Skip to main content
Adipocyte logoLink to Adipocyte
. 2012 Jul 1;1(3):123–131. doi: 10.4161/adip.20035

Distinct roles for α-β hydrolase domain 5 (ABHD5/CGI-58) and adipose triglyceride lipase (ATGL/PNPLA2) in lipid metabolism and signaling

Caleb C Lord 1, J Mark Brown 1,*
PMCID: PMC3492958  NIHMSID: NIHMS398287  PMID: 23145367

Abstract

Catabolism of stored triacylglycerol (TAG) from cytoplasmic lipid droplets is critical for providing energy substrates, membrane building blocks and signaling lipids in most cells of the body. However, the lipolytic machinery dictating TAG hydrolysis varies greatly among different cell types. Within the adipocyte, TAG hydrolysis is dynamically regulated by hormones to ensure appropriate metabolic adaptation to nutritional and physiologic cues. In other cell types such as hepatocytes, myocytes and macrophages, mobilization of stored TAG is regulated quite differently. Within the last decade, mutations in two key genes involved in TAG hydrolysis, α-β hydrolase domain 5 (ABHD5/CGI-58) and adipose triglyceride lipase (ATGL/PNPLA2), were found to cause two distinct neutral lipid storage diseases (NLSD) in humans. These genetic links, along with supporting evidence in mouse models, have prompted a number of studies surrounding the biochemical function(s) of these proteins. Although both CGI-58 and ATGL have been clearly implicated in TAG hydrolysis in multiple tissues and have even been shown to physically interact with each other, recent evidence suggests that they may also have distinct roles. The purpose of this review is to summarize the most recent insights into how CGI-58 and ATGL regulate lipid metabolism and signaling.

Keywords: acyltransferase, insulin resistance, lipase, lipolysis, obesity, phosphatidic acid, triacylglycerol

Introduction

We recently reported that CGI-58 generates signaling lipids that regulate inflammation and insulin action.1 This review expands on the concepts presented in our initial report1 and provides additional discussion of the mechanisms by which CGI-58 and the TAG hydrolase ATGL regulate lipid metabolism and signal transduction. We have shown that antisense oligonucleotide (ASO)-mediated knockdown of CGI-58 in mice results in severe hepatic steatosis, yet paradoxically improves hepatic insulin signaling.1,2 CGI-58 knockdown: (1) prevents HFD-induced activation of hepatic stress kinases, despite elevated plasma TH1 cytokines; (2) prevents the generation of phosphatidic acid (PA) species and other glycerophospholipids in the liver in response to TNFα, thereby attenuating downstream inflammatory signaling; and (3) alters the systemic inflammatory response to endotoxin.1 Inhibition of inflammatory pathways may explain the dissociation of hepatic steatosis and insulin resistance in CGI-58 ASO-treated mice.1,2 Importantly, lipidomic analysis shows several key changes in PA and other glycerophospholipid species with signaling potential in the liver.1 Collectively, we propose that CGI-58 generates signaling lipids that can activate inflammatory pathways and downregulate hepatic insulin signaling (Fig. 1B).1 ATGL/PNPLA2, also known as desnutrin3 or inducible phospholipase A2ζ,4 is a member of the patatin-like phospholipase domain containing (PNPLA) family of proteins, which have diverse lipid esterase and transacylase activities.5 In addition to selective TAG hydrolase activity, ATGL has also been reported to possess phospholipase activity and acyl-CoA-independent acylglycerol transacylase activity, but the physiological functions of these activities are not currently known.4 CGI-58/ABHD5 is a member of the α-β hydrolase domain (ABHD) family of proteins which share a highly conserved GXSXG active site motif. In CGI-58, the catalytic serine nucleophile is replaced by an asparagine,6 and CGI-58 does not possess hydrolase activity to directly mobilize TAG. Rather, it is widely accepted that CGI-58 co-activates the TAG hydrolase activity of ATGL through an incompletely understood mechanism.7-9 Lass and colleagues7 first demonstrated that recombinant CGI-58, or extracts from CGI-58-overexpressing cells, increases the TAG hydrolase activity of both human and mouse ATGL in vitro. Purified CGI-58 increases TAG hydrolase activity in adipose tissue extracts from wild-type mice, but not in extracts from ATGL knockout (ATGL−/−) mice.8 This suggests that CGI-58 specifically co-activates ATGL-mediated lipolysis and that endogenous levels of CGI-58 in adipose tissue are limiting for maximal ATGL lipase activity.8 In addition, CGI-58 possesses intrinsic coA-dependent lysophosphatidic acid acyltransferase (LPAAT) activity in vitro, generating PA.10,11 Since either ATGL-mediated TAG hydrolysis or LPAAT activity could generate signaling lipids, the relative physiological importance of these functions becomes central to understanding the striking effects of CGI-58 knockdown on inflammation and insulin signaling.1 Although CGI-58 co-activation of ATGL-mediated lipolysis has been well-established in vitro,7-9 whether this is the primary physiological function of CGI-58 in vivo remains to be verified. Despite many similarities between CGI-58 and ATGL deficiency, several key differences seem to indicate additional, ATGL-independent functions of CGI-58 (Table 1). Here, we summarize recent insights into the function(s) of CGI-58 and ATGL in vivo, based on studies in humans and mice.

graphic file with name adip-1-123-g1.jpg

Figure 1. CGI-58 and ATGL functions in adipocytes (A) and hepatocytes (B). (A) Within the adipocyte, triacylglycerol (TAG) hydrolysis is dynamically regulated by hormones to ensure appropriate metabolic adaptation to nutritional and physiologic cues. In the basal state, CGI-58 is primarily sequestered by perilipin-1(Plin-1) at the lipid droplet, ATGL is localized at the lipid droplet, and hormone sensitive lipase (HSL) resides in the cytoplasm. Downstream of catecholamine stimulation through β-adrenergic receptors (βAR), cAMP-activated protein kinase A (PKA) phosphorylates Plin-1 and HSL. Plin-1 phosphorylation results in the release of CGI-58. CGI-58 reversibly disperses to the cytoplasm, while a small amount remains at the lipid droplet, interacting with ATGL to co-activate TAG hydrolysis. Meanwhile, phosphorylated HSL translocates to the lipid droplet and interacts closely with Plin-1, preferentially hydrolyzing diacylglycerol (DAG) to monoacylglycerol (MAG). In the final step of lipolysis, MAG is hydrolyzed by MAG lipase (MAGL), leading to glycerol release from the adipocyte. Based on the localization of CGI-58 in the cytoplasm during stimulated lipolysis, CGI-58 may have a physiological function distinct from the co-activation of ATGL-mediated lipolysis. The role of CGI-58 LPA acyltransferase (LPAAT) activity in adipocytes is not known, but CGI-58-derived PA and downstream products might participate in signal transduction. (B) Within the hepatocyte, CGI-58 generates signaling lipids to activate inflammatory kinases and dampen insulin signaling. Downstream of nutrient oversupply or systemic inflammation, CGI-58-derived PA could serve as a precursor to pro-inflammatory signaling lipids that lead to the activation of stress kinases such as IκB kinase (IKK), c-jun N-terminal kinase (JNK) and mammalian target of rapamycin (mTOR). CGI-58-derived PA might directly activate mTOR complex 1(mTORC1), similar to PA generated by phospholipase D (PLD). mTORC1 activates S6 kinase 1 (S6K1) which phosphorylates insulin receptor substrate-1 (IRS-1) at Ser1101 and other residues to inhibit insulin signaling through the insulin receptor (IR). mTOR complex 2 (mTORC2) mediates insulin action by phosphorylating Akt at Ser473. PA generated within the glycerolipid synthesis pathway can impair insulin signaling through mTORC2. CGI-58-derived PA might also inactivate mTORC2 to prevent Akt phosphorylation. Phosphorylated Akt inactivates forkhead box protein O1 (FoxO1), a transcription factor that drives expression of genes involved in gluconeogenesis and very low density lipoprotein (VLDL) secretion. In addition to signaling lipids derived through CGI-58 LPAAT activity, it is possible that ATGL-mediated lipolysis could produce precursors to signaling lipids. Products of ATGL-mediated lipolysis can serve as signaling lipids to activate PPARα, a transcription factor that drives oxidative gene expression. CGI-58’s role in hepatic TAG mobilization and PPARα target gene expression is likely ATGL-dependent (blue lines). Unlike CGI-58, ATGL does not significantly affect hepatic insulin signaling. Thus, CGI-58’s role in the regulation of insulin signaling is proposed to be independent of ATGL (red lines). Direct effects are denoted by solid lines. Potential pathway effects are indicated by dotted lines. CGI-58, comparative gene identification-58; ATGL, adipose triglyceride lipase; LPA, lysophosphatidic acid; PA, phosphatidic acid; PI3Ks, phosphoinositide 3 kinases; PDPK1, phosphoinositide dependent protein kinase 1; GPAT, glycerol-3-phosphate acyltransferases; AGPAT, sn-1-acylglycerol-3-phosphate acyltransferase; PAP, phosphatidic acid phosphatase; DGAT, diglyceride acyltransferase.

Table 1. Comparison of CGI-58 and ATGL mouse models.

Endpoint Mouse model Phenotype vs. control mice
Obesity and adipose tissue
ASO-mediated CGI-58 knockdown2
Complete prevention of HFD-induced obesity
50% lower fat mass on both chow and HFD
No difference in BW on chow diet
Adipose-specific CGI-58 overexpression36
No difference in BW or HFD-induced obesity
No difference in lipolysis
No difference in fat mass or adipocyte size
Whole-body ATGL knockout18,45
Higher rate of HFD-induced obesity
Higher fat mass
Larger adipocyte size
Blunted lipolysis
Adipose-specific ATGL knockout32
Higher rate of HFD-induced obesity
Higher fat mass
Larger adipocyte size
Blunted lipolysis
Adipose-specific ATGL overexpression33
Attenuated HFD-induced obesity
Lower fat mass
Smaller adipocyte size
Lower adipocyte TAG content
Increased lipolysis
Liver lipid
metabolism
Whole-body CGI-58 knockout9
Higher TAG
Lower TAG hydrolase activity
ASO-mediated CGI-58 knockdown1,2
Higher TAG, DAG, and ceramides
Lower TAG hydrolase activity
Lower VLDL-TAG secretion
Lower β-oxidation
Whole-body ATGL knockout9,18,40
Higher TAG
Lower TAG hydrolase activity
Liver-specific ATGL knockout41
Higher TAG
Lower TAG hydrolase activity
No difference in VLDL-TAG secretion
No difference in DAG and ceramides
Adenovirus-mediated ATGL knockdown42
Higher TAG
Lower TAG hydrolase activity
No difference in VLDL-TAG secretion
Adenoviral ATGL overexpression21,44
Lower TAG
No difference in VLDL-TAG secretion
Higher rate of NEFA release into plasma
Higher rate of FA oxidation
Insulin signaling
ASO-mediated CGI-58 knockdown1,2
Improved whole-body glucose tolerance on chow and HFD
Improved whole-body insulin tolerance on HFD
Improved hepatic insulin signaling
Whole-body ATGL knockout18,40,44,45
Improved whole-body glucose tolerance on chow and HFD
Improved whole-body insulin tolerance on chow and HFD
Unchanged or slightly impaired hepatic insulin signaling
No difference in insulin signaling in primary hepatocytes
Adenoviral ATGL overexpression44
Slight improvement in insulin signaling
Lipid signaling ASO-mediated CGI-58 knockdown1,2
Lower PPARα target gene expression in liver
Altered hepatic glycerophospholipids
Prevention of hepatic stress kinase activation and inflammatory signaling
Whole-body ATGL knockout60
Lower PPARα target gene expression in liver
Steatosis and cardiomyopathy prevented by PPARα agonist
Adenovirus-mediated ATGL knockdown42 Lower PPARα target gene expression in liver

ASO, antisense oligonucleotide; HFD, high fat diet; BW, body weight; TAG, triacylglycerol; DAG, diacylglycerol; FA, fatty acid; NEFA, non-esterified fatty acid; VLDL, very low density lipoprotein.

CGI-58 and ATGL Mutations Cause Neutral Lipid Storage Disease in Humans

Neutral lipid storage disease (NLSD) is a rare autosomal recessive, nonlysosomal disorder characterized by ectopic TAG deposition in multiple tissues, including the hallmark accumulation of lipid vacuoles in blood leukocytes (Jordan anomaly).12 NLSD patients can be divided into two subsets: NLSD with ichthyosis (NLSDI), also known as Chanarin-Dorfman syndrome, and NLSD with myopathy (NLSDM).12,13 Mutations in CGI-58 cause NLSDI,14 while ATGL mutations cause NLSDM.15 All known CGI-58 mutations cause ichthyosis, whereas ATGL mutations always cause cardiomyopathy yet do not result in ichthyosis.13 Hepatomegaly and hepatic steatosis occur more frequently with CGI-58 mutations than with ATGL mutations.13 CGI-58 mutations are also commonly associated with neurological disorders, which are not seen with ATGL mutations.13 Interestingly, obesity has not been observed in patients with NLSDI and NLSDM,12,13 suggesting either that adipose lipolysis is not significantly decreased or that other factors compensate to prevent obesity in these patients. In skin fibroblasts from a NLSDI patient lacking functional CGI-58, TAG accumulation appears to be secondary to a defect in phospholipid metabolism.16,17 The phenotypic differences between patients with NLSDI and NLSDM clearly indicate distinct roles for CGI-58 and ATGL, at least in the skin and nervous system. Additional studies in patients with primary CGI-58 and ATGL mutations will be extremely important to clarify the shared and distinct functions of these proteins in the context of human physiology and pathology.

CGI-58 and ATGL in the Skin

Whole-body CGI-58 knockout (CGI-58−/−) mice die within hours after birth due to a skin barrier defect.9 The skin of CGI-58−/− mice shows characteristics of lamellar ichthyosis, similar to NLSDI in humans.9 In contrast, ATGL−/− mice do not develop ichthyosis and survive to 12–14 weeks of age before dying of cardiomyopathy.18 Dermal TAG hydrolase activity is lower in both CGI-58−/− mice and ATGL−/− mice compared with wild-type mice.9 However, epidermal TAG hydrolase activity is 80% lower in CGI-58−/− mice but is unchanged in ATGL−/− mice.9 This difference clearly supports an ATGL-independent function of CGI-58 in the epidermis. Perhaps CGI-58 co-activates an unknown lipase to hydrolyze TAG for the production of skin barrier lipids. Alternatively, CGI-58 could function in glycerophospholipid metabolism to generate signaling lipids that are important for keratinocyte differentiation and skin barrier formation. Interestingly, PNPLA1, a protein that shares substantial homology with ATGL/PNPLA2, was recently discovered to be mutated in autosomal recessive congenital lamellar ichthyosis in humans and dogs.19 Unlike ATGL, PNPLA1 does not exhibit CGI-58-driven TAG hydrolase activity, and PNPLA1 mutations do not cause neutral lipid accumulation in isolated keratinocytes.19 Furthermore, PNPLA1 mutations do not result in neutral lipid vacuoles in blood leukocytes (Jordan anomaly),19 a hallmark of NLSD.12 Rather, levels of PA and phosphatidylethanolamine (PE) are significantly lower in PNPLA1 mutant keratinocytes, suggesting that PNPLA1 may possess acyltransferase activity and could function in glycerophospholipid synthesis or remodeling.19 Based on CGI-58’s LPAAT activity,10,11 it is possible that altered glycerophospholipid metabolism in the skin could contribute to NLSDI. In fact, altered phospholipid metabolism appears to be the primary defect in human NLSDI skin fibroblasts.16,17 However, the role of CGI-58 LPAAT activity in the skin remains unclear, since human CGI-58 mutations associated with ichthyosis do not change the LPAAT activity of the protein in vitro.10 In addition, total levels of PE are not altered in the epidermis of CGI-58−/− mice.9 Further work is needed to elucidate the ATGL-independent function of CGI-58 in the skin. It would be interesting to explore the possibility that mutations in CGI-58 and PNPLA1 may cause ichthyosis through similar glycerophospholipid pathways in the skin. Since ASO-mediated knockdown of CGI-58 does not result in ichthyosis, future studies of CGI-58 in the skin will require conditional knockout mouse models.

CGI-58 and ATGL in White Adipose Tissue

Both CGI-58 and ATGL are highly expressed in white adipose tissue (WAT).2,20,21 ATGL and hormone sensitive lipase (HSL) together account for more than 90% percent of TAG hydrolase activity in mouse WAT.8 In WAT of ATGL−/− mice, TAG hydrolase activity is 80% lower than in wild-type mice, suggesting that ATGL is the rate-limiting enzyme for TAG hydrolysis in WAT, whereas HSL functions primarily as a diacylglycerol hydrolase.18,22 A general model of lipolytic trafficking in adipocytes is depicted in Figure 1A. In the basal state, CGI-58 is primarily sequestered by perilipin-1 (Plin-1) at the lipid droplet (LD),23-25 and ATGL is localized to the LD.26 During hormone-stimulated lipolysis, cAMP-activated protein kinase A (PKA) phosphorylates Plin-1,27-29 resulting in the release of CGI-58 to interact with ATGL at the LD30 or to reversibly disperse to the cytoplasm.23,25 Meanwhile, phosphorylated HSL moves from the cytoplasm to the LD28,31 and interacts closely with Plin-130 to preferentially hydrolyze DAG22 generated by ATGL. In agreement with a rate-limiting role in TAG hydrolysis,8 both whole-body and adipose-specific ATGL knockout mice are mildly obese, with higher fat pad weight and larger adipocyte size, and have blunted lipolysis.18,32 Conversely, adipose-specific ATGL overexpression in HFD-fed mice results in higher TAG hydrolase activity in WAT, attenuated HFD-induced obesity, lower fat pad weight, smaller adipocyte size, and lower TAG content in adipocytes.33 ATGL overexpression has no effect on fat mass in chow-fed mice.33 In addition, ATGL overexpression in WAT increases lipolysis but does not result in elevated serum NEFA or ectopic TAG accumulation, possibly indicating increased FA oxidation in adipose.33 If CGI-58 functions as the co-activator of ATGL-mediated lipolysis in WAT,8 the phenotype of CGI-58 knockdown should be similar to ATGL deficiency. However, in striking contrast to ATGL deficiency, 80–95% knockdown of CGI-58 expression in WAT results in 50% lower fat mass in both chow-fed and HFD-fed mice.2 CGI-58 knockdown does not change body weight in chow-fed mice but completely prevents HFD-induced obesity.2 Neither ATGL nor CGI-58 appears to have an effect on food intake.2,18,32 Although the prevention of adiposity resembles overexpression of ATGL,33 ATGL mRNA levels in WAT are not higher in CGI-58 ASO-treated mice.2 In HFD-fed CGI-58 ASO-treated mice, plasma NEFA levels are 35% lower in both the fed and fasted states, compared with control ASO mice,2 possibly reflecting less ATGL-mediated lipolysis in WAT. Similarly, Radner and colleagues reported 60% lower plasma NEFA in newborn CGI-58−/− mice.9 It is conceivable that CGI-58 could co-activate ATGL to drive TAG hydrolysis, yet also promote expansion of WAT through an ATGL-independent mechanism. For example, ATGL-mediated lipolysis might provide lipid substrates for a separate function of CGI-58 in glycerophospholipid synthesis or signaling lipid generation in adipocytes. Interestingly, Yang and colleagues reported the existence of an alternatively-spliced isoform of mouse CGI-58 lacking a putative lipid binding domain.34 This isoform retains LPAAT activity yet is predominately cytoplasmic and does not co-activate ATGL lipase activity.34 A similar isoform of CGI-58 has not been identified in humans, and the physiological function of this cytoplasmic isoform in mice remains unknown.34 It is noteworthy that in 3T3-L1 adipocytes, the vast majority of CGI-58 disperses from the LD to the cytoplasm following lipolytic stimulation,23,25 while only a small amount remains at the LD, presumably interacting with ATGL.30,35 The fact that the vast majority of CGI-58 moves away from the LD during stimulated lipolysis, and the finding that CGI-58 knockdown prevents HFD-induced obesity seem to suggest that CGI-58 has a physiological function in WAT apart from the co-activation of ATGL-mediated TAG hydrolysis (Fig. 1A). At the same time, it is important to note that CGI-58 knockdown is not adipocyte-specific.2 CGI-58 knockdown in other ASO-targeted tissues, namely the liver and macrophages, could possibly influence adiposity. However, overexpression of CGI-58 using the aP2 promotor (adipose-selective) in mice has no effect on diet-induced obesity, fat pad weight, or adipocyte size.36 Lipolysis in vivo is not altered in response to fasting or treatment with a β3-adrenergic receptor agonist.36 Likewise, neither basal nor isoproterenol-stimulated lipolysis is affected in isolated adipocytes.36 These data suggest that CGI-58 is at least not limiting for ATGL-mediated lipolysis. Although ATGL is clearly a rate-limiting enzyme in adipocyte lipolysis, additional work is needed to clarify the physiological function of CGI-58 in adipocytes and to determine the basis for the opposite effects of CGI-58 and ATGL deficiency on adiposity.

CGI-58 and ATGL in the Liver

In human NLSD, hepatomegaly and hepatic steatosis are more common with CGI-58 mutations than with ATGL mutations.13 The regulation of CGI-58 and ATGL subcellular localization and trafficking is less clearly defined in the liver and other non-adipose tissues that do not express Plin-1. In these tissues, it is likely that other perilipin family proteins such as perilipin-224,37 and perilipin-538,39 play an important role in regulating the interaction of CGI-58 and ATGL. In mice, ATGL deficiency (whole-body knockout,9,18,40 liver-specific knockout41 and adenovirus-mediated knockdown42) has been reported to result in elevated hepatic TAG, although the effect has been variable, ranging from 62% higher40 to 4-fold higher levels of TAG.9 CGI-58 deficiency likewise results in markedly higher hepatic TAG levels compared with control mice.1,2,9 Radner and colleagues9 reported 4-fold higher hepatic TAG in newborn CGI-58−/− mice, and we have measured 2 to 4-fold more hepatic TAG in CGI-58 ASO-treated mice.1,2 Based on more frequent liver complications in humans with NLSDI,13 CGI-58 deficiency might be expected to result in more severe steatosis than ATGL deficiency. However, the variable range of steatosis observed in ATGL studies and the limited number of CGI-58 studies make it difficult to interpret the relative degree of steatosis. Comparison of actual hepatic TAG values is further complicated by experimental differences in the background strains, diets, and ages of the mice. Clearly, both proteins have an important role in TAG metabolism in the liver. Although expression of ATGL is much lower in liver than in WAT,21 ATGL deficiency results in significantly lower total TAG hydrolase activity in the liver.9,18,21,41,42 Likewise, TAG hydrolase activity and intracellular NEFA levels are significantly lower in the liver of CGI-58 ASO-treated mice.2 Interestingly, newborn CGI-58−/− mice have 73% lower TAG hydrolase activity in the liver compared with wild-type mice, while newborn ATGL−/− mice have only 46% lower activity.9 This could indicate a role for CGI-58 in hepatic TAG mobilization beyond the co-activation of ATGL-mediated hydrolysis. In CGI-58 ASO-treated mice fed HFD, fasting-induced plasma levels of β-hydroxybutyrate are significantly lower compared with control mice, suggesting less FA oxidation in the liver.2 In addition, CGI-58 knockdown results in a significantly lower rate of very low density lipoprotein (VLDL) secretion from the liver,2 suggesting that CGI-58-driven TAG mobilization is linked to the secretion of TAG-rich lipoproteins by the liver. This dual role for CGI-58 in promoting FA oxidation and VLDL-TAG secretion has been previously reported using gain-of-function and loss-of-function cell models.20,43 In contrast, ATGL mobilization of hepatic TAG appears to preferentially lead to FA oxidation, without changing VLDL secretion. Haemmerle and colleagues reported that ATGL−/− mice have lower plasma VLDL levels, accounting for lower fasting plasma TAG.18 However, liver-specific ATGL deletion41 or knockdown42 does not alter the rate of VLDL secretion from the liver. This makes it tempting to speculate ATGL deficiency may alter VLDL turnover or clearance instead of hepatic production. In addition, liver-specific overexpression of ATGL in obese mice attenuates hepatic steatosis, but does not change TAG or ApoB secretion from the liver.21 Rather, ATGL mobilization of hepatic TAG is associated with FA oxidation and NEFA release into plasma, as well as elevated fasting-induced ketogenesis.21,42,44 Another important observation is that CGI-58 knockdown results in the accumulation of DAG and ceramides in the liver,1,2 whereas ATGL deficiency has been associated with normal or slightly lower DAG and ceramide content in the liver.40,44 In sum, whether CGI-58 and ATGL have distinct roles in hepatic TAG metabolism remains inconclusive based on complex and subtle differences. However, an ATGL-independent function of CGI-58 in the liver is more clearly suggested by differences in hepatic insulin signaling.

The Role of CGI-58 and ATGL in Insulin Sensitivity

Despite hepatic accumulation of TAG, DAG and ceramides, CGI-58 knockdown improves whole-body glucose tolerance and insulin sensitivity.2 In fact, CGI-58 ASO-treated mice are completely protected against HFD-induced hyperglycemia, glucose intolerance and systemic insulin resistance.2 ATGL−/− mice also have significant improvement in whole-body glucose tolerance and insulin sensitivity,18,40,45 but the underlying mechanisms likely differ significantly. In both ATGL−/− mice and CGI-58 ASO-treated mice, plasma insulin levels are very low compared with wild-type controls.2,18,45 It has been shown that ATGL is necessary for glucose-stimulated insulin secretion (GSIS) from pancreatic β-cells in mice.46 In agreement, plasma insulin does not increase in response to a glucose challenge in ATGL−/− mice.45 Although a role for CGI-58 in GSIS has not been examined, defective GSIS is unlikely in CGI-58 ASO-treated mice because ASOs do not knock down CGI-58 in the pancreas (unpublished data). Furthermore, plasma insulin increases in response to a glucose challenge in CGI-58 ASO-treated mice,2 suggesting that enhanced insulin sensitivity, rather than impaired GSIS, likely accounts for reduced plasma insulin levels. In fact, we have shown that CGI-58 knockdown results in significantly more Akt phosphorylation (Ser473 and Thr308) in the liver in response to insulin injected into the portal vein.1 Insulin-stimulated Akt phosphorylation is not altered in skeletal muscle or WAT,1 suggesting that the liver primarily accounts for enhanced whole-body insulin sensitivity in CGI-58 ASO-treated mice.2 In contrast, Kienesberger and colleagues45 reported that chow-fed ATGL−/− mice have diminished hepatic Akt phosphorylation at Thr308 and impaired Akt activity in response to intraperitoneally administered insulin, while Akt phosphorylation at Ser473 is not affected. The same study showed that insulin-stimulated Akt phosphorylation (Ser473) is increased in skeletal muscle of ATGL−/− mice, suggesting that skeletal muscle, rather than the liver, primarily contributes to whole-body insulin sensitivity in ATGL−/− mice in vivo.45 On the other hand, Hoy and colleagues40 found that in conjunction with insulin administration, glucose uptake was significantly higher in the liver and cardiac muscle of HFD-fed ATGL−/− mice compared with wild-type mice, but was not altered in skeletal muscle. Although this suggested improved hepatic insulin signaling in ATGL−/− mice, insulin-stimulated Akt phosphorylation (Ser473) in the liver was not different compared with wild-type mice.40 Turpin and colleagues44 also reported that Akt phosphorylation is not affected in primary hepatocytes isolated from ATGL−/− mice. In addition, these authors showed that adenoviral overexpression of ATGL in obese mice significantly attenuates hepatic steatosis but only mildly improves hepatic insulin sensitivity.44 Interestingly, both adipose-specific ATGL deficiency32 and overexpression33 have been associated with enhanced hepatic insulin sensitivity, but in both cases the improvement is likely due to an amelioration of hepatic steatosis. In contrast to ATGL deficiency, ASO-mediated CGI-58 knockdown significantly improves Akt phosphorylation in the liver despite marked accumulation of TAG, DAG and ceramides.1,2 Thus, although ATGL and CGI-58 are both implicated in regulating systemic insulin sensitivity, only CGI-58 knockdown improves hepatic insulin signaling.1,2 Whereas liver-specific ATGL deficiency41 or overexpression44 does not alter hepatic inflammation in HFD-fed mice, CGI-58 knockdown in the liver significantly blunts phosphorylation of IκBα, JNK and S6 in response to inflammatory cytokines (TNFα, IL-1β and IL-6).1 In addition, CGI-58 knockdown in the liver is associated with the prevention of HFD-induced stress kinase activation (IKK, mTOR, S6K1 and JNK) and prevention of serine phosphorylation of insulin receptor substrate-1 (IRS1) at Ser1101.1 It is well-known that phospholipase D (PLD)-derived PA can activate the mammalian target of rapamycin complex 1 (mTORC1).47-50 mTORC1 then activates S6 kinase 1 (S6K1) which phosphorylates IRS-1 on serines residues and inhibits insulin signaling.51 Since CGI-58 knockdown prevents HFD-induced phosphorylation of IRS1 (Ser1101) in the liver,1 it is possible that CGI-58-derived PA can directly activate mTORC1 to dampen insulin signaling (Fig. 1B). Recently, Zhang and colleagues elegantly demonstrated that PA in the glycerolipid synthetic pathway can impair mTORC2-mediated phosphorylation of Akt (Ser473) in mouse primary hepatocytes.52 Although we did not examine mTORC2 activity in the context of CGI-58 knockdown, this mechanism could also contribute to improved Akt phosphorylation (Ser473) in the liver of CGI-58 ASO-treated mice.1 Taken together, it is possible that CGI-58-derived PA could both activate mTORC1 and diminish mTORC2 activity (Fig. 1B). Improved insulin signaling could also alter hepatic TAG metabolism in CGI-58 ASO-treated mice. Insulin suppresses VLDL secretion,53 likely through apoB degradation54 and reduced FoxO1-driven MTP expression.55-57 It is tempting to speculate that reduced VLDL secretion in CGI-58 ASO-treated mice2 might partially result from improved Akt-mediated inactivation of FoxO1.1 Collectively, these studies suggest that CGI-58 regulates insulin action in the liver through an ATGL-independent mechanism, either the generation of PA through LPAAT activity or perhaps an unknown function of CGI-58 in signaling pathways.

The Role of CGI-58 and ATGL in Lipid Signaling

We have shown that the attenuation of hepatic inflammation in CGI-58 ASO-treated mice is associated with changes in multiple lipid species with signaling potential.1 Signaling defects are apparent downstream of several cytokine receptors, and it is difficult to identify which lipid species are ultimately responsible for the effects on signaling.1 Although LPAAT activity is the only enzymatic function of CGI-58 described to date,10,11 it is possible that CGI-58 may also modify other lipid substrates in addition to LPA. CGI-58-generated PA could act directly in signaling pathways or serve as a precursor to other signaling glycerophospholipid species (Fig. 1B). In fact, the effects of CGI-58 knockdown on inflammation likely result from changes in multiple lipid species. We were not able to detect a change in total hepatic LPAAT activity in CGI-58 ASO-treated mice.1 Similarly, total LPAAT activity is unchanged in whole liver lysates from CGI-58−/− mice.9 However, this does not rule out an important role for CGI-58 as a signaling LPAAT in the liver. The levels of CGI-58-derived PA could be masked by total cellular PA from other sources and might act locally without a major impact on total glycerophospholipid levels. In addition, CGI-58 activity in vivo could be regulated by mechanisms not detectable in cell lysates. Collectively, these factors may explain why seemingly modest changes in hepatic PA and other glycerophospholipid species have a profound effect on inflammatory signaling in CGI-58 ASO-treated mice.1 It is also possible that signaling lipids could derive from CGI-58’s ability to co-activate ATGL-mediated TAG hydrolysis (Fig. 1B).7-9 The effect of ATGL deficiency on hepatic inflammatory kinase activation has not been reported. However, liver-specific ATGL deficiency does not alter hepatic inflammatory cytokine expression in HFD-fed mice.41 Interestingly, Fuchs and colleagues58 recently reported that ATGL−/− mice are protected from hepatic ER stress and inflammatory gene expression (TNFα and iNOS) in response to tunicamycin. In macrophages, ATGL deficiency results in lower IL-6 expression and impaired migration.59 Further studies are needed to clarify whether specific inflammatory pathways are altered by ATGL deficiency. Haemmerle and colleagues recently showed that ATGL−/− mice have severely blunted PPARα target gene expression in the liver and heart and that ATGL-mediated TAG hydrolysis is required for the activation of hepatic gene transcription by the PPARα-PCG-1 complex.60 In agreement, Ong and colleagues42 reported that adenoviral knockdown of ATGL in mice results in lower hepatic expression of PPARα and target genes. ATGL-mediated lipolysis can also activate PPARα in adipose tissue.32,33 Adipose-specific ATGL knockout mice have reduced PPARα binding to genes involved in thermogenesis and show impaired thermogenesis similar to PPARα−/− mice.32 Conversely, adipose-specific ATGL overexpression results in elevated thermogenic gene expression and FA oxidation in adipose tissue.33 Collectively, these studies suggest that ATGL-mediated TAG hydrolysis generates endogenous ligands of PPARα (Fig. 1B). Remarkably, long-term treatment with a PPARα agonist prevents cardiomyopathy-related death and completely reverses hepatic TAG accumulation in ATGL−/− mice.60 Hepatic PPARα target gene expression is also lower in CGI-58 ASO-treated mice2 (unpublished data), possibly reflecting the loss of PPARα ligands derived from ATGL-mediated TAG hydrolysis. Alternatively, CGI-58 could generate endogenous PPARα ligands through glycerophospholipid pathways.61 In addition to signaling in the liver,1 it is possible that CGI-58 could function in lipid signaling in the nervous system, since neurological disorders are frequently observed in NLSDI patients.12,13 Interestingly, ABHD4, which shares the highest sequence homology (55%) with CGI-58/ABHD5, has been shown to generate precursors for endocannabinoid signaling lipids in the central nervous system.6 In sum, further studies will be needed to examine the role of ATGL in inflammatory signaling and to determine the source of CGI-58-derived signaling lipids in the liver,1 as well as putative signaling functions in other tissues.

Conclusion

In summary, several recent studies provide insight into possible ATGL-independent functions of CGI-58 in humans and mice (Table 1). Despite significant advances in our understanding of the biochemistry of CGI-58, the primary physiological function of CGI-58 still remains unclear. An important next step will be clearly distinguishing which effects of CGI-58 deficiency result from decreased ATGL-mediated lipolysis vs. altered signal transduction. Based on the studies discussed here, several questions remain: does CGI-58 knockdown limit ATGL-mediated lipolysis in vivo? What is the function of CGI-58 in adipose tissue, and why are CGI-58 ASO-treated mice protected from obesity while ATGL−/− mice are obese? Do ATGL and CGI-58 promote hepatic TAG mobilization through distinct or common mechanisms? Does CGI-58 regulate hepatic insulin signaling and inflammation through ATGL-mediated lipolysis or through LPAAT activity? Which CGI-58-derived signaling lipids promote inflammatory signaling in the liver? The answers to these and similar questions will greatly advance our understanding of how CGI-58 and ATGL are related to the development of metabolic disease. Tissue-specific CGI-58 and ATGL knockout mouse models will be essential to understanding cell-autonomous alterations in signaling and metabolism. In conclusion, both CGI-58 and ATGL are critical enzymes in the regulation of TAG metabolism and cellular signaling, yet many additional studies are required to clarify their individual contributions to NLSD, obesity and type 2 diabetes.

Acknowledgments

This work was supported in part by the National Heart, Lung, and Blood Institute (R00-HL096166) and the American Heart Association (11BGIA-7840072) to J.M.B.

Footnotes

References

  • 1.Lord CC, Betters JL, Ivanova PT, Milne SB, Myers DS, Madenspacher J, et al. CGI-58/ABHD5-derived signaling lipids regulate systemic inflammation and insulin action. Diabetes. 2012;61:355–63. doi: 10.2337/db11-0994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Brown JM, Betters JL, Lord C, Ma Y, Han X, Yang K, et al. CGI-58 knockdown in mice causes hepatic steatosis but prevents diet-induced obesity and glucose intolerance. J Lipid Res. 2010;51:3306–15. doi: 10.1194/jlr.M010256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Villena JA, Roy S, Sarkadi-Nagy E, Kim KH, Sul HS. Desnutrin, an adipocyte gene encoding a novel patatin domain-containing protein, is induced by fasting and glucocorticoids: ectopic expression of desnutrin increases triglyceride hydrolysis. J Biol Chem. 2004;279:47066–75. doi: 10.1074/jbc.M403855200. [DOI] [PubMed] [Google Scholar]
  • 4.Jenkins CM, Mancuso DJ, Yan W, Sims HF, Gibson B, Gross RW. Identification, cloning, expression, and purification of three novel human calcium-independent phospholipase A2 family members possessing triacylglycerol lipase and acylglycerol transacylase activities. J Biol Chem. 2004;279:48968–75. doi: 10.1074/jbc.M407841200. [DOI] [PubMed] [Google Scholar]
  • 5.Kienesberger PC, Oberer M, Lass A, Zechner R. Mammalian patatin domain containing proteins: a family with diverse lipolytic activities involved in multiple biological functions. J Lipid Res. 2009;50(Suppl):S63–8. doi: 10.1194/jlr.R800082-JLR200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Simon GM, Cravatt BF. Endocannabinoid biosynthesis proceeding through glycerophospho-N-acyl ethanolamine and a role for alpha/beta-hydrolase 4 in this pathway. J Biol Chem. 2006;281:26465–72. doi: 10.1074/jbc.M604660200. [DOI] [PubMed] [Google Scholar]
  • 7.Lass A, Zimmermann R, Haemmerle G, Riederer M, Schoiswohl G, Schweiger M, et al. Adipose triglyceride lipase-mediated lipolysis of cellular fat stores is activated by CGI-58 and defective in Chanarin-Dorfman Syndrome. Cell Metab. 2006;3:309–19. doi: 10.1016/j.cmet.2006.03.005. [DOI] [PubMed] [Google Scholar]
  • 8.Schweiger M, Schreiber R, Haemmerle G, Lass A, Fledelius C, Jacobsen P, et al. Adipose triglyceride lipase and hormone-sensitive lipase are the major enzymes in adipose tissue triacylglycerol catabolism. J Biol Chem. 2006;281:40236–41. doi: 10.1074/jbc.M608048200. [DOI] [PubMed] [Google Scholar]
  • 9.Radner FP, Streith IE, Schoiswohl G, Schweiger M, Kumari M, Eichmann TO, et al. Growth retardation, impaired triacylglycerol catabolism, hepatic steatosis, and lethal skin barrier defect in mice lacking comparative gene identification-58 (CGI-58) J Biol Chem. 2010;285:7300–11. doi: 10.1074/jbc.M109.081877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Ghosh AK, Ramakrishnan G, Chandramohan C, Rajasekharan R. CGI-58, the causative gene for Chanarin-Dorfman syndrome, mediates acylation of lysophosphatidic acid. J Biol Chem. 2008;283:24525–33. doi: 10.1074/jbc.M801783200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Montero-Moran G, Caviglia JM, McMahon D, Rothenberg A, Subramanian V, Xu Z, et al. CGI-58/ABHD5 is a coenzyme A-dependent lysophosphatidic acid acyltransferase. J Lipid Res. 2010;51:709–19. doi: 10.1194/jlr.M001917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Igal RA, Rhoads JM, Coleman RA. Neutral lipid storage disease with fatty liver and cholestasis. J Pediatr Gastroenterol Nutr. 1997;25:541–7. doi: 10.1097/00005176-199711000-00011. [DOI] [PubMed] [Google Scholar]
  • 13.Schweiger M, Lass A, Zimmermann R, Eichmann TO, Zechner R. Neutral lipid storage disease: genetic disorders caused by mutations in adipose triglyceride lipase/PNPLA2 or CGI-58/ABHD5. Am J Physiol Endocrinol Metab. 2009;297:E289–96. doi: 10.1152/ajpendo.00099.2009. [DOI] [PubMed] [Google Scholar]
  • 14.Lefèvre C, Jobard F, Caux F, Bouadjar B, Karaduman A, Heilig R, et al. 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. doi: 10.1086/324121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Fischer J, Lefèvre C, Morava E, Mussini JM, Laforêt P, Negre-Salvayre A, et al. The gene encoding adipose triglyceride lipase (PNPLA2) is mutated in neutral lipid storage disease with myopathy. Nat Genet. 2007;39:28–30. doi: 10.1038/ng1951. [DOI] [PubMed] [Google Scholar]
  • 16.Igal RA, Coleman RA. Acylglycerol recycling from triacylglycerol to phospholipid, not lipase activity, is defective in neutral lipid storage disease fibroblasts. J Biol Chem. 1996;271:16644–51. doi: 10.1074/jbc.271.28.16644. [DOI] [PubMed] [Google Scholar]
  • 17.Igal RA, Coleman RA. Neutral lipid storage disease: a genetic disorder with abnormalities in the regulation of phospholipid metabolism. J Lipid Res. 1998;39:31–43. [PubMed] [Google Scholar]
  • 18.Haemmerle G, Lass A, Zimmermann R, Gorkiewicz G, Meyer C, Rozman J, et al. Defective lipolysis and altered energy metabolism in mice lacking adipose triglyceride lipase. Science. 2006;312:734–7. doi: 10.1126/science.1123965. [DOI] [PubMed] [Google Scholar]
  • 19.Grall A, Guaguère E, Planchais S, Grond S, Bourrat E, Hausser I, et al. PNPLA1 mutations cause autosomal recessive congenital ichthyosis in golden retriever dogs and humans. Nat Genet. 2012;44:140–7. doi: 10.1038/ng.1056. [DOI] [PubMed] [Google Scholar]
  • 20.Brown JM, Chung S, Das A, Shelness GS, Rudel LL, Yu L. CGI-58 facilitates the mobilization of cytoplasmic triglyceride for lipoprotein secretion in hepatoma cells. J Lipid Res. 2007;48:2295–305. doi: 10.1194/jlr.M700279-JLR200. [DOI] [PubMed] [Google Scholar]
  • 21.Reid BN, Ables GP, Otlivanchik OA, Schoiswohl G, Zechner R, Blaner WS, et al. Hepatic overexpression of hormone-sensitive lipase and adipose triglyceride lipase promotes fatty acid oxidation, stimulates direct release of free fatty acids, and ameliorates steatosis. J Biol Chem. 2008;283:13087–99. doi: 10.1074/jbc.M800533200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Haemmerle G, Zimmermann R, Hayn M, Theussl C, Waeg G, Wagner E, et al. Hormone-sensitive lipase deficiency in mice causes diglyceride accumulation in adipose tissue, muscle, and testis. J Biol Chem. 2002;277:4806–15. doi: 10.1074/jbc.M110355200. [DOI] [PubMed] [Google Scholar]
  • 23.Subramanian V, Rothenberg A, Gomez C, Cohen AW, Garcia A, Bhattacharyya S, et al. Perilipin A mediates the reversible binding of CGI-58 to lipid droplets in 3T3-L1 adipocytes. J Biol Chem. 2004;279:42062–71. doi: 10.1074/jbc.M407462200. [DOI] [PubMed] [Google Scholar]
  • 24.Yamaguchi T, Omatsu N, Matsushita S, Osumi T. CGI-58 interacts with perilipin and is localized to lipid droplets. Possible involvement of CGI-58 mislocalization in Chanarin-Dorfman syndrome. J Biol Chem. 2004;279:30490–7. doi: 10.1074/jbc.M403920200. [DOI] [PubMed] [Google Scholar]
  • 25.Yamaguchi T, Omatsu N, Morimoto E, Nakashima H, Ueno K, Tanaka T, et al. CGI-58 facilitates lipolysis on lipid droplets but is not involved in the vesiculation of lipid droplets caused by hormonal stimulation. J Lipid Res. 2007;48:1078–89. doi: 10.1194/jlr.M600493-JLR200. [DOI] [PubMed] [Google Scholar]
  • 26.Zimmermann R, Strauss JG, Haemmerle G, Schoiswohl G, Birner-Gruenberger R, Riederer M, et al. Fat mobilization in adipose tissue is promoted by adipose triglyceride lipase. Science. 2004;306:1383–6. doi: 10.1126/science.1100747. [DOI] [PubMed] [Google Scholar]
  • 27.Langin D. Control of fatty acid and glycerol release in adipose tissue lipolysis. C R Biol. 2006;329:598–607, discussion 653-5. doi: 10.1016/j.crvi.2005.10.008. [DOI] [PubMed] [Google Scholar]
  • 28.Brasaemle DL. Thematic review series: adipocyte biology. The perilipin family of structural lipid droplet proteins: stabilization of lipid droplets and control of lipolysis. J Lipid Res. 2007;48:2547–59. doi: 10.1194/jlr.R700014-JLR200. [DOI] [PubMed] [Google Scholar]
  • 29.Jaworski K, Sarkadi-Nagy E, Duncan RE, Ahmadian M, Sul HS. Regulation of triglyceride metabolism. IV. Hormonal regulation of lipolysis in adipose tissue. Am J Physiol Gastrointest Liver Physiol. 2007;293:G1–4. doi: 10.1152/ajpgi.00554.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.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. doi: 10.1074/jbc.M610580200. [DOI] [PubMed] [Google Scholar]
  • 31.Holm C. Molecular mechanisms regulating hormone-sensitive lipase and lipolysis. Biochem Soc Trans. 2003;31:1120–4. doi: 10.1042/BST0311120. [DOI] [PubMed] [Google Scholar]
  • 32.Ahmadian M, Abbott MJ, Tang T, Hudak CS, Kim Y, Bruss M, et al. Desnutrin/ATGL is regulated by AMPK and is required for a brown adipose phenotype. Cell Metab. 2011;13:739–48. doi: 10.1016/j.cmet.2011.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Ahmadian M, Duncan RE, Varady KA, Frasson D, Hellerstein MK, Birkenfeld AL, et al. Adipose overexpression of desnutrin promotes fatty acid use and attenuates diet-induced obesity. Diabetes. 2009;58:855–66. doi: 10.2337/db08-1644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Yang X, Lu X, Liu J. Identification of a novel splicing isoform of murine CGI-58. FEBS Lett. 2010;584:903–10. doi: 10.1016/j.febslet.2009.12.058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Miyoshi H, Perfield JW, 2nd, Souza SC, Shen WJ, Zhang HH, Stancheva ZS, et al. Control of adipose triglyceride lipase action by serine 517 of perilipin A globally regulates protein kinase A-stimulated lipolysis in adipocytes. J Biol Chem. 2007;282:996–1002. doi: 10.1074/jbc.M605770200. [DOI] [PubMed] [Google Scholar]
  • 36.Caviglia JM, Betters JL, Dapito DH, Lord CC, Sullivan S, Chua S, et al. Adipose-selective overexpression of ABHD5/CGI-58 does not increase lipolysis or protect against diet-induced obesity. J Lipid Res. 2011;52:2032–42. doi: 10.1194/jlr.M019117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Yamaguchi T, Omatsu N, Omukae A, Osumi T. Analysis of interaction partners for perilipin and ADRP on lipid droplets. Mol Cell Biochem. 2006;284:167–73. doi: 10.1007/s11010-005-9045-y. [DOI] [PubMed] [Google Scholar]
  • 38.Granneman JG, Moore HP, Mottillo EP, Zhu Z. Functional interactions between Mldp (LSDP5) and Abhd5 in the control of intracellular lipid accumulation. J Biol Chem. 2009;284:3049–57. doi: 10.1074/jbc.M808251200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Granneman JG, Moore HP, Mottillo EP, Zhu Z, Zhou L. Interactions of perilipin-5 (Plin5) with adipose triglyceride lipase. J Biol Chem. 2011;286:5126–35. doi: 10.1074/jbc.M110.180711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.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. doi: 10.1210/en.2010-0661. [DOI] [PubMed] [Google Scholar]
  • 41.Wu JW, Wang SP, Alvarez F, Casavant S, Gauthier N, Abed L, et al. Deficiency of liver adipose triglyceride lipase in mice causes progressive hepatic steatosis. Hepatology. 2011;54:122–32. doi: 10.1002/hep.24338. [DOI] [PubMed] [Google Scholar]
  • 42.Ong KT, Mashek MT, Bu SY, Greenberg AS, Mashek DG. Adipose triglyceride lipase is a major hepatic lipase that regulates triacylglycerol turnover and fatty acid signaling and partitioning. Hepatology. 2011;53:116–26. doi: 10.1002/hep.24006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Caviglia JM, Sparks JD, Toraskar N, Brinker AM, Yin TC, Dixon JL, et al. ABHD5/CGI-58 facilitates the assembly and secretion of apolipoprotein B lipoproteins by McA RH7777 rat hepatoma cells. Biochim Biophys Acta. 2009;1791:198–205. doi: 10.1016/j.bbalip.2008.12.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Turpin SM, Hoy AJ, Brown RD, Rudaz CG, Honeyman J, Matzaris M, et al. Adipose triacylglycerol lipase is a major regulator of hepatic lipid metabolism but not insulin sensitivity in mice. Diabetologia. 2011;54:146–56. doi: 10.1007/s00125-010-1895-5. [DOI] [PubMed] [Google Scholar]
  • 45.Kienesberger PC, Lee D, Pulinilkunnil T, Brenner DS, Cai L, Magnes C, et al. Adipose triglyceride lipase deficiency causes tissue-specific changes in insulin signaling. J Biol Chem. 2009;284:30218–29. doi: 10.1074/jbc.M109.047787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Peyot ML, Guay C, Latour MG, Lamontagne J, Lussier R, Pineda M, et al. Adipose triglyceride lipase is implicated in fuel- and non-fuel-stimulated insulin secretion. J Biol Chem. 2009;284:16848–59. doi: 10.1074/jbc.M109.006650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Fang Y, Vilella-Bach M, Bachmann R, Flanigan A, Chen J. Phosphatidic acid-mediated mitogenic activation of mTOR signaling. Science. 2001;294:1942–5. doi: 10.1126/science.1066015. [DOI] [PubMed] [Google Scholar]
  • 48.Hornberger TA, Chu WK, Mak YW, Hsiung JW, Huang SA, Chien S. The role of phospholipase D and phosphatidic acid in the mechanical activation of mTOR signaling in skeletal muscle. Proc Natl Acad Sci U S A. 2006;103:4741–6. doi: 10.1073/pnas.0600678103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Sun Y, Chen J. mTOR signaling: PLD takes center stage. Cell Cycle. 2008;7:3118–23. doi: 10.4161/cc.7.20.6881. [DOI] [PubMed] [Google Scholar]
  • 50.Sun Y, Fang Y, Yoon MS, Zhang C, Roccio M, Zwartkruis FJ, et al. Phospholipase D1 is an effector of Rheb in the mTOR pathway. Proc Natl Acad Sci U S A. 2008;105:8286–91. doi: 10.1073/pnas.0712268105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Tremblay F, Brûlé S, Hee Um S, Li Y, Masuda K, Roden M, et al. Identification of IRS-1 Ser-1101 as a target of S6K1 in nutrient- and obesity-induced insulin resistance. Proc Natl Acad Sci U S A. 2007;104:14056–61. doi: 10.1073/pnas.0706517104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Zhang C, Wendel AA, Keogh MR, Harris TE, Chen J, Coleman RA. Glycerolipid signals alter mTOR complex 2 (mTORC2) to diminish insulin signaling. Proc Natl Acad Sci U S A. 2012;109:1667–72. doi: 10.1073/pnas.1110730109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Grefhorst A, Hoekstra J, Derks TG, Ouwens DM, Baller JF, Havinga R, et al. Acute hepatic steatosis in mice by blocking beta-oxidation does not reduce insulin sensitivity of very-low-density lipoprotein production. Am J Physiol Gastrointest Liver Physiol. 2005;289:G592–8. doi: 10.1152/ajpgi.00063.2005. [DOI] [PubMed] [Google Scholar]
  • 54.Fisher EA, Pan M, Chen X, Wu X, Wang H, Jamil H, et al. The triple threat to nascent apolipoprotein B. Evidence for multiple, distinct degradative pathways. J Biol Chem. 2001;276:27855–63. doi: 10.1074/jbc.M008885200. [DOI] [PubMed] [Google Scholar]
  • 55.Hagan DL, Kienzle B, Jamil H, Hariharan N. Transcriptional regulation of human and hamster microsomal triglyceride transfer protein genes. Cell type-specific expression and response to metabolic regulators. J Biol Chem. 1994;269:28737–44. [PubMed] [Google Scholar]
  • 56.Wetterau JR, Lin MC, Jamil H. Microsomal triglyceride transfer protein. Biochim Biophys Acta. 1997;1345:136–50. doi: 10.1016/s0005-2760(96)00168-3. [DOI] [PubMed] [Google Scholar]
  • 57.Kamagate A, Qu S, Perdomo G, Su D, Kim DH, Slusher S, et al. FoxO1 mediates insulin-dependent regulation of hepatic VLDL production in mice. J Clin Invest. 2008;118:2347–64. doi: 10.1172/JCI32914. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Fuchs C, Claudel T, Kumari P, Haemmerle G, Pollheimer M, Stojakovic T, et al. Absence of adipose triglyceride lipase protects from hepatic ER stress. Hepatology. 2012;56:270–80. doi: 10.1002/hep.25601. [DOI] [PubMed] [Google Scholar]
  • 59.Lammers B, Chandak PG, Aflaki E, Van Puijvelde GH, Radovic B, Hildebrand RB, et al. Macrophage adipose triglyceride lipase deficiency attenuates atherosclerotic lesion development in low-density lipoprotein receptor knockout mice. Arterioscler Thromb Vasc Biol. 2011;31:67–73. doi: 10.1161/ATVBAHA.110.215814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Haemmerle G, Moustafa T, Woelkart G, Büttner S, Schmidt A, van de Weijer T, et al. ATGL-mediated fat catabolism regulates cardiac mitochondrial function via PPAR-α and PGC-1. Nat Med. 2011;17:1076–85. doi: 10.1038/nm.2439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Chakravarthy MV, Lodhi IJ, Yin L, Malapaka RR, Xu HE, Turk J, et al. Identification of a physiologically relevant endogenous ligand for PPARalpha in liver. Cell. 2009;138:476–88. doi: 10.1016/j.cell.2009.05.036. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Adipocyte are provided here courtesy of Taylor & Francis

RESOURCES