and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa,

Size: px
Start display at page:

Download "and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa,"

Transcription

1 1 Lipids, Lysosomes and Autophagy Bharat Jaishy 1 * and E. Dale Abel 1,2 1 Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA. 2 Corresponsdece to: E. Dale Abel, MB.BS., DPhil. Fraternal Order of Eagles Diabetes Research Center Division of Endocrinology and Metabolism Roy J. and Lucille A. Carver College of Medicine, University of Iowa 4312 PBDB, 169 Newton Road, Iowa City, IA Telephone: Fax: DRCadmin@uiowa.edu *Present Address Department of Molecular Genetics University of Texas Southwestern Medical Center Dallas, TX

2 2 ABSTRACT Lipids are essential components of a cell providing energy substrates for cellular processes, signaling intermediates and building blocks for biological membranes. Lipids are constantly recycled and redistributed within a cell. Lysosomes play an important role in this recycling process that involves the recruitment of lipids to lysosomes via autophagy or endocytosis for their degradation by lysosomal hydrolases. The catabolites produced are redistributed to various cellular compartments to support basic cellular function. Several studies demonstrated a bidirectional relationship between lipids and lysosomes that regulate autophagy. While lysosomal degradation pathways regulate cellular lipid metabolism, lipids also regulate lysosome function and autophagy. In this review, we focus on this bidirectional relationship in the context of dietary lipids and provide an overview of recent evidence of how lipid-overload lipotoxicity as observed in obesity and metabolic syndrome impairs lysosomal function and autophagy that may eventually lead to cellular dysfunction or cell death.

3 3 Overview of Dietary Lipid Metabolism: Fatty acids are an essential component of cellular structure and function. They provide backbones for biological membranes, act as potent signaling molecules, coordinate whole body homeostasis as lipid hormones and, importantly, provide fatty acid substrates to generate energy in the form of ATP. Fatty acids (FAs) enter the circulation from various sources (Fig. 1). In adipocytes, sequential hydrolysis of triglycerides (TGs) by three cytosolic lipases, adipose tissue triglyceride lipase (ATGL), hormone-sensitive lipase (HSL) and monoacylglycerol lipase (MGL), releases FAs that circulate bound to albumin. In the intestine, dietary lipids are hydrolyzed by pancreatic lipase into free fatty acid (FFA) and glycerol, which are reesterified into TGs and packaged into lipoprotein particles called chylomicrons in the enterocytes. In liver, TGs and cholesterol are assembled into very low density lipoprotein (VLDL) particles. The lipoprotein lipase-mediated TG hydrolysis of circulating chylomicrons and VLDLs generates FAs and remnants of chylomicrons and VLDLs, respectively (1). The VLDL remnants are converted into low density lipoprotein particles and taken up by liver through lipoprotein receptor-mediated endocytosis along with chylomicron remnants. FA entry into the cell is facilitated by fatty acid transporters such as CD36 and fatty acid transport proteins (FATP) at the cell surface (2). FAs can also be synthesized from carbohydrate precursors by de novo lipogenesis when there is a surplus of carbohydrates. FAs serve multiple functions. As a metabolic fuel, they are either stored or oxidized. FAs are stored as TGs in specialized organelles called lipid droplets (LDs) primarily in white

4 4 adipose tissue (WAT) and liver and, to some extent, in other tissues. LDs also contain cholesterol esters and are surrounded by a phospholipid monolayer and several structural membrane proteins such as perilipins (PLIN), lipases, and the Rab family of small GTPases (3). Alternatively, FAs are transported into the mitochondria or peroxisomes as fatty acyl-coa and undergo β-oxidation, which sequentially removes two carbon units from their chain by a series of cyclic reactions catalyzed by acyl-coa dehydrogenases, enoyl-coa hydratase and 3-ketoacyl-CoA thiolase to generate acetyl- CoA. Acetyl-CoA enters the TCA-cycle generating reducing equivalents nicotinamide adenine dinucleotide and flavin adenine dinucleotide, which drive mitochondrial oxidative phosphorylation to generate ATP (4). FA oxidation is more prominent in tissues with high energy demands such as skeletal muscles and the heart. Synthesis of FAs or their oxidation is tightly regulated in response to organismal nutritional status. In the fed state, glucose is the primary source of energy and fatty acids are stored as TGs in LDs. Upon fasting, although hepatic glucose production is increased, rates of TG hydrolysis also increase to supply FFAs which are utilized as such or converted to ketone bodies by the liver to meet energy requirements of the peripheral tissues during starvation. The intimate regulation of FA metabolism and nutrient availability are essential for organismal lipid homeostasis and are regulated at multiple levels by a complex network of autocrine, paracrine and endocrine signaling. However, chronic feeding of energy rich diets combined with a sedentary life style and genetic predisposition may disrupt these homeostatic networks leading to lipid overload, which contributes in part to insulin resistance in obesity and the metabolic syndrome. Lipid overload eventually

5 5 overwhelms adipose tissue s capacity to store TG that leads to rerouting of lipids in tissues with minimal lipid storage capacity such as skeletal muscle, heart, liver, vascular endothelial cells and pancreas (5-7). Ectopically stored lipids are mostly shunted into non-oxidative pathways generating toxic lipid intermediates such as diacyl glycerol, ceramides, acyl carnitines and long chain fatty acyl-coa, which may cause several deleterious effects collectively known as lipotoxicity that contribute to impaired cellular signaling, mitochondrial dysfunction, endoplasmic reticulum (ER)-stress and cell death (Fig. 2). Consequences of these maladaptations include insulin resistance, β-cell loss, hepatic steatosis and cardiovascular diseases (8-13). Furthermore, chronic lipid oversupply enhances FA oxidation rate and imposes constant pressure on the mitochondrial electron transport chain to increase electron flux, which increases reactive oxygen species (ROS) production from electron transport chain complexes I and III (14). Lipid overload also leads to ROS production from extramitochondrial sources such as NADPH oxidases and other pro-oxidant enzymes. In concert with decreased expression of antioxidative enzymes, these changes precipitate oxidative stress and lead to lipid peroxidation, DNA damage, lysosomal dysfunction, defective autophagy, and activation of inflammatory responses in multiple tissues (15). Emerging evidence suggests that the lysosome mediated catabolic process called autophagy plays a pivotal role in maintaining cellular lipid homeostasis in multiple tissues (16-18). Accordingly, systemic dysregulation of autophagy is observed in animal models of diet-induced obesity, oxidative stress and metabolic syndrome (19-22), suggesting a role for defective autophagy in lipid overload-induced metabolic dysfunction. For lysosome mediated recycling of lipids in general, readers are referred

6 6 to excellent reviews published previously (23, 24). This review focuses on recent advances in our understanding of the bidirectional relationship between autophagy and the metabolism of dietary lipids such as FAs, TGs and cholesterol and highlights recent findings on how lysosomal dysfunction may impair autophagy and exacerbate lipotoxicity in the setting of dietary lipid overload. Understanding the mechanisms of FAinduced lysosomal dysfunction opens a new avenue for therapeutic approaches that may aimed at mitigating lipotoxicity and improving clinical outcomes of obesity. Overview of Autophagy Adaptation to changes in nutrient status is fundamental to the survival of all organisms. Autophagy is an evolutionarily conserved process that is central for cellular survival during periods of nutrient deprivation. Dysregulation of autophagy is associated with diverse diseases including cancer, cardiovascular, neurodegenerative, muscular and metabolic diseases (25). Autophagy is a catabolic process wherein cells degrade their own cytoplasmic materials in lysosomes to supply precursor molecules for processes critical for cell survival. In addition to providing alternative energy sources during starvation, autophagy is also involved in growth and differentiation, metabolic regulation, and macromolecule and organelle quality control. As such, the repertoire of autophagic substrates targeted for degradation is highly heterogeneous and includes proteins, lipids, carbohydrates and damaged organelles such as mitochondria, ER and peroxisomes. Distinct forms of autophagy exist, based on the mode of substrate delivery to lysosomes and distinct molecular mediators. These include chaperone mediated autophagy (CMA), microautophagy, organellar specific autophagy such as mitophagy and macroautophagy, all of which have been comprehensively reviewed (26,

7 7 27). Macroautophagy (hereafter autophagy ) is the major form of autophagy that plays a key role in lipid metabolism and will be the focus of this review. Regulation of Autophagy Autophagy is a multistep process that involves several core autophagy proteins (ATG) and an increasing number of upstream signaling molecules that respond to various intracellular and extracellular stimuli such as nutrient deprivation (28). Initial mechanistic insight into autophagy came from the genetic screening of autophagy-deficient yeast mutants, which formed the basis for the study of more complex mammalian autophagy (29). The mechanistic target of rapamycin complex 1(mTORC1), a nutrient sensing kinase, is a central regulator of autophagy that integrates signals from multiple upstream pathways to modulate autophagy (Fig. 3). In the fed state, growth factor and nutrient signaling activates mtorc1. Active mtorc1 directly interacts unc-51-like autophagy activating kinase 1 and 2 (ULK1/2)-ATG13-focal adhesion kinase family interacting protein of 200 kd (FIP200) complex and phosphorylates ULK1/2 and ATG13 to suppress autophagy to a low basal level (30, 31). However, nutrient depletion or mtorc1 inhibition relieves this inhibitory phosphorylation to initiate activation of autophagy. Additionally, prolonged nutrient depletion activates the AMP-activated protein kinase (AMPK) which simultaneously inhibits mtorc1 and induces ULK1 (32). ULK1/2 phosphorylates ATG13 and FIP200 to activate the ULK1/2-ATG13-FIP200 complex, which in turn activates the Beclin1-class III phosphatidylinositol-3-kinase (PI3K3 or VPS34) complex to initiate autophagosome formation (33). Prior to its activation, Beclin1 dissociates from its inhibitory interaction with B-cell lymphoma 2 (BCL2) and interacts with VPS34, a process mediated by nutrient depletion and BCL2

8 8 phosphorylation by c-jun N-terminal kinase 1 (JNK1) (34). VPS34 catalyzes the formation of phospatidyl inositol-3 phosphate (PI3P) which is essential for the recruitment of several regulatory ATG proteins to the phagophore for autophagosome expansion. Initiation of autophagy is followed by two ubiquitin like conjugation events: one is mediated by ubiquitin-like E1 and E2 ligases ATG7 and ATG10 to form an ATG5- ATG12-ATG16 complex; and the other is mediated by the cysteine protease ATG4 and ubiquitin-like ligases ATG7 and ATG3, resulting in formation lipid-conjugated microtubule-associated protein 1 light chain 3 (LC3-II). The ATG complex and LC3-II are required for the sequestration of autophagic substrates and maturation of autophagosomes. In addition, the adapter protein sequestosome 1 (p62 or SQSTM1) also interacts with autophagic substrates and targets them to the growing autophagosome vesicle (35). Matured autophagosomes fuse with lysosomes to form autolysomes where autophagic substrates are degraded by lysosomal hydrolases into simple molecules and released into the cytosol to complete the process of autophagy. Role of Autophagy in Lipid Metabolism The remarkable similarities between the events regulating autophagy and classical cytosolic lipolysis prompted several studies to examine the link between the two processes. The role of autophagy in lipid metabolism has also garnered considerable interest due to its potential implication in obesity and metabolic syndrome (36). Studies in liver and cultured hepatocytes showed that LDs can be selectively sequestered in autophagosomes and delivered to lysosomes for degradation by lysosomal acid lipases a process known as lipophagy (Fig. 4), which is distinct from classical lipolysis wherein lipids in LDs are directly hydrolyzed by cytosolic lipases without being

9 9 sequestered by autophagosomes for degradation (16). Although, like lipolysis, lipophagy is activated in response to fasting or sustained lipid challenge, our understanding of the molecular mechanism of lipophagy is incomplete. Removal of the LD surface proteins PLIN2 and PLIN3 by CMA represents the preliminary step that opens up the lipid components of the LD core for degradation by lipophagy or cytosolic lipases (37). β-adrenergic receptor agonist-induced activation of lipophagy suggests that PLIN removal allows activation of the small GTPase RAB7 at the exposed LD surface to initiate lysosomal degradation (38). RAB7 directs both autophagosomes and lysosomes to the LD surface to coordinate lipophagy following nutrient deprivation (39). At the transcriptional level, the transcription factor EB (TFEB), activates peroxisome proliferator-activated receptor-α (PPARα) and PPAR-γ Coactivator 1 Alpha (PGC1α) target genes involved in FA catabolism, including lipophagy, to accommodate increased hepatic lipid load following starvation (40). Further study revealed that transcriptional activation of TFEB by camp response element-binding protein (CREB) promotes fasting-induced lipophagy; whereas, activation of farnesoid X receptor upon refeeding inhibits TFEB activity (41). Like TFEB, activation of transcription factor forkhead box O1 (FOXO1) by nutrient depletion was shown to induce lysosomal acid lipase and lipophagy in adipocytes (42). Together, these studies indicate that mobilization of lipid depots by lipophagy is regulated by a complex network of transcriptional and posttranslational signaling in response to starvation. However, the mechanism of lipophagy in response to lipid challenge was not explored in these studies and requires further investigation, especially given the role of lipid overload in diet-induced metabolic diseases.

10 10 It is well established that FFAs generated from autophagy or lipolysis fuels mitochondrial β oxidation. Yet, how these two processes are coordinated and what is their relative contribution to the FFA pool used for mitochondrial β-oxidation is widely debated. Using mouse embryonic fibroblasts, Rambold et al. showed that when LDs were abundant, such as in the fed state, lipolysis of LDs by the cytosolic lipase ATGL supplied FFAs for mitochondrial oxidation, and autophagy was dispensable under these conditions (43). Only after prolonged starvation did bulk autophagy become essential to replenish the LD with TGs to maintain lipolysis mediated LD-to-mitochondria fatty acid transport. These finding are in line with previous findings which demonstrated that autophagy promotes LC3-mediated LD biogenesis in hepatocytes (18, 44). Since the Rambold study did not examine lipophagy, it is not known if lipophagy-derived FFAs follow the same fate as those of bulk-autophagy. Also, it remains to be seen if similar mechanisms exist in other cell types. Nevertheless, these studies support the notion that autophagy regulates lipid catabolism via selective and non-selective pathways based on nutritional cues, which may serve diverse physiological roles in diverse tissues as described below. In contrast to mouse embryonic fibroblasts, inhibition of autophagy by ablating the autophagy gene Atg7 in mouse liver increased hepatic lipid accumulation following starvation or high-fat diet feeding (16). In hepatocytes, inhibition of autophagy by silencing Atg5 or by inhibiting VPS34 significantly increased cellular TG content following oleate exposure. The increased lipid storage was characterized by an increase in LD size and number in autophagy deficient hepatocytes (16). Lipid accumulation was due to reduced lysosomal lipolysis resulting from the decreased delivery of lipid

11 11 substrates into lysosomes and not due to increased de novo lipogenesis. This observation suggests that lipophagy may act as a major lipolytic pathway in liver where cytoplasmic lipolysis has been considered significantly lower than that in adipocytes. As such, lipophagy may play a crucial role in maintaining lipid homeostasis and protecting cells against hepatolipotoxicity. Supporting this notion, hormonal activation of autophagy protects against hepatic lipotoxicity induced by high fat diet in mice or by FFA in hepatocytes (45). In contrast, autophagy deficiency sensitizes heptocytes to oxidative stress-induced apoptotic cell death (46). In addition, lipophagy may also regulate FFA supply required for VLDL production; since inhibition of autophagy suppresses VLDL secretion whereas induction of autophagy upregulates VLDL secretion (47). Unlike in hepatocytes, autophagy promotes adipogenesis and lipid storage in adipocytes. Inhibition of autophagy in 3T3L1 preadipocytes pharmacologically or by genetic knockdown of Atg5 or Atg7 blocked their differentiation into mature WATs and, hence, their lipid-storing capacity. Protein levels of key mediators [PPAR-γ, CCAAT/enhancer binding protein-α (CEBP-α) and β (CEBP-β)] and markers [(fatty acid synthase, steroyl-coenzyme A desaturase and glucose transporter 4] of adipocyte differentiation were reduced in autophagy-deficient adipocytes, suggesting that the impaired lipid storage is secondary to a defect in adipocyte differentiation (17, 48). Studies on 3T3L1 preadipocytes showed that transcriptional activation of Atg4b by CEBP-β is required to induce autophagy during adipocyte differentiation. CEBP-βinduced autophagy then degrades kruppel like factor 2 and 3 two negative regulators of adipogenesis to facilitate 3T3L1 adipocyte differentiation (49). In line with in vitro data, CEBP-β was shown bound to the Atg4b promoter in mouse WAT, and depletion of

12 12 Atg4b increased kruppel like factor 2/3 levels and reduced WAT mass in mice (49). The adipogenic role of autophagy was further validated in adipocyte-specific Atg7 null mice, which showed significantly reduced WAT mass, resulting in a lean, insulin sensitive phenotype. Loss of WAT mass in these mice was accompanied by increased brown adipose tissue (BAT)-like features in white adipose depots such as increased expression of PGC-1α, a key regulator of mitochondrial biogenesis, and uncoupling protein 1 (UCP1); increased levels of mitochondrial enzymes cytochrome oxidase and cytochrome C; increased mitochondrial number; and the presence of rounded nuclei and small multilocular lipid droplets (17, 48). Thus, knockout mice with enlarged BATlike mass exhibited high rates of β-oxidation that might have prevented TG accumulation in skeletal muscles and liver that increased insulin sensitivity. Despite improved FA oxidation and insulin sensitivity, adipose-specific Atg7 knockout mice died prematurely after 6 weeks of age on both regular and high-fat diets, which suggests an essential but undefined role for adipocyte-autophagy on organismal viability (48). Since the studies described above were performed in undifferentiated adipocytes or in mice with germline knockout manipulations, it was not possible to distinguish the effects of autophagy on adipocyte differentiation from effects on mature adipocyte physiology. Recently, Yin et al. showed that palmitate-induced ER stress activates autophagy in mature 3T3L1 adipocytes (50). Pharmacological inhibition of autophagy increased expression of the pro-inflammatory cytokines monocyte chemoattractant protein-1and interleukin-6 (IL-6) and exacerbated palmitate-induced ER stress and cell death (50). These findings imply that autophagy in mature adipocytes may play an adaptive role in mitigating ER stress and low-grade inflammation associated with obesity and insulin

13 13 resistance. In fact, autophagy was induced in adipose tissue from obese individuals, and inhibition of autophagy increased expression of the pro-inflammatory genes IL-1, IL- 6 and IL-8 both in adipocytes and adipose tissue explants of obese individuals compared to non-obese controls (51). Furthermore, overexpression of death-associated kinase 2 (DAPK2) one of the most strongly repressed adipose tissue genes in obese humans and high fat diet-induced obese mice increased autophagic clearance in 3T3L1 adipocytes in nutrient rich conditions (52). Consistent with these observations, in adipocytes isolated from obese patients recovering from bariatric surgery-induced weight loss, both DAPK2 expression and autophagic clearance were markedly increased and adipocyte size was proportionally reduced post-surgery compared to the adipocytes from same patients prior to surgery (52). Despite the progress made in defining the role of autophagy in mature adipocytes, questions remains whether bulk autophagy or selective lipophagy is involved in mediating the changes observed in these studies and what the molecular mechanisms of autophagic regulation are. A conditional deletion of core autophagic genes or genes specifically involved in lipophagy may ultimately clarify of the role of autophagy in mature adipocytes and in adipocyte lipid metabolism. Lipophagy controls whole body energy homeostasis by regulating the activity of hypothalamic neurons. The agouti-related peptide (AgRP) neurons of hypothalamus express AgRP and neuropeptide Y that stimulate food intake and reduce energy expenditure. A recent study showed that autophagy of neuronal lipids in AgRP neurons provides FFAs to induce AgRP expression in response to starvation (53). Inhibition of autophagy by ablating Atg7 in these neurons lowered AgRP expression and resulted in

14 14 leaner mice with higher activity and reduced food intake following starvation, possibly due to increased levels of the catabolic neuropeptide proopiomelanocortin (POMC) in POMC neurons. Consistent with these findings, mice lacking Atg7 in both AgRP and POMC neurons exhibited increased body weight, total fat mass, and lower energy expenditure (54). In skeletal muscles and the heart, autophagy plays important roles in maintaining tissue and energy homeostasis; yet, a comprehensive study on how autophagy regulates lipid metabolism in these tissues is lacking (55, 56). Based on existing evidence, one can assume that TGs or other dietary lipids are degraded via bulk autophagy to provide FFA substrates. However, if lipids are selectively degraded via lipophagy is unknown. Therefore, studies in skeletal muscles and the heart, examining the role of autophagy in mobilizing lipids accumulated in obese, insulin resistant, and type II diabetic models carries great therapeutic implications. At the same time, it important to note that muscles are not well suited to store lipids in bulk as liver or adipose tissue, it is possible that FFA entering muscles are oxidized for energy production or utilized for other physiological functions rather than stored as lipids under normal physiological conditions. Under such conditions, lipid mobilization through autophagy may be too subtle to be detected. Therefore, an obese model of muscle lipid accumulation may provide a good starting point to study lipid catabolism in response to starvation or other autophagic stimuli. In addition to TGs, lipophagy is also involved the hydrolysis of cholesteryl esters (CEs). In normal macrophages, CEs in LDs are hydrolyzed by cytosolic lipases to release free cholesterols which are effluxed from the cell. However, in cholesterol-loaded

15 15 macrophages, CE hydrolysis occurs primarily via lipophagy to generate free cholesterols that are used in reverse cholesterol transport to apolipoprotein A1-rich high density lipoproteins (HDLs) (57). Activation of autophagy upon cholesterol loading is likely a protective mechanism since the inhibition of autophagy in cholesterol-loaded smooth muscle cells significantly induced cell death. In contrast, rapamycin-induced autophagy protected cells from free cholesterol overload-induced cell death (58). These findings imply that inhibition of autophagy may be a key mediator of cholesterol overload-induced vascular cell death in the progression of atherosclerosis. This is an important hypothesis which requires further study to establish the role of autophagy in atherosclerosis and to determine if this could be a potential therapeutic target. Regulation of Autophagic Activity by Lipids With the role of autophagy in lipid catabolism well established, several studies show that an inverse relationship exists where dietary lipid overload changes cellular lipid content or composition to alter autophagic activity, which may have beneficial or detrimental effects in a tissue-specific manner (Table 1). Such lipid-induced changes are distinct from the fundamental role lipids and LDs play in autophagosome biogenesis and in the overall process of autophagy (59, 60). For example, exposure of INS-1E beta cells to palmitate induces autophagy in presence of hyperglycemia (61). Diabetic db/db mice or non-diabetic mice challenged with a high-fat diet exhibited marked induction of autophagosome formation in pancreatic beta cells (62). In these models of lipid overload, induction of autophagy played a protective role against lipotoxicity: inhibition of autophagy in INS-1E beta cells by depleting Atg5 or blocking autolysosome formation accentuated palmitate-induced cell death; likewise, beta cell specific

16 16 deletion of Atg7 in mice induced cell death and led to profound glucose intolerance, partly due to the lack of a compensatory increase in beta cell mass and insulin secretion following high-fat feeding. The loss of autophagy in Atg7-deficient beta cells significantly increased oxidative stress and toxic accumulation of ubiquitinated proteins and damaged organelles that might collectively contribute to beta cell degeneration (63). Inhibition of autophagic flux by chloroquine induced markers of ER stress (C/EBPhomologous protein and phosphorylated eukaryotic initiation factor-2α) and apotosis (cleaved caspse 3) in pancreatic beta cells of high-fat fed mice, which further supports the protective role of autophagy against beta cell lipotoxicity (63). While short-term high-fat feeding induces autophagy in pancreatic beta cells, chronic lipid overload impairs autophagic flux (64, 65). Prolonged exposure of INS1 beta cells and human pancreatic islets to high levels of palmitate and glucose increased the accumulation of autophagosomes and autophagic protein markers, and suppressed autophagic degradation of long-lived proteins. Impairment in autophagic flux led to defective insulin secretion and increased apoptotic cell death. Inhibition of mtorc1 by rapamycin partially restored autophagic flux, insulin secretion, and cell viability by mechanisms that are incompletely understood (64, 65). In addition to the chronicity of lipid overload, the type of fatty acid also determines beta cell lipotoxicity. The saturated fatty acid (SFA) palmitate inhibits beta cell proliferation and induces apoptosis, whereas the monounsaturated fatty acid palmitoleate with identical chain length counteracts the toxic effect of palmitate and promotes cell proliferation (66). Taken together, these in vivo and in vitro data support the notion that lipid overload-induced autophagy plays a protective role against lipotoxicity; however, long-term high-fat feeding or palmitate

17 17 exposure induces lipotoxicity by blocking this adaptive autophagic response. Other factors such as ceramide accumulation, increased ROS production may also induce lipotoxicity independent of autophagy (67). As in beta cells, chronic lipid accumulation inhibits hepatic autophagy. Hepatocytes acutely loaded with oleate failed to mobilize LDs through autophagic pathways (16). Mutliple mechanisms appear to drive lipid-induced inhibtion of hepatic autophagy. One potential mechanism is the sustained activation of mtorc1 in high-fat fed mouse liver that may inhibit autophagic initiation (68). Consistent with this notion, mice on a high-fat diet failed to move LDs to autophagic vesicles in response to starvation (16). In vitro studies showed that high-fat feeding also impairs autophagosome-lysosome fusion the distal step in the autophagic proess by disrupting the normal membrane composition of autophagosomes and lysosomes isolated from mouse liver (69). Suppression of hepatic autophagy may also occur by transcriptional mechanisms. In a mouse model of high-fat diet-induced hyperinsulinemia and insulin resistance, hepatic autophagy was suppressed partly due to transcriptional repression of FOXO1- dependent autophagic genes Vps34, Atg12 and GABA(A) receptor-associated protein like 1(Gabarapl1), which may contribute to autophagy inhibition (70). A separate study in genetic and dietary models of obesity, reported suppression of autophagy in terms of Atg7 expression in liver that paralleled increased ER stress and insulin resistance (71). Restoration of hepatic Atg7 expression in obese mice normalized autophagy, reduced ER stresss, and improved hepatic insulin action and systemic glucose tolerance. Lipid overload in skeletal muscles and the heart is associated with multiple metabolic abnormalities such as oxidative stress, mitochondrial dysfunction, insulin resistance,

18 18 apoptotic cell death and fibrosis (72-75). The role autophagy plays in these cellular stress responses is complex and incompletely understood. Some studies showed that basal autophagy remained intact in skeletal muscles of high-fat fed animals, although fat accumulation and oxidative stress was markedly elevated and insulin signaling pathways impaired (76, 77). In contrast, most studies showed altered autophagic activity in response to lipid overload. High-fat feeding was shown to induce autophagic flux in skeletal muscles marked by an increase in LC3-II, BECLIN1 and ULK1 and a decrease in p62 level in a mouse model of diet induced obesity and insulin resistance (22). Autophagic induction was correlated with increased oxidative stress, manifested by induction of superoxide dismutase activity, lipid peroxidation and mrna levels of key antioxidant enzymes. This increase in high-fat diet-induced autophagy was absent in adiponectin-deficient mice and was restored upon adiponectin reexpression. The insulin-resistant phenotype was recapitulated in an autophagy-deficient cell culture model expressing an Atg5 inactive mutant (22). These data suggest that HFD-induced skeletal muscle autophagy and antioxidant capacity ameliorates HFD-induced insulin resistance. Consistent with the adaptive role of diet-induced autophagy, high fat feeding in skeletal muscle-specific histone deacetylase 1 and 2 (Hdac1 and Hdac2) null mice which normally exhibit autophagy deficiency restored autophagic flux and prevented myopathy in adult mice (78). However, under chronic lipid overload, such as in severely obese, hyperinsulnemic human subjects, autophagic flux was blunted in primary skeletal muscle myotubes, which could contribute to the insulin resistant phenotype (79). Thus in the face of nutrient overload, the skeletal muscle adaptation to autophagy might be biphasic, with an initial increase that might limit insulin resistance. However, if

19 19 nutrient excess is persistent, this adaptative response is lost and declining autophagy might then exacerbate insulin resistance. This model may need to be revised in the light of recent study, which showed that. blocking autophagy by deleting Atg7 in skeletal muscles protects mice from diet-induced systemic obesity and peripheral insulin resistance (80). Mitochondrial dysfunction, induced by lack of autophagy, led to increased expression of fibroblast growth factor 21 (Fgf21) in the muscle and protected mice from diet-induced obesity and insulin resistance. Interestingly, autophagy deficiency in liver also induced Fgf21 expression and provided similar protection from obesity and insulin resistance (80). Together with previous findings, these data portray a complex role of autophagy in lipid-loaded skeletal muscle and warrants further study to reconcile these seemingly contradictory findings, or to more clearly delineate those changes that represent additional mechanisms that might be independent of autophagy per se, but which may impact metabolic homeostasis. The presence of these alternative pathways underscores the pleiotropic adaptations that may defend against diet-induced obesity and insulin resistance. While modest increases in autophagy may improve muscle function, excessive autophagy was linked to muscle atrophy. For example, induction of several key autophagy genes by the transcription factor FOXO3 hyperactivates autophagy and accelerates protein degradation in the muscle (81). A recent in vitro study showed that the FOXO3 pathway of muscle atrophy was induced as a lipotoxic response to SFA overload (82). Coincubation with unsaturated fatty acid completely reversed lipotoxic

20 20 effects of saturated fatty acid, underscoring the fatty acid-specific regulation of autophagy and atrophy in skeletal muscles. In the heart, basal autophagy plays a prosurvial role (83). However, our understanding of the lipid-induced regulation of autophagy is still emerging. He et al. showed that 12 weeks of high fat feeding suppressed cardiac autophagy assessed by the increased level of p62 (84). Several other studies reported impairment in cardiac autophagy following long-term high-fat feeding (85-87). A recent study showed that impaired cardiac autophagy in high fat fed mouse induced lipotoxicity was marked by an increase in ER stress and cardiomyocyte apoptosis, suggesting that autophagy might play a protective role against diet induced cardiac lipotoxicity (88). However, high-fat feeding also induces hyperglycemia, hyperinsulinemia, and cardiac hypertrophy in these mice which could independently impair autophagy. The mechanism of lipid-induced inhibition of autophagy in the heart is complex and multifactorial. A recent study showed that AKT2 is an important component of HFDinduced inhibition of autophagic flux (87). High-fat feeding suppressed the expression of Rab7 a small GTPase involved in lysosomal biogenesis and, thus, reduced autophagosome turnover. Knockdown of Akt2 prevented the suppression of Rab7 by an unknown mechanism and restored normal autophagic flux in high fat fed mouse heart (87). Besides AKT, AMPK-mediated regulation of protein tyrosine phosphatase -1B (PTP1B) also controls lipid-induced inhibition of autophagy (89). Mice fed with a 45% high fat diet for 5 months exhibited impaired autophagosome turnover in the heart in parallel with reduced AMPK phosphorylation. Deletion of PTP1B gene restored normal autophagic flux in high fat fed mice and reversed diet-induced cardiac hypertrophy and

21 21 contractile dysfunction. Pharmacological inhibition of AMPK, however, abrogated beneficial effects of PTP1B ablation on autophagy, cardiac structure and contractile function (89). In contrast to these studies, feeding a high fat diet rich in medium-chain SFAs was found to activate autophagy and promote subsequent development of cardiac hypertrophy and left ventricular contractile dysfunction in the murine heart (90). In this model, induction of autophagy required de novo synthesis of the specific sphingolipid C 14 -ceramide by ceramide synthase 5 (CERS5). Depletion of CerS5 mrna blocked autophagy induction and hypertrophy in isolated adult cardiomyocytes treated with myristate a precursor for C 14 -ceramide (90). This study showed that the specificity of the lipid species is a key determinant of the effect of diet on autophagy, lipotoxicity and cardiac hypertrophy. These studies underscore the importance of defining in even greater detail the composition of diets used in various metabolic studies, which may help reconcile some of the contradictory data in the literature. In vitro studies in H9C2 cardiomyocytes corrobrate some of the in vivo findings on lipid overload-induced cardiac autophagy. Treatment of these cells with palmitate induced apoptosis and increased the accumulation of autophagosomes and lysosomes. Induction of autophagy by rapamycin treatment ameliorated palmitate-induced apoptosis, whereas inhibition of autophagy by 3-MA exacerbated apoptosis (88). Data from multiple studies suggest that lipid overload induces dynamic autophagic response in these cells in a time-dependent manner. For instance, acute palmitate exposure may induce autophagy without inflicting cytotoxicity. Extending palmitate treatment to 4 h, however, inhibited autophagic flux by impairing lysosomal function with no cytotoxicity (91). Prolonged palmitate treatment for 12 h or longer suppressed autophagic flux with

22 22 concomitant increase in cellular lipotoxicity marked by an elevated ROS level, activation of the stress kinases JNK and p38mpak, and a profound increase in apoptotic markers such as cleaved caspase 3 and caspase 7 and their downstream effector poly ADP-ribose polymerase 1 (92). Thus, the initial activation of autophagy may protect cardiomyocytes by preventing accumulation of toxic lipids. The subsequent impairment in autophagic flux offsets this protective adaptation, which leads to cellular lipotoxicity. However, a recent study showed that prolonged treatment with the medium-chain SFA myristate but not the long-chain SFA palmitate induced autophagic flux and promoted hypertrophy in isolated adult primary cardiomyocytes (90). This suggests that myristate or its derivatives induces autophgy and hypertrophy by specific mechanisms that are not shared by other toxic FAs known to inhibit autophagy. Together, in vivo and in vitro studies indicate that multiple pathways mediate lipidinduced regulation of cardiac autophagy depending on composition of diet, type of fatty acids, duration of feeding, and type of cell culture and animal models used. Whether these pathways are linked, to provide a unifying mechanism or they represent independent mechanisms is unknown and requires a more comprehensive study. Lysosomes and Lipids Lysosomes are acidic organelles containing at least 50 different hydrolases that degrade a wide variety of macromolecules including DNA, proteins, carbohydrates and lipids delivered via autophagy and other endocytic pathways. Lysosomes contain membrane-bound vacuolar ATPases that maintain the acidic ph of the lysosomal lumen for efficient functioning of the hydrolases. Lysosomal membrane proteins undergo various modifications including glycosylation that protect them against proteolysis,

23 23 thereby stabilizing lysosomal membranes against acid hydrolases and other degradative agents (93). Like proteins, lipids are an intergral part of lysosomal membranes and play a key role in lysosomal biogenesis and function (reviewed in reference (23)). Reciprocally, lysosomes play a major role in lipid metabolism and the maintenance of cellular lipid homeostasis. A role for lysosomes in the recycling of membrane lipids has been described (24). In this part of the review, we primarily focus on the mutual interrelationship between dietary lipids and lysosomal function. Lysosomes as Regulators of Lipid Metabolism Both exogenous and endogenous lipids are delivered to lysosomes. Exogenous lipids enter the lysosomes via endocytosis. These lipids primarily include lipoproteins and CEs derived from low density lipoproteins (LDL) and remnants of very low density lipoproteins (VLDL) and chylomicrons (94, 95). Endocytosis of lipoproteins is facilitated by members of the LDL receptor family localized in either lipid rafts or clathrin-coated pits (96). Endocytosed LDL particles are transferred as such from late endosomes to the lysosomes where TGs and CEs are hydrolyzed by lysosomal acid lipase (LAL) to generate FFAs and free cholesterols, respectively (97). In contrast, for VLDL and chylomicrons remnants, their apolipoprotein E receptors are recycled back to the plasma membrane leaving remaining lipid particles to be degraded in the lysosome (96, 98). Endogenous lipids or LDs enter the lysosome primarily via autophagy and are hydrolyzed by LAL. Hydrolysis of LD TGs and CEs by LAL generate FFAs and free

24 24 cholesterols, respectively (16, 57). Normally, free cholesterols released from lysosomes are transferred to the ER for reesterification (99). However, as described earlier, in cholesterol-loaded macrophages, free cholesterols derived from lipophagy are incorporated into apolipoprotein A1-rich HDL (57). Biochemical studies on LAL isolated from rat-liver lysosomes suggest that the enzyme catalyzes both the hydrolysis and synthesis of CEs (100). Re-esterification of cholesterol was also observed in endosomes of cultured mouse macrophages exposed to LDL aggregates (101). Whether LAL catalyzes CE formation in these macrophages is not clear. These findings may have an implication in CE storage disease where cholesterol concentration in the membrane was found near saturation a condition under which cholesterolreesterification was upregulated in mouse macrophage endosomes. Deficiency of LAL results in two major diseases: CE storage disease and the more severe Wolman disease. The latter is characterized by the accumulation of both CEs and TGs (102). Besides TGs and CEs, a variety of glycerophospholipids and glycosphingolipids are also hydrolyzed in the lysosomes by specific phospholipases and water soluble acid hydrolases ( ). Besides hydrolyzing and recycling various lipids, lysosomes also act as a nutrient sensor to regulate lipophagy. Zoncu et al. showed that cells sense amino acid content in the lysosomal lumen to determine their nutritional status (108). This information is relayed by lysosomal vacuolar ATPase (v-atpase) proton pump to the Ragulator complex outside the lysosome via human member 9 of the solute carrier family 38, a recently identified lysosomal membrane resident-protein which forms a part of the Ragulator complex (109). Ragulator charges small molecule GTPases RagA/B and

25 25 RagC/D with GTP to activate them. Active Rags recruit mtorc1 to the lysosomal surface where the GTP binding protein ras homolog enriched in brain (RHEB) activates mtorc1 (108, 110). Active mtorc1 inhibits the transcription factor TFEB by direct phosphorylation and blocks PPARα transcriptional activity by promoting its interaction with nuclear receptor corepressor 1 (NCoR1) (111, 112). TFEB is the major transcription factor that globally activates several autophagic and lysosomal genes including LAL (113, 114). PPARα is the master transcriptional activator of several genes involved in fatty acid β-oxidation. Thus, by regulating mtorc1 activity in response to lumenal amino acid content, lysosomes control both lipophagy and mitochondrial fatty acid oxidation by sensing cellular nutrient status. It is interesting but yet unknown why lysosomes use amino acids to relay cellular nutrient status to control lipid metabolism. Regulation of Lysosomal Function by Lipids As in autophagy, lipids play an important role in regulating lysosomal function. While some lipid-induced protein modifications facilitate the transport of lysosomal proteins and enzymes to the lysosome (115), excess lipid supply largely impair lysosomal function by multiple mechanisms (Fig. 5). Lysosomal lipid storage diseases (LSD), or lipidoses, are a constellation of inherited disorders characterized by the accumulation of lipids in late endosomes and lysosomes (116). Many of these diseases are caused by impaired function of lysosomal hydrolyses, membrane transporters and other accessory proteins (Table 2). For instance, in Niemann-Pick disease type C (NPC), defects in the transporter proteins NPC1 and NPC2 impair cholesterol efflux and fill the late endosomes with unesterifed cholesterol (117, 118). How lipid accumulation contributes to the pathogenesis of the disease is still unclear. A previous study linked cholesterol

26 26 accumulation to defects in trafficking between intralumenal vesicles and limiting membrane of the late endosomes (119). Such defects may impair trafficking or reccyling of proteins, lipids and other membrane components between endocytic vesicles and other organelles such as Golgi, ER and plasma membrane, which is often observed in several lysosomal lipid storage diseases ( ). Accumulation of lipids in lysosomes can also inhibit autophagosome turnover. Several studies in LSD, documented an impairment in fusion between autophagosomes and lipid-loaded lysosomes that leads to defective autophagic clearance ( ). Conversely, depletion of cholesterol or inhibition of cholesterol synthesis induces autophagy (127, 128). In vitro fusion assays showed that altered membrane composition of autophagosomes and lysosomes isolated from lipid-loaded fibroblasts or high fat fed mouse liver inhibits the fusion between these organelles, which could account for the impaired autophagy in many lipid-induced lysosomal disorders (69). In addition to macroautophagy, dietary lipid overload also affects CMA. Mice fed with high fat diet showed selective degradation of lysosome associated membrane protein 2A (LAMP2A) in hepatocytes that led to inhibition of CMA in high fat-fed mice but not in chow-fed controls (129). Lipid-induced lysosomal dysfunction is also observed in diet-induced obesity and metabolic disorders (87, 130). Mice fed a high fat diet exhibited increased accumulation of phospholipids in lysosomes that was associated with reduced lysosomal enzyme activity, which was reversed by the chronic activation of AMPK (130). Likewise, high fat feeding in mice reduced the cardiac expression of Rab7 a small GTPase essential for lysosome maturation (131) which impaired autophagosome-lysosome fusion and

27 27 suppressed autophagic flux (87). Cardiac ablation of Atg2 restored Rab7 expression and normalized autophagic flux and prevented cardiac hypertrophy and contractile dysfunction induced by high fat feeding (87). Lysosomal membrane permeabilization (LMP) is a key mechanism by which chronic lipid overload promotes lysosome dysfunction and triggers apoptotic cell death (132, 133). Both high fat feeding and fatty acid exposure induced LMP in adipose tissue that led to the release and activation of the lysosomal protease cathepsin B (CTSB) in the cytosol following macrophage infiltration of adipose tissue and production proinflammatory cytokines (134). High fat feeding also induced LMP and lipotoxicity in the liver of a mouse model of diet-induced non-alcoholic steatohepatitis (NASH) although hepatic autophagy remained largely unchanged (135). In the heart, a high fat diet induced release of lysosomal cathepsin K into the cytosol a hallmark of LMP in parallel with cardiac hypertrophy, contractile dysfunction and the induction of apoptosis. Ablation of cardiac cathepsin K improved cardiac structure and function and suppressed apoptosis in high fat fed mice (136). In macrophages, exposure to atherogenic lipids such as oxidized LDL (oxldl) and cholesterol crystals precipitated lysosomal dysfunction characterized by increased lysosomal ph, reduced proteolytic activity, and increased LMP which led to impaired cholesterol efflux, potentially due to inhibition of autophagy (137). Induction of lysosomal biogenesis by TFEB overexpression attenuated lysosomal dysfunction and restored normal cholesterol efflux in lipid-loaded macrophages. Mechanistic studies on mouse hepatocytes revealed that SFA treatment initiates Bax translocation to lysosomes and lysosomal destabilization that releases cathepsins into the cytosol (138, 139). Cytosolic cathepsins induced mitochondrial

28 28 membrane permeabilization and caspase activation, triggering the intrinsic pathway of apoptosis and liver injury as observed in a dietary mouse model of non-alcoholic fatty liver disease (NAFLD). Genetic and pharmacological inactivation of CTSB was shown to be protective against hepatic steatosis and liver injury in dietary NAFLD mice (132). These studies indicate that LMP may be the key mediator of lipotoxicity. Besides lysosomal pathways, SFA-induced lipotoxicity can also activate non-lysosomal extrinsic pathways of apoptosis (140). Oxidative stress is another important regulator of lysosome function. The high iron content of lysosomes, derived from the degradation of metalloproteins such as ferritin, makes them susceptible to oxidative damage (141, 142). Ferric iron reacts with hydrogen peroxide (H 2 O 2 ) and superoxide anions (O2 - ) to generate the highly reactive hydroxyl radical (OH ) (143). Hydroxyl radicals can oxidize proteins, lipids, and other macromolecules that inhibit their degradation leading to the accumulation of indigestible wastes in lysosomes. Lysosomal wastes are known to impair lysosomal enzyme activity and promote LMP (144). Additionally, ROS and reactive nitrogen species (RNS) generated from various other cellular sources may also affect lysosome function. Previous studies on neutrophils showed that NADPH oxidase isoform 2 (NOX2) is recruited to the late endosomal membrane where it generates superoxides. Initially, superoxides can promote acidification of endosomes by trapping protons in the endosomal lumen. However, excessive superoxides can react with water to generate hydroxyl ions that can ultimately impair acidification of the endosomal vesicles by increasing ph (145). NOX2 activation and increased ROS production has been observed in animal models of diabetes and diet-induced obesity and in humans with

29 29 metabolic syndrome (15, ). Until recently, NOX2-derived ROS was known to regulate phagosomal ph only in inflammatory cells: for antigen presentation in dendritic cells, and for targeting phagosomes for antibacterial autophagy. Due to striking parallels between NOX activation and lysosomal dysfunction in the context of lipid oversupply, we examined if two processes are interconnected. Both high fat fed mouse hearts and palmitate exposed H9C2 cardiomyocytes markedly increased NOX2 activity. NOX2- derived superoxides impaired lysosomal acidification and hydrolase activity that blunted autophagosome degradation despite normal autophagosome-lysosome fusion (91). Genetic and pharmacological inhibition of NOX2 normalized superoxide production, and restored lysosomal function and autophagic flux. Mechanistically, palmitate treatment led to an accumulation of diacylglycerol, which activated protein kinase beta2, to increase NOX2 activity. NOX2-derived superoxide, increased oxidative S-nitrosylation of the Atp6v1a1 subunit of lysosomal vatpase at Cys-277, which masks the free thiol group potentially required for cleaving an inhibitory disulfide bond and activating vatpase (149, 150), leading to impaired lysosomal acidification and defective autophagic clearance. How NOX2-derived superoxides regulate S-nitrosylation of vatpase subunits is under further investigation. Consistent with the role of NOX2 in lysosomal function, Pal et al. found that NOX2-derived ROS impairs both lysosome biogenesis and autophagy in a mouse model of Duchenne muscular dystrophy. Genetic down regulation of NOX2 attenuated ROS production, restored normal autophagy, and improved muscle function in these mice (151). These studies suggest that NOX2 regulates lysosome biology by multiple mechanisms in diverse cells types in a stimulusdependent fashion.

30 30 It is important to note that in line with multiple studies examining FFA-induced lipotoxicity, we observed that only the saturated but not the monounsaturated FAs induced ROS-production and lysosomal dysfunction in cardiomyocytes (91). In fact, studies suggest that monounsaturated FAs may protect against saturated FFA-induced lipotoxicity, possibly by channeling toxic lipids to the relatively benign TG depot (82, 152, 153). Supporting these findings, we found that co-incubation with oleate increased TG accumulation and normalized autophagy in palmitate-treated cardiomyocytes (91). These studies underscore the need for defining the composition of lipids in an animal diet or in vitro experiments to better understand the mechanism of lipotoxicity. Such approaches may also reconcile discrepancies in various studies examining the effect of lipid-overload on lysosome function, autophagy, and other cellular pathways involving lysosome function. Conclusion: Lipids are central to cellular survival and viability by regulating fundamental processes that maintain cellular structure, organelle function and energy homeostasis. Oversupply of dietary lipids overwhelms the ability of adipocytes and peripheral tissues to store lipid, leading to cellular stress responses. Autophagy has emerged as an important process for maintaining cellular and whole body lipid homeostasis under normal conditions, upon nutrient depletion, and during the early phase of lipid challenge. This homeostatic mechanism falters upon chronic lipid overload by multiple mechanisms. Studies of autophagy and lipids over the last decade have greatly increased our understanding of their mutual relationship; however, several key questions still remain to be answered in the short-term and the long-term.

31 31 How are LDs recruited to lysosomes via lipophagy? How is the mechanism of lipophagy distinct from other types of autophagy? What are the molecular signals that govern selective targeting of LDs via lipophagy? What is the molecular switch between non-selective and selective degradation of LDs via autophagy? What are the differences in the utilization of FFAs generated from lipophagy, bulk autophagy and cytosolic lipolysis? Does reactivation of lipophagy protect cells from lipotoxicity when bulk autophagy is inhibited? Does lipophagy in mature adipose tissue the major organ for lipid metabolism play a major role in lipid mobilization as in hepatocytes, given the dogma that classical lipolysis in adipocytes is the major and most efficient lipolytic mechanism? Since lysosomes are the final destination for autophagy and other endocytotic pathways, endo/lysosomal dysfunction not only affects macroautophagy but also impairs CMA, recycling of membrane lipids, sorting of lipids and lipoproteins into appropriate intracellular compartments, and promotes lipotoxic cell death (23, 154, 155). Accordingly, the repertoire of lysosomal functions has greatly increased recently, transforming it from a recycling bag to a hub of nutrient signaling. However, the molecular mechanisms of many of these functions are incompletely understood. From the perspective of lipid metabolism, the following represent some outstanding questions that remain to be answered in future investigations. How do lysosomes process different types of lipids? Do lysosomal hydrolases have equal affinity for all types of lipids?

32 32 How are lysosomal lipid catabolites sorted into different cellular pathways? Are lysosomal membrane proteins and transporters also involved in lipid trafficking within and outside of cells? How do lysosomes coordinate lipophagy and other lipid mobilizing pathways? How do different fatty acid species modulate lysosomal function? Do lysosomes also sense nutrient signals other than amino acids, such as simple FFAs or sugars, and how do these signals crosstalk with amino acid-sensing signaling mechanisms? In conclusion, this review has discussed studies that broaden our understanding of how lysosomes import and process dietary lipids. Moreover, they have also provided additional insights into other cellular functions whereby the lysosome may act as a lipidregulated signaling hub in addition to its role as an organelle for degradation (156, 157). Acknowledgment Relevant studies in the Abel laboratory were supported by NIH grant R01HL108379

33 33 References 1. Young, S. G., and R. Zechner Biochemistry and pathophysiology of intravascular and intracellular lipolysis. Genes Dev 27: Glatz, J. F., J. J. Luiken, and A. Bonen Membrane fatty acid transporters as regulators of lipid metabolism: implications for metabolic disease. Physiological reviews 90: Guo, Y., K. R. Cordes, R. V. Farese, Jr., and T. C. Walther Lipid droplets at a glance. J Cell Sci 122: Rui, L Energy metabolism in the liver. Comprehensive Physiology 4: Savage, D. B., K. F. Petersen, and G. I. Shulman Disordered lipid metabolism and the pathogenesis of insulin resistance. Physiological reviews 87: Wymann, M. P., and R. Schneiter Lipid signalling in disease. Nat Rev Mol Cell Biol 9: Lewis, G. F., A. Carpentier, K. Adeli, and A. Giacca Disordered fat storage and mobilization in the pathogenesis of insulin resistance and type 2 diabetes. Endocr Rev 23: Unger, R. H Minireview: weapons of lean body mass destruction: the role of ectopic lipids in the metabolic syndrome. Endocrinology 144: Wende, A. R., J. D. Symons, and E. D. Abel Mechanisms of lipotoxicity in the cardiovascular system. Curr Hypertens Rep 14: Watt, M. J., and A. J. Hoy Lipid metabolism in skeletal muscle: generation of adaptive and maladaptive intracellular signals for cellular function. American journal of physiology. Endocrinology and metabolism 302: E Trauner, M., M. Arrese, and M. Wagner Fatty liver and lipotoxicity. Biochim Biophys Acta 1801: Chiu, H. C., A. Kovacs, D. A. Ford, F. F. Hsu, R. Garcia, P. Herrero, J. E. Saffitz, and J. E. Schaffer A novel mouse model of lipotoxic cardiomyopathy. J Clin Invest 107: Cunha, D. A., P. Hekerman, L. Ladriere, A. Bazarra-Castro, F. Ortis, M. C. Wakeham, F. Moore, J. Rasschaert, A. K. Cardozo, E. Bellomo, L. Overbergh, C. Mathieu, R. Lupi, T. Hai, A. Herchuelz, P. Marchetti, G. A. Rutter, D. L. Eizirik, and M. Cnop Initiation and execution of lipotoxic ER stress in pancreatic beta-cells. J Cell Sci 121: Fisher-Wellman, K. H., and P. D. Neufer Linking mitochondrial bioenergetics to insulin resistance via redox biology. Trends Endocrinol Metab 23: Furukawa, S., T. Fujita, M. Shimabukuro, M. Iwaki, Y. Yamada, Y. Nakajima, O. Nakayama, M. Makishima, M. Matsuda, and I. Shimomura Increased oxidative stress in obesity and its impact on metabolic syndrome. The Journal of clinical investigation 114: Singh, R., S. Kaushik, Y. Wang, Y. Xiang, I. Novak, M. Komatsu, K. Tanaka, A. M. Cuervo, and M. J. Czaja Autophagy regulates lipid metabolism. Nature 458: Zhang, Y., S. Goldman, R. Baerga, Y. Zhao, M. Komatsu, and S. Jin Adipose-specific deletion of autophagy-related gene 7 (atg7) in mice reveals a role in adipogenesis. Proc Natl Acad Sci U S A 106: Shibata, M., K. Yoshimura, N. Furuya, M. Koike, T. Ueno, M. Komatsu, H. Arai, K. Tanaka, E. Kominami, and Y. Uchiyama The MAP1-LC3 conjugation system is involved in lipid droplet formation. Biochem Biophys Res Commun 382: Yang, L., P. Li, S. Fu, E. S. Calay, and G. S. Hotamisligil. Defective Hepatic Autophagy in Obesity Promotes ER Stress and Causes Insulin Resistance. Cell Metabolism 11: Kim, K. H., Y. T. Jeong, H. Oh, S. H. Kim, J. M. Cho, Y.-N. Kim, S. S. Kim, D. H. Kim, K. Y. Hur, H. K. Kim, T. Ko, J. Han, H. L. Kim, J. Kim, S. H. Back, M. Komatsu, H. Chen, D. C. Chan, M. Konishi, N. Itoh, C. S.

34 34 Choi, and M.-S. Lee Autophagy deficiency leads to protection from obesity and insulin resistance by inducing Fgf21 as a mitokine. Nat Med 19: Nunez, C. E., V. S. Rodrigues, F. S. Gomes, R. F. de Moura, S. C. Victorio, B. Bombassaro, E. A. Chaim, J. C. Pareja, B. Geloneze, L. A. Velloso, and E. P. Araujo Defective regulation of adipose tissue autophagy in obesity. Int J Obes 37: Liu, Y., R. Palanivel, E. Rai, M. Park, T. V. Gabor, M. P. Scheid, A. Xu, and G. Sweeney Adiponectin stimulates autophagy and reduces oxidative stress to enhance insulin sensitivity during high-fat diet feeding in mice. Diabetes 64: Hamer, I., G. Van Beersel, T. Arnould, and M. Jadot Lipids and lysosomes. Curr Drug Metab 13: Kolter, T., and K. Sandhoff Lysosomal degradation of membrane lipids. FEBS Lett 584: Levine, B., and G. Kroemer Autophagy in the pathogenesis of disease. Cell 132: Kaushik, S., and A. M. Cuervo Chaperone-mediated autophagy: a unique way to enter the lysosome world. Trends Cell Biol 22: Li, W. W., J. Li, and J. K. Bao Microautophagy: lesser-known self-eating. Cell Mol Life Sci 69: Weidberg, H., E. Shvets, and Z. Elazar Biogenesis and cargo selectivity of autophagosomes. Annu Rev Biochem 80: Kim, J., and D. J. Klionsky Autophagy, cytoplasm-to-vacuole targeting pathway, and pexophagy in yeast and mammalian cells. Annu Rev Biochem 69: Hosokawa, N., T. Hara, T. Kaizuka, C. Kishi, A. Takamura, Y. Miura, S. Iemura, T. Natsume, K. Takehana, N. Yamada, J. L. Guan, N. Oshiro, and N. Mizushima Nutrient-dependent mtorc1 association with the ULK1-Atg13-FIP200 complex required for autophagy. Mol Biol Cell 20: Jung, C. H., C. B. Jun, S. H. Ro, Y. M. Kim, N. M. Otto, J. Cao, M. Kundu, and D. H. Kim ULK- Atg13-FIP200 complexes mediate mtor signaling to the autophagy machinery. Mol Biol Cell 20: Kim, J., M. Kundu, B. Viollet, and K. L. Guan AMPK and mtor regulate autophagy through direct phosphorylation of Ulk1. Nat Cell Biol 13: Russell, R. C., Y. Tian, H. Yuan, H. W. Park, Y. Y. Chang, J. Kim, H. Kim, T. P. Neufeld, A. Dillin, and K. L. Guan ULK1 induces autophagy by phosphorylating Beclin-1 and activating VPS34 lipid kinase. Nat Cell Biol 15: Lavallard, V. J., A. J. Meijer, P. Codogno, and P. Gual Autophagy, signaling and obesity. Pharmacol Res 66: Pankiv, S., T. H. Clausen, T. Lamark, A. Brech, J. A. Bruun, H. Outzen, A. Overvatn, G. Bjorkoy, and T. Johansen p62/sqstm1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy. J Biol Chem 282: Czaja, M. J Autophagy in health and disease. 2. Regulation of lipid metabolism and storage by autophagy: pathophysiological implications. American journal of physiology. Cell physiology 298: C Kaushik, S., and A. M. Cuervo Degradation of lipid droplet-associated proteins by chaperone-mediated autophagy facilitates lipolysis. Nat Cell Biol 17: Lizaso, A., K. T. Tan, and Y. H. Lee beta-adrenergic receptor-stimulated lipolysis requires the RAB7-mediated autolysosomal lipid degradation. Autophagy 9: Schroeder, B., R. J. Schulze, S. G. Weller, A. C. Sletten, C. A. Casey, and M. A. McNiven The small GTPase Rab7 as a central regulator of hepatocellular lipophagy. Hepatology 61: Settembre, C., R. De Cegli, G. Mansueto, P. K. Saha, F. Vetrini, O. Visvikis, T. Huynh, A. Carissimo, D. Palmer, T. J. Klisch, A. C. Wollenberg, D. Di Bernardo, L. Chan, J. E. Irazoqui, and A. Ballabio

35 35 TFEB controls cellular lipid metabolism through a starvation-induced autoregulatory loop. Nat Cell Biol 15: Seok, S., T. Fu, S. E. Choi, Y. Li, R. Zhu, S. Kumar, X. Sun, G. Yoon, Y. Kang, W. Zhong, J. Ma, B. Kemper, and J. K. Kemper Transcriptional regulation of autophagy by an FXR-CREB axis. Nature 516: Lettieri Barbato, D., G. Tatulli, K. Aquilano, and M. R. Ciriolo FoxO1 controls lysosomal acid lipase in adipocytes: implication of lipophagy during nutrient restriction and metformin treatment. Cell Death Dis 4: e Rambold, A. S., S. Cohen, and J. Lippincott-Schwartz Fatty acid trafficking in starved cells: regulation by lipid droplet lipolysis, autophagy, and mitochondrial fusion dynamics. Dev Cell 32: Shibata, M., K. Yoshimura, H. Tamura, T. Ueno, T. Nishimura, T. Inoue, M. Sasaki, M. Koike, H. Arai, E. Kominami, and Y. Uchiyama LC3, a microtubule-associated protein1a/b light chain3, is involved in cytoplasmic lipid droplet formation. Biochem Biophys Res Commun 393: Mao, Y., J. Cheng, F. Yu, H. Li, C. Guo, and X. Fan Ghrelin Attenuated Lipotoxicity via Autophagy Induction and Nuclear Factor-kappaB Inhibition. Cell Physiol Biochem 37: Wang, Y., R. Singh, Y. Xiang, and M. J. Czaja Macroautophagy and chaperone-mediated autophagy are required for hepatocyte resistance to oxidant stress. Hepatology 52: Skop, V., M. Cahova, Z. Papackova, E. Palenickova, H. Dankova, M. Baranowski, P. Zabielski, J. Zdychova, J. Zidkova, and L. Kazdova Autophagy-lysosomal pathway is involved in lipid degradation in rat liver. Physiol Res 61: Singh, R., Y. Xiang, Y. Wang, K. Baikati, A. M. Cuervo, Y. K. Luu, Y. Tang, J. E. Pessin, G. J. Schwartz, and M. J. Czaja Autophagy regulates adipose mass and differentiation in mice. J Clin Invest 119: Guo, L., J. X. Huang, Y. Liu, X. Li, S. R. Zhou, S. W. Qian, Y. Liu, H. Zhu, H. Y. Huang, Y. J. Dang, and Q. Q. Tang Transactivation of Atg4b by C/EBPbeta promotes autophagy to facilitate adipogenesis. Mol Cell Biol 33: Yin, J., Y. Wang, L. Gu, N. Fan, Y. Ma, and Y. Peng Palmitate induces endoplasmic reticulum stress and autophagy in mature adipocytes: implications for apoptosis and inflammation. Int J Mol Med 35: Jansen, H. J., P. van Essen, T. Koenen, L. A. Joosten, M. G. Netea, C. J. Tack, and R. Stienstra Autophagy activity is up-regulated in adipose tissue of obese individuals and modulates proinflammatory cytokine expression. Endocrinology 153: Soussi, H., S. Reggio, R. Alili, C. Prado, S. Mutel, M. Pini, C. Rouault, K. Clement, and I. Dugail DAPK2 Downregulation Associates With Attenuated Adipocyte Autophagic Clearance in Human Obesity. Diabetes 64: Kaushik, S., J. A. Rodriguez-Navarro, E. Arias, R. Kiffin, S. Sahu, G. J. Schwartz, A. M. Cuervo, and R. Singh Autophagy in hypothalamic AgRP neurons regulates food intake and energy balance. Cell Metab 14: Meng, Q., and D. Cai Defective hypothalamic autophagy directs the central pathogenesis of obesity via the IkappaB kinase beta (IKKbeta)/NF-kappaB pathway. J Biol Chem 286: Sandri, M Autophagy in skeletal muscle. FEBS Lett 584: Nishida, K., S. Kyoi, O. Yamaguchi, J. Sadoshima, and K. Otsu The role of autophagy in the heart. Cell Death Differ 16: Ouimet, M., V. Franklin, E. Mak, X. Liao, I. Tabas, and Y. L. Marcel Autophagy regulates cholesterol efflux from macrophage foam cells via lysosomal acid lipase. Cell Metab 13: Xu, K., Y. Yang, M. Yan, J. Zhan, X. Fu, and X. Zheng Autophagy plays a protective role in free cholesterol overload-induced death of smooth muscle cells. J Lipid Res 51:

36 Dall'Armi, C., K. A. Devereaux, and G. Di Paolo The role of lipids in the control of autophagy. Curr Biol 23: R Shpilka, T., E. Welter, N. Borovsky, N. Amar, M. Mari, F. Reggiori, and Z. Elazar Lipid droplets and their component triglycerides and steryl esters regulate autophagosome biogenesis. EMBO J 34: Choi, S. E., S. M. Lee, Y. J. Lee, L. J. Li, S. J. Lee, J. H. Lee, Y. Kim, H. S. Jun, K. W. Lee, and Y. Kang Protective role of autophagy in palmitate-induced INS-1 beta-cell death. Endocrinology 150: Ebato, C., T. Uchida, M. Arakawa, M. Komatsu, T. Ueno, K. Komiya, K. Azuma, T. Hirose, K. Tanaka, E. Kominami, R. Kawamori, Y. Fujitani, and H. Watada Autophagy is important in islet homeostasis and compensatory increase of beta cell mass in response to high-fat diet. Cell Metab 8: Chu, K. Y., L. O'Reilly, G. Ramm, and T. J. Biden High-fat diet increases autophagic flux in pancreatic beta cells in vivo and ex vivo in mice. Diabetologia 58: Las, G., S. B. Serada, J. D. Wikstrom, G. Twig, and O. S. Shirihai Fatty acids suppress autophagic turnover in beta-cells. J Biol Chem 286: Mir, S. U., N. M. George, L. Zahoor, R. Harms, Z. Guinn, and N. E. Sarvetnick Inhibition of autophagic turnover in beta-cells by fatty acids and glucose leads to apoptotic cell death. J Biol Chem 290: Maedler, K., G. A. Spinas, D. Dyntar, W. Moritz, N. Kaiser, and M. Y. Donath Distinct effects of saturated and monounsaturated fatty acids on beta-cell turnover and function. Diabetes 50: Oh, Y. S Mechanistic insights into pancreatic beta-cell mass regulation by glucose and free fatty acids. Anat Cell Biol 48: Korsheninnikova, E., G. C. van der Zon, P. J. Voshol, G. M. Janssen, L. M. Havekes, A. Grefhorst, F. Kuipers, D. J. Reijngoud, J. A. Romijn, D. M. Ouwens, and J. A. Maassen Sustained activation of the mammalian target of rapamycin nutrient sensing pathway is associated with hepatic insulin resistance, but not with steatosis, in mice. Diabetologia 49: Koga, H., S. Kaushik, and A. M. Cuervo Altered lipid content inhibits autophagic vesicular fusion. FASEB J 24: Liu, H. Y., J. Han, S. Y. Cao, T. Hong, D. Zhuo, J. Shi, Z. Liu, and W. Cao Hepatic autophagy is suppressed in the presence of insulin resistance and hyperinsulinemia: inhibition of FoxO1-dependent expression of key autophagy genes by insulin. J Biol Chem 284: Yang, L., P. Li, S. Fu, E. S. Calay, and G. S. Hotamisligil Defective hepatic autophagy in obesity promotes ER stress and causes insulin resistance. Cell Metab 11: Schaffer, J. E Lipotoxicity: when tissues overeat. Curr Opin Lipidol 14: Badin, P. M., I. K. Vila, K. Louche, A. Mairal, M. A. Marques, V. Bourlier, G. Tavernier, D. Langin, and C. Moro High-fat diet-mediated lipotoxicity and insulin resistance is related to impaired lipase expression in mouse skeletal muscle. Endocrinology 154: Yuzefovych, L. V., S. I. Musiyenko, G. L. Wilson, and L. I. Rachek Mitochondrial DNA damage and dysfunction, and oxidative stress are associated with endoplasmic reticulum stress, protein degradation and apoptosis in high fat diet-induced insulin resistance mice. PloS one 8: e Borradaile, N. M., and J. E. Schaffer Lipotoxicity in the heart. Curr Hypertens Rep 7: Campbell, T. L., A. S. Mitchell, E. M. McMillan, D. Bloemberg, D. Pavlov, I. Messa, J. G. Mielke, and J. Quadrilatero High-fat feeding does not induce an autophagic or apoptotic phenotype in female rat skeletal muscle. Exp Biol Med (Maywood) 240:

37 Turpin, S. M., J. G. Ryall, R. Southgate, I. Darby, A. L. Hevener, M. A. Febbraio, B. E. Kemp, G. S. Lynch, and M. J. Watt Examination of 'lipotoxicity' in skeletal muscle of high-fat fed and ob/ob mice. J Physiol 587: Moresi, V., M. Carrer, C. E. Grueter, O. F. Rifki, J. M. Shelton, J. A. Richardson, R. Bassel-Duby, and E. N. Olson Histone deacetylases 1 and 2 regulate autophagy flux and skeletal muscle homeostasis in mice. Proc Natl Acad Sci U S A 109: Bollinger, L. M., J. J. Powell, J. A. Houmard, C. A. Witczak, and J. J. Brault Skeletal muscle myotubes in severe obesity exhibit altered ubiquitin-proteasome and autophagic/lysosomal proteolytic flux. Obesity (Silver Spring) 23: Kim, K. H., Y. T. Jeong, H. Oh, S. H. Kim, J. M. Cho, Y. N. Kim, S. S. Kim, H. Kim do, K. Y. Hur, H. K. Kim, T. Ko, J. Han, H. L. Kim, J. Kim, S. H. Back, M. Komatsu, H. Chen, D. C. Chan, M. Konishi, N. Itoh, C. S. Choi, and M. S. Lee Autophagy deficiency leads to protection from obesity and insulin resistance by inducing Fgf21 as a mitokine. Nature medicine 19: Mammucari, C., G. Milan, V. Romanello, E. Masiero, R. Rudolf, P. Del Piccolo, S. J. Burden, R. Di Lisi, C. Sandri, J. Zhao, A. L. Goldberg, S. Schiaffino, and M. Sandri FoxO3 controls autophagy in skeletal muscle in vivo. Cell Metab 6: Woodworth-Hobbs, M. E., M. B. Hudson, J. A. Rahnert, B. Zheng, H. A. Franch, and S. R. Price Docosahexaenoic acid prevents palmitate-induced activation of proteolytic systems in C2C12 myotubes. J Nutr Biochem 25: Nakai, A., O. Yamaguchi, T. Takeda, Y. Higuchi, S. Hikoso, M. Taniike, S. Omiya, I. Mizote, Y. Matsumura, M. Asahi, K. Nishida, M. Hori, N. Mizushima, and K. Otsu The role of autophagy in cardiomyocytes in the basal state and in response to hemodynamic stress. Nat Med 13: He, C., M. C. Bassik, V. Moresi, K. Sun, Y. Wei, Z. Zou, Z. An, J. Loh, J. Fisher, Q. Sun, S. Korsmeyer, M. Packer, H. I. May, J. A. Hill, H. W. Virgin, C. Gilpin, G. Xiao, R. Bassel-Duby, P. E. Scherer, and B. Levine Exercise-induced BCL2-regulated autophagy is required for muscle glucose homeostasis. Nature 481: Li, Z. L., J. R. Woollard, B. Ebrahimi, J. A. Crane, K. L. Jordan, A. Lerman, S. M. Wang, and L. O. Lerman Transition from obesity to metabolic syndrome is associated with altered myocardial autophagy and apoptosis. Arterioscler Thromb Vasc Biol 32: Hariharan, N., Y. Maejima, J. Nakae, J. Paik, R. A. Depinho, and J. Sadoshima Deacetylation of FoxO by Sirt1 Plays an Essential Role in Mediating Starvation-Induced Autophagy in Cardiac Myocytes. Circ Res 107: Xu, X., Y. Hua, S. Nair, Y. Zhang, and J. Ren Akt2 knockout preserves cardiac function in high-fat diet-induced obesity by rescuing cardiac autophagosome maturation. J Mol Cell Biol 5: Hsu, H. C., C. Y. Chen, B. C. Lee, and M. F. Chen High-fat diet induces cardiomyocyte apoptosis via the inhibition of autophagy. Eur J Nutr. 89. Kandadi, M. R., E. Panzhinskiy, N. D. Roe, S. Nair, D. Hu, and A. Sun Deletion of protein tyrosine phosphatase 1B rescues against myocardial anomalies in high fat diet-induced obesity: Role of AMPK-dependent autophagy. Biochim Biophys Acta 1852: Russo, S. B., C. F. Baicu, A. Van Laer, T. Geng, H. Kasiganesan, M. R. Zile, and L. A. Cowart Ceramide synthase 5 mediates lipid-induced autophagy and hypertrophy in cardiomyocytes. J Clin Invest 122: Jaishy, B., Q. Zhang, H. S. Chung, C. Riehle, J. Soto, S. Jenkins, P. Abel, L. A. Cowart, J. E. Van Eyk, and E. D. Abel Lipid-induced NOX2 activation inhibits autophagic flux by impairing lysosomal enzyme activity. J Lipid Res 56: Liu, J., F. Chang, F. Li, H. Fu, J. Wang, S. Zhang, J. Zhao, and D. Yin Palmitate promotes autophagy and apoptosis through ROS-dependent JNK and p38 MAPK. Biochem Biophys Res Commun 463:

38 Boya, P Lysosomal function and dysfunction: mechanism and disease. Antioxid Redox Signal 17: Williams, K. J., and K. Chen Recent insights into factors affecting remnant lipoprotein uptake. Curr Opin Lipidol 21: Niemeier, A., D. Niedzielska, R. Secer, A. Schilling, M. Merkel, C. Enrich, P. C. Rensen, and J. Heeren Uptake of postprandial lipoproteins into bone in vivo: impact on osteoblast function. Bone 43: Jones, A. L., G. T. Hradek, C. Hornick, G. Renaud, E. E. Windler, and R. J. Havel Uptake and processing of remnants of chylomicrons and very low density lipoproteins by rat liver. J Lipid Res 25: Brown, M. S., and J. L. Goldstein A receptor-mediated pathway for cholesterol homeostasis. Science 232: Heeren, J., U. Beisiegel, and T. Grewal Apolipoprotein E recycling: implications for dyslipidemia and atherosclerosis. Arterioscler Thromb Vasc Biol 26: Goldstein, J. L., S. E. Dana, J. R. Faust, A. L. Beaudet, and M. S. Brown Role of lysosomal acid lipase in the metabolism of plasma low density lipoprotein. Observations in cultured fibroblasts from a patient with cholesteryl ester storage disease. J Biol Chem 250: Slotte, J. P., and S. Ekman Synthesis and hydrolysis of cholesteryl esters by isolated ratliver lysosomes and cell-free extracts of human lung fibroblasts. Biochim Biophys Acta 879: Wang, Y., A. B. Castoreno, W. Stockinger, and A. Nohturfft Modulation of endosomal cholesteryl ester metabolism by membrane cholesterol. J Biol Chem 280: Anderson, R. A., R. S. Byrum, P. M. Coates, and G. N. Sando Mutations at the lysosomal acid cholesteryl ester hydrolase gene locus in Wolman disease. Proc Natl Acad Sci U S A 91: Kunze, H., B. Hesse, and E. Bohn Hydrolytic degradation of phosphatidylethanolamine and phosphatidylcholine by isolated rat-liver lysosomes. Biochim Biophys Acta 711: Shayman, J. A., R. Kelly, J. Kollmeyer, Y. He, and A. Abe Group XV phospholipase A(2), a lysosomal phospholipase A(2). Prog Lipid Res 50: Matsuzawa, Y., and K. Y. Hostetler Properties of phospholipase C isolated from rat liver lysosomes. J Biol Chem 255: Brown, F. D., N. Thompson, K. M. Saqib, J. M. Clark, D. Powner, N. T. Thompson, R. Solari, and M. J. Wakelam Phospholipase D1 localises to secretory granules and lysosomes and is plasmamembrane translocated on cellular stimulation. Curr Biol 8: Kolter, T., and K. Sandhoff Principles of lysosomal membrane digestion: stimulation of sphingolipid degradation by sphingolipid activator proteins and anionic lysosomal lipids. Annu Rev Cell Dev Biol 21: Zoncu, R., L. Bar-Peled, A. Efeyan, S. Wang, Y. Sancak, and D. M. Sabatini mtorc1 senses lysosomal amino acids through an inside-out mechanism that requires the vacuolar H(+)-ATPase. Science 334: Rebsamen, M., L. Pochini, T. Stasyk, M. E. de Araujo, M. Galluccio, R. K. Kandasamy, B. Snijder, A. Fauster, E. L. Rudashevskaya, M. Bruckner, S. Scorzoni, P. A. Filipek, K. V. Huber, J. W. Bigenzahn, L. X. Heinz, C. Kraft, K. L. Bennett, C. Indiveri, L. A. Huber, and G. Superti-Furga SLC38A9 is a component of the lysosomal amino acid sensing machinery that controls mtorc1. Nature 519: Bar-Peled, L., and D. M. Sabatini SnapShot: mtorc1 signaling at the lysosomal surface. Cell 151: e Settembre, C., R. Zoncu, D. L. Medina, F. Vetrini, S. Erdin, S. Erdin, T. Huynh, M. Ferron, G. Karsenty, M. C. Vellard, V. Facchinetti, D. M. Sabatini, and A. Ballabio A lysosome-to-nucleus signalling mechanism senses and regulates the lysosome via mtor and TFEB. EMBO J 31:

39 Sengupta, S., T. R. Peterson, M. Laplante, S. Oh, and D. M. Sabatini mtorc1 controls fasting-induced ketogenesis and its modulation by ageing. Nature 468: Settembre, C., C. Di Malta, V. A. Polito, M. Garcia Arencibia, F. Vetrini, S. Erdin, S. U. Erdin, T. Huynh, D. Medina, P. Colella, M. Sardiello, D. C. Rubinsztein, and A. Ballabio TFEB links autophagy to lysosomal biogenesis. Science 332: Palmieri, M., S. Impey, H. Kang, A. di Ronza, C. Pelz, M. Sardiello, and A. Ballabio Characterization of the CLEAR network reveals an integrated control of cellular clearance pathways. Hum Mol Genet 20: Braulke, T., and J. S. Bonifacino Sorting of lysosomal proteins. Biochim Biophys Acta 1793: Schulze, H., and K. Sandhoff Lysosomal lipid storage diseases. Cold Spring Harb Perspect Biol Pentchev, P. G., M. E. Comly, H. S. Kruth, M. T. Vanier, D. A. Wenger, S. Patel, and R. O. Brady A defect in cholesterol esterification in Niemann-Pick disease (type C) patients. Proc Natl Acad Sci U S A 82: Sleat, D. E., J. A. Wiseman, M. El-Banna, S. M. Price, L. Verot, M. M. Shen, G. S. Tint, M. T. Vanier, S. U. Walkley, and P. Lobel Genetic evidence for nonredundant functional cooperativity between NPC1 and NPC2 in lipid transport. Proc Natl Acad Sci U S A 101: Sobo, K., I. Le Blanc, P. P. Luyet, M. Fivaz, C. Ferguson, R. G. Parton, J. Gruenberg, and F. G. van der Goot Late endosomal cholesterol accumulation leads to impaired intra-endosomal trafficking. PLoS One 2: e Ikonen, E Cellular cholesterol trafficking and compartmentalization. Nature reviews. Molecular cell biology 9: Choudhury, A., D. K. Sharma, D. L. Marks, and R. E. Pagano Elevated endosomal cholesterol levels in Niemann-Pick cells inhibit rab4 and perturb membrane recycling. Molecular biology of the cell 15: Puri, V., R. Watanabe, M. Dominguez, X. Sun, C. L. Wheatley, D. L. Marks, and R. E. Pagano Cholesterol modulates membrane traffic along the endocytic pathway in sphingolipid-storage diseases. Nature cell biology 1: Fraldi, A., F. Annunziata, A. Lombardi, H. J. Kaiser, D. L. Medina, C. Spampanato, A. O. Fedele, R. Polishchuk, N. C. Sorrentino, K. Simons, and A. Ballabio Lysosomal fusion and SNARE function are impaired by cholesterol accumulation in lysosomal storage disorders. EMBO J 29: Fukuda, T., L. Ewan, M. Bauer, R. J. Mattaliano, K. Zaal, E. Ralston, P. H. Plotz, and N. Raben Dysfunction of endocytic and autophagic pathways in a lysosomal storage disease. Ann Neurol 59: Settembre, C., A. Fraldi, L. Jahreiss, C. Spampanato, C. Venturi, D. Medina, R. de Pablo, C. Tacchetti, D. C. Rubinsztein, and A. Ballabio A block of autophagy in lysosomal storage disorders. Hum Mol Genet 17: Sarkar, S., B. Carroll, Y. Buganim, D. Maetzel, A. H. Ng, J. P. Cassady, M. A. Cohen, S. Chakraborty, H. Wang, E. Spooner, H. Ploegh, J. Gsponer, V. I. Korolchuk, and R. Jaenisch Impaired autophagy in the lipid-storage disorder Niemann-Pick type C1 disease. Cell Rep 5: Cheng, J., Y. Ohsaki, K. Tauchi-Sato, A. Fujita, and T. Fujimoto Cholesterol depletion induces autophagy. Biochem Biophys Res Commun 351: Vilimanovich, U., M. Bosnjak, A. Bogdanovic, I. Markovic, A. Isakovic, T. Kravic-Stevovic, A. Mircic, V. Trajkovic, and V. Bumbasirevic Statin-mediated inhibition of cholesterol synthesis induces cytoprotective autophagy in human leukemic cells. Eur J Pharmacol 765:

40 Rodriguez-Navarro, J. A., S. Kaushik, H. Koga, C. Dall'Armi, G. Shui, M. R. Wenk, G. Di Paolo, and A. M. Cuervo Inhibitory effect of dietary lipids on chaperone-mediated autophagy. Proc Natl Acad Sci U S A 109: E Decleves, A. E., Z. Zolkipli, J. Satriano, L. Wang, T. Nakayama, M. Rogac, T. P. Le, J. L. Nortier, M. G. Farquhar, R. K. Naviaux, and K. Sharma Regulation of lipid accumulation by AMP-activated kinase [corrected] in high fat diet-induced kidney injury. Kidney Int 85: Bucci, C., P. Thomsen, P. Nicoziani, J. McCarthy, and B. van Deurs Rab7: a key to lysosome biogenesis. Molecular biology of the cell 11: Feldstein, A. E., N. W. Werneburg, Z. Li, S. F. Bronk, and G. J. Gores Bax inhibition protects against free fatty acid-induced lysosomal permeabilization. Am J Physiol Gastrointest Liver Physiol 290: G Paris, C., J. Bertoglio, and J. Breard Lysosomal and mitochondrial pathways in miltefosineinduced apoptosis in U937 cells. Apoptosis : an international journal on programmed cell death 12: Gornicka, A., J. Fettig, A. Eguchi, M. P. Berk, S. Thapaliya, L. J. Dixon, and A. E. Feldstein Adipocyte hypertrophy is associated with lysosomal permeability both in vivo and in vitro: role in adipose tissue inflammation. American journal of physiology. Endocrinology and metabolism 303: E Fucho, R., L. Martinez, A. Baulies, S. Torres, N. Tarrats, A. Fernandez, V. Ribas, A. M. Astudillo, J. Balsinde, P. Garcia-Roves, M. Elena, I. Bergheim, S. Lotersztajn, C. Trautwein, H. Appelqvist, A. W. Paton, J. C. Paton, M. J. Czaja, N. Kaplowitz, J. C. Fernandez-Checa, and C. Garcia-Ruiz ASMase regulates autophagy and lysosomal membrane permeabilization and its inhibition prevents early stage nonalcoholic steatohepatitis. Journal of hepatology 61: Hua, Y., Y. Zhang, J. Dolence, G. P. Shi, J. Ren, and S. Nair Cathepsin K knockout mitigates high-fat diet-induced cardiac hypertrophy and contractile dysfunction. Diabetes 62: Emanuel, R., I. Sergin, S. Bhattacharya, J. N. Turner, S. Epelman, C. Settembre, A. Diwan, A. Ballabio, and B. Razani Induction of lysosomal biogenesis in atherosclerotic macrophages can rescue lipid-induced lysosomal dysfunction and downstream sequelae. Arterioscler Thromb Vasc Biol 34: Feldstein, A. E., N. W. Werneburg, A. Canbay, M. E. Guicciardi, S. F. Bronk, R. Rydzewski, L. J. Burgart, and G. J. Gores Free fatty acids promote hepatic lipotoxicity by stimulating TNF-alpha expression via a lysosomal pathway. Hepatology 40: Guicciardi, M. E., M. Leist, and G. J. Gores Lysosomes in cell death. Oncogene 23: Alkhouri, N., C. Carter-Kent, and A. E. Feldstein Apoptosis in nonalcoholic fatty liver disease: diagnostic and therapeutic implications. Expert review of gastroenterology & hepatology 5: Radisky, D. C., and J. Kaplan Iron in cytosolic ferritin can be recycled through lysosomal degradation in human fibroblasts. The Biochemical journal 336 ( Pt 1): Sakaida, I., M. E. Kyle, and J. L. Farber Autophagic degradation of protein generates a pool of ferric iron required for the killing of cultured hepatocytes by an oxidative stress. Molecular pharmacology 37: Kurz, T., A. Terman, and U. T. Brunk Autophagy, ageing and apoptosis: the role of oxidative stress and lysosomal iron. Archives of biochemistry and biophysics 462: Pivtoraiko, V. N., S. L. Stone, K. A. Roth, and J. J. Shacka Oxidative stress and autophagy in the regulation of lysosome-dependent neuron death. Antioxidants & redox signaling 11: Lamb, F. S., J. G. Moreland, and F. J. Miller, Jr Electrophysiology of reactive oxygen production in signaling endosomes. Antioxidants & redox signaling 11:

41 Jiang, F., H. K. Lim, M. J. Morris, L. Prior, E. Velkoska, X. Wu, and G. J. Dusting Systemic upregulation of NADPH oxidase in diet-induced obesity in rats. Redox report : communications in free radical research 16: Lynch, C. M., D. A. Kinzenbaw, X. Chen, S. Zhan, E. Mezzetti, J. Filosa, A. Ergul, J. L. Faulkner, F. M. Faraci, and S. P. Didion Nox2-derived superoxide contributes to cerebral vascular dysfunction in diet-induced obesity. Stroke; a journal of cerebral circulation 44: Fortuno, A., G. San Jose, M. U. Moreno, O. Beloqui, J. Diez, and G. Zalba Phagocytic NADPH oxidase overactivity underlies oxidative stress in metabolic syndrome. Diabetes 55: Feng, Y., and M. Forgac Inhibition of vacuolar H(+)-ATPase by disulfide bond formation between cysteine 254 and cysteine 532 in subunit A. J Biol Chem 269: Nishi, T., and M. Forgac The vacuolar (H+)-ATPases--nature's most versatile proton pumps. Nature reviews. Molecular cell biology 3: Pal, R., M. Palmieri, J. A. Loehr, S. Li, R. Abo-Zahrah, T. O. Monroe, P. B. Thakur, M. Sardiello, and G. G. Rodney Src-dependent impairment of autophagy by oxidative stress in a mouse model of Duchenne muscular dystrophy. Nature communications 5: Coll, T., E. Eyre, R. Rodriguez-Calvo, X. Palomer, R. M. Sanchez, M. Merlos, J. C. Laguna, and M. Vazquez-Carrera Oleate reverses palmitate-induced insulin resistance and inflammation in skeletal muscle cells. J Biol Chem 283: Listenberger, L. L., X. Han, S. E. Lewis, S. Cases, R. V. Farese, Jr., D. S. Ory, and J. E. Schaffer Triglyceride accumulation protects against fatty acid-induced lipotoxicity. Proceedings of the National Academy of Sciences of the United States of America 100: Kaushik, S., A. C. Massey, and A. M. Cuervo Lysosome membrane lipid microdomains: novel regulators of chaperone-mediated autophagy. The EMBO journal 25: Xu, H., and D. Ren Lysosomal physiology. Annual review of physiology 77: Yasuda, M., Y. Tanaka, S. Kume, Y. Morita, M. Chin-Kanasaki, H. Araki, K. Isshiki, S. Araki, D. Koya, M. Haneda, A. Kashiwagi, H. Maegawa, and T. Uzu Fatty acids are novel nutrient factors to regulate mtorc1 lysosomal localization and apoptosis in podocytes. Biochim Biophys Acta 1842: Settembre, C., A. Fraldi, D. L. Medina, and A. Ballabio Signals from the lysosome: a control centre for cellular clearance and energy metabolism. Nat Rev Mol Cell Biol 14: Miyagawa, K., S. Oe, Y. Honma, H. Izumi, R. Baba, and M. Harada Lipid-Induced Endoplasmic Reticulum Stress Impairs Selective Autophagy at the Step of Autophagosome-Lysosome Fusion in Hepatocytes. Am J Pathol. Figure legends: Fig. 1. Overview of dietary triglyceride metabolism. Dietary lipids enter the circulation by three major pathways. (1) Pancreatic lipases digest fats to generate free fatty acids (FFA) and glycerol which are reesterified and released into the circulation packaged into chylomicrons (CM). (2) the liver synthesizes very low-density lipoprotein (VLDL) from

42 42 triglycerides (TG) and cholesteryl esters (CE) and releases them into the circulation. (3) Adipose tissue TG hydrolysis generates FFA which are released into the blood stream bound to albumin. Hydrolysis of CM and VLDL by lipoprotein lipase (LPL) at the endothelial surface generates FFA that enter the peripheral tissue via fatty acid transporters CD36/FATP. The CM and VLDL remnants generated from LPL-mediated hydrolysis are taken up by liver for further processing. In the fed state, FFA are esterified into TG and stored as LDs in adipose tissue and liver or undergo β-oxidation to generate ATP in muscle and other peripheral tissues. In addition, de novo lipogenesis from glucose also generates FAA in the fed state. In the fasted state when lipid supply dwindles, TG hydrolysis generates FFA that are released into the circulation by adipose tissue or used to generate ketone bodies in liver. Albumin-bound FFA (Alb-FFA) and ketone bodies serve as the major source of energy during fasting. Fig. 2. Fatty acid overload-induced oxidative stress and lipotoxicity. Oversupply of FFA may overwhelm mitochondrial capacity to oxidize fatty acids for ATP production, which leads to accumulation of toxic lipid intermediates such as diacyl glycerol (DAG), ceramides and acyl carnitines. Toxic lipid intermediates impair insulin signaling through insulin receptor substrates and blunt insulin-stimulated glucose uptake. DAG also activates NADPH oxidase (NOX) via protein kinase C (PKC) to increase intracellular reactive oxygen species (ROS) production. In addition, dysregulation of mitochondrial electron transport chain (ETC) due to excessive FA β oxidation promotes mitochondrial ROS production. ROS-induced oxidative stress coupled with reduced expression or activity of antioxidant enzymes eventually leads to cellular lipotoxicity manifested by lipid peroxidation, DNA damage, mitochondrial and lysosomal dysfunction, defective

43 43 autophagy, and activation of inflammatory responses in multiple tissues. FA-CoA, fatty acyl-coa; CPT I and CPT II, carnitine palmitoyltransferase I and II; NADH, reduced nicotinamide adenine dinucleotide; FADH 2, reduced flavin adenine dinucleotide. Fig. 3. Molecular mechanism of mammalian autophagy. Under nutrient rich conditions, mtorc1 binds and inactivates Ulk1/2 complex by inhibitory phosphorylation to suppress autophagy. An autophagy stimulus such as starvation relieves inhibition of Ulk1/2 complex by mtorc1. Activation of AMPK upon starvation inhibits mtorc1 and further activates the Ulk1/2 complex to initiate autophagy. The Ulk1/2 complex in turn activates Beclin1-Vps34 (PI3KC3) complex either by phosphorylating Beclin1 or through Ambra1 phosphorylation. The kinase activity of Vps34 is regulated by Beclin1, which in turn is controlled by its inhibitory interaction with Bcl-2. The catalytic activity of Vps34 results in the formation of PI3P: a key lipid molecule that recruits specific membrane and protein components to the growing isolation membrane (phagophore). In addition, autophagosome expansion and maturation requires two ubiquitin-like conjugation systems that results in ATG16-ATG5-ATG12 complex and lipidation of LC3- I to LC3-II. LC3-II is required for the selection of autophagic substrate, determining autophagosome size and membrane curvature, and for the bidirectional movement of autophagosomes towards lysosomes for fusion. Mature autophagosomes fuse with lysosomes to form autolysosomes. The autophagic substrates are degraded by lysosomal hydrolases into simple substrates which are utilized for energy production and biomolecule synthesis to maintain cellular homeostasis. Fig. 4. Degradation of lipid droplets (LDs) via autophagy. Lipid droplets, containing mostly triglycerides (TG) and cholesteryl esters (CE) at the core, undergo chaperone-

44 44 mediated autophagy (CMA) of their structural membrane proteins perilipin 2 and 3 (PLIN2 and PLIN3). PLIN removal activates RAB7 at the exposed LD surface. RAB7 interacts with with autophagy proteins such as p62 and LC3-II to facilitate the engulfment of the LD by the growing autophagosome. LDs are sequestered by autophagosomes either selectively via lipophagy (A and B) or with other autophagic substrates via non-selective macroautophagy (C). Autophagosomes fuse with lysosomes to form autolysosomes wherein lysosomal hydrolases such as lysosomal acid lipases degrade LD TG and CE into free fatty acids (FFA) that are recycled back into the cytosol to support various energetic and structural requirements of the cell. Fig. 5. Lysosomal stress-response pathways induced by dietary lipid overload. Overload of free fatty acid (FFA) supplied exogenously or derived from triglyceride (TG) hydrolysis induces ROS production from mitochondrial electron transport chain complexes, the NADPH oxidase 2 (NOX2) complex, and other reactive oxygen species (ROS) producing enzymes. The ROS molecules hydrogen peroxide (H 2 O 2 ) and superoxide anion (O - 2 ) undergo Fenton s reaction in lysosomes to generate hydroxyl radical (OH ), a highly reactive ROS. ROS induced peroxidation of lysosomal membrane lipids leads to lysosomal membrane permeabilization (LMP) and release of cathepsins into the cytosol. Cytosolic cathepsins induces apoptotic cell death either by direct activation caspases or through mitochondrial membrane permeabilization (MMP)- mediated caspase activation. ROS-induced protein oxidation impairs the function several lysosomal enzymes such as vacuolar ATPase (vatpase) proton pump and lysosomal hydrolases, which raises lysosomal ph and inhibits degradation of autophagic cargo, respectively. Lipid overload also induces lysosomal dysfunction by

45 45 ROS-independent pathways. Incubation with excess FFAs alters the membrane composition of lysosomes and autophagosomes that impairs heterotypic fusion between the two organelles. Macrophages loaded with atherogenic lipids exhibited an increase in lysosomal ph that contributed to a defect in cholesterol efflux that also inhibits chaperone mediated autophagy (CMA) by promoting selective degradation of the lysosome associated membrane protein 2A (LAMP2A). Free fatty acid (FFA)-induced translocation of mtor complex 1 (mtorc1) to the lysosomal membrane has been shown to promote endoplasmic reticulum (ER) stress-dependent apoptosis in podocytes.

46 46 Table 1. Lipid-overload induced regulation of autophagy in different metabolic tissues. (WT= wildtype) Study Model Pancreas Wildtype (WT) or db/db mice Diet or Treatment 60% HFD for 12 weeks Effect on autophagy Induced Observation (Reference) Autophagy maintained beta cell mass, insulin secretion, and normal glucose tolerance (44). INS1 or MIN6 beta cells Liver WT, ob/ob and liver-atg7 null mice 0.4 mm Palmitate and 20 mm Glucose for 14 h or longer 35% HFD for 22 weeks Suppressed Suppressed Impaired autophagic flux led to defective insulin secretion and apoptosis (46, 47). Autophagy suppression increased ER stress and insulin resistance (51). Skeletal muscle WT, Adiponectinnull and musle- HDAC 1/2 null mice WT and muscle- Atg7 null mice Heart WT and Akt2-null mice WT mice 60% HFD for 2 to 12 weeks 60% HFD for 13 weeks 45% HFD for 20 or 25 weeks 60% Milk fat-based diet for 18 weeks Induced Suppressed in Atg7 null mice Suppressed Induced Autophagy induction parallels upregulation of antioxidant genes and ameliorates insulin-resistance and myopathy (60,61) Suppression of autophagy protects from HFD-induced obesity and insulin-resistance (63). Impaired autophagic flux resulted in cardiac hypertrophy, contractile dysfunction, ER stress and apoptosis (70,71). De novo sphingolipid synthesis induced autophagy and cardiomyocytes hypertrophy and contractile dysfunction (73).

47 47 Table 2. Changes in lysosomal function associated with lipid overload. Study Model Effect on lysosomes Effect on autophagy Observation (Reference) Mouse models of Defective endocytic transport. Suppressed turnover due Accumulation of unesterified cholesterol, LSD (e.g., NPC1, MSD, and MPSIIIA). Reduced expression or activity of lysosomal hydrolases. to defective autophagosomelysosome fusion. phospholipids, glycolipids, sphingomyelin ( ), dysfunctional mitochondria and polyubiquitinated proteins (125). Abnormal lysosomal ph. Aberrant sequestration of SNARE complex and Enlarged lysosomal vesicles accumulation of autophagic cargo (123, 126). Mouse models of diet-induced obesity and metabolic syndrome Mouse models of diet-induced LMP Deficiency in Rab7 protein level. Enlarged lysosomal vesicles Bax-induced permeabilization of lysosomal membrane. Release of cathepsin B, D and K into cytosol. Suppressed autophagic flux linked to Rab7- or ER stress-dependent defects in autolysosome formation. Suppressed autophagy due to lack of intact, functional lysosomes Reduced Rab7 expression in the heart (87). Induced ER stress in liver (87, 158). Accumulation of phospholipids, ubiquitinin, and p62-positive protein aggregates in lysosomes in kidney and liver (130, 158). Cytosolic cathepsin-induced apoptotic cell death (132, 138). Cathepsin-induced mitochondrial membrane permeabilization and apoptotic cell death (138). Lipotoxic liver injury in NASH (138). Cardiac hypertrophy and contractile dysfunction (87, 88) Impaired cholesterol efflux in macrophages (137). Cell culture model of lipid-induced oxidative stress and lipotoxicity Increased lysosomal ph. Reduced lysomal cathepsin activity Oxidative post-translational modification of lysosomal Suppressed due to inhibition of autophagosome turnover Increased Nox2-derived superoxide production, s-nitrosylation of lysosomal vatpase, accumulation of defective autolysosomes (91). vatpase Decreased lysosomal Suppressed due to Accumulation of autophagosomes and acidification reduced lysosomal autophagic cargo, reduced mitochondrial Reduced cathepsin L activity acidification and oxidative phosphorylation and ATP hydrolase activity caused production, and β-cell dysfunction marked by by low cellular ATP defective insulin secretion and increase cell levels. death (64).

48 48 Bax-induced lysosomal membrane permeabilization Unknown Upregulation of ceramide and caspase dependent lysosomal membrane permeabilization and apoptotic cell death (132).

49 Figure 1

50 Figure 2

51 Figure 3

52 Figure 4

53 Figure 5

thematic review series

thematic review series thematic review series Thematic Review Series: Lipotoxicity: Many Roads to Cell Dysfunction and Cell Death Lipids, lysosomes, and autophagy Bharat Jaishy 1 and E. Dale Abel 2 Fraternal Order of Eagles

More information

Oxidation of Long Chain Fatty Acids

Oxidation of Long Chain Fatty Acids Oxidation of Long Chain Fatty Acids Dr NC Bird Oxidation of long chain fatty acids is the primary source of energy supply in man and animals. Hibernating animals utilise fat stores to maintain body heat,

More information

Plasma lipoproteins & atherosclerosis by. Prof.Dr. Maha M. Sallam

Plasma lipoproteins & atherosclerosis by. Prof.Dr. Maha M. Sallam Biochemistry Department Plasma lipoproteins & atherosclerosis by Prof.Dr. Maha M. Sallam 1 1. Recognize structures,types and role of lipoproteins in blood (Chylomicrons, VLDL, LDL and HDL). 2. Explain

More information

BIOL2171 ANU TCA CYCLE

BIOL2171 ANU TCA CYCLE TCA CYCLE IMPORTANCE: Oxidation of 2C Acetyl Co-A 2CO 2 + 3NADH + FADH 2 (8e-s donated to O 2 in the ETC) + GTP (energy) + Heat OVERVIEW: Occurs In the mitochondrion matrix. 1. the acetyl portion of acetyl-coa

More information

BCM 221 LECTURES OJEMEKELE O.

BCM 221 LECTURES OJEMEKELE O. BCM 221 LECTURES BY OJEMEKELE O. OUTLINE INTRODUCTION TO LIPID CHEMISTRY STORAGE OF ENERGY IN ADIPOCYTES MOBILIZATION OF ENERGY STORES IN ADIPOCYTES KETONE BODIES AND KETOSIS PYRUVATE DEHYDROGENASE COMPLEX

More information

Companion to Biosynthesis of Ketones & Cholesterols, Regulation of Lipid Metabolism Lecture Notes

Companion to Biosynthesis of Ketones & Cholesterols, Regulation of Lipid Metabolism Lecture Notes Companion to Biosynthesis of Ketones & Cholesterols, Regulation of Lipid Metabolism Lecture Notes The major site of acetoacetate and 3-hydorxybutyrate production is in the liver. 3-hydorxybutyrate is the

More information

LIPID METABOLISM. Sri Widia A Jusman Department of Biochemistry & Molecular Biology FMUI

LIPID METABOLISM. Sri Widia A Jusman Department of Biochemistry & Molecular Biology FMUI LIPID METABOLISM Sri Widia A Jusman Department of Biochemistry & Molecular Biology FMUI Lipid metabolism is concerned mainly with fatty acids cholesterol Source of fatty acids from dietary fat de novo

More information

Cellular control of cholesterol. Peter Takizawa Department of Cell Biology

Cellular control of cholesterol. Peter Takizawa Department of Cell Biology Cellular control of cholesterol Peter Takizawa Department of Cell Biology Brief overview of cholesterol s biological role Regulation of cholesterol synthesis Dietary and cellular uptake of cholesterol

More information

Cholesterol and its transport. Alice Skoumalová

Cholesterol and its transport. Alice Skoumalová Cholesterol and its transport Alice Skoumalová 27 carbons Cholesterol - structure Cholesterol importance A stabilizing component of cell membranes A precursor of bile salts A precursor of steroid hormones

More information

Lehninger 5 th ed. Chapter 17

Lehninger 5 th ed. Chapter 17 Lehninger 5 th ed. Chapter 17 December 26, 2010 Prof. Shimon Schuldiner Email: Shimon.Schuldiner@huji.ac.il Phone: 6585992 CHAPTER 17 Fatty Acid Catabolism Key topics: How fats are digested in animals

More information

Unit IV Problem 3 Biochemistry: Cholesterol Metabolism and Lipoproteins

Unit IV Problem 3 Biochemistry: Cholesterol Metabolism and Lipoproteins Unit IV Problem 3 Biochemistry: Cholesterol Metabolism and Lipoproteins - Cholesterol: It is a sterol which is found in all eukaryotic cells and contains an oxygen (as a hydroxyl group OH) on Carbon number

More information

CHM333 LECTURE 34: 11/30 12/2/09 FALL 2009 Professor Christine Hrycyna

CHM333 LECTURE 34: 11/30 12/2/09 FALL 2009 Professor Christine Hrycyna Lipid Metabolism β-oxidation FA Acetyl-CoA Triacylglycerols (TAGs) and glycogen are the two major forms of stored energy in vertebrates Glycogen can supply ATP for muscle contraction for less than an hour

More information

Metabolism of cardiac muscle. Dr. Mamoun Ahram Cardiovascular system, 2013

Metabolism of cardiac muscle. Dr. Mamoun Ahram Cardiovascular system, 2013 Metabolism of cardiac muscle Dr. Mamoun Ahram Cardiovascular system, 2013 References This lecture Mark s Basic Medical Biochemistry, 4 th ed., p. 890-891 Hand-out Why is this topic important? Heart failure

More information

23.1 Lipid Metabolism in Animals. Chapter 23. Micelles Lipid Metabolism in. Animals. Overview of Digestion Lipid Metabolism in

23.1 Lipid Metabolism in Animals. Chapter 23. Micelles Lipid Metabolism in. Animals. Overview of Digestion Lipid Metabolism in Denniston Topping Caret Copyright! The McGraw-Hill Companies, Inc. Permission required for reproduction or display. Chapter 23 Fatty Acid Metabolism Triglycerides (Tgl) are emulsified into fat droplets

More information

Cell Quality Control. Peter Takizawa Department of Cell Biology

Cell Quality Control. Peter Takizawa Department of Cell Biology Cell Quality Control Peter Takizawa Department of Cell Biology Cellular quality control reduces production of defective proteins. Cells have many quality control systems to ensure that cell does not build

More information

Metabolism of acylglycerols and sphingolipids. Martina Srbová

Metabolism of acylglycerols and sphingolipids. Martina Srbová Metabolism of acylglycerols and sphingolipids Martina Srbová Types of glycerolipids and sphingolipids 1. Triacylglycerols function as energy reserves adipose tissue (storage of triacylglycerol), lipoproteins

More information

Integrative Metabolism: Significance

Integrative Metabolism: Significance Integrative Metabolism: Significance Energy Containing Nutrients Carbohydrates Fats Proteins Catabolism Energy Depleted End Products H 2 O NH 3 ADP + Pi NAD + NADP + FAD + Pi NADH+H + NADPH+H + FADH2 Cell

More information

Cell Injury MECHANISMS OF CELL INJURY

Cell Injury MECHANISMS OF CELL INJURY Cell Injury MECHANISMS OF CELL INJURY The cellular response to injurious stimuli depends on the following factors: Type of injury, Its duration, and Its severity. Thus, low doses of toxins or a brief duration

More information

Lipids digestion and absorption, Biochemistry II

Lipids digestion and absorption, Biochemistry II Lipids digestion and absorption, blood plasma lipids, lipoproteins Biochemistry II Lecture 1 2008 (J.S.) Triacylglycerols (as well as free fatty acids and both free and esterified cholesterol) are very

More information

number Done by Corrected by Doctor Faisal Al-Khatibe

number Done by Corrected by Doctor Faisal Al-Khatibe number 24 Done by Mohammed tarabieh Corrected by Doctor Faisal Al-Khatibe 1 P a g e *Please look over the previous sheet about fatty acid synthesis **Oxidation(degradation) of fatty acids, occurs in the

More information

endomembrane system internal membranes origins transport of proteins chapter 15 endomembrane system

endomembrane system internal membranes origins transport of proteins chapter 15 endomembrane system endo system chapter 15 internal s endo system functions as a coordinated unit divide cytoplasm into distinct compartments controls exocytosis and endocytosis movement of molecules which cannot pass through

More information

Metabolic integration and Regulation

Metabolic integration and Regulation Metabolic integration and Regulation 109700: Graduate Biochemistry Trimester 2/2016 Assistant Prof. Dr. Panida Khunkaewla kpanida@sut.ac.th School of Chemistry Suranaree University of Technology 1 Overview

More information

Lecture 5: Cell Metabolism. Biology 219 Dr. Adam Ross

Lecture 5: Cell Metabolism. Biology 219 Dr. Adam Ross Lecture 5: Cell Metabolism Biology 219 Dr. Adam Ross Cellular Respiration Set of reactions that take place during the conversion of nutrients into ATP Intricate regulatory relationship between several

More information

UNIVERSITY OF PNG SCHOOL OF MEDICINE AND HEALTH SCIENCES DIVISION OF BASIC MEDICAL SCIENCES DISCIPLINE OF BIOCHEMISTRY AND MOLECULAR BIOLOGY

UNIVERSITY OF PNG SCHOOL OF MEDICINE AND HEALTH SCIENCES DIVISION OF BASIC MEDICAL SCIENCES DISCIPLINE OF BIOCHEMISTRY AND MOLECULAR BIOLOGY 1 UNIVERSITY OF PNG SCHOOL OF MEDICINE AND HEALTH SCIENCES DIVISION OF BASIC MEDICAL SCIENCES DISCIPLINE OF BIOCHEMISTRY AND MOLECULAR BIOLOGY GLUCOSE HOMEOSTASIS An Overview WHAT IS HOMEOSTASIS? Homeostasis

More information

ANSC/NUTR 618 LIPIDS & LIPID METABOLISM. Triacylglycerol and Fatty Acid Metabolism

ANSC/NUTR 618 LIPIDS & LIPID METABOLISM. Triacylglycerol and Fatty Acid Metabolism ANSC/NUTR 618 LIPIDS & LIPID METABOLISM II. Triacylglycerol synthesis A. Overall pathway Glycerol-3-phosphate + 3 Fatty acyl-coa à Triacylglycerol + 3 CoASH B. Enzymes 1. Acyl-CoA synthase 2. Glycerol-phosphate

More information

Hypothalamic Autophagy and Regulation of Energy Balance

Hypothalamic Autophagy and Regulation of Energy Balance Hypothalamic Autophagy and Regulation of Energy Balance Rajat Singh, MD Albert Einstein College of Medicine New York NuGOweek 211 Sept 6-9, 211 Autophagy Evolutionarily conserved cellular recycling program

More information

Chapter VIII: Dr. Sameh Sarray Hlaoui

Chapter VIII: Dr. Sameh Sarray Hlaoui Chapter VIII: Dr. Sameh Sarray Hlaoui Lipoproteins a Lipids are insoluble in plasma. In order to be transported they are combined with specific proteins to form lipoproteins: Clusters of proteins and lipids.

More information

LIPID METABOLISM

LIPID METABOLISM LIPID METABOLISM LIPOGENESIS LIPOGENESIS LIPOGENESIS FATTY ACID SYNTHESIS DE NOVO FFA in the blood come from :- (a) Dietary fat (b) Dietary carbohydrate/protein in excess of need FA TAG Site of synthesis:-

More information

Principles of Anatomy and Physiology

Principles of Anatomy and Physiology Principles of Anatomy and Physiology 14 th Edition CHAPTER 25 Metabolism and Nutrition Metabolic Reactions Metabolism refers to all of the chemical reactions taking place in the body. Reactions that break

More information

Biochemistry: A Short Course

Biochemistry: A Short Course Tymoczko Berg Stryer Biochemistry: A Short Course Second Edition CHAPTER 27 Fatty Acid Degradation Dietary Lipid (Triacylglycerol) Metabolism - In the small intestine, fat particles are coated with bile

More information

In The Name Of God. In The Name Of. EMRI Modeling Group

In The Name Of God. In The Name Of. EMRI Modeling Group In The Name Of God In The Name Of God EMRI Modeling Group Cells work together in functionally related groups called tissues Types of tissues: Epithelial lining and covering Connective support Muscle movement

More information

BBSG 501 Section 4 Metabolic Fuels, Energy and Order Fall 2003 Semester

BBSG 501 Section 4 Metabolic Fuels, Energy and Order Fall 2003 Semester BBSG 501 Section 4 Metabolic Fuels, Energy and Order Fall 2003 Semester Section Director: Dave Ford, Ph.D. Office: MS 141: ext. 8129: e-mail: fordda@slu.edu Lecturers: Michael Moxley, Ph.D. Office: MS

More information

Lipids and Membranes

Lipids and Membranes Lipids and Membranes Presented by Dr. Mohammad Saadeh The requirements for the Pharmaceutical Biochemistry I Philadelphia University Faculty of pharmacy Membrane transport D. Endocytosis and Exocytosis

More information

Summary of Endomembrane-system

Summary of Endomembrane-system Summary of Endomembrane-system 1. Endomembrane System: The structural and functional relationship organelles including ER,Golgi complex, lysosome, endosomes, secretory vesicles. 2. Membrane-bound structures

More information

Biosynthesis of Fatty Acids. By Dr.QUTAIBA A. QASIM

Biosynthesis of Fatty Acids. By Dr.QUTAIBA A. QASIM Biosynthesis of Fatty Acids By Dr.QUTAIBA A. QASIM Fatty Acids Definition Fatty acids are comprised of hydrocarbon chains terminating with carboxylic acid groups. Fatty acids and their associated derivatives

More information

Lipid Metabolism. Remember fats?? Triacylglycerols - major form of energy storage in animals

Lipid Metabolism. Remember fats?? Triacylglycerols - major form of energy storage in animals Remember fats?? Triacylglycerols - major form of energy storage in animals Your energy reserves: ~0.5% carbs (glycogen + glucose) ~15% protein (muscle, last resort) ~85% fat Why use fat for energy? 1 gram

More information

Lipid Metabolism. Catabolism Overview

Lipid Metabolism. Catabolism Overview Lipid Metabolism Pratt & Cornely, Chapter 17 Catabolism Overview Lipids as a fuel source from diet Beta oxidation Mechanism ATP production Ketone bodies as fuel 1 High energy More reduced Little water

More information

Medical Biochemistry and Molecular Biology department

Medical Biochemistry and Molecular Biology department Medical Biochemistry and Molecular Biology department Cardiac Fuels [Sources of energy for the Cardiac muscle] Intended learning outcomes of the lecture: By the end of this lecture you would be able to:-

More information

Metabolic Syndrome. DOPE amines COGS 163

Metabolic Syndrome. DOPE amines COGS 163 Metabolic Syndrome DOPE amines COGS 163 Overview - M etabolic Syndrome - General definition and criteria - Importance of diagnosis - Glucose Homeostasis - Type 2 Diabetes Mellitus - Insulin Resistance

More information

MILK BIOSYNTHESIS PART 3: FAT

MILK BIOSYNTHESIS PART 3: FAT MILK BIOSYNTHESIS PART 3: FAT KEY ENZYMES (FROM ALL BIOSYNTHESIS LECTURES) FDPase = fructose diphosphatase Citrate lyase Isocitrate dehydrogenase Fatty acid synthetase Acetyl CoA carboxylase Fatty acyl

More information

Fatty Acid Degradation. Catabolism Overview. TAG and FA 11/11/2015. Chapter 27, Stryer Short Course. Lipids as a fuel source diet Beta oxidation

Fatty Acid Degradation. Catabolism Overview. TAG and FA 11/11/2015. Chapter 27, Stryer Short Course. Lipids as a fuel source diet Beta oxidation Fatty Acid Degradation Chapter 27, Stryer Short Course Catabolism verview Lipids as a fuel source diet Beta oxidation saturated Unsaturated dd chain Ketone bodies as fuel Physiology High energy More reduced

More information

1Why lipids cannot be transported in blood alone? 2How we transport Fatty acids and steroid hormones?

1Why lipids cannot be transported in blood alone? 2How we transport Fatty acids and steroid hormones? 1Why lipids cannot be transported in blood alone? 2How we transport Fatty acids and steroid hormones? 3How are dietary lipids transported? 4How lipids synthesized in the liver are transported? 5 Lipoprotien

More information

Lecture 36. Key Concepts. Overview of lipid metabolism. Reactions of fatty acid oxidation. Energy yield from fatty acid oxidation

Lecture 36. Key Concepts. Overview of lipid metabolism. Reactions of fatty acid oxidation. Energy yield from fatty acid oxidation Lecture 36 Lipid Metabolism 1 Fatty Acid Oxidation Ketone Bodies Key Concepts Overview of lipid metabolism Reactions of fatty acid oxidation Energy yield from fatty acid oxidation Formation of ketone bodies

More information

Molecular Cell Biology Problem Drill 16: Intracellular Compartment and Protein Sorting

Molecular Cell Biology Problem Drill 16: Intracellular Compartment and Protein Sorting Molecular Cell Biology Problem Drill 16: Intracellular Compartment and Protein Sorting Question No. 1 of 10 Question 1. Which of the following statements about the nucleus is correct? Question #01 A. The

More information

Fatty Acid and Triacylglycerol Metabolism 1

Fatty Acid and Triacylglycerol Metabolism 1 Fatty Acid and Triacylglycerol Metabolism 1 Mobilization of stored fats and oxidation of fatty acids Lippincott s Chapter 16 What is the first lecture about What is triacylglycerol Fatty acids structure

More information

OVERVIEW M ET AB OL IS M OF FR EE FA TT Y AC ID S

OVERVIEW M ET AB OL IS M OF FR EE FA TT Y AC ID S LIPOLYSIS LIPOLYSIS OVERVIEW CATABOLISM OF FREE FATTY ACIDS Nonesterified fatty acids Source:- (a) breakdown of TAG in adipose tissue (b) action of Lipoprotein lipase on plasma TAG Combined with Albumin

More information

Integration Of Metabolism

Integration Of Metabolism Integration Of Metabolism Metabolism Consist of Highly Interconnected Pathways The basic strategy of catabolic metabolism is to form ATP, NADPH, and building blocks for biosyntheses. 1. ATP is the universal

More information

What systems are involved in homeostatic regulation (give an example)?

What systems are involved in homeostatic regulation (give an example)? 1 UNIVERSITY OF PNG SCHOOL OF MEDICINE AND HEALTH SCIENCES DIVISION OF BASIC MEDICAL SCIENCES DISCIPLINE OF BIOCHEMISTRY AND MOLECULAR BIOLOGY GLUCOSE HOMEOSTASIS (Diabetes Mellitus Part 1): An Overview

More information

Fatty acid breakdown

Fatty acid breakdown Fatty acids contain a long hydrocarbon chain and a terminal carboxylate group. Most contain between 14 and 24 carbon atoms. The chains may be saturated or contain double bonds. The complete oxidation of

More information

CHY2026: General Biochemistry. Lipid Metabolism

CHY2026: General Biochemistry. Lipid Metabolism CHY2026: General Biochemistry Lipid Metabolism Lipid Digestion Lipid Metabolism Fats (triglycerides) are high metabolic energy molecules Fats yield 9.3 kcal of energy (carbohydrates and proteins 4.1 kcal)

More information

Autophagy in Insulin Resistance. Abstract. Introduction. Takeshi Yoshizaki. KEY WORDS: Autophagy, insulin resistance, aging, inflammation

Autophagy in Insulin Resistance. Abstract. Introduction. Takeshi Yoshizaki. KEY WORDS: Autophagy, insulin resistance, aging, inflammation Received: Oct. 10, 2012 Accepted: Oct. 17, 2012 Published online: Oct. 31, 2012 Review Article Autophagy in Insulin Resistance Takeshi Yoshizaki Department of Medicine, Shiga University of Medical Science

More information

Multiple choice: Circle the best answer on this exam. There are 12 multiple choice questions, each question is worth 3 points.

Multiple choice: Circle the best answer on this exam. There are 12 multiple choice questions, each question is worth 3 points. CHEM 4420 Exam 4 Spring 2015 Dr. Stone Page 1 of 6 Name Use complete sentences when requested. There are 120 possible points on this exam. Therefore there are 20 bonus points. Multiple choice: Circle the

More information

BEIGE AND BROWN FAT: BASIC BIOLOGY AND NOVEL THERAPEUTICS Dr. Carl Ascoli

BEIGE AND BROWN FAT: BASIC BIOLOGY AND NOVEL THERAPEUTICS Dr. Carl Ascoli BEIGE AND BROWN FAT: BASIC BIOLOGY AND NOVEL THERAPEUTICS Dr. Carl Ascoli Symposium Co-Chairs: Bruce M. Spiegelman (Harvard/Dana Farber) and Sven Enerbäck (U.Gothenburg) April 17-23, 2015 Snowbird Resort,

More information

Chapt. 10 Cell Biology and Biochemistry. The cell: Student Learning Outcomes: Describe basic features of typical human cell

Chapt. 10 Cell Biology and Biochemistry. The cell: Student Learning Outcomes: Describe basic features of typical human cell Chapt. 10 Cell Biology and Biochemistry Cell Chapt. 10 Cell Biology and Biochemistry The cell: Lipid bilayer membrane Student Learning Outcomes: Describe basic features of typical human cell Integral transport

More information

Lysosomes, Peroxisomes and Centrioles. Hüseyin Çağsın

Lysosomes, Peroxisomes and Centrioles. Hüseyin Çağsın Lysosomes, Peroxisomes and Centrioles Hüseyin Çağsın Lysosomes Outline Endosomes Molecule transport to the lysosomes Endocytosis Exocytosis Autophagy Vacuoles Peroxisomes Centrioles Lysosomes Lysosomes

More information

1- Which of the following statements is TRUE in regards to eukaryotic and prokaryotic cells?

1- Which of the following statements is TRUE in regards to eukaryotic and prokaryotic cells? Name: NetID: Exam 3 - Version 1 October 23, 2017 Dr. A. Pimentel Each question has a value of 4 points and there are a total of 160 points in the exam. However, the maximum score of this exam will be capped

More information

General Principles of Endocrine Physiology

General Principles of Endocrine Physiology General Principles of Endocrine Physiology By Dr. Isabel S.S. Hwang Department of Physiology Faculty of Medicine University of Hong Kong The major human endocrine glands Endocrine glands and hormones

More information

Lipid metabolism in familial hypercholesterolemia

Lipid metabolism in familial hypercholesterolemia Lipid metabolism in familial hypercholesterolemia Khalid Al-Rasadi, BSc, MD, FRCPC Head of Biochemistry Department, SQU Head of Lipid and LDL-Apheresis Unit, SQUH President of Oman society of Lipid & Atherosclerosis

More information

Biochemistry: A Short Course

Biochemistry: A Short Course Tymoczko Berg Stryer Biochemistry: A Short Course Second Edition CHAPTER 28 Fatty Acid Synthesis 2013 W. H. Freeman and Company Chapter 28 Outline 1. The first stage of fatty acid synthesis is transfer

More information

Cholesterol metabolism. Function Biosynthesis Transport in the organism Hypercholesterolemia

Cholesterol metabolism. Function Biosynthesis Transport in the organism Hypercholesterolemia Cholesterol metabolism Function Biosynthesis Transport in the organism Hypercholesterolemia - component of all cell membranes - precursor of bile acids steroid hormones vitamin D Cholesterol Sources: dietary

More information

Energy Production In A Cell (Chapter 25 Metabolism)

Energy Production In A Cell (Chapter 25 Metabolism) Energy Production In A Cell (Chapter 25 Metabolism) Large food molecules contain a lot of potential energy in the form of chemical bonds but it requires a lot of work to liberate the energy. Cells need

More information

Zool 3200: Cell Biology Exam 4 Part I 2/3/15

Zool 3200: Cell Biology Exam 4 Part I 2/3/15 Name: Key Trask Zool 3200: Cell Biology Exam 4 Part I 2/3/15 Answer each of the following questions in the space provided, explaining your answers when asked to do so; circle the correct answer or answers

More information

A particular set of insults induces apoptosis (part 1), which, if inhibited, can switch to autophagy. At least in some cellular settings, autophagy se

A particular set of insults induces apoptosis (part 1), which, if inhibited, can switch to autophagy. At least in some cellular settings, autophagy se A particular set of insults induces apoptosis (part 1), which, if inhibited, can switch to autophagy. At least in some cellular settings, autophagy serves as a defence mechanism that prevents or retards

More information

Intermediary metabolism. Eva Samcová

Intermediary metabolism. Eva Samcová Intermediary metabolism Eva Samcová Metabolic roles of tissues Four major tissues play a dominant role in fuel metabolism : liver, adipose, muscle, and brain. These tissues do not function in isolation.

More information

PROTEIN TRAFFICKING. Dr. SARRAY Sameh, Ph.D

PROTEIN TRAFFICKING. Dr. SARRAY Sameh, Ph.D PROTEIN TRAFFICKING Dr. SARRAY Sameh, Ph.D Overview Proteins are synthesized either on free ribosomes or on ribosomes bound to endoplasmic reticulum (RER). The synthesis of nuclear, mitochondrial and peroxisomal

More information

Lipoproteins Metabolism Reference: Campbell Biochemistry and Lippincott s Biochemistry

Lipoproteins Metabolism Reference: Campbell Biochemistry and Lippincott s Biochemistry Lipoproteins Metabolism Reference: Campbell Biochemistry and Lippincott s Biochemistry Learning Objectives 1. Define lipoproteins and explain the rationale of their formation in blood. 2. List different

More information

Introduction to Metabolism Cell Structure and Function

Introduction to Metabolism Cell Structure and Function Introduction to Metabolism Cell Structure and Function Cells can be divided into two primary types prokaryotes - Almost all prokaryotes are bacteria eukaryotes - Eukaryotes include all cells of multicellular

More information

Summary of fatty acid synthesis

Summary of fatty acid synthesis Lipid Metabolism, part 2 1 Summary of fatty acid synthesis 8 acetyl CoA + 14 NADPH + 14 H+ + 7 ATP palmitic acid (16:0) + 8 CoA + 14 NADP + + 7 ADP + 7 Pi + 7 H20 1. The major suppliers of NADPH for fatty

More information

REVIEWS. Targeting autophagy in obesity: from pathophysiology to management

REVIEWS. Targeting autophagy in obesity: from pathophysiology to management REVIEWS Targeting autophagy in obesity: from pathophysiology to management Yingmei Zhang 1,2 *, James R. Sowers 3 and Jun Ren 1,2 * Abstract Obesity poses a severe threat to human health, including the

More information

lysosomes Ingested materials Defective cell components Degrades macromolecules of all types:

lysosomes Ingested materials Defective cell components Degrades macromolecules of all types: lysosomes Digests Ingested materials Defective cell components Degrades macromolecules of all types: Proteins Nucleic acids Carbohydrates Lipids Single membrane bound vesicle, contains up to 50 digestive

More information

Roles of Lipids. principal form of stored energy major constituents of cell membranes vitamins messengers intra and extracellular

Roles of Lipids. principal form of stored energy major constituents of cell membranes vitamins messengers intra and extracellular Roles of Lipids principal form of stored energy major constituents of cell membranes vitamins messengers intra and extracellular = Oxidation of fatty acids Central energy-yielding pathway in animals. O

More information

Leptin Intro/Signaling. ATeamP: Angelo, Anthony, Charlie, Gabby, Joseph

Leptin Intro/Signaling. ATeamP: Angelo, Anthony, Charlie, Gabby, Joseph Leptin Intro/Signaling ATeamP: Angelo, Anthony, Charlie, Gabby, Joseph Overview Intro to Leptin Definition & Sources Physiology Bound vs. Free Receptors Signaling JAK/STAT MAPK PI3K ACC Experimental findings

More information

BIOL212 Biochemistry of Disease. Metabolic Disorders - Obesity

BIOL212 Biochemistry of Disease. Metabolic Disorders - Obesity BIOL212 Biochemistry of Disease Metabolic Disorders - Obesity Obesity Approx. 23% of adults are obese in the U.K. The number of obese children has tripled in 20 years. 10% of six year olds are obese, rising

More information

6. How Are Fatty Acids Produced? 7. How Are Acylglycerols and Compound Lipids Produced? 8. How Is Cholesterol Produced?

6. How Are Fatty Acids Produced? 7. How Are Acylglycerols and Compound Lipids Produced? 8. How Is Cholesterol Produced? Lipid Metabolism Learning bjectives 1 How Are Lipids Involved in the Generationand Storage of Energy? 2 How Are Lipids Catabolized? 3 What Is the Energy Yield from the xidation of Fatty Acids? 4 How Are

More information

Protein Trafficking in the Secretory and Endocytic Pathways

Protein Trafficking in the Secretory and Endocytic Pathways Protein Trafficking in the Secretory and Endocytic Pathways The compartmentalization of eukaryotic cells has considerable functional advantages for the cell, but requires elaborate mechanisms to ensure

More information

Chapter 1 The Lipid Droplet: a Dynamic Organelle, not only Involved in the Storage and Turnover of Lipids

Chapter 1 The Lipid Droplet: a Dynamic Organelle, not only Involved in the Storage and Turnover of Lipids Chapter 1 The Lipid Droplet: a Dynamic Organelle, not only Involved in the Storage and Turnover of Lipids Sven-Olof Olofsson, Pontus Boström, Jens Lagerstedt, Linda Andersson, Martin Adiels, Jeanna Perman,

More information

BIOL 4374/BCHS 4313 Cell Biology Exam #1 February 13, 2001

BIOL 4374/BCHS 4313 Cell Biology Exam #1 February 13, 2001 BIOL 4374/BCHS 4313 Cell Biology Exam #1 February 13, 2001 SS# Name This exam is worth a total of 100 points. The number of points each question is worth is shown in parentheses. Good luck! 1. (2) The

More information

Chapter 14. Energy conversion: Energy & Behavior

Chapter 14. Energy conversion: Energy & Behavior Chapter 14 Energy conversion: Energy & Behavior Why do you Eat and Breath? To generate ATP Foods, Oxygen, and Mitochodria Cells Obtain Energy by the Oxidation of Organic Molecules Food making ATP making

More information

Lecture: 26 OXIDATION OF FATTY ACIDS

Lecture: 26 OXIDATION OF FATTY ACIDS Lecture: 26 OXIDATION OF FATTY ACIDS Fatty acids obtained by hydrolysis of fats undergo different oxidative pathways designated as alpha ( ), beta ( ) and omega ( ) pathways. -oxidation -Oxidation of fatty

More information

Fatty acids synthesis

Fatty acids synthesis Fatty acids synthesis The synthesis start from Acetyl COA the first step requires ATP + reducing power NADPH! even though the oxidation and synthesis are different pathways but from chemical part of view

More information

Niacin Metabolism: Effects on Cholesterol

Niacin Metabolism: Effects on Cholesterol Niacin Metabolism: Effects on Cholesterol By Julianne R. Edwards For Dr. William R. Proulx, PhD, RD Associate Professor of Nutrition and Dietetics In partial fulfillments for the requirements of NUTR342

More information

Homework Hanson section MCB Course, Fall 2014

Homework Hanson section MCB Course, Fall 2014 Homework Hanson section MCB Course, Fall 2014 (1) Antitrypsin, which inhibits certain proteases, is normally secreted into the bloodstream by liver cells. Antitrypsin is absent from the bloodstream of

More information

cholesterol structure Cholesterol FAQs Cholesterol promotes the liquid-ordered phase of membranes Friday, October 15, 2010

cholesterol structure Cholesterol FAQs Cholesterol promotes the liquid-ordered phase of membranes Friday, October 15, 2010 cholesterol structure most plasma cholesterol is in the esterified form (not found in cells or membranes) cholesterol functions in all membranes (drives formation of lipid microdomains) cholesterol is

More information

Ahmad Ulnar. Faisal Nimri ... Dr.Faisal

Ahmad Ulnar. Faisal Nimri ... Dr.Faisal 24 Ahmad Ulnar Faisal Nimri... Dr.Faisal Fatty Acid Synthesis - Occurs mainly in the Liver (to store excess carbohydrates as triacylglycerols(fat)) and in lactating mammary glands (for the production of

More information

Regulating Hepatic Cellular Cholesterol

Regulating Hepatic Cellular Cholesterol Under circumstances of cholesterol deficiency, Sterol Regulatory Element Binding Proteins (SREBPs) via binding to DNA nuclear response elements set off genomic production of proteins and enzymes that induce

More information

ANSC/NUTR 618 Lipids & Lipid Metabolism

ANSC/NUTR 618 Lipids & Lipid Metabolism I. Overall concepts A. Definitions ANC/NUTR 618 Lipids & Lipid Metabolism 1. De novo synthesis = synthesis from non-fatty acid precursors a. Carbohydrate precursors (glucose, lactate, and pyruvate) b.

More information

Lipoproteins Metabolism

Lipoproteins Metabolism Lipoproteins Metabolism LEARNING OBJECTIVES By the end of this Lecture, the student should be able to describe: What are Lipoproteins? Describe Lipoprotein Particles. Composition of Lipoproteins. The chemical

More information

Biochemistry 7/11/ Bio-Energetics & ATP. 5.1) ADP, ATP and Cellular Respiration OVERVIEW OF ENERGY AND METABOLISM

Biochemistry 7/11/ Bio-Energetics & ATP. 5.1) ADP, ATP and Cellular Respiration OVERVIEW OF ENERGY AND METABOLISM Biochemistry 5. Bio-Energetics & ATP 5.1) ADP, ATP and Cellular Respiration Prof. Dr. Klaus Heese OVERVIEW OF ENERGY AND METABOLISM 1. The food we eat, (carbohydrates/ glucose /sugar, lipids/fat, proteins),

More information

Chemistry 1120 Exam 4 Study Guide

Chemistry 1120 Exam 4 Study Guide Chemistry 1120 Exam 4 Study Guide Chapter 12 12.1 Identify and differentiate between macronutrients (lipids, amino acids and saccharides) and micronutrients (vitamins and minerals). Master Tutor Section

More information

Metabolism. Chapter 5. Catabolism Drives Anabolism 8/29/11. Complete Catabolism of Glucose

Metabolism. Chapter 5. Catabolism Drives Anabolism 8/29/11. Complete Catabolism of Glucose 8/29/11 Metabolism Chapter 5 All of the reactions in the body that require energy transfer. Can be divided into: Cell Respiration and Metabolism Anabolism: requires the input of energy to synthesize large

More information

Signal Transduction Cascades

Signal Transduction Cascades Signal Transduction Cascades Contents of this page: Kinases & phosphatases Protein Kinase A (camp-dependent protein kinase) G-protein signal cascade Structure of G-proteins Small GTP-binding proteins,

More information

ANSC/NUTR 618 LIPIDS & LIPID METABOLISM Lipoprotein Metabolism

ANSC/NUTR 618 LIPIDS & LIPID METABOLISM Lipoprotein Metabolism ANSC/NUTR 618 LIPIDS & LIPID METABOLISM Lipoprotein Metabolism I. Chylomicrons (exogenous pathway) A. 83% triacylglycerol, 2% protein, 8% cholesterol plus cholesterol esters, 7% phospholipid (esp. phosphatidylcholine)

More information

THE GLUCOSE-FATTY ACID-KETONE BODY CYCLE Role of ketone bodies as respiratory substrates and metabolic signals

THE GLUCOSE-FATTY ACID-KETONE BODY CYCLE Role of ketone bodies as respiratory substrates and metabolic signals Br. J. Anaesth. (1981), 53, 131 THE GLUCOSE-FATTY ACID-KETONE BODY CYCLE Role of ketone bodies as respiratory substrates and metabolic signals J. C. STANLEY In this paper, the glucose-fatty acid cycle

More information

Lipid Metabolism * OpenStax

Lipid Metabolism * OpenStax OpenStax-CNX module: m46462 1 Lipid Metabolism * OpenStax This work is produced by OpenStax-CNX and licensed under the Creative Commons Attribution License 3.0 By the end of this section, you will be able

More information

Cells extract energy from their environment and use the energy for a host of biological activities including biosynthesis.

Cells extract energy from their environment and use the energy for a host of biological activities including biosynthesis. ATP=cellular energy Cells extract energy from their environment and use the energy for a host of biological activities including biosynthesis. The reactions of energy extraction and energy use are called

More information

Chapter 12 Nutrition

Chapter 12 Nutrition Chapter 12 Nutrition Nutrients macronutrients: large required daily quantities carbohydrates, lipids, proteins micronutrients: small required daily quantities vitamins, minerals Also required: water and

More information

1. to understand how proteins find their destination in prokaryotic and eukaryotic cells 2. to know how proteins are bio-recycled

1. to understand how proteins find their destination in prokaryotic and eukaryotic cells 2. to know how proteins are bio-recycled Protein Targeting Objectives 1. to understand how proteins find their destination in prokaryotic and eukaryotic cells 2. to know how proteins are bio-recycled As a protein is being synthesized, decisions

More information

OVERVIEW OF ENERGY AND METABOLISM

OVERVIEW OF ENERGY AND METABOLISM Biochemistry 5. Bio-Energetics & ATP 5.1) ADP, ATP and Cellular Respiration OVERVIEW OF ENERGY AND METABOLISM 1. The food we eat, (carbohydrates/ glucose /sugar, lipids/fat, proteins), are our only source

More information