312

The Lipid Droplet as a Potential Therapeutic Target in NAFLD David L. Silver, PhD1

1 Signature Research Program in Cardiovascular & Metabolic Disorders,

Duke-NUS Graduate Medical School, Singapore Semin Liver Dis 2013;33:312–320.

Abstract

Keywords

► nonalcoholic fatty liver disease ► lipid droplets ► triglyceride

Address for correspondence David L. Silver, PhD, Signature Research Program in Cardiovascular & Metabolic Disorders, Duke-NUS Graduate Medical School, 8 College Road, Singapore 169857 (e-mail: [email protected]).

Nonalcoholic fatty liver disease (NAFLD) is a growing problem worldwide. Nonalcoholic fatty liver disease is characterized by an abnormal accumulation of triglyceride-rich lipid droplets (LDs) in the liver, which can lead to liver inflammation and metabolic disturbances. Lipid droplets are dynamic organelles that have recently gained considerable scientific interest. Their formation and growth are regulated processes requiring the participation of many endoplasmic reticulum- (ER-) and LD-associated proteins, which may serve as potential therapeutic targets for NAFLD. Protein families such as fatinducing transmembrane proteins 1 and 2 (FITM1/FIT1 and FITM2/FIT2), the CIDE family of proteins, and the perilipin family, play important roles in LD biology. In this review, the authors discuss current views on LD formation and growth, and how various proteins may affect LD metabolism and lipoprotein assembly in the pathogenesis of NAFLD.

NAFLD and Triglyceride Lipid Droplets Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease in the Western world.1 Nonalcoholic fatty liver disease is characterized by an excess accumulation of triglyceride (TG) within the hepatocyte, in organelles called lipid droplets (LDs). Nonalcoholic fatty liver disease is not a singly defined disease, but can manifest as a spectrum of disorders ranging from hepatic steatosis to nonalcoholic steatohepatitis (NASH). Accordingly, the severity of NAFLD varies widely from patient to patient, often incidentally detected in some asymptomatic individuals, while leading to liver inflammation, cirrhosis, and even hepatocellular carcinoma in others.2 A recent data analysis of the literature revealed the strongest risk factors for NAFLD to be central obesity, type 2 diabetes, dyslipidemia, and hypertension.3 Despite its widespread occurrence, current treatments for NAFLD remain limited. A recently published meta-analysis of randomized trials found that weight loss and thiazolidinediones improved liver histology and cardiometabolic profiles, although weight gain for the latter intervention was common.4 Other current and novel therapeutics for NAFLD have been previously discussed4–9 and are beyond the scope of this review. With the

Issue Theme Lipids and the Liver; Guest Editor, David E. Cohen, MD, PhD

prevalence of NAFLD on the rise,10 it is no surprise that the disease has garnered immense interest in the scientific community. Several comprehensive reviews have been published on the topic in recent years.2,3,11–17 In this review, we will present a LD-centric perspective on the pathogenesis of NAFLD. Because excessive deposition of lipid within hepatocyte cytosolic LDs characterizes NAFLD, we will take an indepth look at the mechanisms of LD biogenesis and growth.

Lipid Droplet Composition: More than a Droplet of Fat Once perceived as relatively simple storage particles for TG, LDs are now widely recognized as dynamic organelles. Lipid droplets are found in the cytoplasm, and are made up of a hydrophobic core of neutral and minimally charged lipids surrounded by a phospholipid monolayer. The mammalian LD phospholipid monolayer consists mainly of phosphatidylcholine (PC), followed in descending order of abundance by phosphatidylethanolamine (PE), phosphatidylinositol (PI), lyso-PC, and lyso-PE.18 Peripherally associated with or embedded within the phospholipid monolayer is an ecosystem of proteins that play important roles in LD biology. Lipid

Copyright © 2013 by Thieme Medical Publishers, Inc., 333 Seventh Avenue, New York, NY 10001, USA. Tel: +1(212) 584-4662.

DOI http://dx.doi.org/ 10.1055/s-0033-1358521. ISSN 0272-8087.

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

Vera J. Goh, BSc1

droplets are phylogenetically conserved organelles found in all eukaryotes. Lipid droplet composition has been examined using lipidomic and proteomic approaches in diverse eukaryotes including yeast, worm, fly, and mammals.18–28 Among the LD-associated proteins in mammals, the perilipin (Plin) family stands out as the most well characterized to date. There are five members of the Plin family, namely Plin1/ perilipin, Plin2/adipophilin, Plin3/tail-interacting protein of 47 kDa (TIP47), Plin4/S3–12, and Plin5/OXPAT. Plin proteins differ in terms of tissue expression: Plin2 and Plin3 are expressed ubiquitously; Plin1, Plin4, and Plin5 are expressed in a tissuerestricted manner. Plin1 and Plin2 are primarily found on the LD surface, whereas Plin3, Plin4, and Plin5 are stable in the cytoplasm and target to the LD surface upon fatty acid loading of cells, a stimulus that induces rapid LD accumulation.29–31 Plin proteins have been shown to distribute preferentially to LDs of different sizes in adipocytes,32 although it is unclear what mechanisms govern Plin targeting to LDs, or whether Plins have distinct functional roles on specific populations of LDs. For example, Plin3 coats very small-sized LDs in steatotic hepatocytes, whereas Plin1 and Plin2 associate with larger LDs.33 In general, Plin proteins are known to play important roles in stabilizing LDs and regulating lipolysis in the cell. The few and small LDs found in normal human and mouse livers contain Plin2 and Plin3, whereas Plin1 is additionally found only in steatotic livers.33 The expression of Plin1 in steatotic livers may be due to transcriptional transactivation by PPARgamma,34,35 a master regulator of adipogenesis that directly regulates the expression of Plin1 in adipose tissue. Indeed, induction of other PPARgamma targets has been seen in steatotic livers of rodents and humans.36–44 Plin1 and Plin2, but not Plin3, correlate positively with the amount of hepatocytic LDs in the steatotic liver.33 In a hepatoma cell model fed with oleic acid, de novo synthesized LDs stained for Plin2 and Plin3, but not Plin1, indicating that expression of these proteins preceded that of Plin1 during steatogenesis.33 Interestingly, like normal hepatocytes, enterocytes only express Plin2 and Plin3,45 suggesting a role for these proteins in lipoprotein assembly. Further differentiation between Plin functions was discovered by coherent anti-Stokes Raman scattering (CARS) microscopy, when Plin3 was found to coat cytosolic LDs in enterocytes after an acute fat challenge, whereas Plin2 was found to coat cytosolic LDs only after chronic fat challenge. The functional significance that Plin2 and Plin3 impart to hepatocytic LDs under acute or chronic fat challenge is unknown, but it is likely that each has multiple functions in regulating LD metabolism. Similar to its postulated function in enterocytes, Plin3 also seems to play a role in LD accumulation in the hepatocyte. Plin3 antisense oligonucleotide treatment in mice reduced histological features of hepatic steatosis. At the same time, reduction in Plin3 levels significantly blunted hepatic TG secretion, suggesting a role in hepatic lipoprotein metabolism.46

Lipid Droplets in Lipoprotein Assembly Lipoprotein assembly begins with the cotranslational lipidation of apoB during its translocation into the ER lumen. Very

Goh, Silver

low-density lipoprotein (VLDL) is the primary lipoprotein synthesized and secreted by the liver. Its assembly is believed to occur via a two-step lipidation pathway whereby apoB-100 is first cotranslationally lipidated to form a dense pre-VLDL particle, then secondarily lipidated when the VLDL precursors combine with ER intraluminal TG-rich LDs to form maturing VLDL particles.47,48 Microsomal triglyceride transfer protein (MTTP), a heterodimer predominantly expressed in hepatocytes and enterocytes, lipidates the elongating apoB-100 polypeptide chain.49 In addition to apoB-100 lipidation in the liver, the 97kDa lipid-binding and transfer subunit of MTTP is also essential for the lipidation of the intestinespecific truncated form of apoB called apoB-48 during chylomicron synthesis.50 The lipidation of apoB stabilizes the polypeptide chain and prevents it from being degraded via the ER-associated degradation (ERAD) pathway. Apo B can also be degraded even when apoB lipidation is normal by a process called post-ER presecretory proteolysis.51 Humans with inactivating mutations in the MTTP gene suffer from a condition called familial abetalipoproteinemia. Patients with this disease have almost undetectable plasma apoB levels and develop hepatic steatosis. Human apoB mRNA can be translated into the full-length apoB-100 polypeptide chain in hepatocytes, or edited to translate apoB-48 containing only the N-terminal 48% of the polypeptide. Both truncating and nontruncating mutations in the human APOB gene cause familial hypobetalipoproteinemia (FHBL). Familial hypobetalipoproteinemia patients have defective apoB secretion and hepatic steatosis,52,53 and in some cases, intestinal fat malabsorption.53,54 Because MTTP and apoB represent two protein factors essential for the assembly and secretion of neutral lipid-containing VLDL, disrupting the function of either protein leads to hepatocyte TG retention and fatty liver. An important LD-associated protein affecting VLDL assembly and secretion is CIDEB. CIDEB is part of the cell deathinducing DNA fragmentation factor 45-like effector (CIDE) family, which also includes CIDEA and CIDEC/Fsp27. CIDEB is detected in many tissues, but is most highly expressed in the liver where it exists as both an ER- and LD-associated protein. CIDEB interacts with apoB and is involved in VLDL lipidation and maturation in mice.55 Peng Li and colleagues found that CIDEB null mice displayed reduced VLDL secretion with a concomitant increase in hepatic TG content as compared with control littermates. They proposed a model in which CIDEB is localized to the cytosolic side of the ER and LDs, and upon binding to apoB in VLDL precursors, facilitates the transfer of TG from cytosolic LDs or de novo synthesized TG within the ER bilayer onto the pre-VLDL. It remains to be tested if nascent LDs that remain attached to the ER are the source of apoB lipidation and sites of apoB-CIDEB interactions. In the subsequent steps of VLDL assembly, maturing VLDL particles travel from the ER to the Golgi via coat complex II(COPII-) coated vesicles.56 There, they acquire more phospholipid and neutral lipids to form mature VLDL particles.56,57 Formation of the COPII-coated vesicles, termed VLDL transport vesicles (VTVs), has been suggested to occur via CIDEB interaction with COPII components, Sar1 and Sec24.58 In a recent publication by Siddiqi et al, proteomic and Western Seminars in Liver Disease

Vol. 33

No. 4/2013

313

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

Lipid Droplet as a Potential Therapeutic Target in NAFLD

Lipid Droplet as a Potential Therapeutic Target in NAFLD

Goh, Silver

blotting data show the presence of CIDEB in VTVs, but no other ER-derived vesicles. Furthermore, blocking CIDEB in an in vitro ER budding assay inhibited VTV budding.58 These combined studies indicate that CIDEB is important in both assembly of nascent VLDL and export of nascent VLDL to the Golgi. It is still unclear precisely how cytosolic LD metabolism is coordinated with lipoprotein synthesis and secretion, but some evidence exists that suggests a tug of war between the distribution of TG to cytosolic LDs versus distribution to the ER lumen. In support of this idea, it has recently been shown that CIDEB and Plin2 were found to play opposing roles in VLDL lipidation.59 CIDEB null mice were observed to have increased Plin2 levels, suggesting that ablation of CIDEB decreased VLDL lipidation, shunting neutral lipids synthesized in the ER toward the formation of cytosolic LDs. In further evidence for the opposing effects of Plin2 and CIDEB on VLDL synthesis, liver-specific knockdown of Plin2 in CIDEB null mice reduced the hepatic TG content and increased VLDL-TG secretion to wild-type levels.59 Accordingly, Plin2 knockout mice displayed reduced hepatic TG content and resistance to HFD-induced hepatic steatosis, possibly due to increased TG secretion from the liver.60 Chang and colleagues postulated that Plin2 could play a role in regulating LD biogenesis, whereby ablation of the protein would lead to accumulation of TG within its site of synthesis in the ER, inducing TG to be preferentially transferred toward ER intraluminal VLDL assembly. Although this idea has not been directly shown, it is supported by the increase in MTTP levels and hepatocyte microsomal TG content in Plin2 knockout mice.60 However, reduction of hepatic Plin2 levels using antisense oligonucleotide injections in two other mouse models of hepatic steatosis61 did not corroborate the previous observation in CIDEB / Plin2 knockdown mice of increased TG export as VLDL. Instead, expression of lipogenic genes was decreased along with VLDL secretion, suggesting that the protective effects of Plin2 deficiency against liver steatosis in these models were due to reduced lipogenesis rather than increased VLDL secretion. In a hepatoma cell line, knocking down Plin2 reduced the formation of LDs while increasing VLDL secretion.62 Increasing Plin2 levels in this cell modelinduced storage of TG in cytosolic LDs while reducing its secretion as VLDL. In view of contradicting results from different experimental approaches, it is more likely that LDs can both facilitate VLDL production as well as limit it by sequestering fatty substrates away from the ER. This underscores a recent finding that some LDs remain attached to the ER, while others do not, indicating the existence of distinct populations of LDs within a given cell. Such distinct populations could have opposing effects on VLDL assembly. Clearly, more work needs to be done to fully understand the role of the Plin proteins in modulating LD formation and VLDL secretion from the liver.

as Saccharomyces cerevisiae. Their conservation throughout evolution likely points to a conserved function in storing TG. Stored TG is used for phospholipid biosynthesis during logarithmic phase growth in yeast, or for storing excess energy in higher eukaryotes. Indeed, LDs provide the largest storage depot for energy. They store energy in the form of esterified fatty acids that can be mobilized to peripheral tissues for ATP production. Lipid droplets also help to maintain cellular lipid homeostasis in several ways. First, they function as reservoirs for sterols and fatty acids, which are important building blocks for the synthesis of cell membranes. Second, LDs are thought to compartmentalize lipids to prevent cellular damage caused by their excess. In particular, unesterified fatty acids may serve as bioactive signaling molecules, detergents, or may drive ceramide synthesis within cells, leading to effects often referred to as lipotoxicity.63–66 Acyl CoA:diacylglycerol acyltransferase 1 (DGAT1) is an enzyme that catalyzes the final, committed step in the TGsynthesizing pathway preceding LD formation. As Farese and coworkers discovered, human DGAT1 deficiency results in a congenital diarrheal disorder,67 which in turn is postulated to be due to the lipotoxic effects of accumulating DGAT1 substrates, DAG, and fatty acids, on enterocyte function. DGAT1 is one of two unrelated genes in mammals. The other DGAT in mammals is DGAT2 that mediates the same enzymatic reaction of fatty acid esterification with DAG to form TG. Interestingly, only DGAT1 and not DGAT2 is expressed in human intestine. Conversely, murine intestine contains both DGATs, and loss of murine DGAT1 alone does not lead to the diarrheal phenotype seen in humans.68,69 The gastrointestinal phenotype of loss of both DGATs in mice has yet to be reported. Another function of LDs is their purported role in regulating protein metabolism. In leukocytes, LDs are formed in response to infectious and inflammatory stimuli, and thought to serve as niduses for the biosynthesis of eicosanoids such as prostaglandins and leukotrienes.70–73 Lipid droplets may also serve as parking spots for hydrophobic proteins, as many proteins have previously been observed to localize to LDs.20 Another function of LDs is their possible role in the ERAD of misfolded ER membrane proteins via the ubiquitin-proteasome pathway. Cytosolic LDs harbor some proteins involved in ERAD, such as the ubiquitin-like (UBX) domain-containing protein Ubx2/Ubxd8 and the ancient ubiquitous protein 1 (AUP1),74–76 and may play a role in ER quality control. Furthermore, cytosolic LD-associated apoB has been shown to represent a lipidated apoB pool targeted for ERAD,77,78 possibly via an ER-to-cytoplasm escape hatch mechanism,79 which is separate from the poorly lipidated apoB degradation at the translocon.80 This suggests that LDs may help to regulate the amount and quality of secreted apoB by participating in protein degradation. However, there is also evidence suggesting that LDs are dispensable for the ERAD of proteins in yeast.81 Finally, LDs participate in some pathogen-host interactions, such as when they serve as assembly platforms for virion synthesis during hepatitis C virus (HCV) replication.82 In HCV-infected hepatocytes, the HCV core is thought to promote virus-induced steatosis by decreasing LD turnover.83

Lipid Droplet Function Lipid droplets are found ubiquitously in almost all eukaryotic cells including the most ancient extant species of yeast such Seminars in Liver Disease

Vol. 33

No. 4/2013

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

314

Lipid Droplet Formation Lipid droplets are formed at the ER where neutral lipid synthesis enzymes like acyl-CoA:cholesterol acyltransferases (ACATs) and DGATs reside, catalyzing the formation of sterol esters and TG, respectively. There have been several proposed models of LD formation, although one particular model has gained considerable popularity. This model posits that LDs form from a “lens” in the cytoplasmic leaflet of the ER membrane that pinches out from the ER, taking with it the newly synthesized TG that accumulates within the ER bilayer. In a recent publication, such nascent LDs may have been visualized during fatty acid loading of drosophila S2 cells,84 but are not commonly reported. This paucity in evidence for visualizing nascent LD structures may be due to technical challenges or the short-lived nature of these structures. It would likely be energetically unfavorable for TG to cluster at one location between the ER membrane leaflets, and TG should instead travel freely within the bilayer to reduce overall stress on the membrane.85 Based on the “lens” hypothesis of LD formation, it is therefore logical to hypothesize the role of a TG-binding protein localized to the ER membrane that could sequester TG at one point and physically facilitate the partitioning of TG into a LD. Fatty acids composing PC and lyso-PC in LDs are uniquely more unsaturated than those found in the ER,19 suggesting that these rare species of phospholipids either partition onto LDs after being synthesized in the ER, or are synthesized through desaturation and phospholipase action on the LDs themselves. It is still unknown what factors participate in physically partitioning TG into a forming LD, and how phospholipids move from the ER membrane to form a monolayer delineating the budding particle. A promising candidate protein family postulated to play a role in de novo LD biogenesis is the fat-inducing transmembrane (FIT) protein family, comprised of FITM1/FIT1 and FITM2/FIT2. Fat-inducing transmembrane proteins were first identified from a genetic screen using PPARalpha-agonist treated mouse livers. PPARalpha activation induced FIT1 and FIT2 gene expression in murine liver but not heart. Fatinducing transmembrane proteins are localized to the ER and have six transmembrane domains with both N- and C-termini facing the cytosol.86 Fat-inducing transmembrane proteins were found to be important in the partitioning of neutral lipid into cytosolic LDs, but do not participate in neutral lipid synthesis.87 Furthermore, overexpression of FIT2 protein in human embryonic kidney cells, murine liver,87 and muscle88 induced the formation of LDs, while shRNA-mediated FIT2 knockdown in cultured adipocytes drastically reduced the number of LDs.87 Using purified FIT proteins in detergent micelles, it was shown that both proteins bind directly to TG and DAG, with FIT2 having a higher affinity and capacity to bind these neutral lipids than FIT1. It is perhaps not coincidental that overexpression of FIT2 in cells produces larger LDs than overexpression of FIT1, as size of the LDs directly correlates with TG binding affinity to FIT. Binding preference for both FIT proteins was higher for TG and lower for DAG. Mutagenesis studies identified a gain-of-function mutant of FIT2 that generates larger LDs than wild-type FIT2, and a

Goh, Silver

partial loss-of-function mutant that produces smaller LDs than wild-type FIT2. Importantly, the gain-of-function mutant exhibited a significant increase in TG and DAG binding, while the partial loss-of-function mutant showed reduced binding.89 These findings are consistent with the conclusion that TG binding by FIT proteins is important in FIT-mediated LD formation. One possible model of FIT function is that FIT proteins play a role in the initial local accumulation of TG within the leaflets of the ER and thus act as regulatory proteins for determining where and when a nascent LD will form. Although the ability to bind TG is essential to FIT protein function in generating LDs, it is unknown whether this TG-binding property alone is sufficient for FIT to induce LD formation. It is also possible that either FIT proteins bind to other LD-stabilizing proteins that promote LD formation or they harbor more complex activity in partitioning TG or phospholipids onto a growing LD. That said, evidence for direct binding of phospholipids to FIT is lacking.89 Lipoprotein-secreting liver and intestine can form lipid-containing particles that bud into the cytosol or the ER lumen.49 That FIT proteins bind directly to TG and are ER transmembrane proteins raises the possibility that they could mediate both types of budding (►Fig. 1). Fat-inducing transmembrane proteins have distinct tissue distributions, with FIT1 being expressed primarily in muscle and to a lesser extent in heart. Although FIT1 mRNA is expressed in mouse liver, the tissue from which it was initially discovered, surprisingly FIT1 protein has not been found in liver. FIT2 is ubiquitously expressed in mouse and human tissues, with the highest levels in adipose tissue.87 The high level of FIT2 in both white and brown adipose tissue of mouse is likely due to its direct regulation by PPARgamma.90,91 Indeed, FIT2 expression is induced during adipogenesis of 3T3-L1 cells.87 The tissue distribution of FIT proteins suggests unique functions in regulating LD formation. The small and large LDs formed by FIT1 and FIT2 overexpression, respectively, are reminiscent of the characteristic LDs found in muscle and adipocytes. These observations suggest that FIT proteins might be in part responsible for the size distribution of LDs in various tissues. Normal skeletal muscle contains small LDs adjacent to mitochondria; adipose tissue contains large or unilocular LDs. The FIT protein family is highly evolutionarily conserved, with FIT orthologs found in mammals, birds, amphibians, worms, insects, and yeast.87 In fact, Saccharomyces cerevisiae has two homologs of FIT2, named SCS3 and YFT2. Expression of yeast homologs of FIT2 in human embryonic kidney cells promotes LD formation, while expression of human FIT2 in yeast rescues the inositol auxotrophic phenotype of SCS3deleted yeast.92 This indicates that despite the passage of greater than 170 million years and crossing the species barrier, yeast orthologs and human FIT retain some common function. It has recently been shown using synthetic genetic interaction arrays that yeast strains deleted for either or both genes display shared and unique phenotypes involving phospholipid biosynthesis and lipid metabolism.92 However, deletion of SCS3 or YFT2 did not abrogate LD formation in yeast, indicating that FIT2 may in fact play a more fundamental role Seminars in Liver Disease

Vol. 33

No. 4/2013

315

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

Lipid Droplet as a Potential Therapeutic Target in NAFLD

Lipid Droplet as a Potential Therapeutic Target in NAFLD

Goh, Silver

Fig. 1 Model of cytosolic lipid droplet (LD) and very low-density lipoprotein (VLDL) assembly in the liver. This model proposes that FIT2 binds triglyceride (TG) at the endoplasmic reticulum (ER) and generates a nascent LD by concentrating TG between the leaflets of the ER. The production of a nascent LD or a mature LD that remains attached to the ER might supply the TG for apoB lipidation. Thus, FIT2 might facilitate both extraluminal and intraluminal LD budding. During this process, other important LD-associated proteins like CIDEB and Plin2 could regulate the directional flux of LD formation either into the ER lumen or into the cytosol. MTTP, microsomal triglyceride transfer protein.

in lipid metabolism at the ER, which precedes the development of its function in LD formation.

Lipid Droplet Growth: Expansion and Fusion Lipid droplets are dynamic organelles that contract in volume through lipolysis of the TG core, and expand in volume when excess fatty acids are present in the cell. Two models for LD growth are strongly supported by experimental evidence—in situ TG synthesis at the LD84 and fusion of two LDs. With the intent of identifying genes involved in LD formation and growth, Guo et al used a genome-wide RNAi screen in drosophila S2 cells. This screen revealed that genes encoding enzymes of the phospholipid biosynthesis pathway determined LD size and number.93 Lipid droplet growth requires the addition of surface phospholipids, of which PC is the most abundant18 and acts like a surfactant to prevent LD coalescence.94 It is thought that when PC is limiting, LDs fuse to form a larger LD, decreasing the surface area to volume ratio of the LD. CTP:phosphocholine cytidylyltransferase (CCT) is the rate-limiting enzyme of PC synthesis, which converts phosphocholine to CDP-choline. CDP-choline then condenses with DAG to form PC via the action of ER-membrane protein CDP-choline:1,2 diacylglycerol cholinephosphotransferase (CPT). Krahmer and colleagues have shown that the source of additional PC required during LD surface expansion is provided by the Kennedy pathway, which in turn is activated by the targeting of CCT to the expanding LD surface.94 Even so, LDs do not contain CPT activity that is Seminars in Liver Disease

Vol. 33

No. 4/2013

confined to the ER, and it is still unknown how synthesized PC in the ER membrane reaches the expanding LD surface. Alternatively, PC can also be synthesized by the PE-methyltransferase (PEMT) pathway or by fatty acid remodelling of existing phospholipids.95 In the PEMT pathway, which is highly expressed in the liver, PEMT on the ER and mitochondria catalyzes the conversion of PE to PC in close proximity to LDs.96 Again, it remains unknown whether the PC formed via this pathway is used for LD expansion, or how it would reach the expanding LD surface. Phosphatidylcholine formation from fatty acid remodeling on existing phospholipids takes place via the sequential actions of phospholipase A2 (PLA2) and lysophospholipid acyltransferases (LPCATs). This is supported by the discovery of robust LPCAT activity on LDs,97 and the observations that LD PC contains more monounsaturated fatty acids93 than the PC derived from the PEMT pathway,98 or the PC found on ER membranes.18,19,99 In a recent publication by Krahmer et al, just over 100 LD proteins were identified with high confidence using a methodology combining proteomics and correlative profiling on LDs from insect cells.21 Enzymes of lipid metabolism such as acyl CoA synthetases, lipases, and CTP:phosphocholine cytidyltransferases (CCTs) were identified on LDs. Importantly, LDs were found to contain all the enzymes required for de novo in situ synthesis of TG,21,84 supporting the hypothesis that LDs can grow while being completely detached from the ER. Wilfling et al reported that membrane bridges form between the ER and LD that help provide a conduit for the relocalization of TG-synthesizing enzymes from the ER to a

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

316

Lipid Droplet as a Potential Therapeutic Target in NAFLD

317

differentiation and lipogenesis in adipocytes, while suppressing lipogenesis in nonadipose tissues.

Future Perspectives The discovery of the FIT proteins, Fsp27, and many other proteins involved in LD biology, as well as the localization of enzymes involved in TG and phospholipid biosynthesis to the LD, are important steps toward a deeper understanding of LD biogenesis and expansion. It will be important to determine a detailed biochemical mechanism for FIT protein function in LD biogenesis, and for the LD fusion process involving Fsp27. Whether FIT plays a role in regulating lipoprotein secretion in the liver and intestine must also be ascertained. Questions regarding the physiological contribution of FIT proteins to the generation of LDs in tissues must similarly be addressed using in vivo knockout models. Additionally, it is important to investigate the prevalence of de novo LD formation versus LD expansion/fusion under lipogenic conditions in the liver. An ever-expanding amount of research is being focused on LD biology. It is inevitable that these and other important questions regarding LD biogenesis, LD fusion and expansion, and LD protein targeting will be answered. It is hoped that multiple therapeutic targets to treat NAFLD and other lipid storage disorders will be identified and characterized from research on LD biology.

Acknowledgments This work was supported in part by grants from the Singapore Ministry of Health’s National Medical Research Council CBRG/0012/2012 (to D.L.S.), and the Agency for Science, Technology and Research (ASTAR) National Science Scholarship award (to V.J.G.).

References 1 Lazo M, Hernaez R, Eberhardt MS, et al. Prevalence of nonalco-

holic fatty liver disease in the United States: the Third National Health and Nutrition Examination Survey, 1988-1994. Am J Epidemiol 2013;178(1):38–45 2 Vernon G, Baranova A, Younossi ZM. Systematic review: the

epidemiology and natural history of non-alcoholic fatty liver disease and non-alcoholic steatohepatitis in adults. Aliment Pharmacol Ther 2011;34(3):274–285 3 Souza MR, Diniz MdeF, Medeiros-Filho JE, Araújo MS. Metabolic

syndrome and risk factors for non-alcoholic fatty liver disease. Arq Gastroenterol 2012;49(1):89–96 4 Musso G, Cassader M, Rosina F, Gambino R. Impact of current

treatments on liver disease, glucose metabolism and cardiovascular risk in non-alcoholic fatty liver disease (NAFLD): a systematic review and meta-analysis of randomised trials. Diabetologia 2012;55(4):885–904 5 Mark N, de Alwis W, Day CP. Current and future therapeutic

strategies in NAFLD. Curr Pharm Des 2010;16(17):1958–1962 6 Nakajima K. Multidisciplinary pharmacotherapeutic options for

nonalcoholic Fatty liver disease. Int J Hepatol 2012;2012:950693 7 Eguchi A, Povero D, Alkhouri N, Feldstein AE. Novel therapeutic

targets for nonalcoholic fatty liver disease. Expert Opin Ther Targets 2013;17(7):773–779 Seminars in Liver Disease

Vol. 33

No. 4/2013

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

subset of forming LDs. This results in two different LD populations: one that contains TG synthesis enzymes and can expand via in situ synthesis of TG, and another that lacks these enzymes and remains small during fatty acid loading.84 CIDEA and CIDEC/Fsp27 of the CIDE family discussed above play essential roles in mediating LD fusion. CIDEA and CIDEC/ Fsp27 are direct targets of PPARgamma37,100,101 and are primarily expressed in adipose tissue where they promote the formation of large unilocular LDs.102,103 This function however is not restricted to adipocytes, and both proteins are also significantly upregulated in steatotic livers where they are thought to induce hepatic TG accumulation.37,104,105 A human mutation in Fsp27 resulting in the expression of a truncated protein has been reported to cause partial lipodystrophy and insulin-resistant diabetes in the afflicted individual.106 It has been shown that Fsp27 enhances LD clustering and then fusion107 by mediating the transfer of TG from smaller to larger LDs at LD contact sites (LDCS) where the proteins are highly enriched.108 The transfer of neutral lipid from the smaller to the larger LD was postulated to occur because of higher internal pressure within the smaller organelle. Live cell imaging of LD fusion events further revealed that after lipid transfer, the smaller LD would be absorbed by the larger one to form an even larger LD.108 It remains to be determined how the phospholipid monolayer of the smaller droplet fuses with the monolayer of the larger droplet. As suggested by Gong et al, enrichment of CIDEA and Fsp27 at LDCS could promote the formation of a passageway between the LDs that allows neutral lipids to be exchanged,108 but the functional significance of CIDEA localization to the LDCS remains to be determined. Indeed, in a recent publication by Sun et al, the CIDE-N domain of Fsp27 was found to form homodimers in a configuration important for Fsp27-mediated lipid transfer.109 In this model, Plin1 is suggested to play a role in Fsp27-mediated lipid transfer by interacting with Fsp27, changing its conformation to enhance its activity.109 Human mutations in the BSCL2 gene encoding the ER resident membrane protein seipin cause a disorder called congenital generalized lipodystrophy type 2 (CGL2).110 CGL2 manifests as a near complete loss of adipose tissue, insulin resistance, and hepatic steatosis. Seipin null mice develop a severe lipodystrophy syndrome with fatty liver resembling the human condition.111,112 It has been proposed that seipin functions in LD biogenesis. However, knockdown of seipin in mammalian fibroblast-like cells induces TG biosynthesis and proliferation of small clusters of LDs, while overexpression resulted in reduction in TG biosynthesis and LD accumulation.113 Other important studies indicate an important role of seipin in the regulation of lipogenesis and adipogenesis.114–117 As suggested by Yang and coworkers, seipin could play a more fundamental role in phospholipid metabolism such that in its absence, phosphatidic acid accumulation leads to the downstream effects of impaired adipogenesis, increased TG synthesis, and LD fusion.113,118,119 Indeed, knockdown of seipin in the fat body of drosophila is associated with increased phosphatidic acid (PA) accumulation rather than LD formation.120 Collectively, these results indicate that seipin’s role in lipid homeostasis is highly tissue-specific, promoting

Goh, Silver

Lipid Droplet as a Potential Therapeutic Target in NAFLD

Goh, Silver

8 Lomonaco R, Sunny NE, Bril F, Cusi K. Nonalcoholic fatty liver

30 Wolins NE, Quaynor BK, Skinner JR, et al. OXPAT/PAT-1 is a PPAR-

disease: current issues and novel treatment approaches. Drugs 2013;73(1):1–14 Xiao J, Guo R, Fung ML, Liong EC, Tipoe GL. Therapeutic approaches to non-alcoholic fatty liver disease: past achievements and future challenges. Hepatobiliary Pancreat Dis Int 2013;12(2): 125–135 Welsh JA, Karpen S, Vos MB. Increasing prevalence of nonalcoholic fatty liver disease among United States adolescents, 19881994 to 2007-2010. J Pediatr 2013;162(3):496–500, e1 Perlemuter G, Bigorgne A, Cassard-Doulcier AM, Naveau S. Nonalcoholic fatty liver disease: from pathogenesis to patient care. Nat Clin Pract Endocrinol Metab 2007;3(6):458–469 Byrne CD, Olufadi R, Bruce KD, Cagampang FR, Ahmed MH. Metabolic disturbances in non-alcoholic fatty liver disease. Clin Sci (Lond) 2009;116(7):539–564 Smith BW, Adams LA. Non-alcoholic fatty liver disease. Crit Rev Clin Lab Sci 2011;48(3):97–113 Karlas T, Wiegand J, Berg T. Gastrointestinal complications of obesity: non-alcoholic fatty liver disease (NAFLD) and its sequelae. Best Pract Res Clin Endocrinol Metab 2013;27(2):195–208 Tuyama AC, Chang CY. Non-alcoholic fatty liver disease. J Diabetes 2012;4(3):266–280 Moore JB. Non-alcoholic fatty liver disease: the hepatic consequence of obesity and the metabolic syndrome. Proc Nutr Soc 2010;69(2):211–220 Musso G, Gambino R, Cassader M. Non-alcoholic fatty liver disease from pathogenesis to management: an update. Obes Rev 2010;11(6):430–445 Bartz R, Li WH, Venables B, et al. Lipidomics reveals that adiposomes store ether lipids and mediate phospholipid traffic. J Lipid Res 2007;48(4):837–847 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(46): 44507–44512 Cermelli S, Guo Y, Gross SP, Welte MA. The lipid-droplet proteome reveals that droplets are a protein-storage depot. Curr Biol 2006; 16(18):1783–1795 Krahmer N, Hilger M, Kory N, et al. Protein correlation profiles identify lipid droplet proteins with high confidence. Mol Cell Proteomics 2013;12(5):1115–1126 Fujimoto T, Parton RG. Not just fat: the structure and function of the lipid droplet. Cold Spring Harb Perspect Biol 2011;3(3): Zhang P, Na H, Liu Z, et al. Proteomic study and marker protein identification of Caenorhabditis elegans lipid droplets. Mol Cell Proteomics 2012;11(8):317–328 Zhang H, Wang Y, Li J, et al. Proteome of skeletal muscle lipid droplet reveals association with mitochondria and apolipoprotein a-I. J Proteome Res 2011;10(10):4757–4768 Larsson S, Resjö S, Gomez MF, James P, Holm C. Characterization of the lipid droplet proteome of a clonal insulin-producing β-cell line (INS-1 832/13). J Proteome Res 2012;11(2):1264–1273 Beller M, Riedel D, Jänsch L, et al. Characterization of the drosophila lipid droplet subproteome. Mol Cell Proteomics 2006;5(6):1082–1094 Grillitsch K, Connerth M, Köfeler H, et al. Lipid particles/droplets of the yeast Saccharomyces cerevisiae revisited: lipidome meets proteome. Biochim Biophys Acta 2011;1811(12):1165–1176 Ivashov VA, Grillitsch K, Koefeler H, et al. Lipidome and proteome of lipid droplets from the methylotrophic yeast Pichia pastoris. Biochim Biophys Acta 2013;1831(2):282–290 Ohsaki Y, Maeda T, Maeda M, Tauchi-Sato K, Fujimoto T. Recruitment of TIP47 to lipid droplets is controlled by the putative hydrophobic cleft. Biochem Biophys Res Commun 2006;347(1): 279–287

induced lipid droplet protein that promotes fatty acid utilization. Diabetes 2006;55(12):3418–3428

9

10

11

12

13 14

15 16

17

18

19

20

21

22 23

24

25

26

27

28

29

Seminars in Liver Disease

Vol. 33

No. 4/2013

31 Bickel PE, Tansey JT, Welte MA. PAT proteins, an ancient family of

lipid droplet proteins that regulate cellular lipid stores. Biochim Biophys Acta 2009;1791(6):419–440 32 Wolins NE, Quaynor BK, Skinner JR, Schoenfish MJ, Tzekov A,

Bickel PE. S3-12, Adipophilin, and TIP47 package lipid in adipocytes. J Biol Chem 2005;280(19):19146–19155 33 Straub BK, Stoeffel P, Heid H, Zimbelmann R, Schirmacher P.

Differential pattern of lipid droplet-associated proteins and de novo perilipin expression in hepatocyte steatogenesis. Hepatology 2008;47(6):1936–1946 34 Okumura T. Role of lipid droplet proteins in liver steatosis. J

Physiol Biochem 2011;67(4):629–636 35 Dalen KT, Schoonjans K, Ulven SM, et al. Adipose tissue expres-

sion of the lipid droplet-associating proteins S3-12 and perilipin is controlled by peroxisome proliferator-activated receptor-gamma. Diabetes 2004;53(5):1243–1252 36 Motomura W, Inoue M, Ohtake T, et al. Up-regulation of ADRP in

fatty liver in human and liver steatosis in mice fed with high fat diet. Biochem Biophys Res Commun 2006;340(4):1111–1118 37 Matsusue K, Kusakabe T, Noguchi T, et al. Hepatic steatosis in

leptin-deficient mice is promoted by the PPARgamma target gene Fsp27. Cell Metab 2008;7(4):302–311 38 Gavrilova O, Haluzik M, Matsusue K, et al. Liver peroxisome

proliferator-activated receptor gamma contributes to hepatic steatosis, triglyceride clearance, and regulation of body fat mass. J Biol Chem 2003;278(36):34268–34276 39 Inoue M, Ohtake T, Motomura W, et al. Increased expression of

PPARgamma in high fat diet-induced liver steatosis in mice. Biochem Biophys Res Commun 2005;336(1):215–222 40 Panasyuk G, Espeillac C, Chauvin C, et al. PPARγ contributes to

PKM2 and HK2 expression in fatty liver. Nat Commun 2012;3:672 41 Yamazaki T, Shiraishi S, Kishimoto K, Miura S, Ezaki O. An increase

in liver PPARγ2 is an initial event to induce fatty liver in response to a diet high in butter: PPARγ2 knockdown improves fatty liver induced by high-saturated fat. J Nutr Biochem 2011;22(6): 543–553 42 Schadinger SE, Bucher NL, Schreiber BM, Farmer SR. PPAR-

gamma2 regulates lipogenesis and lipid accumulation in steatotic hepatocytes. Am J Physiol Endocrinol Metab 2005;288(6): E1195–E1205 43 Zhang YL, Hernandez-Ono A, Siri P, et al. Aberrant hepatic

expression of PPARgamma2 stimulates hepatic lipogenesis in a mouse model of obesity, insulin resistance, dyslipidemia, and hepatic steatosis. J Biol Chem 2006;281(49):37603–37615 44 Greco D, Kotronen A, Westerbacka J, et al. Gene expression in

human NAFLD. Am J Physiol Gastrointest Liver Physiol 2008; 294(5):G1281–G1287 45 Lee B, Zhu J, Wolins NE, Cheng JX, Buhman KK. Differential

association of adipophilin and TIP47 proteins with cytoplasmic lipid droplets in mouse enterocytes during dietary fat absorption. Biochim Biophys Acta 2009;1791(12):1173–1180 46 Carr RM, Patel RT, Rao V, et al. Reduction of TIP47 improves

hepatic steatosis and glucose homeostasis in mice. Am J Physiol Regul Integr Comp Physiol 2012;302(8):R996–R1003 47 Fisher EA, Ginsberg HN. Complexity in the secretory pathway: the

assembly and secretion of apolipoprotein B-containing lipoproteins. J Biol Chem 2002;277(20):17377–17380 48 Olofsson SO, Stillemark-Billton P, Asp L. Intracellular assembly of

VLDL: two major steps in separate cell compartments. Trends Cardiovasc Med 2000;10(8):338–345 49 Raabe M, Véniant MM, Sullivan MA, et al. Analysis of the role of

microsomal triglyceride transfer protein in the liver of tissuespecific knockout mice. J Clin Invest 1999;103(9):1287–1298

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

318

Goh, Silver

50 Jamil H, Dickson JK Jr, Chu CH, et al. Microsomal triglyceride

71 D’Avila H, Melo RC, Parreira GG, Werneck-Barroso E, Castro-Faria-

transfer protein. Specificity of lipid binding and transport. J Biol Chem 1995;270(12):6549–6554 Ginsberg HN, Fisher EA. The ever-expanding role of degradation in the regulation of apolipoprotein B metabolism. J Lipid Res 2009;50(Suppl):S162–S166 Hooper AJ, van Bockxmeer FM, Burnett JR. Monogenic hypocholesterolaemic lipid disorders and apolipoprotein B metabolism. Crit Rev Clin Lab Sci 2005;42(5-6):515–545 Linton MF, Farese RV Jr, Young SG. Familial hypobetalipoproteinemia. J Lipid Res 1993;34(4):521–541 Tarugi P, Averna M, Di Leo E, et al. Molecular diagnosis of hypobetalipoproteinemia: an ENID review. Atherosclerosis 2007;195(2):e19–e27 Ye J, Li JZ, Liu Y, et al. Cideb, an ER- and lipid droplet-associated protein, mediates VLDL lipidation and maturation by interacting with apolipoprotein B. Cell Metab 2009;9(2):177–190 Gusarova V, Seo J, Sullivan ML, Watkins SC, Brodsky JL, Fisher EA. Golgi-associated maturation of very low density lipoproteins involves conformational changes in apolipoprotein B, but is not dependent on apolipoprotein E. J Biol Chem 2007;282(27): 19453–19462 Gusarova V, Brodsky JL, Fisher EA. Apolipoprotein B100 exit from the endoplasmic reticulum (ER) is COPII-dependent, and its lipidation to very low density lipoprotein occurs post-ER. J Biol Chem 2003;278(48):48051–48058 Tiwari S, Siddiqi S, Siddiqi SA. CideB protein is required for the biogenesis of very low density lipoprotein (VLDL) transport vesicle. J Biol Chem 2013;288(7):5157–5165 Li X, Ye J, Zhou L, Gu W, Fisher EA, Li P. Opposing roles of cell death-inducing DFF45-like effector B and perilipin 2 in controlling hepatic VLDL lipidation. J Lipid Res 2012;53(9):1877–1889 Chang BH, Li L, Paul A, et al. Protection against fatty liver but normal adipogenesis in mice lacking adipose differentiationrelated protein. Mol Cell Biol 2006;26(3):1063–1076 Imai Y, Varela GM, Jackson MB, Graham MJ, Crooke RM, Ahima RS. Reduction of hepatosteatosis and lipid levels by an adipose differentiation-related protein antisense oligonucleotide. Gastroenterology 2007;132(5):1947–1954 Magnusson B, Asp L, Boström P, et al. Adipocyte differentiationrelated protein promotes fatty acid storage in cytosolic triglycerides and inhibits secretion of very low-density lipoproteins. Arterioscler Thromb Vasc Biol 2006;26(7):1566–1571 Summers SA. Ceramides in insulin resistance and lipotoxicity. Prog Lipid Res 2006;45(1):42–72 Chavez JA, Summers SA. Lipid oversupply, selective insulin resistance, and lipotoxicity: molecular mechanisms. Biochim Biophys Acta 2010;1801(3):252–265 Cusi K. Role of insulin resistance and lipotoxicity in non-alcoholic steatohepatitis. Clin Liver Dis 2009;13(4):545–563 Cantley JL, Yoshimura T, Camporez JP, et al. CGI-58 knockdown sequesters diacylglycerols in lipid droplets/ER-preventing diacylglycerol-mediated hepatic insulin resistance. Proc Natl Acad Sci U S A 2013;110(5):1869–1874 Haas JT, Winter HS, Lim E, et al. DGAT1 mutation is linked to a congenital diarrheal disorder. J Clin Invest 2012;122(12): 4680–4684 Ables GP, Yang KJ, Vogel S, et al. Intestinal DGAT1 deficiency reduces postprandial triglyceride and retinyl ester excursions by inhibiting chylomicron secretion and delaying gastric emptying. J Lipid Res 2012;53(11):2364–2379 Buhman KK, Smith SJ, Stone SJ, et al. DGAT1 is not essential for intestinal triacylglycerol absorption or chylomicron synthesis. J Biol Chem 2002;277(28):25474–25479 Bozza PT, Yu W, Penrose JF, Morgan ES, Dvorak AM, Weller PF. Eosinophil lipid bodies: specific, inducible intracellular sites for enhanced eicosanoid formation. J Exp Med 1997;186(6):909–920

Neto HC, Bozza PT. Mycobacterium bovis bacillus Calmette-Guérin induces TLR2-mediated formation of lipid bodies: intracellular domains for eicosanoid synthesis in vivo. J Immunol 2006;176(5): 3087–3097 D’Avila H, Freire-de-Lima CG, Roque NR, et al. Host cell lipid bodies triggered by Trypanosoma cruzi infection and enhanced by the uptake of apoptotic cells are associated with prostaglandin E2 generation and increased parasite growth. J Infect Dis 2011; 204(6):951–961 Vieira-de-Abreu A, Assis EF, Gomes GS, et al. Allergic challengeelicited lipid bodies compartmentalize in vivo leukotriene C4 synthesis within eosinophils. Am J Respir Cell Mol Biol 2005; 33(3):254–261 Wang CW, Lee SC. The ubiquitin-like (UBX)-domain-containing protein Ubx2/Ubxd8 regulates lipid droplet homeostasis. J Cell Sci 2012;125(Pt 12):2930–2939 Mantzaris MD, Tsianos EV, Galaris D. Interruption of triacylglycerol synthesis in the endoplasmic reticulum is the initiating event for saturated fatty acid-induced lipotoxicity in liver cells. FEBS J 2011;278(3):519–530 Jo Y, Hartman IZ, DeBose-Boyd RA. Ancient ubiquitous protein-1 mediates sterol-induced ubiquitination of 3-hydroxy-3-methylglutaryl CoA reductase in lipid droplet-associated endoplasmic reticulum membranes. Mol Biol Cell 2013;24(3):169–183 Suzuki M, Otsuka T, Ohsaki Y, et al. Derlin-1 and UBXD8 are engaged in dislocation and degradation of lipidated ApoB-100 at lipid droplets. Mol Biol Cell 2012;23(5):800–810 Forte TM, Shu X, Ryan RO. The ins (cell) and outs (plasma) of apolipoprotein A-V. J Lipid Res 2009;50(Suppl):S150–S155 Ploegh HL. A lipid-based model for the creation of an escape hatch from the endoplasmic reticulum. Nature 2007;448(7152): 435–438 Zhou M, Fisher EA, Ginsberg HN. Regulated co-translational ubiquitination of apolipoprotein B100. A new paradigm for proteasomal degradation of a secretory protein. J Biol Chem 1998;273(38):24649–24653 Olzmann JA, Kopito RR. Lipid droplet formation is dispensable for endoplasmic reticulum-associated degradation. J Biol Chem 2011;286(32):27872–27874 Herker E, Ott M. Emerging role of lipid droplets in host/pathogen interactions. J Biol Chem 2012;287(4):2280–2287 Harris C, Herker E, Farese RV Jr, Ott M. Hepatitis C virus core protein decreases lipid droplet turnover: a mechanism for coreinduced steatosis. J Biol Chem 2011;286(49):42615–42625 Wilfling F, Wang H, Haas JT, et al. Triacylglycerol synthesis enzymes mediate lipid droplet growth by relocalizing from the ER to lipid droplets. Dev Cell 2013;24(4):384–399 Farese RV Jr, Walther TC. Lipid droplets finally get a little R-E-S-PE-C-T. Cell 2009;139(5):855–860 Gross DA, Snapp EL, Silver DL. Structural insights into triglyceride storage mediated by fat storage-inducing transmembrane (FIT) protein 2. PLoS ONE 2010;5(5):e10796 Kadereit B, Kumar P, Wang WJ, et al. Evolutionarily conserved gene family important for fat storage. Proc Natl Acad Sci U S A 2008;105(1):94–99 Miranda DA, Koves TR, Gross DA, et al. Re-patterning of skeletal muscle energy metabolism by fat storage-inducing transmembrane protein 2. J Biol Chem 2011;286(49):42188–42199 Gross DA, Zhan C, Silver DL. Direct binding of triglyceride to fat storage-inducing transmembrane proteins 1 and 2 is important for lipid droplet formation. Proc Natl Acad Sci U S A 2011;108(49): 19581–19586 Villanueva CJ, Vergnes L, Wang J, et al. Adipose subtype-selective recruitment of TLE3 or Prdm16 by PPARγ specifies lipid storage versus thermogenic gene programs. Cell Metab 2013;17(3): 423–435

51

52

53 54

55

56

57

58

59

60

61

62

63 64

65 66

67

68

69

70

72

73

74

75

76

77

78 79

80

81

82 83

84

85 86

87

88

89

90

Seminars in Liver Disease

Vol. 33

No. 4/2013

319

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

Lipid Droplet as a Potential Therapeutic Target in NAFLD

Lipid Droplet as a Potential Therapeutic Target in NAFLD

Goh, Silver

91 Lefterova MI, Zhang Y, Steger DJ, et al. PPARgamma and C/EBP

106 Rubio-Cabezas O, Puri V, Murano I, et al; LD Screening Consor-

factors orchestrate adipocyte biology via adjacent binding on a genome-wide scale. Genes Dev 2008;22(21):2941–2952 Moir RD, Gross DA, Silver DL, Willis IM. SCS3 and YFT2 link transcription of phospholipid biosynthetic genes to ER stress and the UPR. PLoS Genet 2012;8(8):e1002890 Guo Y, Walther TC, Rao M, et al. Functional genomic screen reveals genes involved in lipid-droplet formation and utilization. Nature 2008;453(7195):657–661 Krahmer N, Guo Y, Wilfling F, et al. Phosphatidylcholine synthesis for lipid droplet expansion is mediated by localized activation of CTP:phosphocholine cytidylyltransferase. Cell Metab 2011;14(4): 504–515 Penno A, Hackenbroich G, Thiele C. Phospholipids and lipid droplets. Biochim Biophys Acta 2013;1831(3):589–594 Hörl G, Wagner A, Cole LK, et al. Sequential synthesis and methylation of phosphatidylethanolamine promote lipid droplet biosynthesis and stability in tissue culture and in vivo. J Biol Chem 2011;286(19):17338–17350 Moessinger C, Kuerschner L, Spandl J, Shevchenko A, Thiele C. Human lysophosphatidylcholine acyltransferases 1 and 2 are located in lipid droplets where they catalyze the formation of phosphatidylcholine. J Biol Chem 2011;286(24):21330–21339 DeLong CJ, Shen YJ, Thomas MJ, Cui Z. Molecular distinction of phosphatidylcholine synthesis between the CDP-choline pathway and phosphatidylethanolamine methylation pathway. J Biol Chem 1999;274(42):29683–29688 Shi X, Li J, Zou X, et al. Regulation of lipid droplet size and phospholipid composition by stearoyl-CoA desaturase. J Lipid Res 2013;54(9):2504–2514 Kim YJ, Cho SY, Yun CH, Moon YS, Lee TR, Kim SH. Transcriptional activation of Cidec by PPARgamma2 in adipocyte. Biochem Biophys Res Commun 2008;377(1):297–302 Viswakarma N, Yu S, Naik S, et al. Transcriptional regulation of Cidea, mitochondrial cell death-inducing DNA fragmentation factor alpha-like effector A, in mouse liver by peroxisome proliferator-activated receptor alpha and gamma. J Biol Chem 2007; 282(25):18613–18624 Puri V, Czech MP. Lipid droplets: FSP27 knockout enhances their sizzle. J Clin Invest 2008;118(8):2693–2696 Nishino N, Tamori Y, Tateya S, et al. FSP27 contributes to efficient energy storage in murine white adipocytes by promoting the formation of unilocular lipid droplets. J Clin Invest 2008;118(8): 2808–2821 Jinno Y, Nakakuki M, Sato A, et al. Cide-a and Cide-c are induced in the progression of hepatic steatosis and inhibited by eicosapentaenoic acid. Prostaglandins Leukot Essent Fatty Acids 2010; 83(2):75–81 Zhou L, Xu L, Ye J, et al. Cidea promotes hepatic steatosis by sensing dietary fatty acids. Hepatology 2012;56(1):95–107

tium. Partial lipodystrophy and insulin resistant diabetes in a patient with a homozygous nonsense mutation in CIDEC. EMBO Mol Med 2009;1(5):280–287 Jambunathan S, Yin J, Khan W, Tamori Y, Puri V. FSP27 promotes lipid droplet clustering and then fusion to regulate triglyceride accumulation. PLoS ONE 2011;6(12):e28614 Gong J, Sun Z, Wu L, et al. Fsp27 promotes lipid droplet growth by lipid exchange and transfer at lipid droplet contact sites. J Cell Biol 2011;195(6):953–963 Sun Z, Gong J, Wu H, et al. Perilipin1 promotes unilocular lipid droplet formation through the activation of Fsp27 in adipocytes. Nat Commun 2013;4:1594 Magré J, Delépine M, Khallouf E, et al; BSCL Working Group. Identification of the gene altered in Berardinelli-Seip congenital lipodystrophy on chromosome 11q13. Nat Genet 2001;28(4): 365–370 Chen W, Chang B, Saha P, et al. Berardinelli-seip congenital lipodystrophy 2/seipin is a cell-autonomous regulator of lipolysis essential for adipocyte differentiation. Mol Cell Biol 2012;32(6): 1099–1111 Cui X, Wang Y, Tang Y, et al. Seipin ablation in mice results in severe generalized lipodystrophy. Hum Mol Genet 2011;20(15): 3022–3030 Fei W, Li H, Shui G, et al. Molecular characterization of seipin and its mutants: implications for seipin in triacylglycerol synthesis. J Lipid Res 2011;52(12):2136–2147 Payne VA, Grimsey N, Tuthill A, et al. The human lipodystrophy gene BSCL2/seipin may be essential for normal adipocyte differentiation. Diabetes 2008;57(8):2055–2060 Chen W, Yechoor VK, Chang BH, Li MV, March KL, Chan L. The human lipodystrophy gene product Berardinelli-Seip congenital lipodystrophy 2/seipin plays a key role in adipocyte differentiation. Endocrinology 2009;150(10):4552–4561 Yang W, Thein S, Guo X, et al. Seipin differentially regulates lipogenesis and adipogenesis through a conserved core sequence and an evolutionarily acquired C-terminus. Biochem J 2013; 452(1):37–44 Cui X, Wang Y, Meng L, et al. Overexpression of a short human seipin/BSCL2 isoform in mouse adipose tissue results in mild lipodystrophy. Am J Physiol Endocrinol Metab 2012;302(6): E705–E713 Fei W, Shui G, Zhang Y, et al. A role for phosphatidic acid in the formation of “supersized” lipid droplets. PLoS Genet 2011;7(7): e1002201 Fei W, Du X, Yang H. Seipin, adipogenesis and lipid droplets. Trends Endocrinol Metab 2011;22(6):204–210 Tian Y, Bi J, Shui G, et al. Tissue-autonomous function of Drosophila seipin in preventing ectopic lipid droplet formation. PLoS Genet 2011;7(4):e1001364

92

93

94

95 96

97

98

99

100

101

102 103

104

105

Seminars in Liver Disease

Vol. 33

No. 4/2013

107

108

109

110

111

112

113

114

115

116

117

118

119 120

This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited.

320

Copyright of Seminars in Liver Disease is the property of Thieme Medical Publishing Inc. and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use.

The lipid droplet as a potential therapeutic target in NAFLD.

Nonalcoholic fatty liver disease (NAFLD) is a growing problem worldwide. Nonalcoholic fatty liver disease is characterized by an abnormal accumulation...
249KB Sizes 0 Downloads 0 Views