Although apolipoprotein E (apoE) is well known to play a major role in lipid metabolism, its role in glucose and energy homeostasis remains unclear. Herein, we established apoE-deficient genetically obese Ay (apoE−/−;Ay/+) mice. ApoE deficiency in Ay mice prevented the development of obesity, with decreased fat accumulation in the liver and adipose tissues. ApoE−/−;Ay/+ mice exhibited better glucose tolerance than apoE+/+;Ay/+ mice. Insulin tolerance testing and hyperinsulinemic-euglycemic clamp study revealed marked improvement of insulin sensitivity, despite increased plasma free fatty acid levels. These metabolic phenotypes were reversed by adenoviral replenishment of apoE protein, indicating circulating apoE to be involved in increased adiposity and obesity-related metabolic disorders. Uptake of apoE-lacking VLDL into the liver and adipocytes was markedly inhibited, but adipocytes in apoE−/−;Ay/+ mice exhibited normal differentiation, suggesting that apoE-dependent VLDL transport is involved in the development of obesity, i.e., surplus fat accumulation. Interestingly, apoE−/−;Ay/+ mice exhibited decreased food intake and increased energy expenditure. Pair-feeding experiments indicate these phenomena to both contribute to the obesity-resistant phenotypes associated with apoE deficiency. Thus, apoE is involved in maintaining energy homeostasis. ApoE-dependent excess fat accumulation is a promising therapeutic target for the metabolic syndrome.

Obesity causes various metabolic disorders, including diabetes, dyslipidemia, and hypertension and has become a major public health concern in most industrialized countries in recent decades (1). Obesity results mainly from excess energy intake and physical inactivity, and the molecular mechanisms of body weight control have been intensively studied from various aspects, including appetite, energy expenditure, glucose and lipid metabolism, and adiposity (2). Excess fat accumulation is associated with metabolic disorders including insulin resistance, glucose intolerance, and dyslipidemia (3). In addition, we reported that dissipating excess energy improves diabetes and obesity in mice (4). On the other hand, lipoatrophy also leads to these metabolic disorders in mice and humans. Fat-less mice (5,6) as well as patients with lipodystrophy (7) exhibit severe insulin resistance and diabetes. Thus, having an appropriate adipose tissue amount is important for maintaining glucose homeostasis via adipocyte metabolic activities including production and secretion of adipocytokines.

Apolipoprotein E (apoE) is a structural component of all lipoprotein particles except LDL and serves as a high-affinity ligand for lipoprotein receptors (8). ApoE plays important roles in hepatic uptake of chylomicron and VLDL remnants (9,10) as well as hepatic secretion of VLDL (11). In contrast, involvement of apoE in adiposity, insulin sensitivity, and glucose metabolism is somewhat controversial. ApoE−/− mice treated with gold thioglucose become obese and diabetic (12). High-fat chow induces obesity in apoE−/− mice in a manner similar to that in wild-type C57BL/6 mice, while apoE deficiency produced resistance to some obesity-related phenotypic features, including hyperinsulinemia and hyperglycemia (13). On the other hand, Chiba et al. reported that apoE deficiency in ob/ob mice resulted in neither increased body weight nor adiposity, but glucose metabolism and insulin sensitivity were not examined (14). They attributed decreased adiposity in ob/ob;apoE−/− mice to impaired adipocyte differentiation based on in vitro findings obtained using bone marrow stromal cells and 3T3-L1 cells. However, impaired adipocyte differentiation induces severe insulin resistance and diabetes (5,6), markedly different from the metabolic phenotypes of apoE−/− mice. Thus, it remains unclear how, and even whether, apoE is involved in adiposity and glucose metabolism.

Herein, we recognized that adult apoE−/− mice are leaner and more glucose tolerant than wild-type mice. In contrast, younger apoE−/− mice exhibit normal adiposity, i.e., they are not lipoatrophic. These findings led us to postulate that apoE is involved in surplus fat accumulation, resulting in the development of insulin resistance, but does not play a major role in the fat accumulation required for adipocyte function. To clarify whether apoE is involved in the development of insulin resistance associated with obesity and, if so, to identify the underlying mechanisms, we established apoE-deficient genetically obese Ay (apoE−/−;Ay/+) mice. Without impairing adipocyte differentiation in vivo, apoE deficiency in Ay mice prevented obesity, glucose intolerance, and insulin resistance. Interestingly, apoE−/−;Ay/+ mice exhibited decreased food intake and increased energy expenditure, both of which contribute to the obesity-resistant phenotypes associated with apoE deficiency. Thus, apoE is a key molecule for development of obesity, i.e., excess fat accumulation, and is a possible therapeutic target for the metabolic syndrome.

Animal studies were conducted in accordance with the institutional guidelines for animal experiments at Tohoku University. ApoE+/−;Ay/+ mice were obtained by mating male KKAy mice (Nippon CLEA, Shizuoka, Japan) and female apoE-deficient mice with the C57BL/6J background (15) (The Jackson Laboratory, Bar Harbor, ME). Male apoE+/−;Ay/+ mice were mated with female apoE+/− mice with the C57BL/6J background. Mice of three genotyopes, apoE+/+;Ay/+, apoE+/−;Ay/+, and apoE−/−;Ay/+, were selected. Littermates were used in each experiment. These mice were housed individually and started on a high-fat diet consisting of 15.3% (wt/wt) fat (Quick Fat; Nippon CLEA, Shizuoka, Japan) at 6 weeks of age. Experiments were performed 5 weeks after high-fat loading. Viruses were administered intravenously at a dose of 2 × 108 plaque-forming units 4 weeks after high-fat loading. For pair-feeding experiments, apoE+/+;Ay/+ mice were allotted into two groups at 4 weeks of age. One group was given their daily food allotments based on the previous days’ consumption by apoE−/−;Ay/+ littermate mice.

Recombinant adenovirus preparation.

Recombinant adenovirus, containing the human apoE2, E3, E4, or bacterial β-galactosidase gene (Adex1CAlacZ) cDNA under the CAG promoter, was prepared as described previously (16).

Oxygen consumption.

Oxygen consumption was measured with an O2/CO2 metabolism measuring system (model MK-5000RQ; Muromachikikai, Tokyo, Japan) as described previously (4).

Histological analysis.

Livers and adipose tissues were removed, fixed with 10% formalin, and embedded in paraffin. Tissue sections were stained with hematoxylin-eosin or 0.1% (wt/vol) Oil Red O. Total adipocyte areas were traced manually and analyzed. Brown and white adipocyte areas were measured in 100 or more cells per mouse in each group.

Triglyceride contents of the liver and adipose tissue.

Frozen livers or adipose tissues were homogenized, and triglycerides were extracted with CHCl3:CH3OH (2:1 vol:vol), dried, and resuspended in 2-propanol. Triglyceride contents were measured using Lipidos liquid (Toyobo, Osaka, Japan).

Tyrosine phosphorylation of insulin receptor substrate 1.

Mice that had been fasted for 16 h were injected with 100 μl normal saline (0.9% NaCl), with or without 10 units/kg body wt of insulin via the tail vein. Hindlimb muscles were removed 300 s later and immediately homogenized. After centrifugation, the resulting supernatants were used for immunoprecipitation with anti–insulin receptor substrate 1 (IRS-1) antibody (17). Immunoprecipitates were subjected to SDS-PAGE and then immunoblotted using antiphosphotyrosine antibody (4G10) as described previously (17).

Blood analysis.

Blood glucose, plasma insulin, leptin, adiponectin, tumor necrosis factor (TNF)-α, total cholesterol, triglyceride, and free fatty acid (FFA) concentrations were determined as described previously (4). Plasma lipoproteins were analyzed by high-performance liquid chromatography (HPLC) using molecular sieve columns (Skylight Biotech, Akita, Japan) (18).

Glucose and insulin tolerance tests.

Glucose and insulin tolerance tests were performed as described previously with slight modification (19). Glucose tolerance tests were performed on fasted (10 h) mice. Mice were given oral glucose (1 g/kg body wt), and blood glucose was assayed immediately before and at 15, 30, 60, and 120 min postadministration. Insulin tolerance tests were performed on fed mice. Mice were injected with human regular insulin (1.5 units/kg body wt; Eli Lilly, Kobe, Japan) into the intraperitoneal space, and blood glucose was assayed immediately before and at 20, 40, 60, and 80 min postinjection.

Hyperinsulinemic-euglycemic clamp.

Hyperinsulinemic-euglycemic clamp studies were performed as described previously (20). Chronically cannulated, conscious and unrestrained mice were fasted for 6 h before the study. Insulin (500 mU · kg−1 · min−1) was infused throughout the clamp study. Blood glucose was monitored every 5 min via carotid arterial catheter samples. Glucose was infused at a variable rate to maintain blood glucose at 120 mg/dl. The glucose infusion rate and endogenous glucose production were calculated as described (20).

Quantitative RT-PCR–based gene expression.

Total RNA was isolated from 0.1 g mouse adipose tissue with Isogen (Wako Pure Chemical, Osaka, Japan), and cDNA was synthesized with a Cloned AMV First Strand Synthesis Kit (Invitrogen, Rockville, MD) using 5 μg total RNA. cDNA synthesized from total RNA was evaluated using real-time quantitative PCR (Light Cycler Quick System 350S; Roche Diagnostics, Mannheim, Germany). The relative amount of mRNA was calculated with 28S rRNA as the invariant control. The primers used are described in Supplemental Table 1 (online appendix [available at http://diabetes.diabetesjournals.org]).

β-VLDL uptake into cultured adipocytes.

Murine 3T3-L1 preadipocytes were maintained and differentiated into adipocytes as described previously (21). β-VLDL (d < 1.006 g/ml) was purified from the blood of apoE−/− mice with high-fat loading and labeled with fluorescent lipid DiI (1,1′-dioctadecyl-3,3,3′,3′-tetramethylindocarbocyanine perchlorate) as described previously (22). After labeling with DiI, β-VLDL was incubated with an equal amount of recombinant human apoE3 for 1 h at 37°C (23). DiI-labeled β-VLDL (4 μg protein/ml) with or without apoE had been incubated with fully differentiated 3T3-L1 adipocytes for 8 h at 37°C. Cells were rinsed twice with PBS and solubilized with 0.1 N NaOH/1% SDS. Fluorescent intensities of cell lysates were measured with a fluorescent spectrophotometer (F-2000; Hitachi, Tokyo, Japan) (24).

Triglyceride secretion rate.

To assess hepatic triglyceride secretion, 500 mg/kg Triton WR-1339 (Sigma), which blocks lipolysis of triglycerides in peripheral tissue, was injected into the tail veins of the 6 h–fasted apoE+/+;Ay/+ mice, and blood samples were taken immediately before and 30, 60, and 90 min after injection (25).

Hepatic uptake of β-VLDL.

Male C57BL/6N mice at 6 weeks of age were injected via tail vein with fluorescent β-VLDL (5 μg/mouse) with or without apoE in 0.2 ml PBS. Thirty minutes after injection, the mice were killed and their livers excised for lipid extraction and fluorescence measurement as described previously (26).

Statistical analysis.

All data were expressed as means ± SEM. The statistical significance of differences was assessed by the unpaired t test and one-factor ANOVA.

ApoE deficiency inhibited the development of obesity.

This study was prompted by the unexpected observation that aged apoE−/− mice are leaner and more insulin sensitive than apoE+/+ mice of the same age with the C57BL/6J background. Therefore, we weighed apoE−/− and apoE+/+ mice from 6 to 42 weeks of age, while maintaining the animals on high-fat chow. Body weights were similar through 10 weeks of age, but apoE−/− mice had significantly lower body weights after 14 weeks of age (Fig. 1A). Glucose tolerance in 30-week-old apoE−/− mice was moderately better than that in apoE+/+ littermate mice (Fig. 1B). Thus, apoE deficiency prevents the development of obesity and associated glucose intolerance, while its effects on glucose tolerance are small.

Next, to elucidate the roles of apoE in development of the metabolic disorders associated with obesity, we established apoE+/+;Ay/+, apoE+/−;Ay/+, and apoE−/−;Ay/+ mice. These mice were fed a 15% fat chow from 6 weeks of age. First, we compared body weights. As shown in Fig. 1C, mice gained weight in an apoE gene dosage-dependent manner; apoE deficiency significantly suppressed weight gain. Several organs and tissues were weighed at 11 weeks of age. Weights of the liver and both brown and white adipose tissues were significantly lower in an apoE gene dosage-dependent manner (Fig. 1D). In contrast, weights of other organs, which are minimally involved in lipid accumulation, were similar in apoE+/+;Ay/+ and apoE−/−; Ay/+ mice (Supplemental Table 2 [online appendix]). Triglyceride contents of the liver and mesenteric adipose tissue were significantly smaller in apoE−/−;Ay/+ than in apoE+/+;Ay/+ mice (apoE+/+;Ay/+ vs. apoE−/−;Ay/+ mice: liver, 0.467 ± 0.069 vs. 0.365 ± 0.036 mg/protein, P = 0.02; white adipose tissue, 12.4 ± 1.4 vs. 5.8 ± 1.3 mg/protein, P = 0.002). Histological analyses revealed that apoE deficiency inhibited hepatic fat accumulation, while abundant lipid droplets were present in the livers of apoE+/+;Ay/+ mice (Fig. 1E and F). In addition, brown (Fig. 1G) and white (Fig. 1H) adipose tissues of apoE−/−;Ay/+ mice were significantly smaller than those of apoE+/+;Ay/+ mice. ApoE deficiency also decreased cell diameters in both brown (Fig. 1I) and white (Fig. 1J) adipose tissues. These findings suggest that apoE deficiency results in resistance to obesity via suppression of fat accumulation in the liver and fat tissues under obesity-inducing conditions.

ApoE deficiency induced greater glucose tolerance and insulin sensitivity in obese states.

As described previously (15), lipid metabolism was markedly impaired with apoE deficiency (Supplemental Fig. 1A [online appendix]). Plasma cholesterol, triglyceride, and FFA levels were markedly higher in apoE−/−;Ay/+ than in apoE+/+;Ay/+ mice. HPLC analysis of plasma lipid profiles revealed that chylomicron, VLDL, and LDL fractions were markedly increased in apoE−/−;Ay/+ compared with apoE+/+;Ay/+ mice (Supplemental Fig. 1B [online appendix]).

Next, we examined the effects of apoE deficiency on glucose metabolism as well as insulin sensitivity in genetically obese Ay mice. From age 8 weeks onward, fasting blood glucose was markedly increased in apoE+/+;Ay/+ mice on high-fat chow, but this glucose elevation was significantly inhibited by apoE deficiency (Fig. 2A). Fasting blood insulin levels at 11 weeks of age were lower, by 78%, in apoE−/−;Ay/+ than in apoE+/+;Ay/+ mice (Fig. 2B). Glucose (Fig. 2C) and insulin (Fig. 2D) tolerance tests documented that ApoE−/−;Ay/+ mice were more glucose tolerant and insulin sensitive. We further examined insulin sensitivity using hyperinsulinemic-euglycemic clamp procedures. ApoE+/+;Ay/+ mice fed high-fat chow exhibited severe insulin resistance, while in apoE−/−;Ay/+ mice, glucose infusion rates were markedly higher, by 54-fold (Fig. 2E). Thus, despite hyperlipidemia, especially increased plasma FFA levels, apoE deficiency markedly improves glucose tolerance and insulin sensitivity. In addition, insulin-stimulated tyrosine phosphorylation of IRS-1 in muscle was enhanced in apoE−/−;Ay/+ compared with apoE+/+;Ay/+ mice (Fig. 2F). Thus, apoE deficiency apparently prevents surplus fat accumulation in adipose tissue and insulin resistance in muscle, resulting in better glucose tolerance.

Adipocytes are differentiated normally in apoE−/−;Ay/+ mice in vivo.

We next determined plasma adipocytokine profiles (Fig. 3A). In apoE−/−;Ay/+ mice, plasma leptin levels were slightly decreased and TNF-α levels were markedly lower than those in apoE+/+;Ay/+ mice, while plasma adiponectin levels were significantly higher. Thus, apoE deficiency improved obesity-induced alterations in adipocytokine profiles. In addition, quantitative RT-PCR revealed that expressions of F4/80, monocyte chemoattractant protein-1, inducible nitric oxide synthase, and interleukin-6 in mesenteric adipose tissue were significantly lower in apoE−/−;Ay/+ than in apoE+/+;Ay/+ mice (Fig. 3B), suggesting inhibition of inflammation and macrophage invasion into adipose tissue. Obesity is reportedly associated with macrophage infiltration of adipose tissue, which is likely to promote insulin resistance (27,28). Inhibition of macrophage invasion of adipose tissue may be involved in the higher insulin sensitivity and glucose tolerance observed in apoE−/−;Ay/+ mice.

As described above, plasma adiponectin levels were increased in apoE−/−;Ay/+ mice, suggesting normal adipocyte differentiation in vivo. Furthermore, mRNA expressions for adipocyte-related proteins, such as CCAAT/enhancer binding protein-α, peroxisome proliferator–activated receptor-γ, and aP2, were similar among adipose tissues from apoE+/+;Ay/+, apoE+/−;Ay/+, and apoE−/−;Ay/+ mice (Fig. 3C). mRNA expressions levels of these three genes were also similar in adipose tissues from younger apoE+/+;Ay/+ and apoE−/−;Ay/+ mice, 4 weeks of age, when body weights were not significantly different (data not shown). In addition, apoE deficiency did not alter sterol regulatory element–binding protein 1 (SREBP1) expression (Fig. 3C). These findings indicate that adipocytes are normally differentiated in apoE−/−;Ay/+ mice in vivo.

Adenoviral apoE replenishment induced obesity and diabetes in genetically obese mice.

To confirm that the metabolic phenotypes observed in apoE−/−;Ay/+ mice were, in fact, mediated by apoE deficiency, we examined metabolic effects of adenovirus-mediated apoE expression in the livers of apoE−/−;Ay/+ mice. Replenishment of apoE protein (human apoE3) resulted in markedly decreased plasma cholesterol, triglyceride, and FFA levels (Supplemental Fig. 2A [online appendix]), indicating functional expression of apoE. HPLC analyses of plasma lipid profiles revealed that adenoviral replenishment of apoE protein in apoE−/−;Ay/+ mice markedly decreased the chylomicron and VLDL fractions (Supplemental Fig. 2B [online appendix]).

Increases in body weight for 7 days after adenoviral administration were significantly greater with the apoE adenovirus than with the LacZ control adenovirus (Fig. 4A). Liver weights tended to be increased (Fig. 4B), and those of brown adipose (Fig. 4C) and epididymal, mesenteric, and retroperitoneal white adipose (Fig. 4D) tissues were significantly increased with apoE adenoviral administration. Histological analyses revealed that apoE replenishment increased sizes of brown (Fig. 4E and F) and white (Fig. 4G and H) adipocytes. In addition, glucose tolerance tests revealed that apoE replenishment worsened glucose tolerance in apoE−/−;Ay/+ mice (Fig. 4I). These findings show clearly that circulating apoE contributes to increased adiposity and the glucose intolerance associated with obesity. Furthermore, plasma leptin levels were significantly increased on day 7 after adenoviral administration. TNF-α and adiponectin levels tended to be increased and decreased, respectively (Fig. 4J), suggesting that circulating apoEs are involved in obesity-induced alterations in adipocytokine levels.

ApoE occurs in three major isoforms (apoE2, -E3, and -E4) in humans. ApoE3, the most common isoform, is considered to be the wild type. To compare the roles of the three human apoE isoforms in obesity and diabetes, recombinant adenoviruses encoding human apoE2 and -E4 as well as apoE3 were injected into apoE−/−;Ay/+ mice. Administration of these apoE adenoviruses resulted in similar expression amounts of apoE proteins (data not shown), and similar increases in body weights (Supplemental Fig. 3A [online appendix]) and blood glucose levels (Supplemental Fig. 3B [online appendix]). These findings suggest that the three apoE isoforms contribute similarly to fat accumulation and glucose tolerance.

ApoE-less VLDL was uptaken into adipocytes, and the liver was impaired.

Why does apoE deficiency inhibit obesity in genetically obese mice? We next examined the uptake of β-VLDL, with or without apoE, into fully differentiated 3T3-L1 adipocytes. β-VLDL obtained from apoE−/− mice was labeled with DiI, followed by incubation with or without recombinant human apoE3. As shown in Fig. 5A, uptake of apoE-deficient VLDL was markedly lower, by 85%, than that of apoE-positive (after incubation with human apoE3) VLDL. These findings suggest that impaired VLDL uptake into adipocytes contributes to decreased adiposity in apoE−/−;Ay/+ mice. Thus, VLDL uptake into adipocytes is likely to play a role in excess fat deposition and, thereby, in the development of diabetes associated with obesity.

ApoE deficiency reportedly reduces hepatic VLDL secretion, resulting in fatty liver findings (11). In our model as well, hepatic triglyceride secretion was inhibited in apoE−/−;Ay/+ mice compared with apoE+/+;Ay/+ mice, by 48% (Fig. 5B). However, interestingly, apoE−/−;Ay/+ mice displayed less fat accumulation in the liver than apoE+/+;Ay/+ mice (Fig. 1D–F). To elucidate the underlying mechanism, we examined β-VLDL uptake into the liver. Fluorescence-labeled β-VLDL, with or without apoE, was intravenously injected into wild-type C57BL/6 mice. Fluorescence values in the liver were then measured. Uptake of apoE-deficient β-VLDL was markedly lower, by 49%, than that of apoE-positive VLDL (Fig. 5C). Thus, despite decreased secretion, decreased β-VLDL uptake with apoE deficiency may contribute to prevention of hepatic steatosis. Therefore, apoE is likely to be involved in excess fat uptake into hepatocytes as well as adipocytes. Taken together with the findings that adenoviral apoE replenishment decreased the VLDL fraction (Supplemental Fig. 2B [online appendix]), our results indicate that apoE-dependent VLDL transport into tissues, including the liver and adipose tissue, is involved in the development of obesity, resulting in glucose intolerance and insulin resistance.

ApoE deficiency decreased food intake and increased energy expenditure in genetically obese mice.

Next, to elucidate the systemic mechanism underlying the obesity prevention associated with apoE deficiency, we first measured food intakes in apoE+/+;Ay/+ and apoE−/−;Ay/+ mice. Interestingly, apoE deficiency in Ay mice significantly suppressed food intake (Fig. 5D). Then, to eliminate the secondary effects of reduced food intake in apoE−/−;Ay/+ mice, ApoE+/+;Ay/+ mice were allotted the same amounts of food consumption as apoE−/−;Ay/+ mice, followed by weight measurement and glucose tolerance testing. Pair-feeding blunted the body weight increments in apoE+/+;Ay/+ mice, while the weights of pair-fed apoE+/+;Ay/+ mice were significantly greater than those of apoE−/−;Ay/+ mice (Fig. 5E). ApoE−/−;Ay/+ mice exhibited better glucose tolerance than pair-fed apoE+/+;Ay/+ mice (Fig. 5F). Thus, the inhibition of obesity and glucose intolerance by apoE deficiency is not attributable solely to decreased food intake.

Next, we measured basal metabolic rates. As shown in Fig. 5G, resting oxygen consumption in both the light and the dark phase at 5 weeks of age was significantly greater in apoE−/−;Ay/+ than in apoE+/+;Ay/+ mice. Taken together with the pair-feeding experiment results, these findings show that decreased food intake and increased energy expenditure both contribute to the prevention of obesity and insulin resistance with apoE deficiency and that apoE is involved in regulation of energy metabolism.

To examine the effects of apoE deficiency on insulin resistance associated with obesity, apoE−/− mice were interbred with KK-Ay mice. ApoE−/−;Ay/+ mice showed resistance to the development of obesity and glucose intolerance. Insulin sensitivity was markedly greater in apoE−/−;Ay/+ than in apoE+/+;Ay/+ mice. Recently, several attempts to induce obesity in apoE−/− mice have been reported, but the results have been somewhat controversial (1214). In the present study, in addition to inhibition of adiposity and insulin resistance with apoE deficiency, adenoviral apoE replenishment reversed inhibition of obesity and glucose intolerance. These findings directly demonstrate apoE involvement in the development of obesity and obesity-related disorders of glucose metabolism and insulin sensitivity. Chiba et al. (14) previously reported that ob/ob;apoE−/− mice are also resistant to obesity. They attributed decreased adiposity in ob/ob;apoE−/− mice to impaired adipocyte differentiation based on in vitro findings that expression levels of aP2 and peroxisome proliferator–activated receptor-γ were markedly lower when bone marrow stromal cells and 3T3-L1 cells were cultured with apoE-less VLDL. However, in the present study, adipocytes in apoE−/−;Ay/+ mice expressed these adipocyte-related proteins normally in vivo. Furthermore, apoE−/−;Ay/+ mice showed better insulin sensitivity and less hepatic lipid accumulation, accompanied by improved adipocytokine profiles, than apoE+/+;Ay/+ mice. Thus, decreased adiposity and improved insulin sensitivity in apoE−/−;Ay/+ mice can be explained by factors other than adipocyte differentiation.

While body weight and adiposity were similar in young apoE−/− and apoE+/+ mice, apoE deficiency ameliorated obesity and insulin resistance under obesity-inducing conditions, such as aging, genetic susceptibility, and dietary loading, suggesting that apoE is involved in obesity development, i.e., excess fat accumulation, while being less involved in basal fat accumulation. Lack of apoE in β-VLDL markedly impaired β-VLDL transport into adipocytes. ApoE is an important component of VLDL. There are several receptors, including VLDL receptor (VLDLR) and LDL receptor–related protein, which recognize VLDL in an apoE-dependent manner (22). Among them, VLDLRs reportedly have similar affinities for apoE2, -E3, and -E4 isoforms (29). In addition, VLDLR-deficient mice reportedly exhibit obesity resistance with high-fat chow loading (30). Taken together with our findings that replenishment of apoE2, -E3, and -E4 isoforms contributes similarly to fat accumulation and glucose tolerance in apoE−/−;Ay/+ mice, the apoE-VLDLR interaction plays an important role in the development of obesity. Furthermore, it is well known that high levels of plasma VLDL are associated with obesity and type 2 diabetes (31). Thus, receptor-mediated VLDL transport into adipocytes in an apoE-dependent manner is involved mainly in excess lipid uptake into adipocytes. Lipid uptake, required for adipocyte differentiation and metabolic activities, might be mediated mainly by apoE-independent lipid transport pathways or de novo lipid synthesis in adipocytes.

In addition, transport of apoE-deficient β-VLDL into the liver was markedly impaired compared with that of apoE-positive β-VLDL. Despite decreased triglyceride secretion, apoE deficiency decreased hepatic fat accumulation in Ay, but not in wild-type, mice (11). Hepatic expressions of SREBP1c and fatty acid synthase were similar in apoE−/−;Ay/+ and apoE+/+;Ay/+ mice (data not shown), suggesting no apparent decrease in hepatic fatty acid synthesis in apoE−/−;Ay/+ mice. These findings suggest that apoE-dependent uptake of β-VLDL into hepatocytes is involved in the development of hepatic steatosis in Ay mice. The machinery that transports β-VLDL into the liver, including the receptor(s) playing a major role, is a potential target for elucidating the mechanism underlying hepatic steatosis.

Intriguingly, in apoE−/−;Ay/+ mice, food intake was decreased and energy expenditure enhanced compared with apoE+/+;Ay/+ mice. Pair-feeding experiments revealed that both these phenomena result in obesity resistance in apoE−/−;Ay/+ mice. There appear to be several possible explanations for these alterations in energy metabolism. First, hyperlipidemia induced by apoE deficiency might contribute to decreased food intake and increased energy expenditure. However, LDLR−/− mice, which also exhibit hyperlipidemia, reportedly become more obese and diabetic in response to high-fat and high-carbonate diets than wild-type mice (13). In addition, food intake was similar in LDLR−/− mice and LDLR+/+ mice (13). Therefore, although hyperlipidemia is more severe in apoE−/− than in LDLR−/− mice, involvement of hyperlipidemia in food intake regulation might be unlikely in our model. Second, we speculate that leptin sensitization is involved in decreased food intake and increased energy expenditure. Obesity is well known to be associated with poor responses to leptin despite hyperleptinemia, a state defined as leptin resistance (32). Lower plasma leptin levels with lower body weight in apoE−/−;Ay/+ mice compared with apoE+/+;Ay/+ mice suggests greater leptin sentivity in the former. Therefore, decreased food intake and increased energy expenditure in apoE−/−;Ay/+ mice might be explained by leptin sensitization. We have recently reported that alterations in metabolism in adipose tissue affect food intake amounts (19). However, ob/ob;apoE−/− mice are also reportedly resistant to obesity (14). Since ob/ob mice are leptin deficient, the obesity resistance in ob/ob;apoE−/− mice is not mediated by leptin signaling, e.g., leptin sensitization, although food intake and energy expenditure were not measured in the earlier study (14). Thus, mechanisms other than leptin sensitization might be involved in decreased food intake and increased energy expenditure in apoE−/−;Ay/+ mice. Third, direct effects of apoE on neurons are also possible. ApoE, produced by glial cells, is a major apolipoprotein in the brain and mediates the transport of cholesterol and phospholipids, and its receptors are abundantly expressed on neurons (33). Furthermore, numerous studies have shown that apoE plays multiple roles in the nervous system. In the central and peripheral nervous systems, apoE promotes neurite outgrowth and regeneration (34). ApoE protects neurons from oxidative injury (35) and modulates amyloid-β deposition (36), interactions with Alzheimer amyloid precursor protein (37), and transmission of signals to neurons (38). In this context, modulation of neurons by apoE might be involved in energy metabolism. ApoE is reportedly expressed in tissues other than the liver, including the brain (33) and adipose tissue (39). ApoE deficiency in these tissues may affect the metabolic phenotypes of apoE−/−;Ay/+ mice observed herein. Intensive research, including tissue-specific disruption of apoE or its receptor, is required to examine this hypothesis.

ApoE is involved in surplus fat accumulation and energy metabolism, including regulation of food intake and energy expenditure. Thus, excess fat accumulation via an apoE-dependent pathway might play a role in development of the metabolic syndrome. In addition to dissipation of surplus energy (4), apoE-dependent excess lipid transport is a potentially novel therapeutic target for the metabolic syndrome.

FIG. 1.

ApoE deficiency suppressed body weight gain and fat accumulation. A and B: Body weights were determined from 6 to 40 weeks of age (A) and glucose tolerance tests were performed (B) in apoE+/+ mice (○) and apoE−/− mice (•; n = 6 per group) at 30 weeks of age with the C57BL/6 background. C: Body weights were determined from 6 to 20 weeks of age in apoE+/+;Ay/+ (○), apoE+/−;Ay/+ (▵), and apoE−/−;Ay/+ (•) mice fed 15% fat chow. d–J: The liver and fat tissues of apoE+/+;Ay/+, apoE+/−;Ay/+, and apoE−/−;Ay/+ mice at 11 weeks of age. D: The liver, brown adipose, and mesenteric and retroperitoneal white adipose tissues from apoE+/+;Ay/+ (□), apoE+/−;Ay/+ (), and apoE−/−;Ay/+ (▪) mice (n = 6–8 per group) were weighed. EJ: Histological findings of the liver stained with hematoxylin-eosin (E) and Oil Red O (F), as well as hematoxylin- eosin staining of brown adipose (G) and mesenteric white adipose (H) tissues in apoE+/+;Ay/+ (left), apoE+/−;Ay/+ (middle), and apoE−/−;Ay/+ (right) mice. Brown adipose (I) and mesenteric white adipose (J) cell diameters were measured in apoE+/+;Ay/+ (□), apoE+/−;Ay/+ (), and apoE−/−;Ay/+ (□) mice. Representative histological findings are shown. Data are presented as means ± SE. *P < 0.05, **P < 0.01 by the unpaired t test and one-way ANOVA.

FIG. 1.

ApoE deficiency suppressed body weight gain and fat accumulation. A and B: Body weights were determined from 6 to 40 weeks of age (A) and glucose tolerance tests were performed (B) in apoE+/+ mice (○) and apoE−/− mice (•; n = 6 per group) at 30 weeks of age with the C57BL/6 background. C: Body weights were determined from 6 to 20 weeks of age in apoE+/+;Ay/+ (○), apoE+/−;Ay/+ (▵), and apoE−/−;Ay/+ (•) mice fed 15% fat chow. d–J: The liver and fat tissues of apoE+/+;Ay/+, apoE+/−;Ay/+, and apoE−/−;Ay/+ mice at 11 weeks of age. D: The liver, brown adipose, and mesenteric and retroperitoneal white adipose tissues from apoE+/+;Ay/+ (□), apoE+/−;Ay/+ (), and apoE−/−;Ay/+ (▪) mice (n = 6–8 per group) were weighed. EJ: Histological findings of the liver stained with hematoxylin-eosin (E) and Oil Red O (F), as well as hematoxylin- eosin staining of brown adipose (G) and mesenteric white adipose (H) tissues in apoE+/+;Ay/+ (left), apoE+/−;Ay/+ (middle), and apoE−/−;Ay/+ (right) mice. Brown adipose (I) and mesenteric white adipose (J) cell diameters were measured in apoE+/+;Ay/+ (□), apoE+/−;Ay/+ (), and apoE−/−;Ay/+ (□) mice. Representative histological findings are shown. Data are presented as means ± SE. *P < 0.05, **P < 0.01 by the unpaired t test and one-way ANOVA.

Close modal
FIG. 2.

ApoE deficiency improved glucose tolerance and insulin sensitivity in genetically obese mice. A: Fasting blood glucose levels were examined in apoE+/+;Ay/+ (○), apoE+/−;Ay/+ (▵), and apoE−/−;Ay/+ (•) mice biweekly from 6 to 12 weeks of age. B: Fasting plasma insulin levels of apoE+/+;Ay/+ (□), apoE+/−;Ay/+ (), and apoE−/−;Ay/+ (▪) mice were measured. C and D: Glucose tolerance (C) and insulin tolerance (D) tests were performed in apoE+/+;Ay/+ (○), apoE+/−;Ay/+ (▵), and apoE−/−;Ay/+ (•) mice. Data were expressed as percentages of blood glucose levels immediately before intraperitoneal insulin loading (n = 4–6 per group). E: Glucose infusion rates during hyperinsulinemic-euglycemic clamp studies were measured in apoE+/+;Ay/+ (□), apoE+/−;Ay/+ (), and apoE−/−;Ay/+ (▪) mice (n = 3 per group). F: Insulin-stimulated tyrosine phosphorylation of IRS-1 proteins in muscle. Mice that had been fasted for 16 h received an intravenous injection of 100 μl normal saline with or without insulin (10 units/kg body wt). Hindlimb muscles were removed 300 s later, and lysates were immunoprecipitated with anti–IRS-1 antibody. Immunoprecipitates were subjected to SDS-PAGE and immunoblotted with anti-phosphotyrosine antibody (4G10). Representative histological findings and immunoblots are presented. These metabolic studies in BF were performed using 11-week-old mice. In AE, data are presented graphically as means ± SE. *P < 0.05, **P < 0.01 by the unpaired t test and one-way ANOVA.

FIG. 2.

ApoE deficiency improved glucose tolerance and insulin sensitivity in genetically obese mice. A: Fasting blood glucose levels were examined in apoE+/+;Ay/+ (○), apoE+/−;Ay/+ (▵), and apoE−/−;Ay/+ (•) mice biweekly from 6 to 12 weeks of age. B: Fasting plasma insulin levels of apoE+/+;Ay/+ (□), apoE+/−;Ay/+ (), and apoE−/−;Ay/+ (▪) mice were measured. C and D: Glucose tolerance (C) and insulin tolerance (D) tests were performed in apoE+/+;Ay/+ (○), apoE+/−;Ay/+ (▵), and apoE−/−;Ay/+ (•) mice. Data were expressed as percentages of blood glucose levels immediately before intraperitoneal insulin loading (n = 4–6 per group). E: Glucose infusion rates during hyperinsulinemic-euglycemic clamp studies were measured in apoE+/+;Ay/+ (□), apoE+/−;Ay/+ (), and apoE−/−;Ay/+ (▪) mice (n = 3 per group). F: Insulin-stimulated tyrosine phosphorylation of IRS-1 proteins in muscle. Mice that had been fasted for 16 h received an intravenous injection of 100 μl normal saline with or without insulin (10 units/kg body wt). Hindlimb muscles were removed 300 s later, and lysates were immunoprecipitated with anti–IRS-1 antibody. Immunoprecipitates were subjected to SDS-PAGE and immunoblotted with anti-phosphotyrosine antibody (4G10). Representative histological findings and immunoblots are presented. These metabolic studies in BF were performed using 11-week-old mice. In AE, data are presented graphically as means ± SE. *P < 0.05, **P < 0.01 by the unpaired t test and one-way ANOVA.

Close modal
FIG. 3.

ApoE deficiency affected plasma adipocytokine profiles and expressions of the mRNAs for inflammation- and differentiation-related proteins in adipose tissue. A: Plasma adipocytokines (left, leptin; middle, TNF-α; right, adiponectin) of high-fat chow–fed apoE+/+;Ay/+ (□), apoE+/−;Ay/+ (), and apoE−/−;Ay/+ (▪) mice were measured after a 10-h fast at 11 weeks of age. B and C: Relative amounts of mRNA in mesenteric white adipose tissue from apoE+/+;Ay/+ (□) and apoE−/−;Ay/+ (▪) mice were determined by quantitative RT-PCR and corrected with 28S rRNA as the internal standard. Total RNA in white adipose tissue was isolated, after a 10-h fast, from 11-week-old mice. Levels of the mRNA levels for inflammation (B)- and differentiation (C)-related proteins in adipose tissue were assayed (n = 6 per group). Data are presented as means ± SE. *P < 0.05, **P < 0.01 by one-way ANOVA (A) and the unpaired t test (B and C).

FIG. 3.

ApoE deficiency affected plasma adipocytokine profiles and expressions of the mRNAs for inflammation- and differentiation-related proteins in adipose tissue. A: Plasma adipocytokines (left, leptin; middle, TNF-α; right, adiponectin) of high-fat chow–fed apoE+/+;Ay/+ (□), apoE+/−;Ay/+ (), and apoE−/−;Ay/+ (▪) mice were measured after a 10-h fast at 11 weeks of age. B and C: Relative amounts of mRNA in mesenteric white adipose tissue from apoE+/+;Ay/+ (□) and apoE−/−;Ay/+ (▪) mice were determined by quantitative RT-PCR and corrected with 28S rRNA as the internal standard. Total RNA in white adipose tissue was isolated, after a 10-h fast, from 11-week-old mice. Levels of the mRNA levels for inflammation (B)- and differentiation (C)-related proteins in adipose tissue were assayed (n = 6 per group). Data are presented as means ± SE. *P < 0.05, **P < 0.01 by one-way ANOVA (A) and the unpaired t test (B and C).

Close modal
FIG. 4.

Adenoviral apoE replenishment induced obesity and diabetes in genetically obese mice. ApoE−/−;Ay/+ mice were intravenously administered LacZ (□ or ○) or apoE (▪ or •) adenovirus at 10 weeks of age after 4 weeks of high-fat loading. A: Body weight changes for 7 days after adenoviral administration were examined. BD: Weights of the liver (B) and brown adipose (C) as well as white adipose (D: left, epidydimal fat; middle, mesenteric fat; right, retroperitoneal fat) tissues were determined on day 7 after adenoviral injection (n = 6 per group). EH: Histological findings of brown adipose (E and F) and mesenteric white adipose (G and H) tissues. Representative hematoxylin-eosin stained tissue samples are presented (E and G). Cell diameters were measured in these tissues (F and H). I: Glucose tolerance tests were performed on day 7 after adenoviral injection (n = 5–8 per group). J: Plasma adipocytokine levels (left, leptin; middle, TNF-α; right, adiponectin) were assayed on day 7 after adenoviral injection (n = 5–8 per group). Data are presented as means ± SE. *P < 0.05, **P < 0.01 by the unpaired t test.

FIG. 4.

Adenoviral apoE replenishment induced obesity and diabetes in genetically obese mice. ApoE−/−;Ay/+ mice were intravenously administered LacZ (□ or ○) or apoE (▪ or •) adenovirus at 10 weeks of age after 4 weeks of high-fat loading. A: Body weight changes for 7 days after adenoviral administration were examined. BD: Weights of the liver (B) and brown adipose (C) as well as white adipose (D: left, epidydimal fat; middle, mesenteric fat; right, retroperitoneal fat) tissues were determined on day 7 after adenoviral injection (n = 6 per group). EH: Histological findings of brown adipose (E and F) and mesenteric white adipose (G and H) tissues. Representative hematoxylin-eosin stained tissue samples are presented (E and G). Cell diameters were measured in these tissues (F and H). I: Glucose tolerance tests were performed on day 7 after adenoviral injection (n = 5–8 per group). J: Plasma adipocytokine levels (left, leptin; middle, TNF-α; right, adiponectin) were assayed on day 7 after adenoviral injection (n = 5–8 per group). Data are presented as means ± SE. *P < 0.05, **P < 0.01 by the unpaired t test.

Close modal
FIG. 5.

ApoE deficiency inhibited β-VLDL uptake into adipocytes and the liver, decreased food intake, and increased energy expenditure. A: Uptakes of fluorescence-labeled β-VLDL, with (□) or without (▪) apoE, into cultured adipocytes were measured. β-VLDL was isolated from apoE−/− mouse sera, followed by labeling with DiI and pretreatment with or without human recombinant apoE3. Fully differentiated 3T3-L1 adipocytes were incubated with apoE-positive or apoE-less β-VLDL for 8 h, followed by measurement of fluorescence uptake into adipocytes. Data are presented as the relative amounts of β-VLDL uptake compared with apoE-positive β-VLDL uptake (n = 6 per group). B: Triglyceride secretion rates from the liver after administration of Triton WR-1339 were measured in 11-week-old apoE+/+;Ay/+ (○) and apoE−/−;Ay/+ (•) mice fed a high-fat diet for 5 weeks. C: Hepatic uptake of β-VLDL with or without apoE. Fluorescence-labeled β-VLDL with or without apoE was intravenously injected into 11-week-old apoE+/+;Ay/+ mice fed a high-fat diet for 5 weeks, followed by measurement of fluorescence levels in the liver 30 min after injection. Data are presented as the relative amounts of β-VLDL uptake compared with apoE-positive β-VLDL uptake (n = 4 per group). D: Daily food intake amounts from weaning through 8 weeks of age are presented. E and F: ApoE+/+;Ay/+ mice were allotted into two groups at 4 weeks of age, and one group of apoE+/+;Ay/+mice was given their daily food allotments based on the previous days’ consumption by apoE−/−;Ay/+ littermate mice. Body weights (E) were determined and glucose tolerance tests (F) were performed in apoE+/+;Ay/+ mice (○), pair-fed apoE+/+;Ay/+ mice (□), and apoE−/−;Ay/+ mice (•) (n = 6–8 per group). G: Resting oxygen consumption in the light and dark phases was measured at 5 weeks of age with open-circuit indirect calorimetry. n = 4 per group. Data are presented as means ± SE. *P < 0.05, **P < 0.01 by the unpaired t test. In E and F ##P < 0.01 for pair-feeding apoE+/+;Ay/+ mice versus apoE+/+;Ay/+ mice; *P < 0.05, **P < 0.01 for pair-feeding apoE+/+;Ay/+ mice versus apoE−/−;Ay/+ mice, by one-way ANOVA.

FIG. 5.

ApoE deficiency inhibited β-VLDL uptake into adipocytes and the liver, decreased food intake, and increased energy expenditure. A: Uptakes of fluorescence-labeled β-VLDL, with (□) or without (▪) apoE, into cultured adipocytes were measured. β-VLDL was isolated from apoE−/− mouse sera, followed by labeling with DiI and pretreatment with or without human recombinant apoE3. Fully differentiated 3T3-L1 adipocytes were incubated with apoE-positive or apoE-less β-VLDL for 8 h, followed by measurement of fluorescence uptake into adipocytes. Data are presented as the relative amounts of β-VLDL uptake compared with apoE-positive β-VLDL uptake (n = 6 per group). B: Triglyceride secretion rates from the liver after administration of Triton WR-1339 were measured in 11-week-old apoE+/+;Ay/+ (○) and apoE−/−;Ay/+ (•) mice fed a high-fat diet for 5 weeks. C: Hepatic uptake of β-VLDL with or without apoE. Fluorescence-labeled β-VLDL with or without apoE was intravenously injected into 11-week-old apoE+/+;Ay/+ mice fed a high-fat diet for 5 weeks, followed by measurement of fluorescence levels in the liver 30 min after injection. Data are presented as the relative amounts of β-VLDL uptake compared with apoE-positive β-VLDL uptake (n = 4 per group). D: Daily food intake amounts from weaning through 8 weeks of age are presented. E and F: ApoE+/+;Ay/+ mice were allotted into two groups at 4 weeks of age, and one group of apoE+/+;Ay/+mice was given their daily food allotments based on the previous days’ consumption by apoE−/−;Ay/+ littermate mice. Body weights (E) were determined and glucose tolerance tests (F) were performed in apoE+/+;Ay/+ mice (○), pair-fed apoE+/+;Ay/+ mice (□), and apoE−/−;Ay/+ mice (•) (n = 6–8 per group). G: Resting oxygen consumption in the light and dark phases was measured at 5 weeks of age with open-circuit indirect calorimetry. n = 4 per group. Data are presented as means ± SE. *P < 0.05, **P < 0.01 by the unpaired t test. In E and F ##P < 0.01 for pair-feeding apoE+/+;Ay/+ mice versus apoE+/+;Ay/+ mice; *P < 0.05, **P < 0.01 for pair-feeding apoE+/+;Ay/+ mice versus apoE−/−;Ay/+ mice, by one-way ANOVA.

Close modal

J.G., H.K., and Y.I. contributed equally to this work.

Additional information for this article can be found in an online appendix at http://diabetes.diabetesjournals.org.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked “advertisement” in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

This work was supported by a Grant-in-Aid for Scientific Research (B2, 15390282) to H.K.; a Grant-in-Aid for Scientific Research (17790599) to Y.I. from the Ministry of Education, Science, Sports and Culture of Japan; and a Grant-in-Aid for Scientific Research (H16-genome-003) to Y.O. from the Ministry of Health, Labor and Welfare of Japan. This work was also supported by the 21st Century COE Programs “CRESCENDO” to H.K. and “the Center for Innovative Therapeutic Development for Common Diseases” to Y.O. from the Ministry of Education Science, Sports and Culture.

We thank I. Sato, J. Fushimi, K. Kawamura, and M. Hoshi for technical support.

1.
Eckel RH, Grundy SM, Zimmet PZ: The metabolic syndrome.
Lancet
365
:
1415
–1428,
2005
2.
Fruhbeck G, Gomez-Ambrosi J: Control of body weight: a physiologic and transgenic perspective.
Diabetologia
46
:
143
–172,
2003
3.
Friedman JM: A war on obesity, not the obese.
Science
299
:
856
–858,
2003
4.
Ishigaki Y, Katagiri H, Yamada T, Ogihara T, Imai J, Uno K, Hasegawa Y, Gao J, Ishihara H, Shimosegawa T, Sakoda H, Asano T, Oka Y: Dissipating excess energy stored in the liver is a potential treatment strategy for diabetes associated with obesity.
Diabetes
54
:
322
–332,
2005
5.
Moitra J, Mason MM, Olive M, Krylov D, Gavrilova O, Marcus-Samuels B, Feigenbaum L, Lee E, Aoyama T, Eckhaus M, Reitman ML, Vinson C: Life without white fat: a transgenic mouse.
Genes Dev
12
:
3168
–3181,
1998
6.
Shimomura I, Hammer RE, Richardson JA, Ikemoto S, Bashmakov Y, Goldstein JL, Brown MS: Insulin resistance and diabetes mellitus in transgenic mice expressing nuclear SREBP-1c in adipose tissue: model for congenital generalized lipodystrophy.
Genes Dev
12
:
3182
–3194,
1998
7.
Oral EA, Simha V, Ruiz E, Andewelt A, Premkumar A, Snell P, Wagner AJ, DePaoli AM, Reitman ML, Taylor SI, Gorden P, Garg A: Leptin-replacement therapy for lipodystrophy.
N Engl J Med
346
:
570
–578,
2002
8.
Hussain MM, Maxfield FR, Mas-Oliva J, Tabas I, Ji ZS, Innerarity TL, Mahley RW: Clearance of chylomicron remnants by the low density lipoprotein receptor-related protein/alpha 2-macroglobulin receptor.
J Biol Chem
266
:
13936
–13940,
1991
9.
Chappell DA, Medh JD: Receptor-mediated mechanisms of lipoprotein remnant catabolism.
Prog Lipid Res
37
:
393
–422,
1998
10.
Cooper AD: Hepatic uptake of chylomicron remnants.
J Lipid Res
38
:
2173
–2192,
1997
11.
Kuipers F, Jong MC, Lin Y, Eck M, Havinga R, Bloks V, Verkade HJ, Hofker MH, Moshage H, Berkel TJ, Vonk RJ, Havekes LM: Impaired secretion of very low density lipoprotein-triglycerides by apolipoprotein E-deficient mouse hepatocytes.
J Clin Invest
100
:
2915
–2922,
1997
12.
Lyngdorf LG, Gregersen S, Daugherty A, Falk E: Paradoxical reduction of atherosclerosis in apoE-deficient mice with obesity-related type 2 diabetes.
Cardiovasc Res
59
:
854
–862,
2003
13.
Schreyer SA, Vick C, Lystig TC, Mystkowski P, LeBoeuf RC: LDL receptor but not apolipoprotein E deficiency increases diet-induced obesity and diabetes in mice.
Am J Physiol Endocrinol Metab
282
:
E207
–E214,
2002
14.
Chiba T, Nakazawa T, Yui K, Kaneko E, Shimokado K: VLDL induces adipocyte differentiation in ApoE-dependent manner.
Arterioscler Thromb Vasc Biol
23
:
1423
–1429,
2003
15.
Zhang SH, Reddick RL, Piedrahita JA, Maeda N: Spontaneous hypercholesterolemia and arterial lesions in mice lacking apolipoprotein E.
Science
258
:
468
–471,
1992
16.
Ishigaki Y, Oikawa S, Suzuki T, Usui S, Magoori K, Kim DH, Suzuki H, Sasaki J, Sasano H, Okazaki M, Toyota T, Saito T, Yamamoto TT: Virus-mediated transduction of apolipoprotein E (ApoE)-sendai develops lipoprotein glomerulopathy in ApoE-deficient mice.
J Biol Chem
275
:
31269
–31273,
2000
17.
Anai M, Funaki M, Ogihara T, Terasaki J, Inukai K, Katagiri H, Fukushima Y, Yazaki Y, Kikuchi M, Oka Y, Asano T: Altered expression levels and impaired steps in the pathway to phosphatidylinositol 3-kinase activation via insulin receptor substrates 1 and 2 in Zucker fatty rats.
Diabetes
47
:
13
–23,
1998
18.
Usui S, Hara Y, Hosaki S, Okazaki M: A new on-line dual enzymatic method for simultaneous quantification of cholesterol and triglycerides in lipoproteins by HPLC.
J Lipid Res
43
:
805
–814,
2002
19.
Yamada T, Katagiri H, Ishigaki Y, Ogihara T, Imai J, Uno K, Hasegawa Y, Gao J, Ishihara H, Niijima A, Mano H, Aburatani H, Asano T, Oka Y: Signals from intra-abdominal fat modulate insulin and leptin sensitivity through different mechanisms: neuronal involvement in food-intake regulation.
Cell Metab
3
:
223
–229,
2006
20.
Uno K, Katagiri H, Yamada T, Ishigaki Y, Ogihara T, Imai J, Hasegawa Y, Gao J, Kaneko K, Iwasaki H, Ishihara H, Sasano H, Inukai K, Mizuguchi H, Asano T, Shiota M, Nakazato M, Oka Y: Neuronal pathway from the liver modulates energy expenditure and systemic insulin sensitivity.
Science
312
:
1656
–1659,
2006
21.
Min J, Okada S, Kanzaki M, Elmendorf JS, Coker KJ, Ceresa BP, Syu LJ, Noda Y, Saltiel AR, Pessin JE: Synip: a novel insulin-regulated syntaxin 4-binding protein mediating GLUT4 translocation in adipocytes.
Mol Cell
3
:
751
–760,
1999
22.
Takahashi S, Kawarabayasi Y, Nakai T, Sakai J, Yamamoto T: Rabbit very low density lipoprotein receptor: a low density lipoprotein receptor-like protein with distinct ligand specificity.
Proc Natl Acad Sci U S A
89
:
9252
–9256,
1992
23.
Descamps O, Bilheimer D, Herz J: Insulin stimulates receptor-mediated uptake of apoE-enriched lipoproteins and activated alpha 2-macroglobulin in adipocytes.
J Biol Chem
268
:
974
–981,
1993
24.
Wilsie LC, Chanchani S, Navaratna D, Orlando RA: Cell surface heparan sulfate proteoglycans contribute to intracellular lipid accumulation in adipocytes.
Lipids Health Dis
4
:
2
,
2005
25.
Siri P, Candela N, Zhang YL, Ko C, Eusufzai S, Ginsberg HN, Huang LS: Post-transcriptional stimulation of the assembly and secretion of triglyceride-rich apolipoprotein B lipoproteins in a mouse with selective deficiency of brown adipose tissue, obesity, and insulin resistance.
J Biol Chem
276
:
46064
–46072,
2001
26.
Fujino T, Asaba H, Kang MJ, Ikeda Y, Sone H, Takada S, Kim DH, Ioka RX, Ono M, Tomoyori H, Okubo M, Murase T, Kamataki A, Yamamoto J, Magoori K, Takahashi S, Miyamoto Y, Oishi H, Nose M, Okazaki M, Usui S, Imaizumi K, Yanagisawa M, Sakai J, Yamamoto TT: Low-density lipoprotein receptor-related protein 5 (LRP5) is essential for normal cholesterol metabolism and glucose-induced insulin secretion.
Proc Natl Acad Sci U S A
100
:
229
–234,
2003
27.
Xu H, Barnes GT, Yang Q, Tan G, Yang D, Chou CJ, Sole J, Nichols A, Ross JS, Tartaglia LA, Chen H: Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance.
J Clin Invest
112
:
1821
–1830,
2003
28.
Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW Jr: Obesity is associated with macrophage accumulation in adipose tissue.
J Clin Invest
112
:
1796
–1808,
2003
29.
Takahashi S, Oida K, Ookubo M, Suzuki J, Kohno M, Murase T, Yamamoto T, Nakai T: Very low density lipoprotein receptor binds apolipoprotein E2/2 as well as apolipoprotein E3/3.
FEBS Lett
386
:
197
–200,
1996
30.
Goudriaan JR, Tacken PJ, Dahlmans VE, Gijbels MJ, van Dijk KW, Havekes LM, Jong MC: Protection from obesity in mice lacking the VLDL receptor.
Arterioscler Thromb Vasc Biol
21
:
1488
–1493,
2001
31.
Reaven GM: Banting lecture 1988: Role of insulin resistance in human disease.
Diabetes
37
:
1595
–1607,
1988
32.
Considine RV, Sinha MK, Heiman ML, Kriauciunas A, Stephens TW, Nyce MR, Ohannesian JP, Marco CC, McKee LJ, Bauer TL, et al.: Serum immunoreactive-leptin concentrations in normal-weight and obese humans.
N Engl J Med
334
:
292
–295,
1996
33.
Mahley RW, Nathan BP, Pitas RE: Apolipoprotein E: structure, function, and possible roles in Alzheimer’s disease.
Ann N Y Acad Sci
777
:
139
–145,
1996
34.
Masliah E, Mallory M, Ge N, Alford M, Veinbergs I, Roses AD: Neurodegeneration in the central nervous system of apoE-deficient mice.
Exp Neurol
136
:
107
–122,
1995
35.
Chen Y, Lomnitski L, Michaelson DM, Shohami E: Motor and cognitive deficits in apolipoprotein E-deficient mice after closed head injury.
Neuroscience
80
:
1255
–1262,
1997
36.
Bales KR, Verina T, Dodel RC, Du Y, Altstiel L, Bender M, Hyslop P, Johnstone EM, Little SP, Cummins DJ, Piccardo P, Ghetti B, Paul SM: Lack of apolipoprotein E dramatically reduces amyloid beta-peptide deposition.
Nat Genet
17
:
263
–264,
1997
37.
Barger SW, Harmon AD: Microglial activation by Alzheimer amyloid precursor protein and modulation by apolipoprotein E.
Nature
388
:
878
–881,
1997
38.
Trommsdorff M, Gotthardt M, Hiesberger T, Shelton J, Stockinger W, Nimpf J, Hammer RE, Richardson JA, Herz J: Reeler/disabled-like disruption of neuronal migration in knockout mice lacking the VLDL receptor and ApoE receptor 2.
Cell
97
:
689
–701,
1999
39.
Zechner R, Moser R, Newman TC, Fried SK, Breslow JL: Apolipoprotein E gene expression in mouse 3T3–L1 adipocytes and human adipose tissue and its regulation by differentiation and lipid content.
J Biol Chem
266
:
10583
–10588,
1991

Supplementary data