy between resting and stimulated conditions.

Lipases

IMTG breakdown (i.e. lipolysis) is controlled by different enzymes, and their activity is finely regulated. The lipolytic machinery is highly expressed in red oxidative compared to white glycolytic muscle (Figure 2), and this is correlated with both IMTG content and oxidative capacity (unpublished data). The first step of IMTG breakdown is catalyzed by adipose triglyceride lipase (ATGL), which converts TAG into DAG. Then, hormone-sensitive lipase (HSL) converts DAG into MAG, and MAG are finally hydrolyzed by the monoacylglycerol lipase (MGL) [76]. At the end, lipolysis of one TAG molecule releases three fatty acid molecules and one glycerol molecule. Although HSL and MGL were the first lipases identified in skeletal muscle, and are both highly expressed, ATGL appears to be the first and rate limiting step of skeletal muscle lipolysis.

Figure 2: Lipolytic proteins are highly expressed in oxidative compared to glycolytic muscles.Representative blot of protein expression of the major lipolytic proteins in glycolytic (EDL), mixed (TA) and oxidative (Sol) skeletal muscles. EDL, Extensor digitorum longus; TA, tibialis anterior; Sol, soleus; ATGL, adipose triglyceride lipase; HSL, hormone-sensitive lipase; G0S2, G0/G1 switch gene 2; PLIN5, perilipin 5.
Figure 2:

Lipolytic proteins are highly expressed in oxidative compared to glycolytic muscles.

Representative blot of protein expression of the major lipolytic proteins in glycolytic (EDL), mixed (TA) and oxidative (Sol) skeletal muscles. EDL, Extensor digitorum longus; TA, tibialis anterior; Sol, soleus; ATGL, adipose triglyceride lipase; HSL, hormone-sensitive lipase; G0S2, G0/G1 switch gene 2; PLIN5, perilipin 5.

Monoacylglycerol lipase

MGL was discovered in 1976 by Tornqvist and colleagues, isolated from rat adipose tissue, and shown to specifically hydrolyze MAG but not TAG and DAG [77, 78]. MGL-knockout mice exhibit an increased MAG content associated with a down-regulation of free fatty acid and glycerol release from white adipose tissue [79]. MGL deletion also protects from diet-induced insulin-resistance and its inhibition in diabetic mice improves glucose tolerance [79, 80]. However, the physiological role of MGL in skeletal muscle lipolysis has not been investigated so far.

Hormone-sensitive lipase and adipose triglyceride lipase

Recent studies from our group have shown that diabetic status is associated with changes of ATGL and HSL activity and expression level in humans [81]. An increase of ATGL and a decrease of HSL expression/activity in skeletal muscle of obese and T2D patients was observed when compared to lean healthy individuals [82]. ATGL activity is negatively associated with whole-body insulin sensitivity measured by a hyperinsulinemic-euglycemic clamp [81]. Furthermore, ATGL protein expression has been shown to be increased in obese and T2D patients [81]. On the other hand, HSL expression and activity is down-regulated in obese insulin resistant subjects [82]. Of interest, this could be a primary defect acquired in obesity that persists in cultured human primary myotubes from obese type 2 diabetic individuals [83].

HSL was first identified in 1964 as a lipase sensitive to catecholamines in adipose tissue [84]. HSL presents TAG and DAG hydrolase activities, but displays a 10-times higher affinity for DAG rather than for TAG [76]. Confocal imaging studies have shown that HSL translocates to LDs in skeletal muscle upon stimulation by epinephrine or in response to contraction [85]. HSL-knockout mice accumulate DAG, but not TAG, in white adipose tissue and skeletal muscle, strengthening its role as a DAG hydrolase in vivo [86]. The observation that HSL-knockout mice do not accumulate TAG in skeletal muscle led to the conclusion that another enzyme is responsible for TAG-specific hydrolysis in skeletal muscle.

ATGL is highly expressed in skeletal muscle and white adipose tissue [87, 88], and ATGL mutations in humans lead to neutral lipid storage disease with myopathy [89]. Genetic modulations of ATGL expression in rodents helped to unravel its physiological role in vivo. Importantly, ATGL knockout mice display a massive TAG accumulation in cardiac and skeletal muscles [88]. In contrast, ATGL overexpression in human primary myotubes strongly decreases TAG content, while FA release and oxidation are increased [81]. More recent studies confirmed the important role of ATGL in skeletal muscle through muscle-specific modulations of its expression in mice [90, 91]. Collectively, these findings strengthen the role of ATGL as a TAG-specific hydrolase in skeletal muscle.

Mouse and cell models were helpful to define a causal relationship to strengthen the correlative findings linking ATGL to insulin resistance in human studies. Thus, ATGL knockout mice display a strong accumulation of TAG in skeletal muscle [88], as stated above, and are protected against high-fat diet induced insulin-resistance [92]. ATGL overexpression in human primary myotubes blunted insulin signalling and action. This phenotype was rescued when HSL was simultaneously overexpressed with ATGL, thus highlighting an important role for the balance between ATGL and HSL activity to maintain insulin sensitivity [81].

Enzymatic co-factors

ATGL activity is regulated by an enzymatic coactivator called comparative gene identification 58 (CGI58), which has also been shown to enhance TAG hydrolase activity in skeletal muscle [39]. CGI58 mutations in humans lead to the development of the Chanarin-Dorfman syndrome, associated with a severe myopathy [93]. CGI58 overexpression in human primary myotubes reduces TAG content and induces FA release and oxidation, whereas its downregulation leads to TAG accumulation and reduces lipolysis [39]. It was demonstrated more recently that ATGL activity is also inhibited by G0/G1 switch gene 2 (G0S2) in adipocytes [94, 95]. Unpublished data from our group suggest that G0S2 also inhibits ATGL activity in human and mouse skeletal muscle, and further studies are necessary to better characterize its functional role.

Concluding remarks

Even if skeletal muscle lipids are undoubtedly linked to insulin resistance, which lipid species are responsible for the impairment of insulin signaling and action is still a matter of ongoing debate. Unlike what was originally proposed, IMTG per se do not seem to induce insulin resistance, and IMTG content is a predictor for T2D development only in sedentary individuals. In highly trained individuals, IMTG content is increased to match the energy demand of the exercising muscle, thus disconnecting IMTG from insulin resistance. In this case, muscle oxidative capacity seems to be a better predictor of insulin resistance than lipid content. This is explained by the fact that a mismatch between skeletal muscle lipolysis and oxidative capacity leads to the accumulation of lipotoxic species, such as DAG and CER, impairing insulin action. Of importance, other molecular mechanisms besides lipotoxicity might contribute to insulin resistance in metabolic diseases as reviewed extensively [4]. For instance, saturated fat and/or endotoxin-mediated inflammation could also trigger insulin resistance in liver and skeletal muscle [96, 97]. Endoplasmic reticulum and oxidative stress may also play a role in some organs while their direct role in mediating insulin resistance in skeletal muscle is still debated [98]. Although no direct evidence of apoptosis are visible in mouse skeletal muscle in response to lipid infusion and chronic high fat feeding [4], ceramides can induce apoptosis by activating caspase 3 in a number of cell type such as beta cells [99]. Thus all these mechanisms are not mutually exclusive in mediating skeletal muscle insulin resistance and could be interconnected. Although it is now widely accepted that DAG and CER are key mediators of insulin resistance in skeletal muscle, the molecular mechanisms by which these lipotoxic lipids emerge in skeletal muscle remain unclear and need further investigation.

Expert opinion

During the last two decades the field has evolved with the concept that ectopic lipid storage in skeletal muscle can cause insulin resistance. It is now clear from a number of studies that not all lipids are alike and have the potential to inhibit insulin signaling and action. Thus non esterified fatty acids incoming to the muscle are neutralized within LD as IMTG. One current view, supported by in vitro data in human primary myotubes and in vivo work in transgenic mice, proposes that disturbances in LD dynamics could facilitates the emergence of toxic lipids, i.e. lipotoxicity, and insulin resistance. This includes alterations in lipases expression/activity and in LD proteins such as perilipins like PLIN2 and PLIN5. Because studies describing the link between muscle PLIN expression and insulin resistance are mostly correlative, future studies should examine the underlying molecular mechanisms in vitro as well as in vivo in mice with muscle-specific manipulations of PLIN in particular, and LD proteins in general. This area of research may help identify novel players in insulin resistance and design drug therapies to combat this strong risk factor of T2D in obese individuals.

Outlook

  • LD is now recognized as a complex organelle with a specific proteome. Understanding if and how the LD proteome influences its dynamic and potential FA flux in and out the LD is a key challenge for the next decade. It is predictable that novel specific LD proteins will be identified in skeletal muscle and may behaves as potential druggable targets to combat insulin resistance.

  • Although a number of clinical studies now underscore the potential importance of IMAT in metabolic diseases and aging, little is known on this ectopic fat depot and the biology of intramuscular adipocytes. It will be important in the near furture to investigate if IMAT is causally related to muscle dysfunction and how this depot emerges in the context of metabolic diseases and aging.

Highlights

  • Intramuscular lipids are stored either as adipocytes between muscle fibers or as LD within muscle fibers

  • Skeletal muscle lipid content is a stronger predictor of insulin resistance than circulating fatty acids in sedentary individuals

  • The pathophysiological role of adipocytes located in skeletal muscle remains to be elucidated

  • DAG and CER impair insulin signaling by activating PKC and PP2A

  • Elevated ATGL activity in skeletal muscle contributes to lipotoxicity and insulin resistance

  • HSL expression is reduced in skeletal muscle and primary myotubes from obese and T2D subjects

  • Disturbances in LD dynamics and/or LD protein expression influences lipotoxicity and insulin sensitivity

  • PLIN5 protein expression in skeletal muscle is a determinant of insulin sensitivity


Corresponding author: Cedric Moro, Ph.D., INSERM, UMR 1048, Institut des Maladies Métaboliques et Cardiovasculaires, CHU Rangueil, BP 84225, 1 Avenue Jean Poulhès, 31432 Toulouse Cedex 4, France, Phone: +33(0)561325626, Fax: +33(0)561325623, E-mail: ; and University of Toulouse, UMR1048, Paul Sabatier University, Toulouse, France

Acknowledgments

The authors are grateful to Dr. François Crampes for outstanding discussion and critical reading of the manuscript. Some of the work that is discussed here was supported by grants from the National Research Agency ANR-09-JCJC-0019-01 and ANR-12-JSV1-0010-01, and from the Société Francophone du Diabète.

References

1. Kissebah AH, Krakower GR. Regional adiposity and morbidity. Physiol Rev 1994;74:761–811.10.1152/physrev.1994.74.4.761Search in Google Scholar PubMed

2. Defronzo RA, Simonson D, Ferrannini E, Barrett E. Insulin resistance: a universal finding in diabetic states. Bull Schweiz Akad Med Wiss 1981:223–38.Search in Google Scholar

3. DeFronzo RA, Hendler R, Simonson D. Insulin resistance is a prominent feature of insulin-dependent diabetes. Diabetes 1982;31:795–801.10.2337/diab.31.9.795Search in Google Scholar PubMed

4. Samuel VT, Shulman GI. Mechanisms for insulin resistance: common threads and missing links. Cell 2012;148:852–71.10.1016/j.cell.2012.02.017Search in Google Scholar PubMed PubMed Central

5. Samocha-Bonet D, Dixit VD, Kahn CR, Leibel RL, Lin X, Nieuwdorp M, Pietilainen KH, Rabasa-Lhoret R, Roden M, Scherer PE, Klein S, Ravussin E. Metabolically healthy and unhealthy obese–the 2013 Stock Conference report. Obes Rev 2014;15:697–708.10.1111/obr.12199Search in Google Scholar PubMed PubMed Central

6. El-Assaad W, Buteau J, Peyot ML, Nolan C, Roduit R, Hardy S, Joly E, Dbaibo G, Rosenberg L, Prentki M. Saturated fatty acids synergize with elevated glucose to cause pancreatic beta-cell death. Endocrinology 2003;144:4154–63.10.1210/en.2003-0410Search in Google Scholar PubMed

7. Krssak M, Brehm A, Bernroider E, Anderwald C, Nowotny P, Dalla Man C, Cobelli C, Cline GW, Shulman GI, Waldhausl W, Roden M. Alterations in postprandial hepatic glycogen metabolism in type 2 diabetes. Diabetes 2004;53:3048–56.10.2337/diabetes.53.12.3048Search in Google Scholar PubMed

8. Petersen KF, Dufour S, Befroy D, Lehrke M, Hendler RE, Shulman GI. Reversal of nonalcoholic hepatic steatosis, hepatic insulin resistance, and hyperglycemia by moderate weight reduction in patients with type 2 diabetes. Diabetes 2005;54:603–8.10.2337/diabetes.54.3.603Search in Google Scholar PubMed PubMed Central

9. Coen PM, Hames KC, Leachman EM, DeLany JP, Ritov VB, Menshikova EV, Dube JJ, Stefanovic-Racic M, Toledo FG, Goodpaster BH. Reduced skeletal muscle oxidative capacity and elevated ceramide but not diacylglycerol content in severe obesity. Obesity (Silver Spring) 2013;21:2362–71.10.1002/oby.20381Search in Google Scholar PubMed PubMed Central

10. Moro C, Galgani JE, Luu L, Pasarica M, Mairal A, Bajpeyi S, Schmitz G, Langin D, Liebisch G, Smith SR. Influence of gender, obesity, and muscle lipase activity on intramyocellular lipids in sedentary individuals. J Clin Endocrinol Metab 2009;94:3440–7.10.1210/jc.2009-0053Search in Google Scholar PubMed PubMed Central

11. Perseghin G, Scifo P, De Cobelli F, Pagliato E, Battezzati A, Arcelloni C, Vanzulli A, Testolin G, Pozza G, Del Maschio A, Luzi L. Intramyocellular triglyceride content is a determinant of in vivo insulin resistance in humans: a 1H–13C nuclear magnetic resonance spectroscopy assessment in offspring of type 2 diabetic parents. Diabetes 1999;48:1600–6.10.2337/diabetes.48.8.1600Search in Google Scholar PubMed

12. Coen PM, Dube JJ, Amati F, Stefanovic-Racic M, Ferrell RE, Toledo FG, Goodpaster BH. Insulin resistance is associated with higher intramyocellular triglycerides in type I but not type II myocytes concomitant with higher ceramide content. Diabetes 2010;59:80–8.10.2337/db09-0988Search in Google Scholar

13. Virkamaki A, Korsheninnikova E, Seppala-Lindroos A, Vehkavaara S, Goto T, Halavaara J, Hakkinen AM, Yki-Jarvinen H. Intramyocellular lipid is associated with resistance to in vivo insulin actions on glucose uptake, antilipolysis, and early insulin signaling pathways in human skeletal muscle. Diabetes 2001;50:2337–43.10.2337/diabetes.50.10.2337Search in Google Scholar

14. DeFronzo RA, Tripathy D. Skeletal muscle insulin resistance is the primary defect in type 2 diabetes. Diabetes Care 2009;32 Suppl 2:S157–63.10.2337/dc09-S302Search in Google Scholar

15. Ferrannini E, Simonson DC, Katz LD, Reichard G, Jr., Bevilacqua S, Barrett EJ, Olsson M, DeFronzo RA. The disposal of an oral glucose load in patients with non-insulin-dependent diabetes. Metabolism 1988;37:79–85.10.1016/0026-0495(88)90033-9Search in Google Scholar

16. Vettor R, Milan G, Franzin C, Sanna M, De Coppi P, Rizzuto R, Federspil G. The origin of intermuscular adipose tissue and its pathophysiological implications. Am J Physiol Endocrinol Metab 2009;297:E987–98.10.1152/ajpendo.00229.2009Search in Google Scholar PubMed

17. Amati F, Dube JJ, Alvarez-Carnero E, Edreira MM, Chomentowski P, Coen PM, Switzer GE, Bickel PE, Stefanovic-Racic M, Toledo FG, Goodpaster BH. Skeletal muscle triglycerides, diacylglycerols, and ceramides in insulin resistance: another paradox in endurance-trained athletes? Diabetes 2011;60:2588–97.10.2337/db10-1221Search in Google Scholar PubMed PubMed Central

18. Goodpaster BH, Thaete FL, Kelley DE. Thigh adipose tissue distribution is associated with insulin resistance in obesity and in type 2 diabetes mellitus. Am J Clin Nutr 2000;71:885–92.10.1093/ajcn/71.4.885Search in Google Scholar PubMed

19. Yim JE, Heshka S, Albu J, Heymsfield S, Kuznia P, Harris T, Gallagher D. Intermuscular adipose tissue rivals visceral adipose tissue in independent associations with cardiovascular risk. Int J Obes (Lond) 2007;31:1400–5.10.1038/sj.ijo.0803621Search in Google Scholar PubMed PubMed Central

20. Boettcher M, Machann J, Stefan N, Thamer C, Haring HU, Claussen CD, Fritsche A, Schick F. Intermuscular adipose tissue (IMAT): association with other adipose tissue compartments and insulin sensitivity. J Magn Reson Imaging 2009;29:1340–5.10.1002/jmri.21754Search in Google Scholar PubMed

21. Goodpaster BH, Krishnaswami S, Resnick H, Kelley DE, Haggerty C, Harris TB, Schwartz AV, Kritchevsky S, Newman AB. Association between regional adipose tissue distribution and both type 2 diabetes and impaired glucose tolerance in elderly men and women. Diabetes Care 2003;26:372–9.10.2337/diacare.26.2.372Search in Google Scholar PubMed

22. Arrighi N, Moratal C, Clement N, Giorgetti-Peraldi S, Peraldi P, Loubat A, Kurzenne JY, Dani C, Chopard A, Dechesne CA. Characterization of adipocytes derived from fibro/adipogenic progenitors resident in human skeletal muscle. Cell Death Dis 2015;6:e1733.10.1038/cddis.2015.79Search in Google Scholar PubMed PubMed Central

23. van Loon LJ. Use of intramuscular triacylglycerol as a substrate source during exercise in humans. J Appl Physiol (1985) 2004;97:1170–87.10.1152/japplphysiol.00368.2004Search in Google Scholar PubMed

24. Kiens B. Skeletal muscle lipid metabolism in exercise and insulin resistance. Physiol Rev 2006;86:205–43.10.1152/physrev.00023.2004Search in Google Scholar PubMed

25. Dagenais GR, Tancredi RG, Zierler KL. Free fatty acid oxidation by forearm muscle at rest, and evidence for an intramuscular lipid pool in the human forearm. J Clin Invest 1976;58:421–31.10.1172/JCI108486Search in Google Scholar PubMed PubMed Central

26. Kanaley JA, Shadid S, Sheehan MT, Guo Z, Jensen MD. Relationship between plasma free fatty acid, intramyocellular triglycerides and long-chain acylcarnitines in resting humans. J Physiol 2009;587:5939–50.10.1113/jphysiol.2009.180695Search in Google Scholar PubMed PubMed Central

27. Guo Z, Burguera B, Jensen MD. Kinetics of intramuscular triglyceride fatty acids in exercising humans. J Appl Physiol (1985) 2000;89:2057–64.10.1152/jappl.2000.89.5.2057Search in Google Scholar PubMed

28. Roepstorff C, Steffensen CH, Madsen M, Stallknecht B, Kanstrup IL, Richter EA, Kiens B. Gender differences in substrate utilization during submaximal exercise in endurance-trained subjects. Am J Physiol Endocrinol Metab 2002;282:E435–47.10.1152/ajpendo.00266.2001Search in Google Scholar PubMed

29. Krssak M, Falk Petersen K, Dresner A, DiPietro L, Vogel SM, Rothman DL, Roden M, Shulman GI. Intramyocellular lipid concentrations are correlated with insulin sensitivity in humans: a 1H NMR spectroscopy study. Diabetologia 1999;42:113–6.10.1007/s001250051123Search in Google Scholar PubMed

30. McGarry JD. Banting lecture 2001: dysregulation of fatty acid metabolism in the etiology of type 2 diabetes. Diabetes 2002;51:7–18.10.2337/diabetes.51.1.7Search in Google Scholar PubMed

31. Goodpaster BH, He J, Watkins S, Kelley DE. Skeletal muscle lipid content and insulin resistance: evidence for a paradox in endurance-trained athletes. J Clin Endocrinol Metab 2001;86:5755–61.10.1210/jcem.86.12.8075Search in Google Scholar PubMed

32. Tarnopolsky MA, Rennie CD, Robertshaw HA, Fedak-Tarnopolsky SN, Devries MC, Hamadeh MJ. Influence of endurance exercise training and sex on intramyocellular lipid and mitochondrial ultrastructure, substrate use, and mitochondrial enzyme activity. Am J Physiol Regul Integr Comp Physiol 2007;292:R1271–78.10.1152/ajpregu.00472.2006Search in Google Scholar PubMed

33. Jansson E, Kaijser L. Substrate utilization and enzymes in skeletal muscle of extremely endurance-trained men. J Appl Physiol (1985) 1987;62:999–1005.10.1152/jappl.1987.62.3.999Search in Google Scholar PubMed

34. Jong-Yeon K, Hickner RC, Dohm GL, Houmard JA. Long- and medium-chain fatty acid oxidation is increased in exercise-trained human skeletal muscle. Metabolism 2002;51:460–4.10.1053/meta.2002.31326Search in Google Scholar PubMed

35. Moro C, Bajpeyi S, Smith SR. Determinants of intramyocellular triglyceride turnover: implications for insulin sensitivity. Am J Physiol Endocrinol Metab 2008;294:E203–13.10.1152/ajpendo.00624.2007Search in Google Scholar PubMed

36. Sacchetti M, Saltin B, Olsen DB, van Hall G. High triacylglycerol turnover rate in human skeletal muscle. J Physiol 2004;561:883–91.10.1113/jphysiol.2004.075135Search in Google Scholar PubMed PubMed Central

37. van Loon LJ, Goodpaster BH. Increased intramuscular lipid storage in the insulin-resistant and endurance-trained state. Pflugers Arch 2006;451:606–16.10.1007/s00424-005-1509-0Search in Google Scholar PubMed

38. van Loon LJ, Koopman R, Manders R, van der Weegen W, van Kranenburg GP, Keizer HA. Intramyocellular lipid content in type 2 diabetes patients compared with overweight sedentary men and highly trained endurance athletes. Am J Physiol Endocrinol Metab 2004;287:E558–65.10.1152/ajpendo.00464.2003Search in Google Scholar PubMed

39. Badin PM, Loubiere C, Coonen M, Louche K, Tavernier G, Bourlier V, Mairal A, Rustan AC, Smith SR, Langin D, Moro C. Regulation of skeletal muscle lipolysis and oxidative metabolism by the co-lipase CGI-58. J Lipid Res 2012;53:839–48.10.1194/jlr.M019182Search in Google Scholar PubMed PubMed Central

40. Yu C, Chen Y, Cline GW, Zhang D, Zong H, Wang Y, Bergeron R, Kim JK, Cushman SW, Cooney GJ, Atcheson B, White MF, Kraegen EW, Shulman GI. Mechanism by which fatty acids inhibit insulin activation of insulin receptor substrate-1 (IRS-1)-associated phosphatidylinositol 3-kinase activity in muscle. J Biol Chem 2002;277:50230–6.10.1074/jbc.M200958200Search in Google Scholar PubMed

41. Fillmore N, Keung W, Kelly SE, Proctor SD, Lopaschuk GD, Ussher JR. Accumulation of ceramide in slow-twitch muscle contributes to the development of insulin resistance in the obese JCR:LA-cp rat. Exp Physiol 2015;100:730–41.10.1113/EP085052Search in Google Scholar PubMed

42. Holloway GP, Han XX, Jain SS, Bonen A, Chabowski A. Chronic muscle stimulation improves insulin sensitivity while increasing subcellular lipid droplets and reducing selected diacylglycerol and ceramide species in obese Zucker rats. Diabetologia 2014;57:832–40.10.1007/s00125-014-3169-0Search in Google Scholar PubMed

43. Szendroedi J, Yoshimura T, Phielix E, Koliaki C, Marcucci M, Zhang D, Jelenik T, Muller J, Herder C, Nowotny P, Shulman GI, Roden M. Role of diacylglycerol activation of PKCtheta in lipid-induced muscle insulin resistance in humans. Proc Natl Acad Sci USA 2014;111:9597–602.10.1073/pnas.1409229111Search in Google Scholar PubMed PubMed Central

44. Straczkowski M, Kowalska I, Nikolajuk A, Dzienis-Straczkowska S, Kinalska I, Baranowski M, Zendzian-Piotrowska M, Brzezinska Z, Gorski J. Relationship between insulin sensitivity and sphingomyelin signaling pathway in human skeletal muscle. Diabetes 2004;53:1215–21.10.2337/diabetes.53.5.1215Search in Google Scholar PubMed

45. Dube JJ, Amati F, Toledo FG, Stefanovic-Racic M, Rossi A, Coen P, Goodpaster BH. Effects of weight loss and exercise on insulin resistance, and intramyocellular triacylglycerol, diacylglycerol and ceramide. Diabetologia 2011;54:1147–56.10.1007/s00125-011-2065-0Search in Google Scholar PubMed PubMed Central

46. Helge JW, Dobrzyn A, Saltin B, Gorski J. Exercise and training effects on ceramide metabolism in human skeletal muscle. Exp Physiol 2004;89:119–27.10.1113/expphysiol.2003.002605Search in Google Scholar PubMed

47. Itani SI, Ruderman NB, Schmieder F, Boden G. Lipid-induced insulin resistance in human muscle is associated with changes in diacylglycerol, protein kinase C, and IkappaB-alpha. Diabetes 2002;51:2005–11.10.2337/diabetes.51.7.2005Search in Google Scholar PubMed

48. Lessard SJ, Lo Giudice SL, Lau W, Reid JJ, Turner N, Febbraio MA, Hawley JA, Watt MJ. Rosiglitazone enhances glucose tolerance by mechanisms other than reduction of fatty acid accumulation within skeletal muscle. Endocrinology 2004;145:5665–70.10.1210/en.2004-0659Search in Google Scholar PubMed

49. Timmers S, Nabben M, Bosma M, van Bree B, Lenaers E, van Beurden D, Schaart G, Westerterp-Plantenga MS, Langhans W, Hesselink MK, Schrauwen-Hinderling VB, Schrauwen P. Augmenting muscle diacylglycerol and triacylglycerol content by blocking fatty acid oxidation does not impede insulin sensitivity. Proc Natl Acad Sci USA 2012;109:11711–6.10.1073/pnas.1206868109Search in Google Scholar PubMed PubMed Central

50. Bergman BC, Hunerdosse DM, Kerege A, Playdon MC, Perreault L. Localisation and composition of skeletal muscle diacylglycerol predicts insulin resistance in humans. Diabetologia 2012;55:1140–50.10.1007/s00125-011-2419-7Search in Google Scholar PubMed PubMed Central

51. Dresner A, Laurent D, Marcucci M, Griffin ME, Dufour S, Cline GW, Slezak LA, Andersen DK, Hundal RS, Rothman DL, Petersen KF, Shulman GI. Effects of free fatty acids on glucose transport and IRS-1-associated phosphatidylinositol 3-kinase activity. J Clin Invest 1999;103:253–9.10.1172/JCI5001Search in Google Scholar PubMed PubMed Central

52. Li Y, Soos TJ, Li X, Wu J, Degennaro M, Sun X, Littman DR, Birnbaum MJ, Polakiewicz RD. Protein kinase C Theta inhibits insulin signaling by phosphorylating IRS1 at Ser(1101). J Biol Chem 2004;279:45304–7.10.1074/jbc.C400186200Search in Google Scholar PubMed

53. Badin PM, Vila IK, Louche K, Mairal A, Marques MA, Bourlier V, Tavernier G, Langin D, Moro C. High-fat diet-mediated lipotoxicity and insulin resistance is related to impaired lipase expression in mouse skeletal muscle. Endocrinology 2013;154:1444–53.10.1210/en.2012-2029Search in Google Scholar PubMed

54. Ussher JR, Koves TR, Cadete VJ, Zhang L, Jaswal JS, Swyrd SJ, Lopaschuk DG, Proctor SD, Keung W, Muoio DM, Lopaschuk GD. Inhibition of de novo ceramide synthesis reverses diet-induced insulin resistance and enhances whole-body oxygen consumption. Diabetes 2010;59:2453–64.10.2337/db09-1293Search in Google Scholar PubMed PubMed Central

55. Holland WL, Knotts TA, Chavez JA, Wang LP, Hoehn KL, Summers SA. Lipid mediators of insulin resistance. Nutr Rev 2007;65:S39–46.10.1111/j.1753-4887.2007.tb00327.xSearch in Google Scholar PubMed

56. Bikman BT, Summers SA. Ceramides as modulators of cellular and whole-body metabolism. J Clin Invest 2011;121:4222–30.10.1172/JCI57144Search in Google Scholar PubMed PubMed Central

57. Powell DJ, Turban S, Gray A, Hajduch E, Hundal HS. Intracellular ceramide synthesis and protein kinase Czeta activation play an essential role in palmitate-induced insulin resistance in rat L6 skeletal muscle cells. Biochem J 2004;382:619–29.10.1042/BJ20040139Search in Google Scholar PubMed PubMed Central

58. Goudriaan JR, Dahlmans VE, Teusink B, Ouwens DM, Febbraio M, Maassen JA, Romijn JA, Havekes LM, Voshol PJ. CD36 deficiency increases insulin sensitivity in muscle, but induces insulin resistance in the liver in mice. J Lipid Res 2003;44:2270–7.10.1194/jlr.M300143-JLR200Search in Google Scholar PubMed

59. Hajri T, Han XX, Bonen A, Abumrad NA. Defective fatty acid uptake modulates insulin responsiveness and metabolic responses to diet in CD36-null mice. J Clin Invest 2002;109:1381–9.10.1172/JCI0214596Search in Google Scholar

60. Kim JK, Gimeno RE, Higashimori T, Kim HJ, Choi H, Punreddy S, Mozell RL, Tan G, Stricker-Krongrad A, Hirsch DJ, Fillmore JJ, Liu ZX, Dong J, Cline G, Stahl A, Lodish HF, Shulman GI. Inactivation of fatty acid transport protein 1 prevents fat-induced insulin resistance in skeletal muscle. J Clin Invest 2004;113:756–63.10.1172/JCI200418917Search in Google Scholar

61. Liu L, Zhang Y, Chen N, Shi X, Tsang B, Yu YH. Upregulation of myocellular DGAT1 augments triglyceride synthesis in skeletal muscle and protects against fat-induced insulin resistance. J Clin Invest 2007;117:1679–89.10.1172/JCI30565Search in Google Scholar PubMed PubMed Central

62. Levin MC, Monetti M, Watt MJ, Sajan MP, Stevens RD, Bain JR, Newgard CB, Farese RV, Sr., Farese RV, Jr. Increased lipid accumulation and insulin resistance in transgenic mice expressing DGAT2 in glycolytic (type II) muscle. Am J Physiol Endocrinol Metab 2007;293:E1772–81.10.1152/ajpendo.00158.2007Search in Google Scholar PubMed

63. Timmers S, de Vogel-van den Bosch J, Hesselink MK, van Beurden D, Schaart G, Ferraz MJ, Losen M, Martinez-Martinez P, De Baets MH, Aerts JM, Schrauwen P. Paradoxical increase in TAG and DAG content parallel the insulin sensitizing effect of unilateral DGAT1 overexpression in rat skeletal muscle. PLoS One 2011;6:e14503.10.1371/journal.pone.0014503Search in Google Scholar PubMed PubMed Central

64. Sparks LM, Bosma M, Brouwers B, van de Weijer T, Bilet L, Schaart G, Moonen-Kornips E, Eichmann TO, Lass A, Hesselink MK, Schrauwen P. Reduced incorporation of fatty acids into triacylglycerol in myotubes from obese individuals with type 2 diabetes. Diabetes 2014;63:1583–93.10.2337/db13-1123Search in Google Scholar PubMed PubMed Central

65. Olofsson SO, Bostrom P, Andersson L, Rutberg M, Perman J, Boren J. Lipid droplets as dynamic organelles connecting storage and efflux of lipids. Biochim Biophys Acta 2009;1791:448–58.10.1016/j.bbalip.2008.08.001Search in Google Scholar PubMed

66. Fujimoto T, Ohsaki Y. Cytoplasmic lipid droplets: rediscovery of an old structure as a unique platform. Ann NY Acad Sci 2006;1086:104–15.10.1196/annals.1377.010Search in Google Scholar PubMed

67. Tauchi-Sato K, Ozeki S, Houjou T, Taguchi R, Fujimoto T. The surface of lipid droplets is a phospholipid monolayer with a unique Fatty Acid composition. J Biol Chem 2002;277:44507–12.10.1074/jbc.M207712200Search in Google Scholar PubMed

68. Gjelstad IM, Haugen F, Gulseth HL, Norheim F, Jans A, Bakke SS, Raastad T, Tjonna AE, Wisloff U, Blaak EE, Riserus U, Gaster M, Roche HM, Birkeland KI, Drevon CA. Expression of perilipins in human skeletal muscle in vitro and in vivo in relation to diet, exercise and energy balance. Arch Physiol Biochem 2012;118:22–30.10.3109/13813455.2011.630009Search in Google Scholar PubMed

69. Mason RR, Mokhtar R, Matzaris M, Selathurai A, Kowalski GM, Mokbel N, Meikle PJ, Bruce CR, Watt MJ. PLIN5 deletion remodels intracellular lipid composition and causes insulin resistance in muscle. Mol Metab 2014;3:652–63.10.1016/j.molmet.2014.06.002Search in Google Scholar PubMed PubMed Central

70. Granneman JG, Moore HP, Krishnamoorthy R, Rathod M. Perilipin controls lipolysis by regulating the interactions of AB-hydrolase containing 5 (Abhd5) and adipose triglyceride lipase (Atgl). J Biol Chem 2009;284:34538–44.10.1074/jbc.M109.068478Search in Google Scholar PubMed PubMed Central

71. Granneman JG, Moore HP, Granneman RL, Greenberg AS, Obin MS, Zhu Z. Analysis of lipolytic protein trafficking and interactions in adipocytes. J Biol Chem 2007;282:5726–35.10.1074/jbc.M610580200Search in Google Scholar PubMed

72. Subramanian V, Rothenberg A, Gomez C, Cohen AW, Garcia A, Bhattacharyya S, Shapiro L, Dolios G, Wang R, Lisanti MP, Brasaemle DL. Perilipin A mediates the reversible binding of CGI-58 to lipid droplets in 3T3-L1 adipocytes. J Biol Chem 2004;279:42062–71.10.1074/jbc.M407462200Search in Google Scholar

73. Wang H, Sztalryd C. Oxidative tissue: perilipin 5 links storage with the furnace. Trends Endocrinol Metab 2011;22:197–203.10.1016/j.tem.2011.03.008Search in Google Scholar

74. Wang H, Sreenivasan U, Hu H, Saladino A, Polster BM, Lund LM, Gong DW, Stanley WC, Sztalryd C. Perilipin 5, a lipid droplet-associated protein, provides physical and metabolic linkage to mitochondria. J Lipid Res 2011;52:2159–68.10.1194/jlr.M017939Search in Google Scholar

75. Bosma M, Minnaard R, Sparks LM, Schaart G, Losen M, de Baets MH, Duimel H, Kersten S, Bickel PE, Schrauwen P, Hesselink MK. The lipid droplet coat protein perilipin 5 also localizes to muscle mitochondria. Histochem Cell Biol 2012;137:205–16.10.1007/s00418-011-0888-xSearch in Google Scholar

76. Lass A, Zimmermann R, Oberer M, Zechner R. Lipolysis – a highly regulated multi-enzyme complex mediates the catabolism of cellular fat stores. Prog Lipid Res 2011;50:14–27.10.1016/j.plipres.2010.10.004Search in Google Scholar

77. Tornqvist H, Belfrage P. Purification and some properties of a monoacylglycerol-hydrolyzing enzyme of rat adipose tissue. J Biol Chem 1976;251:813–9.10.1016/S0021-9258(17)33857-7Search in Google Scholar

78. Karlsson M, Contreras JA, Hellman U, Tornqvist H, Holm C. cDNA cloning, tissue distribution, and identification of the catalytic triad of monoglyceride lipase. Evolutionary relationship to esterases, lysophospholipases, and haloperoxidases. J Biol Chem 1997;272:27218–23.10.1074/jbc.272.43.27218Search in Google Scholar PubMed

79. Taschler U, Radner FP, Heier C, Schreiber R, Schweiger M, Schoiswohl G, Preiss-Landl K, Jaeger D, Reiter B, Koefeler HC, Wojciechowski J, Theussl C, Penninger JM, Lass A, Haemmerle G, Zechner R, Zimmermann R. Monoglyceride lipase deficiency in mice impairs lipolysis and attenuates diet-induced insulin resistance. J Biol Chem 2011;286:17467–77.10.1074/jbc.M110.215434Search in Google Scholar PubMed PubMed Central

80. Zhao S, Mugabo Y, Iglesias J, Xie L, Delghingaro-Augusto V, Lussier R, Peyot ML, Joly E, Taib B, Davis MA, Brown JM, Abousalham A, Gaisano H, Madiraju SR, Prentki M. alpha/beta-Hydrolase domain-6-accessible monoacylglycerol controls glucose-stimulated insulin secretion. Cell Metab 2014;19: 993–1007.10.1016/j.cmet.2014.04.003Search in Google Scholar PubMed

81. Badin PM, Louche K, Mairal A, Liebisch G, Schmitz G, Rustan AC, Smith SR, Langin D, Moro C. Altered skeletal muscle lipase expression and activity contribute to insulin resistance in humans. Diabetes 2011;60:1734–42.10.2337/db10-1364Search in Google Scholar PubMed PubMed Central

82. Jocken JW, Moro C, Goossens GH, Hansen D, Mairal A, Hesselink MK, Langin D, van Loon LJ, Blaak EE. Skeletal muscle lipase content and activity in obesity and type 2 diabetes. J Clin Endocrinol Metab 2010;95:5449–53.10.1210/jc.2010-0776Search in Google Scholar

83. Kase ET, Feng YZ, Badin PM, Bakke SS, Laurens C, Coue M, Langin D, Gaster M, Thoresen GH, Rustan AC, Moro C. Primary defects in lipolysis and insulin action in skeletal muscle cells from type 2 diabetic individuals. Biochim Biophys Acta 2015;1851:1194–201.10.1016/j.bbalip.2015.03.005Search in Google Scholar

84. Vaughan M, Berger JE, Steinberg D. Hormone-sensitive lipase and monoglyceride lipase activities in adipose tissue. J Biol Chem 1964;239:401–9.10.1016/S0021-9258(18)51692-6Search in Google Scholar

85. Prats C, Donsmark M, Qvortrup K, Londos C, Sztalryd C, Holm C, Galbo H, Ploug T. Decrease in intramuscular lipid droplets and translocation of HSL in response to muscle contraction and epinephrine. J Lipid Res 2006;47:2392–9.10.1194/jlr.M600247-JLR200Search in Google Scholar PubMed

86. Haemmerle G, Zimmermann R, Hayn M, Theussl C, Waeg G, Wagner E, Sattler W, Magin TM, Wagner EF, Zechner R. Hormone-sensitive lipase deficiency in mice causes diglyceride accumulation in adipose tissue, muscle, and testis. J Biol Chem 2002;277:4806–15.10.1074/jbc.M110355200Search in Google Scholar PubMed

87. Zimmermann R, Strauss JG, Haemmerle G, Schoiswohl G, Birner-Gruenberger R, Riederer M, Lass A, Neuberger G, Eisenhaber F, Hermetter A, Zechner R. Fat mobilization in adipose tissue is promoted by adipose triglyceride lipase. Science 2004;306:1383–6.10.1126/science.1100747Search in Google Scholar PubMed

88. Haemmerle G, Lass A, Zimmermann R, Gorkiewicz G, Meyer C, Rozman J, Heldmaier G, Maier R, Theussl C, Eder S, Kratky D, Wagner EF, Klingenspor M, Hoefler G, Zechner R. Defective lipolysis and altered energy metabolism in mice lacking adipose triglyceride lipase. Science 2006;312:734–7.10.1126/science.1123965Search in Google Scholar PubMed

89. Fischer J, Lefevre C, Morava E, Mussini JM, Laforet P, Negre-Salvayre A, Lathrop M, Salvayre R. The gene encoding adipose triglyceride lipase (PNPLA2) is mutated in neutral lipid storage disease with myopathy. Nat Genet 2007;39:28–30.10.1038/ng1951Search in Google Scholar PubMed

90. Sitnick MT, Basantani MK, Cai L, Schoiswohl G, Yazbeck CF, Distefano G, Ritov V, DeLany JP, Schreiber R, Stolz DB, Gardner NP, Kienesberger PC, Pulinilkunnil T, Zechner R, Goodpaster BH, Coen P, Kershaw EE. Skeletal muscle triacylglycerol hydrolysis does not influence metabolic complications of obesity. Diabetes 2013;62:3350–61.10.2337/db13-0500Search in Google Scholar PubMed PubMed Central

91. Dube JJ, Sitnick MT, Schoiswohl G, Wills RC, Basantani MK, Cai L, Pulinilkunnil T, Kershaw EE. Adipose triglyceride lipase deletion from adipocytes, but not skeletal myocytes, impairs acute exercise performance in mice. Am J Physiol Endocrinol Metab 2015;308:E879–90.10.1152/ajpendo.00530.2014Search in Google Scholar PubMed PubMed Central

92. Hoy AJ, Bruce CR, Turpin SM, Morris AJ, Febbraio MA, Watt MJ. Adipose triglyceride lipase-null mice are resistant to high-fat diet-induced insulin resistance despite reduced energy expenditure and ectopic lipid accumulation. Endocrinology 2011;152:48–58.10.1210/en.2010-0661Search in Google Scholar PubMed

93. Lefevre C, Jobard F, Caux F, Bouadjar B, Karaduman A, Heilig R, Lakhdar H, Wollenberg A, Verret JL, Weissenbach J, Ozguc M, Lathrop M, Prud’homme JF, Fischer J. Mutations in CGI-58, the gene encoding a new protein of the esterase/lipase/ thioesterase subfamily, in Chanarin-Dorfman syndrome. Am J Hum Genet 2001;69:1002–12.10.1086/324121Search in Google Scholar PubMed PubMed Central

94. Yang X, Lu X, Lombes M, Rha GB, Chi YI, Guerin TM, Smart EJ, Liu J. The G(0)/G(1) switch gene 2 regulates adipose lipolysis through association with adipose triglyceride lipase. Cell Metab 2010;11:194–205.10.1016/j.cmet.2010.02.003Search in Google Scholar PubMed PubMed Central

95. Schweiger M, Paar M, Eder C, Brandis J, Moser E, Gorkiewicz G, Grond S, Radner FP, Cerk I, Cornaciu I, Oberer M, Kersten S, Zechner R, Zimmermann R, Lass A. G0/G1 switch gene-2 regulates human adipocyte lipolysis by affecting activity and localization of adipose triglyceride lipase. J Lipid Res 2012;53:2307–17.10.1194/jlr.M027409Search in Google Scholar PubMed PubMed Central

96. Everard A, Cani PD. Diabetes, obesity and gut microbiota. Best Pract Res Clin Gastroenterol 2013;27:73–83.10.1016/j.bpg.2013.03.007Search in Google Scholar PubMed

97. Holland WL, Bikman BT, Wang LP, Yuguang G, Sargent KM, Bulchand S, Knotts TA, Shui G, Clegg DJ, Wenk MR, Pagliassotti MJ, Scherer PE, Summers SA. Lipid-induced insulin resistance mediated by the proinflammatory receptor TLR4 requires saturated fatty acid-induced ceramide biosynthesis in mice. J Clin Invest 2011;121:1858–70.10.1172/JCI43378Search in Google Scholar PubMed PubMed Central

98. Hotamisligil GS. Inflammation and endoplasmic reticulum stress in obesity and diabetes. Int J Obes (Lond) 2008;32 Suppl 7:S52–4.10.1002/9780470985571.ch8Search in Google Scholar PubMed

99. Turpin SM, Lancaster GI, Darby I, Febbraio MA, Watt MJ. Apoptosis in skeletal muscle myotubes is induced by ceramides and is positively related to insulin resistance. Am J Physiol Endocrinol Metab 2006;291:E1341–50.10.1152/ajpendo.00095.2006Search in Google Scholar PubMed

Received: 2015-10-1
Accepted: 2015-11-18
Published Online: 2015-12-19
Published in Print: 2016-4-1

©2016 by De Gruyter

Downloaded on 28.4.2024 from https://www.degruyter.com/document/doi/10.1515/hmbci-2015-0045/html
Scroll to top button